Scolaris Content Display Scolaris Content Display

Antiangiogenic therapy with anti‐vascular endothelial growth factor modalities for diabetic macular oedema

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Ahmadieh 2008 {published data only}

Ahmadieh H, Ramezani A, Shoeibi N, Bijanzadeh B, Tabatabaei A, Azarmina M, et al. Intravitreal bevacizumab with or without triamcinolone for refractory diabetic macular edema; a placebo‐controlled, randomized clinical trial. Graefe's Archive for Clinical and Experimental Ophthalmology 2008;246(4):483‐9.

BOLT {published data only}

Michaelides M, Kaines A, Hamilton RD, Fraser‐Bell S, Rajendram R, Quhill F, et al. A prospective randomized trial of intravitreal bevacizumab or laser therapy in the management of diabetic macular edema (BOLT study) 12‐month data: report 2. Ophthalmology 2010;117(6):1078‐86.

DA VINCI {published data only}

Do DV, Nguyen QD, Boyer D, Schmidt‐Erfurth U, Brown DM, Vitti R, et al. One‐year outcomes of the DA VINCI Study of VEGF Trap‐Eye in eyes with diabetic macular edema. Ophthalmology 2012;119(8):1658‐65.
Do DV, Schmidt‐Erfurth U, Gonzalez VH, Gordon CM, Tolentino M, Berliner AJ, et al. The DAVINCI Study: phase 2 primary results of VEGF Trap‐Eye in patients with diabetic macular edema. Ophthalmology 2011;118(9):1819‐26.

DRCRnet {published data only}

Diabetic Retinopathy Clinical Research Network, Elman MJ, Aiello LP, Beck RW, Bressler NM, Bressler SB, et al. Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology 2010;117(6):1064–77.
Elman MJ, Bressler NM, Qin H, Beck RW, Ferris FL, Friedman SM, et al. Expanded 2‐year follow‐up of ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology 2011;118(4):609‐14.

Macugen 2005 {published data only}

Cunningham ET, Adamis AP, Altaweel M, Aiello LP, Bressler NM, D'Amico DJ, et al. A phase II randomized double‐masked trial of pegaptanib, an anti‐vascular endothelial growth factor aptamer, for diabetic macular edema. Ophthalmology 2005;112(10):1747‐57.

Macugen 2011 {published data only}

Loftus JV, Sultan MB, Pleil AM, Macugen 1013 Study Group. Changes in vision‐ and health‐related quality of life in patients with diabetic macular edema treated with pegaptanib sodium or sham. Investigative Ophthalmology and Visual Science 2011;52(10):7498‐505.
Sultan MB, Zhou D, Loftus J, Dombi T, Ice KS, Macugen 1013 Study Group. A phase 2/3, multicenter, randomized, double‐masked, 2‐year trial of pegaptanib sodium for the treatment of diabetic macular edema. Ophthalmology 2011;118(6):1107‐18.

READ2 {published data only}

Nguyen QD, Shah SM, Heier JS, Do DV, Lim J, Boyer D, et al. Primary End Point (Six Months) Results of the Ranibizumab for Edema of the mAcula in diabetes (READ‐2) study. Ophthalmology 2009;116(11):2175‐81.
Nguyen QD, Shah SM, Khwaja AA, Channa R, Hatef E, Do DV, et al. Two‐year outcomes of the ranibizumab for edema of the mAcula in diabetes (READ‐2) study. Ophthalmology 2010;117(11):2146‐51.

RESOLVE {published data only}

Massin P, Bandello F, Garweg JG, Hansen LL, Harding SP, Larsen M, et al. Safety and efficacy of ranibizumab in diabetic macular edema (RESOLVE Study): a 12‐month, randomized, controlled, double‐masked, multicenter phase II study. Diabetes Care 2010;33(11):2399‐405.

RESTORE {published data only}

Mitchell P, Bandello F, Schmidt‐Erfurth U, Lang GE, Massin P, Schlingemann RO, et al. The RESTORE study: ranibizumab monotherapy or combined with laser versus laser monotherapy for diabetic macular edema. Ophthalmology 2011;118(4):615–62.

RISE‐RIDE {published data only}

Nguyen QD, Brown DM, Marcus DM, Boyer DS, Patel S, Feiner L, et al. Ranibizumab for Diabetic Macular Edema: Results from 2 Phase III Randomized Trials: RISE and RIDE. Ophthalmology 2012;119(4):789‐801.

Soheilian 2007 {published data only}

Soheilian M, Garfami KH, Ramezani A, Yaseri M, Peyman GA. Two‐year results of a randomized trial of intravitreal bevacizumab alone or combined with triamcinolone versus laser in diabetic macular edema. Retina 2012;32(2):314‐21.
Soheilian M, Ramezani A, Bijanzadeh B, Yaseri M, Ahmadieh H, Dehghan MH, et al. Intravitreal bevacizumab (avastin) injection alone or combined with triamcinolone versus macular photocoagulation as primary treatment of diabetic macular edema. Retina 2007;27(9):1187‐95.
Soheilian M, Ramezani A, Obudi A, Bijanzadeh B, Salehipour M, Yaseri M, et al. Randomized trial of intravitreal bevacizumab alone or combined with triamcinolone versus macular photocoagulation in diabetic macular edema. Ophthalmology 2009;116(6):1142‐50.

References to studies excluded from this review

DRCRnet 2007 {published data only}

Diabetic Retinopathy Clinical Research Network, Scott IU, Edwards AR, Beck RW, Bressler NM, Chan CK, et al. A phase II randomized clinical trial of intravitreal bevacizumab for diabetic macular edema. Ophthalmology 2007;114(10):1860‐7.

DRCRnet 2011 {published data only}

Diabetic Retinopathy Clinical Research Network, Googe J, Brucker AJ, Bressler NM, Qin H, Aiello LP, et al. Randomized trial evaluating short‐term effects of intravitreal ranibizumab or triamcinolone acetonide on macular edema after focal/grid laser for diabetic macular edema in eyes also receiving panretinal photocoagulation. Retina 2011;31(6):1009‐27.

DRCRnet 2012 {published data only}

Diabetic Retinopathy Clinical Research Network, Elman MJ, Qin H, Aiello LP, Beck RW, Bressler NM, et al. Intravitreal ranibizumab for diabetic macular edema with prompt versus deferred laser treatment: three‐year randomized trial results. Ophthalmology 2012, Sep 19; Epub ahead of print.

Faghihi 2008 {published data only}

Faghihi H, Roohipoor R, Mohammadi SF, Hojat‐Jalali K, Mirshahi A, Lashay A, et al. Intravitreal bevacizumab versus combined bevacizumab‐triamcinolone versus macular laser photocoagulation in diabetic macular edema. European Journal of Ophthalmology 2008;18(6):941‐8.

Lim 2012 {published data only}

Lim JW, Lee HK, Shin MC. Comparison of intravitreal bevacizumab alone or combined with triamcinolone versus triamcinolone in diabetic macular edema: a randomized clinical trial. Ophthalmologica 2012;227(2):100‐6.

Paccola 2008 {published data only}

Paccola L, Costa RA, Folgosa MS, Barbosa JC, Scott IU, Jorge R. Intravitreal triamcinolone versus bevacizumab for treatment of refractory diabetic macular oedema (IBEME study). British Journal of Ophthalmology 2008;92(1):76‐80.

Solaiman 2010 {published data only}

Solaiman KA, Diab MM, Abo‐Elenin M. Intravitreal bevacizumab and/or macular photocoagulation as a primary treatment for diffuse diabetic macular edema. Retina 2010;30(10):1638‐45.

NCT00387582 {published data only}

Efficacy study of lucentis in the treatment of diabetic macular edema ‐ a phase II, single center, randomized study to evaluate the efficacy of ranibizumab versus focal laser treatment in subjects with diabetic macular edema. clinicaltrials.gov/show/NCT00387582 (accessed 15 Oct 2012).

NCT00901186 {published data only}

A randomized, open label, multicenter, laser‐controlled phase II study assessing the efficacy and safety of ranibizumab (intravitreal Injections) vs. laser treatment in patients with visual impairment due to diabetic macular edema. clinicaltrials.gov/show/NCT00901186 (accessed 15 Oct 2012).

NCT00989989 {published data only}

A randomized, double‐masked, multicenter, laser controlled phase III study assessing the efficacy and safety of ranibizumab in patients with diabetic macular edema. clinicaltrials.gov/show/NCT00989989 (accessed 15 Oct 2012).

NCT00997191 {published data only}

Intravitreal bevacizumab and intravitreal triamcinolone associated to laser photocoagulation for diabetic macular edema (IBeTA). clinicaltrials.gov/show/NCT00997191 (accessed 15 Oct 2012).

NCT01077401 {published data only}

A phase 3, randomized, controlled, double‐masked, multi‐center, comparative, in parallel roups (for 24 weeks), to compare the efficacy and safety of 0.3 mg pegaptanib sodium, with sham injections, and open study (for 30 weeks) to confirm the safety of 0.3 mg pegaptanib sodium in subjects with diabetic macular edema (DME). clinicaltrials.gov/show/NCT01100307 (accessed 15 Oct 2012).

NCT01100307 {published data only}

Ranibizumab for edema of the macula in diabetes: protocol 3 with high dose ‐ the READ 3 study. clinicaltrials.gov/show/NCT01077401 (accessed on 15 Oct 2012).

NCT01112085 {published data only}

Phase 2 study of microdoses of ranibizumab in diabetic macular edema ‐ the MINIMA 2 study. clinicaltrials.gov/show/NCT01112085 (accessed 15 Oct 2012).

NCT01131585 {published data only}

A 12‐month, two‐armed, randomized, double‐masked, multicenter, phase IIIb study assessing the efficacy and safety of laser photocoagulation as adjunctive to ranibizumab intravitreal injections vs. laser photocoagulation monotherapy in patients with visual impairment due to diabetic macular edema followed by a 12 month follow up period. clinicaltrials.gov/show/NCT01131585 (accessed 15 Oct 2012).

NCT01171976 {published data only}

Diabetic macula oedema. A prospective randomised study comparing the detailed functional and anatomical changes of repeated pan Anti‐VEGF therapy with ranibizumab versus conventional macular laser therapy. clinicaltrials.gov/show/NCT01223612 (accessed 15 Oct 2012).

NCT01223612 {published data only}

Diabetic macula oedema. A prospective randomised study comparing the detailed functional and anatomical changes of repeated pan Anti‐VEGF therapy with ranibizumab versus conventional macular laser therapy. clinicaltrials.gov/show/NCT01223612 (accessed 15 Oct 2012).

NCT01445899 {published data only}

An open‐label dose escalation study of PF‐04523655 (Stratum I) combined with a prospective, randomized, double‐masked, multi‐center, controlled study (Stratum II) evaluating the efficacy and safety of PF‐04523655 alone and in combination with ranibizumab versus ranibizumab alone in diabetic macular edema (MATISSE STUDY). clinicaltrials.gov/ct2/show/study/NCT01445899 (accessed 15 Oct 2012).

NCT01476449 {published data only}

Monthly ranibizumab versus treat and extend ranibizumab for diabetic macular edema. clinicaltrials.gov/ct2/show/NCT01476449 (accessed 15 Oct 2012).

NCT01487629 {published data only}

Bevacizumab versus ranibizumab for the treatment of diabetic macular edema. clinicaltrials.gov/ct2/show/study/NCT01487629 (accessed 15 Oct 2012).

NCT01552408 {published data only}

A phase I/II, randomized, study for diabetic macular edema using 0.3mg ranibizumab combined with targeted PRP monthly for 4 months, then PRN vs. 0.3mg ranibizumab 4 months monotherapy, then as needed(DME‐AntiVEgf) DAVE. clinicaltrials.gov/ct2/show/study/NCT01552408 (accessed 15 Oct 2012).

NCT01565148 {published data only}

A randomized, multi‐center, phase II study of the safety, tolerability, and bioactivity of repeated intravitreal injections of iCo‐007 as monotherapy or in combination with ranibizumab or laser photocoagulation in the treatment of diabetic macular edema with involvement of the foveAL center (the iDEAL Study). clinicaltrials.gov/ct2/show/study/NCT01565148 (accessed 15 Oct 2012).

NCT01572350 {published data only}

Randomized multicenter clinical trial of three parallel groups to estimate the safety and efficacy of triamcinolone acetonide combined with laser, bevacizumab combined with laser versus laser alone for the treatment of diffuse non‐tractional diabetic macular edema. clinicaltrials.gov/ct2/show/study/NCT01572350 (accessed 15 Oct 2012).

NCT01610557 {published data only}

A phase II randomized study to compare anti‐VEGF agents in the treatment of diabetic macular edema (CADME). clinicaltrials.gov/ct2/show/study/NCT01610557 (accessed 15 Oct 2012).

NCT01627249 {published data only}

A comparative effectiveness study of intravitreal aflibercept, bevacizumab and ranibizumab for diabetic macular edema. clinicaltrials.gov/ct2/show/study/NCT01627249 (accessed 15 Oct 2012).

AAO 2012

Ho AC, Scott IU, Kim S, Brown GC, Brown MM, Ip MS, et al. Anti‐vascular endothelial growth factor pharmacotherapy for diabetic macular edema: a report by the american academy of ophthalmology. Ophthalmology 2012;119(10):2179‐88.

Aiello 2005

Aiello LP. Angiogenic pathways in diabetic retinopathy. New England Journal of Medicine 2005;353(8):839‐41.

Antcliff 1999

Antcliff RJ, Marshall J. The pathogenesis of edema in diabetic maculopathy. Seminars in Ophthalmology 1999;14(4):223‐32.

Arevalo 2007

Arevalo JF, Fromow‐Guerra J, Quiroz‐Mercado H, Sanchez JG, Wu L, Maia M, et al. Pan‐American Collaborative Retina Study Goup. Primary intravitreal bevacizumab (Avastin) for diabetic macular edema: results from the pan‐American Collaborative Retina Study Group at 6‐month follow‐up. Ophthalmology 2007;114(4):743‐50.

Arevalo 2009

Arevalo JF, Garcia‐Amaris RA. Intravitreal bevacizumab for diabetic retinopathy. Current Diabetes Reviews 2009;5(1):39‐46.

Aroca 2004

Aroca PR, Salvat M, Fernandez J, Mendez I. Risk factors for diffuse and focal macular edema. Journal of Diabetic Complications 2004;18(4):211‐5.

ATC 1994

Anonymous. Collaborative overview of randomised trials of antiplatelet therapy‐‐I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various categories of patients. Antiplatelet Trialists' Collaboration. BMJ 1994;308(6921):81‐106.

Avery 2006

Avery RL, Pieramici DJ, Rabena MD. Intravitreal bevacizumab (Avastin) for neovascular age related macular degeneration. Ophthalmology 2006;113(3):363‐72.

Borm 2009

Borm GF, Lemmers O, Fransen J, Donders R. The evidence provided by a single trial is less reliable than its statistical analysis suggests. Journal of Clinical Epidemiology 2009;62(7):711‐5.

Boscia 2010

Boscia F. Current approaches to the management of diabetic retinopathy and diabetic macular oedema. Drugs 2010;70(16):2171‐200.

BRAVO 2010

Campochiaro PA, Heier JS, Feiner L, Gray S, Saroj N, Rundle AC, et al. Ranibizumab for macular edema following branch retinal vein occlusion: six‐month primary end point results of a phase III study. Ophthalmology 2010;117(6):1102‐12.

BRAVO 2011

Brown DM, Campochiaro PA, Bhisitkul RB, Ho AC, Gray S, Saroj N, et al. Sustained benefits from ranibizumab for macular edema following branch retinal vein occlusion: 12‐month outcomes of a phase III study. Ophthalmology 2011;118(8):1594‐602.

Brazier 2012

Brazier J, Rowen D, Mavranezouli I, Tsuchiya A, Young T, Yang Y, et al. Developing and testing methods for deriving preference‐based measures of health from condition‐specific measures (and other patient‐based measures of outcome). Health Technology Assessment 2012;16(32):1‐114.

Bresnick 1976

Bresnick GH, Engerman R, Davis MD, de Venecia G, Myers FL. Patterns of ischemia in diabetic retinopathy. Transactions. Section on Ophthalmology. American Academy of Ophthalmology and Otolaryngology 1976;81(4 Pt 1):OP694‐709.

Bresnick 1983

Bresnick GH. Diabetic maculopathy: a critical review highlighting diffuse macular edema. Ophthalmology 1983;90(11):1301‐17.

Bresnick 1984

Bresnick GH, Condit R, Syriala S, Palta M, Groo A, Korth K. Abnormalities of the foveal avascular zone in diabetic retinopathy. Archives of Ophthalmology 1984;102(9):1286‐93.

Bressler 2012

Bressler NM, Boyer DS, Williams DF, Butler S, Francom SF, Brown B, et al. Cerebrovascular accidents in patients treated for choroidal neovascularization with ranibizumab in randomized controlled trials. Retina 2012;32(9):1821‐8.

Brown 2004

Brown JC, Solomon SD, Bressler SB, Schachat AP, DiBernardo C, Bressler NM. Detection of diabetic foveal edema: contact lens biomicroscopy compared with optical coherence tomography. Archives of Ophthalmology 2004;122(3):330‐5.

CATT 2011

CATT Research Group, Martin DF, Maguire MG, Ying GS, Grunwald JE, Fine SL, et al. Ranibizumab and bevacizumab for neovascular age‐related macular degeneration. New England Journal of Medicine 2011;364(20):1897‐908.

Ciulla 2003

Ciulla TA, Amador AG, Zinman B. Diabetic retinopathy and diabetic macular edema: pathophysiology, screening, and novel therapies. Diabetes Care 2003;26(9):2653‐64.

Ciulla 2004

Ciulla TA, Walker JD, Fong DS, Criswell MH. Corticosteroids in posterior segment disease: an update on new delivery systems and new indications. Current Opinions in Ophthalmology 2004;15(3):211‐20.

CLEAR‐IT 2

Heier JS, Boyer D, Nguyen QD, Marcus D, Roth DB, Yancopoulos G, et al. The 1‐year results of CLEAR‐IT 2, a phase 2 study of vascular endothelial growth factor trap‐eye dosed as‐needed after 12‐week fixed dosing. Ophthalmology 2011;118(6):1098‐106.

CRUISE 2010

Brown DM, Campochiaro PA, Singh RP, Li Z, Gray S, Saroj N, et al. Ranibizumab for macular edema following central retinal vein occlusion: six‐month primary end point results of a phase III study. Ophthalmology 2010;117(6):1124‐33.

CRUISE 2011

Campochiaro PA, Brown DM, Awh CC, Lee SY, Gray S, Saroj N, et al. Sustained benefits from ranibizumab for macular edema following central retinal vein occlusion: twelve‐month outcomes of a phase III study. Ophthalmology 2011;118(10):2041‐9.

Cunha‐Vaz 1998

Cunha‐Vaz J. Diabetic macular edema. European Journal of Ophthalmology 1998;8(3):127‐30.

Cunningham 2005

Cunningham ET, Adamis AP, Altaweel M, Aiello LP, Bressler NM, D'Amico DJ, et al. Macugen Diabetic Retinopathy Study Group. A phase II randomized double‐masked trial of pegaptanib, an anti‐vascular endothelial growth factor aptamer, for diabetic macular edema. Ophthalmology 2005;112(10):1747‐57.

Czosky‐Murray 2008

Czoski‐Murray C, Carlton J, Brazier J, Young T, Papo NL, Kang HK. Valuing condition‐specific health states using simulation contact lenses. Value Health 2009;12:793‐9.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Dewan 2012

Dewan V, Lambert D, Edler J, Kymes S, Apte RS. Cost‐effectiveness analysis of ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology 2012;119(8):1679‐84.

ETDRS 1985

Early Treatment Diabetic Retinopathy Study Research Group. Photocoagulation for diabetic macular edema. Early Treatment Diabetic Retinopathy Study report number 1. Archives of Ophthalmology 1985;103(12):1796‐806.

Evers 2005

Evers S, Goossens M, de Vet H, van Tulder M, Ament A. Criteria list for assessment of methodological quality of economic evaluations: Consensus on Health Economic Criteria. International Journal of Technology Assessment in Health Care 2005;21(2):240‐5.

Ferrara 2004

Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti‐VEGF antibody for treating cancer. Nature Reviews. Drug Discovery 2004;3(5):391‐400.

Ferrara 2006

Ferrara N, Damico L, Shams N, Lowman H, Kim R. Development of ranibizumab, an anti‐vascular endothelial growth factor antigen binding fragment, as therapy for neovascular age‐related macular degeneration. Retina 2006;26(8):859‐70.

Ferris 1984

Ferris FL, Fine SL, Hyman L. Age‐related macular degeneration and blindness due to neovascular maculopathy. Archives of Ophthalmology 1984;102(11):1640‐2.

Ford 2012

Ford JA, Elders A, Shyangdan D, Royle P, Waugh N. The relative clinical effectiveness of ranibizumab and bevacizumab in diabetic macular oedema: an indirect comparison in a systematic review. BMJ 2012;345:e5182.

Frank 2004

Frank RN. Diabetic Retinopathy. New England Journal of Medicine 2004;350(1):48‐58.

Glanville 2006

Glanville JM, Lefebvre C, Miles JN, Camosso‐Stefinovic J. How to identify randomized controlled trials in MEDLINE: ten years on. Journal of the Medical Library Association 2006;94(2):130‐6.

Goyal 2011

Goyal S, Lavalley M, Subramanian ML. Meta‐analysis and review on the effect of bevacizumab in diabetic macular edema. Graefe's Archive for Clinical and Experimental Ophthalmology 2011;249(1):15‐27.

Gragoudas 2004

Gragoudas ES, Adamis AP, Cunningham ET Jr. Feinsod M, Guyer DR. VEGF Inhibition Study in Ocular Neovascularization Clinical Trial Group. Pegaptanib for neovascular age‐related macular degeneration. New England Journal of Medicine 2004;351(27):2805‐16.

Grover 2008

Grover D, Li TJ, Chong CC. Intravitreal steroids for macular edema in diabetes. Cochrane Database of Systematic Reviews 2008, Issue 1. [DOI: 10.1002/14651858.CD005656.pub2]

Guyatt 2011

Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence‐‐imprecision. Journal of Clinical Epidemiology 2011;64(12):1283‐93.

Haller 2010

Haller JA, Kuppermann BD, Blumenkranz MS, Williams GA, Weinberg DV, Chou C, et al. Randomized controlled trial of an intravitreous dexamethasone drug delivery system in patients with diabetic macular edema. Archives of Ophthalmology 2010;128(3):289‐96.

Harbour 1996

Harbour JW, Smiddy WE, Flynn HW, Rubsamen RE. Vitrectomy for diabetic macular edema associated with a thickened and taut posterior hyaloid membrane. American Journal of Ophthalmology 1996;121(4):405‐13.

Higgins 2011a

Higgins JPT, Deeks JJ, Altman DG (editors). Chapter 16: Special topics in statistics. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Higgins 2011b

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Iturralde 2006

Iturralde D Spaide RF, Meyerle CB, Klancnik JM, Yannuzzi LA, Fisher YL, et al. Intravitreal bevacizumab (Avastin) treatment of macular edema in central retina vein occlusion: a short‐term study. Retina 2006;26(3):279‐84.

Klein 1984

Klein R, Klein BE, Moss SE, Davis MD, DeMets DL. The Wisconsin epidemiologic study of diabetic retinopathy. IV. Diabetic macular edema. Ophthalmology 1984;91(12):1464‐74.

Kuppermann 2010

Kuppermann BD, Chou C, Weinberg DV, Whitcup SM, Haller JA, Blumenkranz MS. Intravitreous dexamethasone effects on different patterns of diabetic macular edema. Archives of Ophthalmology 2010;128(5):642‐3.

Kylstra 1999

Kylstra JA, Brown JC, Jaffe GJ, Cox TA, Gallemore R, Greven CM, et al. The importance of fluorescein angiography in planning laser treatment of diabetic macular edema. Ophthalmology 1999;106(11):2068‐73.

Ladas 1993

Ladas ID, Theodossiadis GP. Long‐term effectiveness of modified grid laser photocoagulation for diffuse diabetic macular edema. Acta Ophthalmologica 1993;71(3):393‐7.

Lee 1991

Lee CM, Olk RJ. Modified grid laser photocoagulation for diffuse diabetic macular edema. Long‐term visual results. Ophthalmology 1991;98(10):1594‐602.

Lewis 1992

Lewis H, Abrams GW, Blumenkranz MS, Campo RV. Vitrectomy for diabetic macular traction and edema associated with posterior hyaloidal traction. Ophthalmology 1992;99(5):753‐9.

Lloyd 2008

Lloyd A, Nafees B, Gavriel S, Rousculp MD, Boye KS, Ahmad A. Health utility values associated with diabetic retinopathy. Diabetic Medicine 2008;25(5):618‐24.

Manousaridis 2012

Manousaridis K, Talks J. Macular ischaemia: a contraindication for anti‐VEGF treatment in retinal vascular disease?. British Journal of Ophthalmology 2012;96(2):179‐84.

MEDCAC 2012

Anti‐vascular endothelial growth factor treatment for diabetic macular edema. www.cms.gov/medicare‐coverage‐database/details/technology‐assessments‐details.aspx?TAId=85&bc=AAAQAAAAAAAA& (accessed 24 Sept 2012).

Mitchell 2012

Mitchell P, Annemans L, Gallagher M, Hasan R, Thomas S, Gairy K, et al. Cost‐effectiveness of ranibizumab in treatment of diabetic macular oedema (DME) causing visual impairment: evidence from the RESTORE trial. British Journal of Ophthalmology 2012;96(5):688‐93.

NICE 2008

National Institute for Health and Clinical Excellence. Pegaptanib and ranibizumab for the treatment of age‐related macular degeneration. http://guidance.nice.org.uk/TA155 (accessed 21 Oct 2012).

NICE 2011

National Institute for Health and Clinical Excellence. Ranibizumab for the treatment of diabetic macular oedema Issued: November 2011 NICE technology appraisal guidance 237. www.nice.org.uk/nicemedia/live/13125/55324/55324.pdf (accessed 20 Sept 2012).

NICE 2012

National Institute for Health and Clinical Excellence. Ranibizumab for the treatment of diabetic macular oedema (rapid review of TA237). http://guidance.nice.org.uk/TA/Wave23/41 (accessed 21 Oct 2012).

Nicholson 2010

Nicholson BP, Schachat AP. A review of clinical trials of anti‐VEGF agents for diabetic retinopathy. Graefe's Archive for Clinical and Experimental Ophthalmology 2010;248(7):915‐30.

O' Malley 2012

O'Malley PG. Comparative effectiveness of anti‐growth factor therapies for diabetic macular edema: summary of primary findings and conclusions. Archives of Internal Medicine 2012;172(13):1014‐5.

O'Doherty 2008

O'Doherty M, Dooley I, Hickey‐Dwyer M. Interventions for diabetic macular oedema: a systematic review of the literature. British Journal of Ophthalmology 2008;92(12):1581‐90.

Philips 2004

Philips Z, Ginnelly L, Sculpher M, Claxton K, Golder S, Riemsma R, et al. Review of guidelines for good practice in decision‐analytic modelling in health technology assessment. Health Technology Assessment 2004;8(36):1‐158.

Presta 1997

Presta LG, Chen H, O'Connor SJ, Chisholm V, Meng YG, Krummen L, et al. Humanization of an anti‐vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Research 1997;57(20):4593‐9.

RevMan 2011 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.1. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2011.

Rosenfeld 2006

Rosenfeld PJ, Rich RM, Lalwani GA. Ranibizumab: Phase III clinical trial results. Ophthalmology Clinics of North America 2006;19(3):361‐72.

Salam 2010

Salam A, DaCosta J, Sivaprasad S. Anti‐vascular endothelial growth factor agents for diabetic maculopathy. British Journal of Ophthalmology 2010;94(7):821‐6.

Schmucker 2012

Schmucker C, Ehlken C, Agostini HT, Antes G, Ruecker G, Lelgemann M, et al. A safety review and meta‐analyses of bevacizumab and ranibizumab: off‐label versus gold standard. PLoS One 2012;7(8):e42701.

Smiddy 2007

Smiddy WE. Relative cost of a line of vision in age‐related macular degeneration. Ophthalmology 2007;114(5):847‐54.

Smiddy 2011

Smiddy WE. Economic considerations of macular edema therapies. Ophthalmology 2011;118(9):827‐33.

Spaide 2006

Spaide RF, Fisher YL. Intravitreal bevacizumab (Avastin) treatment of proliferative diabetic retinopathy complicated by vitreous hemorrhage. Retina 2006;26(3):275‐8.

Ticho 1973

Ticho U, Patz A. The role of capillary perfusion in the management of diabetic macular edema. American Journal of Ophthalmology 1973;76(6):880‐6.

Torjesen 2012

Torjesen I. Why using Avastin for eye disease is so difficult. BMJ 2012;344:e3012.

Tranos 2004

Tranos PG, Wickremasinghe SS, Stangos NT, Topouzis F, Tsinopoulos I, Pavesio CE. Macular edema. Survey of Ophthalmology 2004;49(5):470‐90.

Vedula 2008

Vedula SS, Krzystzolyk MG. Antiangiogenic therapy with anti‐vascular endothelial growth factor modalities for neovascular age‐related macular degeneration. Cochrane Database of Systematic Reviews 2008, Issue 2. [DOI: 10.1002/14651858.CD005139.pub2]

Wang 2012

Wang H, Sun X, Liu K, Xu X. Intravitreal ranibizumab (lucentis) for the treatment of diabetic macular edema: a systematic review and meta‐analysis of randomized clinical control trials. Current Eye Research 2012;37(8):661‐70.

White 2008

White IR, Higgins JP, Wood AM. Allowing for uncertainty due to missing data in meta‐analysis ‐ Part 1. Two‐stage methods. Statistics in Medicine 2008;27(5):711‐27.

Williams 2004

Williams R, Airey M, Baxter H, Forrester J, Kennedy‐Martin T, Girach A. Epidemiology of diabetic retinopathy and macular oedema: a systematic review. Eye 2004;18(10):963‐83.

Zechmeister‐Koss 2012

Zechmeister‐Koss I, Huic M. Vascular endothelial growth factor inhibitors (anti‐VEGF) in the management of diabetic macular oedema: a systematic review. British Journal of Ophthalmology 2012;96(2):167‐78.

References to other published versions of this review

Parravano 2009

Parravano M, Menchini F, Virgili G. Antiangiogenic therapy with anti‐vascular endothelial growth factor modalities for diabetic macular oedema. Cochrane Database of Systematic Reviews 2009, Issue 4. [DOI: 10.1002/14651858.CD007419.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Ahmadieh 2008

Methods

Randomisation was performed using a random block permutation method according to a computer‐generated randomisation list. The block lengths varied randomly. A random allocation sequence was performed by a biostatistician. Details of the series were unknown to the investigators.
Participants were masked to the treatment method, but not specified if care providers were masked to treatment method. VA assessment and OCT were performed by optometrists who were masked to the groups.
Exclusion after randomisation: One patient was excluded in the bevacizumab/triamcinolone group because of vitreous haemorrhage. no loss to follow‐up mentioned.

Participants

Country: Iran.

Number randomised: 101 participants, 115 eyes. Age: 59.7 ± 8.3 (39 to 74) years.

Sex: 50 (49.5%) M, 51 (50.5%) F.
Inclusion criteria: Eyes with clinically significant macular oedema unresponsive to previous macular laser photocoagulation, with the last session being more than 3 months prior, were included.

Exclusion criteria: Visual acuity > 20/40, history of cataract surgery within the past 6 months, prior intraocular injection or vitrectomy, glaucoma or ocular hypertension, PDR with high‐risk characteristics, vitreous haemorrhage, significant media opacity, and presence of traction on the macula. Monocular patients were excluded. Pregnancy and serum creatinine level > 3 mg/100 were also among the exclusion criteria.

Interventions

Treatment: Participants were randomly assigned to one of the three study arms: 1) three injections of IVB (1.25 mg/0.05 mL) at 6‐week intervals, 2) combined IVB and IVT (1.25 mg/0.05 mL and 2 mg/0.05 mL respectively) followed by two injections of IVB at 6‐week intervals. Control: 3) sham injection (control group). Duration: 24 weeks.

Outcomes

The primary outcome measure was change in CMT compared to baseline. CMT was defined by the average thickness of a central macular region 1,000 µm in diameter centred on the patientÍs foveola. Secondary outcome measures included change in best‐corrected logMAR visual acuity, IOP rise, cataract progression, intraocular inflammation, and any other serious adverse effect.

Notes

None

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

See 'Characteristics of included studies' table.

Allocation concealment (selection bias)

Low risk

See 'Characteristics of included studies' table.

Blinding (performance bias and detection bias)
All outcomes

Low risk

See 'Characteristics of included studies' table.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No incomplete outcome data were reported, but number of patients at 24 weeks follow‐up was not specified.

Selective reporting (reporting bias)

High risk

The study protocol is mentioned. However, dichotomous visual acuity outcomes are not provided.

Other bias

High risk

28 eyes of 14 patients (14%) with bilateral CSMO were included in the analysis.

BOLT

Methods

Patients were randomised into 2 groups by means of an in‐house computerised randomisation program.

The research investigator was not involved in the randomisation process.

Patients were stratified for BCVA, with the aim being that both groups would have comparable mean baseline BCVAs. If both eyes were eligible for enrolment, the eye with the worst VA was randomised.

Participants

Country: UK.

Number randomised: 80 patients (42 bevacizumab group, 38 laser).

Age: 64.9 ± 9.4 yrs bevacizumab group; 63.5 ± 8.1 yrs laser group.

Sex:M/F 30/12 bevacizumab group; 25/13 laser group.

Inclusion criteria:

1) patients of either gender aged >18 years; (2) diabetes mellitus (type 1 or 2); (3) BCVA in the study eye between 35 and 69 ETDRS letters at 4 m (Snellen equivalent ≥ 6/60 or ≤ 6/12); (4) centre‐involving CSMO with CMT on optical coherence tomography (OCT) of > 270 mm; (5) media clarity, pupillary dilation, and participant co‐operation sufficient for adequate fundus imaging; (6) at least 1 prior macular laser; (7) IOP > 30 mmHg; (8) ability to return for regular study visits; (9) fellow eye BCVA ≥ 3/60; and 10) fellow eye has received no anti‐VEGF treatment within the previous 3 months and no expectation of such treatment during the study.

Exclusion criteria:

(1) macular ischaemia (foveal avascular zone [FAZ] ≥ 1000 mm GLD or severe perifoveal intercapillary loss on FFA); (2) MO due to a cause other than DMO; (3) coexistent ocular disease: (i) a pre‐existing ocular condition that was likely to preclude VA improvement despite resolution of macular oedema (e.g., foveal atrophy, dense subfoveal hard exudates, marked cataract, amblyopia) or (ii) an ocular condition that may affect macular oedema or alter VA during the course of the study (e.g., retinal vascular occlusion, ocular inflammatory disease, neovascular glaucoma, Irvine‐Gass syndrome); (4) any treatment for DMO in the preceding 3 months; (5) panretinal photocoagulation within 3 months of enrolment or anticipated 6 months thereafter; (6) proliferative diabetic retinopathy except for tufts of new vessels elsewhere < 1 disc in area with no vitreous haemorrhage; (7) haemoglobin A1c (HbA1c) > 11.0%; (8) medical history of chronic renal failure requiring dialysis or kidney transplantation; (9) BP > 170/100 mmHg; (10) any thromboembolic event within 6 months, unstable angina, or evidence of active ischaemia on ECG at time of screening; (11) major surgery within 28 days of randomisation or planned during the subsequent 12 months; (12) participation in an investigational drug trial within 30 days of randomisation (or any time during the study); (13) systemic anti‐VEGF or pro‐VEGF treatment within 3 months of enrolment; (14) pregnancy, breast feeding, or intention to become pregnant within the study period; (15) intraocular surgery within 3 months of randomisation; (16) aphakia; (17) uncontrolled glaucoma; and (18) significant external ocular disease.

Interventions

Participants were randomly assigned to bevacizumab or laser treatment.

Duration: 12 months.

Outcomes

The primary outcome measure of the trial was a comparison of the mean ETDRS BCVA at 12 months between the iv bevacizumab and laser arms.

The secondary outcome measures relating to efficacy were a comparison between both groups at 12 months with regard to (i) mean CMT; (ii) mean change in CMT; (iii) mean change in ETDRS BCVA; (iv) the proportion of patients who gained > 15 and > 10 ETDRS letters (improvement); (v) the proportion of patients who lost < 15 ETDRS letters (stabilisation); (vi) the proportion of patients who lost > 30 ETDRS letters; and (vii) ETDRS grading of retinopathy severity.

The secondary outcome measures relating to safety were a comparison between both arms at 12 months with regard to (i) GLD of the FAZ, area of the FAZ, and PFCL (the methodology of determining these parameters has been previously described in detail); 24 (ii) RNFL thickness; (iii) other ocular side effects; and (iv) systemic side effects, including thromboembolic events, BP, and ECG findings.

Notes

None

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generated by means of in‐house computerised randomisation program.

Allocation concealment (selection bias)

Low risk

The doctor had to phone the CTU in order to obtain a randomisation from the statistician.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "Although the patient and the study physician were not masked to the therapeutic modality, the study optometrist, OCT technician, photographer, graders performing assessment of the FAZ and ETDRS retinopathy grading, and study statistician were all masked to the patient randomization." Comment: masking outcome assessors should suffice to avoid bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Two patients in the laser group did not complete 12 months of follow‐up (1 patient moved away, and 1 patient could not be contacted). They were last reviewed at the 32‐week time point,with these data being carried forward and an intention‐to‐treat analysis undertaken. All 42 patients in the ivB group completed the study."

Selective reporting (reporting bias)

Low risk

We could not find a protocol but primary outcomes were stated in the methods and were those routinely used in the field.

Other bias

Low risk

No other bias identified.

DA VINCI

Methods

Quote: "Randomized, double‐masked, active controlled multicenter phase 2 clinical trial. Thirty‐nine sites in the United States, Canada, and Austria participated in the trial, and patients were enrolled between December 2008 and June 2009. The primary objective was to assess the efficacy of various doses and dose intervals of intravitreal VEGF Trap‐Eye (aflibercept injection) on BCVA."

Participants

Quote: "The study enrolled adult patients 18 years of age or older with type 1 or 2 diabetes mellitus with clinically significant DME with center involvement of the fovea, defined as a central subfield measurement of 250 micron or more on time‐domain OCT (Stratus OCT)". "In addition, patients had an ETDRS BCVA letter score at 4m of 73 to 24 (20/40 to 20/320) in the study eye."

Patients were excluded for a number of ocular comorbidities and recent ocular treatment, as well as uncontrolled diabetes.

Interventions

Quote: "Eyes were assigned randomly using a 1:1:1:1:1 ratio to one of the following treatment regimens: (1) 0.5 mg VEGF Trap‐Eye every 4 weeks (0.5q4); (2) 2 mg VEGF Trap‐Eye every 4 weeks (2q4); (3) 2 mg VEGF Trap‐Eye every 8 weeks after 3 initial monthly doses (2q8); (4) 2 mg VEGF Trap‐Eye, with dosing as needed after 3 initial monthly doses (2PRN); (5) laser photocoagulation using a modified ETDRS protocol at baseline and then as needed (but no more frequently than every 16 weeks). Eyes in the laser group also received a sham injection every 4 weeks."

Outcomes

Quote: "The primary end point was the change inBCVA from baseline to week 24. Secondary objectives were to assess the effects of intravitreal VEGF Trap‐Eye on retinal thickness assessed by optical coherence tomography (OCT) and to assess safety and tolerability of intravitreal VEGF Trap‐Eye in eyes with DME. Secondary outcomes were the change in BCVA from baseline at week 52, the proportion of eyes that gained at least 15 ETDRS letters in BCVA compared with baseline at weeks24 and 52, the change in central retinal thickness (CRT; central subfield on OCT) from baseline to weeks 24 and 52, and the number of focal laser treatments given."

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quoted text provided by the producer: "The randomization was handled by an IVRS vendor. The study statistician at REGENERON provided the randomization plan and reviewed and approved the dummy rand table. Study Data Management at REGENERON tested the randomization function extensively along with the Clinical team."

Allocation concealment (selection bias)

Low risk

Quoted text provided by the producer: "Sites called into IVRS to randomize patients and received the randomization number and drug kit assignment at the completion of the call. The site also received a confirmation email. Neither of these contained the actual randomization assignment. The randomization assignments were kept by the IVRS vendor in a secure, access‐controlled database and were delivered to REGENERON by the IVRS vendor at the primary endpoint database lock."

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "Treatments (study drug injection, sham injection, laser or sham laser photocoagulation) were performed by an unmasked physician. A separate masked physician was assigned to assess adverse events (AEs) and retreatment and rescue criteria and to supervise the masked assessment of efficacy. Every effort was made to ensure that all other study site personnel remained masked to treatment assignment to facilitate an unbiased assessment of efficacy and safety."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Forty‐three patients discontinued the study after receiving at least 1 treatment for the following reasons: lost to follow‐up (n 11), withdrew consent (n 11), death (n 6), treatment failures (n 2), AE (n 7), protocol deviation (n 2), other (n 4). Discontinuations were distributed evenly among all the treatment groups."

Comment: LOCF used.

Selective reporting (reporting bias)

Low risk

Primary outcome declared and consistent with our review.

Other bias

Low risk

No other bias identified.

DRCRnet

Methods

Study participants with 1 study eye were assigned randomly on the DRCR.net study website (using a permuted blocks design stratified by study eye visual acuity) with equal probability to 1 of 4 treatment groups. For study participants with 2 study eyes, the right eye was assigned randomly with equal probability to 1 of the 4 groups as indicated above. If the right eye was assigned to a treatment group other than the sham + prompt laser group, then the left eye was assigned to the sham + prompt laser group. If the right eye was assigned to the sham + prompt laser group, then the left eye was assigned randomly to 1 of the other 3 groups. Thus, there were more eyes in the sham + prompt laser group than in the other 3 groups.

Participants

Country: multicentre. Number randomised: 854 eyes (691 patients).

Age:

63 (57‐69) sham + prompt laser

62 (56‐70) ranibizumab + prompt laser

64 (58‐70) ranibizumab + deferred laser

62 (55‐70) triamcinolone + prompt laser

Sex: Females 123 (42%) sham + prompt laser

85 (45%) ranibizumab + prompt laser

78 (41%) ranibizumab + deferred laser

86 (46%) triamcinolone + prompt laser

Inclusion criteria: eligible patients were at least 18 years old with type 1 or 2 diabetes. The major eligibility criteria for a study eye included the following: (1) best‐corrected Electronic‐Early Treatment Diabetic Retinopathy Study (E‐ETDRS Visual Acuity Test11) visual acuity letter score 78 to 24 (20/32 to 20/320), (2) definite retinal thickening due to DME on clinical examination involving the centre of the macula assessed to be the main cause of visual loss, and (3) retinal thickness measured on time domain optical coherence tomography (OCT) ≥ 250 mm in the central subfield.

Exclusion criteria: Principal exclusion criteria included the following: (1) treatment for DMO within the prior 4 months, (2) panretinal photocoagulation within the prior 4 months or anticipated need for panretinal photocoagulation within the next 6 months, (3) major ocular surgery within the prior 4 months, (4) history of open‐angle glaucoma or steroid‐induced IOP elevation that required IOP‐lowering treatment, and (5) IOP > 25 mmHg. Patients were excluded if their systolic blood pressure was > 180 mmHg or diastolic blood pressure was > 110 mmHg, or if a myocardial infarction, other cardiac event requiring hospitalisation, cerebrovascular accident, transient Ischaemic attack, or treatment for acute congestive heart failure occurred within 4 months before randomisation.

Interventions

Treatment:

(1) sham injection plus prompt (within 3‐10 days after injection) focal/grid photocoagulation (sham + prompt laser group).

(2) 0.5 mg intravitreal ranibizumab plus prompt (within 3‐10 days after injection) focal/grid photocoagulation (ranibizumab + prompt laser group).

(3) 0.5 mg intravitreal ranibizumab with deferred (≥ 24 weeks) focal/grid photocoagulation (ranibizumab + deferred laser group).

(4) 4 mg intravitreal triamcinolone plus prompt (within 3 to 10 days after injection) focal/ grid photocoagulation (triamcinolone + prompt laser group.

Duration: 12 months (24 months).

Outcomes

BCVA and safety at 1 year.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation sequence was computer‐generated by the DRCR.net coordinating centre.

Allocation concealment (selection bias)

Low risk

Randomisation assignments were obtained through the DRCR.net study website, therefore no study personnel had access to the list or to the next assignment before it was assigned.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Participants: masked to treatment assignment.

Outcome assessor: VA assessment and OCT were performed by certified examiners masked for treatments.

Treating physician: unmasked

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Patients randomised in each group were: 293 laser, 187 ranibizumab + prompt laser, 188 ranibizumab + deferred laser and 186 IVTA + laser. At 1 year complete patients were 274, 171, 178, 176 respectively (91‐95%).

At 2 years complete patients were 211, 136, 139, 142 respectively (72‐76%).

Causes of missing were balanced across groups.

Selective reporting (reporting bias)

Low risk

We could not find a protocol but primary outcomes were stated in the methods and were those routinely used in the field.

Other bias

Low risk

No other source of bias identified.

Macugen 2005

Methods

Method of allocation: Dynamic minimisation procedure using a stochastic treatment allocation algorithm based on the variance method. Randomisation was stratified by study site, size of the thickened retina area (< 2.5 disc areas versus > 2.5 disc areas), and baseline BCVA (letter score > 58 versus letter score < 58). An independent fundus photograph and angiogram reading centre confirmed eligibility and appropriate retinal thickness classification both for study entry and for randomisation and stratification using baseline fluorescein angiography and OCT.
Masking of participants: Yes. Study participants receiving sham or study medication were treated identically in all regards, including ocular antisepsis procedures and subconjunctival anaesthetic, except that participants receiving active treatment had pegaptanib injected into the vitreous, whereas those receiving sham had a needleless syringe pressed against the conjunctiva and sclera. The injection procedure prevented participants from seeing the syringe and needle, to minimize the risk of unmasking.
Masking of care provider: Yes. In all but 3 participants, the injection was administered by a staff member other than the study ophthalmologist responsible for all other aspects of the protocol, to maintain investigator masking.
Masking of outcome assessor: Certified visual acuity and OCT examiners were masked both to randomisation and to findings of the previous measurement.
Exclusions after randomisation: None reported.
Loss to follow‐up: Nine participants were discontinued from the study before week 36 (3 mg subgroup: 2 patients by request at weeks 12 and 16 and 1 by other reason at week 1; sham subgroup: 5 patients by request at weeks 6, 11, 18, 30, and 33 and 1 due to death at week 8).

Participants

Country: USA
Number randomised: 172 participants.
Age: in 0.3 mg group: 61.9 ± 10.0 years, in 1 mg group: 62.8 ± 10.1 years, in 3 mg group: 61.3 ± 9.8 years, in sham group: 64.0 ± 9.3 years.
Sex: in 0.3 mg group M24/F20, in 1 mg group M22/F22, in 3 mg M19/F23, in sham group M23/F19.
Inclusion criteria: Macular oedema involving the centre of the macula demonstrated on OCT with corresponding leakage from microaneurysms, retinal telangiectasis, or both on fluorescein angiography; an area of retinal thickening of at least half a disc area involving the central macula as confirmed by graders at an independent fundus photograph and angiogram reading centre (University of Wisconsin, Madison, Wisconsin). Eligible patients had BCVA letter scores between 68 and 25 inclusive (approximate Snellen equivalent, 20/50 ‐20/320) in the study eye and at least 35 (20/100 or better) in the fellow eye, following a protocol.
VA measurement that used ETDRS charts. Other major inclusion criteria included IOP of no greater than 23 mmHg and assessment by the treating ophthalmologist that focal photocoagulation could be deferred safely for 16 weeks. Additional inclusion criteria included an electrocardiogram that demonstrated no abnormalities judged to be clinically relevant and serological test results that suggested no clinically meaningful hematological, liver, or renal abnormalities.
Exclusion criteria: A history of panretinal or focal photocoagulation; neodymium: yttrium‐aluminum‐garnet laser or peripheral retinal cryoablation within the previous 6 months; any abnormality thought likely to confound VA assessments or fundus photography, including cataract; vitreoretinal traction within 1 disc diameter of the fovea confirmed either clinically or on OCT; vitreous incarceration in a previous wound or incision; any retinal vein occlusion involving the macula; and atrophy/ scarring/fibrosis or hard exudates involving the centre of the macula that would preclude improvement in VA. Other exclusion criteria included a history of any intraocular surgery within the previous 12 months, myopia of > 8 diopters, axial length of > 25 mm, and the likelihood of requiring either scatter (panretinal) photocoagulation within the ensuing 9 months or cataract surgery within 12 months. Individuals also were excluded who had evidence of severe cardiac disease, clinically significant peripheral vascular disease (previous surgery, amputation, or symptoms of claudication), uncontrolled hypertension (treated systolic BP > 155 or diastolic BP > 95), or stroke within the preceding 12 months.

Interventions

Treatments: Patients were allocated to 0.3 mg, 1 mg, or 3 mg iv pegaptanib group.
Control: Sham injection.
Duration: 36 weeks.

Outcomes

Efficacy: VA, central retinal thickness on OCT, change in retinal thickness derived by comparing measurements at baseline with those at week 36 or final examination if before week 36. The proportion of participants for whom focal photocoagulation was applied at week 12 or later, size of the area of retinal thickness measured by photography, and fluorescein angiographic findings concerning macular capillary leakage and cystoid spaces also were determined.
Safety: Safety end points included all adverse events and serious adverse events, whether they were deemed related to treatment or not, as well as all laboratory test abnormalities.

Notes

Sponsored by Eyetech Pharmaceuticals, Inc., New York, New York, and Pfizer Inc., New York, New York

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Dynamic minimisation procedure using a stochastic treatment allocation algorithm based on the variance method.

Allocation concealment (selection bias)

Low risk

An independent fundus photograph and angiogram reading centre confirmed eligibility and appropriate retinal thickness classification both for study entry and for randomisation and stratification using baseline fluorescein angiography and OCT.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Patients, investigators and outcome assessors were masked for all outcome measures.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Also see Results section, Risk of bias in included studies. Nine participants were discontinued from the study before week 36. None in pegaptanib groups 0.3 mg and 1 mg, 3 in pegaptanib 3 mg group (3 mg subgroup: 2 patients by request at weeks 12 and 16 and 1 by other reason at week 1), 6 in sham group (5 patients by request at weeks 6, 11, 18, 30, and 33 and 1 due to death at week 8).

Selective reporting (reporting bias)

Low risk

The study protocol is available and all (primary and secondary) outcomes that are of interest in the study have been reported in the pre‐specified way.

Other bias

Low risk

No other source of bias identified.

Macugen 2011

Methods

Method of allocation: patients were centrally allocated to receive either pegaptanib 0.3 mg or sham injections (1:1) using a dynamic minimization procedure stratified by the site, haemoglobin A1c (7.6% versus 7.6%), systolic BP (140 versus 140 mmHg), diastolic BP (80 versus 80 mmHg), and baseline BCVA (54 versus 54 letters); the dynamic minimisation used a stochastic treatment allocation algorithm based on the variance method.

The study investigator remained masked to treatment and responsible for the patient's care. BCVA was measured at 4 m by the study refractionist/ophthalmologist, who was masked to the patient's treatment and to the patient's previous VA assessments.

Exclusions after randomisation: None reported.
Loss to follow‐up: 48/144 [33.3%] of pegaptanib‐treated and 45/142 [31.7%] in sham‐treated discontinued from the study, with 5/144 (3.5%) of those treated with pegaptanib and 7/142 (4.9%) treated with sham discontinuing owing to adverse vents (safety population).

Participants

Country: USA.
Number randomised: 288 patients.

Age: 62.3 ± 9.3 pegaptanib 0.3 mg group 62.5 ± 10.2 sham group.

Male/female: 81/52 pegaptanib 0.3 mg group 68/59 sham group.

Inclusion criteria: 18 years of age of either gender with type 1 or 2 diabetes and DMO involving the centre of the macula not associated with ischaemia. A foveal thickness of 250 micron (centre point thickness measured on OCT; BCVA with a letter score of 65 through 35 (20/50 –20/200 Snellen equivalents); intraocular pressure (IOP) 21 mmHg; and clear ocular media and adequate pupillary dilation to allow good quality stereoscopic fundus photography were required in the study eye. Additionally, eligible patients could not have undergone yttrium‐aluminum‐garnet laser, peripheral retinal cryoablation, laser retinopexy for retinal tears, or focal or grid photocoagulation within the prior 16 weeks or scatter (panretinal) photocoagulation 6 months before baseline or likely to be needed within 9 months. Only patients for whom focal or grid laser photocoagulation could be deferred in the study eye for 18 weeks in the opinion of the treating ophthalmologist could be entered in the study.

Exclusion criteria: Patients with macular ischaemia were excluded if there was a nonperfusion area of 1 disc area involving the foveal avascular zone (2 quadrants centred around the foveal avascular zone).

Interventions

Treatment: Patients received pegaptanib 0.3 mg or sham injections every 6 weeks in year 1 (total 9 injections) and could receive focal/grid photocoagulation beginning at week 18. During year 2, patients received injections as often as every 6 weeks per prespecified criteria.

Control: sham.

Duration: 12 months (24 months).

Outcomes

The primary efficacy endpoint of the trial was the proportion of patients with a 10‐letter (2‐line) improvement from baseline in VA at week 54.

Secondary efficacy endpoints included the proportion of patients with a 10‐letter improvement from baseline in VA at week 102; changes from baseline in mean VA over time; and, at weeks 54 and 102, the proportion of patients with a 15‐letter (3‐line) improvement in VA, the proportion of patients with a change in degree of retinopathy of 2 steps based on the 12‐step scale of retinopathy, the proportion of patients with a decrease in retinal thickness at the centre point by 25% and 50%, the proportion of patients requiring focal or grid laser, and change in NEIVFQ‐25 and EQ‐5D from baseline.

Notes

No other source of bias identified.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Blinding (performance bias and detection bias)
All outcomes

Low risk

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

At 1 year 116/144 treated patients and 114/142 controls completed the 54 week visit (80%). Adverse events led to discontinuation of 5 treated and 7 control patients.

At 2 years 66 patients in each group completed the 102 week visit.

ITT analysis with LOCF was used leading to the analysis of 133 treated and 127 control patients.

Selective reporting (reporting bias)

Low risk

Other bias

Low risk

READ2

Methods

Randomised study but randomisation sequence generation and methods for its allocation not reported.

Participants

Country: Multicentre USA.

Number:126 participants.

Age: 62 yrs in the 3 groups.

Sex: Female % 69 F (group 1), 55 F (group 2), 52 F (group 3).

Inclusion criteria: Patients (aged >18 years) with type 1 or 2 diabetes and DMO were eligible if they had reduction in visual acuity between 20/40 and 20/320 and met the following criteria: (1) centre subfield thickness measured by OCT ≥ 250 mm, (2) glycosylated haemoglobin ≥ 6% within 12 months before randomisation, (3) no potential contributing causes to reduced VA other than DMO, (4) reasonable expectation that scatter laser photocoagulation would not be required for the next 6 months.

Exclusion criteria: Patients were excluded if they had received focal/grid laser treatment within 3 months, intraocular injection of steroid within 3 months, or intraocular injection of a VEGF antagonist within 2 months. If both eyes were eligible, the eye with the greater centre subfield thickness was entered.

Interventions

Patients were randomised 1:1:1 to receive 0.5 mg ranibizumab at baseline and months 1, 3, and 5 (group 1), focal or grid laser photocoagulation at baseline and month 3 if needed (group 2), or a combination of 0.5 mg ranibizumab and focal or grid laser at baseline and month 3 (group 3). Starting at month 6, if retreatment criteria were met, all patients could be treated with ranibizumab.

Duration: primary outcome at 6 months, extension to 24 months.

Outcomes

The primary outcome measure was the change in BCVA between baseline and month 24.

Secondary vision‐related outcome measures were the change in BCVA between baseline and month 24 and the percentage of patients with 3 or more lines or 2 or more lines improvement at month 24. Secondary anatomic outcomes were the change in foveal thickness between baseline and month 24 and the percentage of patients with elimination of 90% or 50% excess foveal thickness.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear method of sequence generation and information could not be obtained from the authors.

Allocation concealment (selection bias)

Unclear risk

Unclear method of allocation concealment and information could not be obtained from the authors.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear if masked and who was masked and information could not be obtained from the authors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Patients randomised in each group were: 33 ranibizumab, 34 ranibizumab + laser, 34 laser.

At 1 year complete patients were 29, 29, 30 (85‐88%).

At 2 years complete patients were 24, 26, 24 (71‐76%).

Causes of missing were balanced across groups.

Selective reporting (reporting bias)

Unclear risk

We could not find a protocol but primary outcomes were stated in the methods and were those routinely used in the field.

Other bias

Unclear risk

No other source of bias identified

RESOLVE

Methods

Eligible patients were randomised 1:1:1 to either ranibizumab (0.3 mg or 0.5 mg) or sham treatment according to a computer‐generated randomised allocation schedule (kept at a secure site and accessible only to the injecting physician; stratification by centre and by the thickness of MO as assessed by the central reading centre at Visit 1 (< 400 μm versus > 400 μm). Based on the patient strata the injecting physician would take the treatment allocation card and tear‐off the cover and follow instructions to choose vial from the box as indicated (3 boxes, randomisation block size 3). The randomisation data were kept strictly confidential until database lock; not accessible to anyone involved in the study with the exception of injecting physician(s) and drug accountability monitor.

Participants

Country: Multicentre Europe.

Number randomised: 207 screened; 151 patients randomised.

Age: 63 (32‐85) yrs ranibizumab; 65 (41‐82) sham.

Sex: 56 M (54.9%) ranibizumab, 25 M (51%) sham, 46 F (45.1%) ranibizumab, 24 F (49%) sham.

Inclusion criteria: Patients (aged >18 years) with type 1 or 2 diabetes and DMO were eligible if they had a VA between 20/40 and 20/160, CRT ≥ 300 μm, HbA1C < 12%, decreased vision attributed to foveal thickening from DMO, that was not explained by any other cause, and clinically significant DMO in at least one eye confirmed by a central reading centre (Bern Photographic Reading Centre, University Bern, Bern, Switzerland) using stereoscopic fundus photographs, fluorescein angiography, and OCT (Stratus OCT; Carl Zeiss Meditec, Jena, Germany). Eyes were deemed eligible if, in the judgment of the investigator, laser photocoagulation could be safely withheld in the study eye for at least 3 months after random assignment.

Exclusion criteria: Patients were excluded if they had unstable medical status including glycaemic control and blood pressure or panretinal laser photocoagulation performed within 6 months before study entry, and grid/central laser photocoagulation was excluded except for patients with only mild laser burns at least 1,000 μm from the centre of the fovea performed > 6 months preceding day 1.

Interventions

Ranibizumab (0.3 mg, n  51 or 0.5 mg, n  51) or sham treatment (n 49).

Duration: 12 months.

Outcomes

The primary end point was the mean average change in BCVA from baseline to month 1 through month 12 (chosen as the primary end point because it is less sensitive to monthly variations and reflects the treatment impact over the entire treatment period).

Secondary end points included mean change in BCVA and CMT from baseline to month 12, categorised BCVA outcome, and safety.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Computer generated randomised allocation schedule"

Allocation concealment (selection bias)

Low risk

See 'Characteristics of included studies' table.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Masked patients, physician and outcome assessor

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Complete patients at 1 year were 92/102 ranibizumab and 40/49 sham. Causes of missingness were balanced.

ITT analysis with LOCF was used.

Selective reporting (reporting bias)

Unclear risk

We could not find a protocol but primary outcomes were stated in the methods and were those routinely used in the field.

Other bias

Low risk

No other source of bias identified.

RESTORE

Methods

A randomisation list was produced by, or under the responsibility of, Novartis Drug Supply Management using a validated system that automated the random assignment of treatment arms to randomisation numbers in the specified ratio.

Participants

Country: multicentre (Europe, Australia, Canada, Turkey).

Number of patients: 345.

Mean age: 62.9 ±.9.29 yrs ranibizumab 0.5 mg 64.0 ± 8.15 yrs ranibizumab 0.5 mg + laser 63.5 ± 8.81 yrs laser.

Women 43 (37.1) ranibizumab 0.5 mg 48 (40.7) ranibizumab 0.5 mg + laser 53 (47.7) laser

Inclusion criteria: patients 18 years of age with either type 1 or 2 diabetes mellitus (as per American Diabetes Association or World Health Organization guidelines), glycosylated haemoglobin (HbA1c) 10%, and visual impairment due to DMO. The key inclusion criteria were (1) stable medication for the management of diabetes within 3 months before randomisation and expected to remain stable during the study; (2) visual impairment due to focal or diffuse DMO (definition in Table 1) in at least 1 eye that was eligible for laser treatment in the opinion of the investigator; (3) BCVA letter score between 78 and 39, both inclusive, based on ETDRS‐like VA testing charts administered at a starting distance of 4 metres (approximate Snellen equivalent 20/32–20/160); and (4) decreased vision due to DMO and not other causes, in the investigator’s opinion (at visit 1).

Exclusion criteria: concomitant conditions in the study eye that could prevent the improvement in VA on the study treatment in the investigator’s opinion; (2) active intraocular inflammation or infection in either eye; (3) uncontrolled glaucoma in either eye (e.g., IOP 24 mmHg on medication, or from the investigator’s judgment); (4) panretinal laser photocoagulation (within 6 months) or focal/grid laser photocoagulation (within 3 months) before study entry; (5) treatment with antiangiogenic drugs in the study eye within 3 months before randomisation; (6) history of stroke; and (7) systolic BP 160 mmHg or diastolic BP 100 mmHg, untreated hypertension, or change in antihypertensive treatment within 3 months preceding baseline.

Interventions

3 treatment arms: Intravitreal ranibizumab (0.5 mg) injection sham laser, adjunctive administration of intravitreal ranibizumab (0.5 mg) injection active laser, or laser treatment sham injections for 12 months.

Outcomes

The primary objective of this study was to demonstrate superiority of ranibizumab 0.5 mg as monotherapy or combined with laser therapy over laser alone (the current standard of care) with respect to mean average change in BCVA from baseline over 12 months.

Secondary objectives were to evaluate (1) if ranibizumab 0.5 mg as monotherapy or adjunctive to laser was superior to laser alone in the proportion of patients with VA improvement and with BCVA letter score 73 (20/40 Snellen equivalent) at month 12; (2) the time course of mean change in BCVA letter score and central retinal (subfield) thickness (CRT); (3) patient‐reported outcomes relative to those associated with laser treatment; and (4) the safety of intravitreal injections of ranibizumab 0.5 mg, as monotherapy or adjunctive to laser therapy relative to laser treatment.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A randomisation list was produced by, or under the responsibility of, Novartis Drug Supply Management using a validated system that automated the random assignment of treatment arms to randomisation numbers in the specified ratio.

Allocation concealment (selection bias)

Low risk

Central randomisation using an eCRF after each patient was included by study investigators.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Masked patients, physician and outcome assessor.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Patients randomised in each group were: 116 ranibizumab, 118 ranibizumab + laser, 111 laser.

At 1 year complete patients were 87.9%, 87.3% and 88.3% respectively.

There were 2 deaths in each of the 3 treatment arms.

ITT analysis with LOCF was used.

Selective reporting (reporting bias)

Low risk

We could not find a protocol but primary outcomes were stated in the methods and were those routinely used in the field.

Other bias

Unclear risk

No other source of bias identified.

RISE‐RIDE

Methods

Multicentre study in USA and South America.

Quote: "Randomization was stratified by study eye BCVA (55 versus 55 ETDRS letters), baseline HbA1c (8% versus 8%), prior DME therapy in the study eye (yes versus no), and study site. Dynamic randomization was used to obtain approximately a 1:1:1 ratio among groups. Randomization was done via interactive phone system. The sponsor developed the specifications for the randomization, and a third party programmed and held the randomization algorithm. "

Quote: "Ocular assessments, including the need for macular laser, were made by evaluating ophthalmologists masked to patients’ treatment assignments. Study treatments were administered by treating ophthalmologists unmasked to treatment assignments but masked to ranibizumab dose. To improve patient masking, all patients received subconjunctival anesthesia before sham or active injections. Study site personnel (except treating physicians and assistants), central reading centre personnel, and the sponsor and its agents (except drug accountability monitors) were masked to treatment assignment. Treating physicians were masked to the assigned dose of ranibizumab. An independent statistical coordinating center performed the unmasked interim analyses for the data monitoring committee."

Participants

759 patients were enrolled and randomised to study treatment (377 in RISE and 382 in RIDE; sham 257, ranibizumab 0.3 mg 250, ranibizumab 0.5 mg 250.

Interventions

Quote: "The median number of ranibizumab injections was 24. The mean number of macular laser treatments over 24 months was 1.8 and 1.6 in the sham groups and 0.3 to 0.8 in the ranibizumab groups. Substantially more sham‐treated patients received macular laser under the protocol‐specified criteria or underwent panretinal photocoagulation for proliferative diabetic retinopathy."

Outcomes

Quote: "The primary efficacy measure was the proportion of patients gaining 15 ETDRS letters in BCVA score from baseline at 24 months (corresponding to 3 lines on the eye chart). Secondary outcomes at 24 months were mean change from baseline BCVA score over time, proportion of patients with BCVA Snellen equivalent of 20/40, mean change from baseline BCVA score over time in patients with focal edema as assessed on FA, proportion of patients losing 15 letters in BCVA score from baseline, mean change from baseline in OCT CFT over time, proportion of patients with a 3‐step progression from baseline in ETDRS retinopathy severity on FP, proportion of patients with resolution of
leakage on FA, and the mean number of macular laser treatments over time."

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

See 'Characteristics of included studies' table

Allocation concealment (selection bias)

Low risk

See 'Characteristics of included studies' table

Blinding (performance bias and detection bias)
All outcomes

Low risk

Patients and outcome assessors masked, unmasked treating physician.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The 2‐year study period was completed by 83.3% of patients in RISE and by 84.6% in RIDE. Causes of missingness not reported.

Selective reporting (reporting bias)

High risk

All visual acuity cut‐offs and secondary outcomes available at 2 years, but not at 1 year.

Other bias

Low risk

No other bias identified.

Soheilian 2007

Methods

Randomisation was performed using random block permutation method according to a computer‐generated randomisation list. The block length varied randomly. Random allocation sequence was performed by a biostatistician. The detail of series was unknown by the study investigators.

Masking of participant: A sham laser procedure (20 seconds) was performed by aiming the laser beam on the macula for the eyes in the IVB and IVB/IVT groups. In the MPC group, a sham injection was done by a needleless syringe pressed against the conjunctiva. To keep the masking process, patients were prevented from seeing the syringes.

Outcome assessor: Yes; best‐corrected VA measurement and OCT were performed by certified examiners masked both to the randomisation and to the findings of previous measurements.

Provider: Yes; all procedures were run by staff members other than the study investigators to preserve investigator masking.

Exclusions after randomisation: None.

Losses to follow‐up:

1) at 9 months, 25 participants, 6 in the IVB group (4 high‐risk retinopathy, 2 lost); 12 patients IVB/IVTA (3 high risk retinopathy, 4 cataract, 1 neovascular glaucoma; 2 deceased providing 3 eyes, 1 lost); 7 patients in photocoagulation group (3 high risk retinopathy, 1 cataract, 2 deaths, 1 lost).

2) at 24 months, in a subsequent publication in 2012, the authors reported 39 (78%), 36 (72%) and 38 (76%) eyes in the three arms; 8 patients (12 eyes) missing were dead for causes unrelated to treatment, but other causes of death were not reported.

Participants

Country: Iran

Number randomised: 129 participants, 150 eyes.

Age: 60.5 ± 5.9: IVB group, 62.3 ± 6.8 IVB/IVTA, 61.0 ± 5.3 photocoagulation group.

Sex: F/M 27/23 IVB, 22/28 IVB/IVTA, 22/28 macular laser.

Inclusion criteria: Eligible cases were 150 eyes of 129 patients with clinically significant DMO based on ETDRS criteria. 

Exclusion criteria: Previous panretinal or focal laser photocoagulation, prior intraocular surgery or injection, history of glaucoma or ocular hypertension, VA of 20/40 or better or worse than 20/300, presence of iris neovascularisation, high‐risk proliferative diabetic retinopathy, and significant media opacity. Monocularity, pregnancy, serum creatinine 3 mg/dL, and uncontrolled diabetes mellitus were also among the exclusion criteria.

Interventions

Treatment: IVB group, patients who received 1.25 mg IVB (50 eyes); the IVB/IVT group, patients who received 1.25 mg of IVB and 2 mg of IVT (50 eyes); and the photocoagulation group, patients who underwent focal or modified grid laser (50 eyes). 

Control: The photocoagulation group, patients who underwent focal or modified grid laser (50 eyes).

Retreatment was performed at 12‐week intervals whenever indicated.

Duration: 36 weeks.

Outcomes

Primary outcome measure was change in best‐corrected VA (logMAR) at week 24 (data available at 36 weeks). 

Secondary outcomes were VA change, central macular thickness change by optical coherence tomography and potential injection‐related complications.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated sequence.

Allocation concealment (selection bias)

Unclear risk

Investigatior concealment mentioned but not described.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Masked participants, investigator and outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were 6, 7 and 12 missing patients out of 50 at 36 weeks in the IVB, IVB/IVTA and photocoagulation groups and causes where not clearly unrelated to visual acuity outcome, except for 2 deaths. In a subsequent publication in 2012 the authors reported 39 (78%), 36 (72%) and 38 (76%) eyes in the three arms; 8 patients (12 eyes) missing were dead for causes unrelated to treatment, but other causes of death were not reported.

Selective reporting (reporting bias)

Low risk

The primary outcomes are continuous measures and no arbitrary cut‐point were used.

Other bias

High risk

There was an imbalance of baseline visual acuity in the three group (IVB: 0.71 logMAR, IVB/IVTA 0.73 logMAR, photocoagulation 0.55 logMAR).

Although there was a potential unit of analysis issue (150 eyes of 129 patients, 16% of patients with both eyes included), comparisons were made in a marginal regression model (based on generalised estimating equation methods) adjusted for the baseline values and to eliminate any possible correlation effects between the 2 eyes of patients in bilateral enrolled cases. However, we could not take correlation into account when analysing dichotomous visual acuity definitions.

BCVA: best‐corrected visual acuity
BP: blood pressure
CMT: central macular thickness
CRT: central retinal thicknessCSMO: clinically significant macular oedema
CTU:
DMO: diabetic macular oedema (DME:US spelling edema)
ECG: electrocardiogram
ETDRS: Early Treatment Diabetic Retinopathy Study
FAZ: foveal avascular zone
GLD: greatest linear dimension
IOP: intraocular pressure
ITT: intention‐to‐treat
iv: intravenous
IVB: intravitreal bevacizumab
IVT: intravitreal triamcinolone
LOCF: last observation carried forward
OCT: optical coherence tomography
PDR: proliferative diabetic retinopathy
PFCL: perifoveal capillary loss
VA: visual acuity
VEGF: vascular endothelial growth factor

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

DRCRnet 2007

Follow‐up at 12 weeks only.

DRCRnet 2011

Follow‐up at 14 weeks only. RCT comparing ranibizumab (2 injections), triamcinolone (1 injection) to sham in patients with DMO undergoing grid and panretinal laser photocoagulation.

DRCRnet 2012

Follow‐up of DRCRnet comparing prompt to deferred laser in patients treated for ranibizumab for DMO: does not report on comparison of ranibizumab with laser.

Faghihi 2008

Follow‐up at 16 weeks only.

Lim 2012

Bevacizumab compared to intravitreal triamcinolone.

Paccola 2008

Single injection of intravitreal triamcinolone acetonide (4 mg/0.1 mL) compared to single injection of intravitreal bevacizumab (1.5 mg/0.06 mL). Duration: 24 weeks.

Solaiman 2010

Single intravitreal injection of bevacizumab (inadequate dose); follow‐up 6 months.

DMO: diabetic macular oedema
RCT: randomised controlled trial

Characteristics of ongoing studies [ordered by study ID]

NCT00387582

Trial name or title

Lucentis in the Treatment of Macular Edema ‐ A Phase II, Single Center, Randomized Study to Evaluate the Efficacy of Ranibizumab Versus Focal Laser Treatment in Subjects With Diabetic Macular Edema

Methods

Allocation: Randomised
Endpoint Classification: Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label
Primary Purpose: Treatment

Participants

49, country: USA

Interventions

Experimental: I
Lucentis injections for the first three months of the study and then per the protocol for the duration of the trial.

Active Comparator: II
Argon Laser treatment at enrolment and then per the protocol for the duration of the study.

Outcomes

Primary Outcome Measures [Time Frame: 6 and 12 months]:
Prevention of vision loss at one year as evidenced by ETDRS visual acuity.

Secondary Outcome Measures:
Reduction in retinal thickening based on OCT.

Starting date

Study Start Date:July 2006
Study Completion Date:February 2009

Contact information

Roy A. Goodart, M.D., Principal Investigator, Rocky Mountain Retina Consultants

Notes

NCT00901186

Trial name or title

A Randomized, Open Label, Multicenter, Laser‐controlled Phase II Study Assessing the Efficacy and Safety of Ranibizumab (Intravitreal Injections) vs. Laser Treatment in Patients With Visual Impairment Due to Diabetic Macular Edema

Methods

Allocation: Randomised
Endpoint Classification: Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label
Primary Purpose: Treatment

Participants

84, country: Spain

Interventions

Drug: Ranibizumab
Procedure: Laser

Outcomes

Primary Outcome Measures:
Change BCVA with ranibizumab 0.5 mg versus laser 12‐month.

Secondary Outcome Measures:
Improvement in BCVA with ranibizumab (0.5 mg) versus laser 12‐month measure
Mean BCVA change with ranibizumab (0.5 mg) versus laser
% of patients with VA > 73 letters with ranibizumab (0.5 mg) versus laser
Time and mean change in central retinal thickness by OCT with ranibizumab (0.5 mg) versus laser
Monitoring and registry of all adverse events, serious adverse events, VA, concomitant medications, ophthalmologic exams (including count of fingers and movement of the hands), IOP, vital constants and analytical parameters.

Starting date

Study First Received:May 11, 2009
Last Updated:November 16, 2011

Contact information

Novartis (Novartis Pharmaceuticals)

Notes

Sponsor: Novartis (Novartis Pharmaceuticals)

NCT00989989

Trial name or title

Efficacy and Safety of Ranibizumab (Intravitreal Injections) in Patients With Visual Impairment Due to Diabetic Macular Edema (REVEAL)

Methods

Allocation: Randomised
Intervention Model: Parallel Assignment
Masking: Double Masked (Patient, Investigator)
Primary Purpose: Treatment

Participants

395, country: China, Hong Kong, Korea, Japan, Singapore, Taiwan

Interventions

Experimental: Group 1‐ adjunctive group Drug: ranibizumab Procedure: laser photocoagulation
Experimental: Group 2‐ monotherapy group Drug: ranibizumab
Active Comparator: Group 3‐ laser control group Procedure: laser photocoagulation

Outcomes

Primary Outcome Measures:
To demonstrate superiority of ranibizumab 0.5 mg as adjunctive or mono‐therapy to laser treatment in the mean change from baseline in BCVA over a 12‐month treatment period.

Secondary Outcome Measures:
To evaluate the effects of ranibizumab (0.5 mg) adjunctive and monotherapy on CRT and other anatomical changes relative to laser treatment
To evaluate whether ranibizumab (0.5 mg) as adjunctive or mono‐therapy is superior to laser treatment in the number of patients with VA above 73 letters and in the number of patients with improvement in BCVA
To evaluate the time course of BCVA changes on ranibizumab (0.5 mg) adjunctive and monotherapy relative to laser treatment
To evaluate the effects of ranibizumab (0.5 mg) adjunctive and mono‐therapy on overall health status through patient‐reported outcomes (PROs) relative to laser treatment
To evaluate the safety of intravitreal injections of ranibizumab (0.5 mg) as adjunctive and monotherapy in patients with DMO overall and relative to laser treatment

Starting date

Study Start Date:September 2009
Study Completion Date:August 2011

Contact information

Novartis

Notes

NCT00997191

Trial name or title

Intravitreal Bevacizumab and Intravitreal Triamcinolone Associated to Laser Photocoagulation for Diabetic Macular Edema (IBeTA)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label
Primary Purpose: Treatment

Participants

12, country: Brasil

Interventions

Procedure: Laser photocoagulation
Drug: Intravitreal triamcinolone
Drug: Intravitreal bevacizumab

Outcomes

Primary Outcome Measures [Time Frame: One Year]:
BCVA

Secondary Outcome Measures:
Macular Mapping Test
Multifocal Electroretinogram
CMT

Starting date

Study Start Date:October 2009
Estimated Study Completion Date:October 2011

Contact information

Bianka Yukari Nakase Yamasato Katayama, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo

Notes

NCT01077401

Trial name or title

A Phase 3, Randomized, Controlled, Double‐Masked, Multi‐Center, Comparative, In Parallel Groups (For 24 Weeks), To Compare The Efficacy And Safety Of 0.3 MG Pegaptanib Sodium, With Sham Injections, And Open Study (For 30 Weeks) To Confirm The Safety Of 0.3 MG Pegaptanib Sodium In Subjects With Diabetic Macular Edema (DME)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Double Masked

Participants

NA

Interventions

Drug: pegaptanib sodium
Other: sham injection

Outcomes

NA

Starting date

Contact information

Notes

NCT01100307

Trial name or title

Ranibizumab for Edema of the Macula in Diabetes: Protocol 3 With High Dose ‐ the READ 3 Study

Methods

Allocation: Randomised Endpoint Classification: Safety/Efficacy StudyIntervention Model: Parallel AssignmentMasking: Double Masked

Participants

92, country: USA

Interventions

Drug: pegaptanib sodium
Other: sham injection

Outcomes

Primary Outcome Measures [Time Frame: 3,6, 9 and 12 months]:

Adverse events
The primary outcome measures for safety and tolerability are the following:
The incidence and severity of systemic and ocular adverse events that are associated with repeated intravitreal injections of two doses of RBZ in patients with DMO such as cardiovascular events, intraocular reactions (inflammation), vitreous haemorrhage, retinal detachment, endophthalmitis (intraocular infection),increased IOP, and cataract formation, among others.

Starting date

Study Start Date:February 2010
Estimated Study Completion Date:March 2013

Contact information

Dr. Diana V. Do, Johns Hopkins University

Notes

Sponsor: Johns Hopkins University

NCT01112085

Trial name or title

MIcrodoses of raNIbizumab in Diabetic MAcular Edema (MINIMA‐2)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Single Masked (Patient)

Participants

Estimated Enrollment:72, country: Mexico

Interventions

Experimental: Ranibizumab 0.05 mg. Intravitreal injections of 0.05 mg ranibizumab over 6 months then additional treatment with ranibizumab 0.05 mg as needed (according to re‐treatment criteria)

Experimental: Ranibizumab 0.5 mg. Intravitreal injections of 0.5 mg ranibizumab over 6 months then additional treatment with ranibizumab 0.5 mg as needed (according to re‐treatment criteria)

Outcomes

Primary Outcome Measures [Time Frame: 6 months and 12 months]:
BCVA
Improvement in vision of BCVa of 15 or more letters, or a final vision of 20/25 (50 letters) or better if BCVA was 20/40 (40 letters)

Secondary Outcome Measures [Time Frame: 6 months and 12 months]:
Mean change in CRT and volume by OCT
Changes in CRT and volume assessed by OCT

Starting date

Study Start Date:April 2010
Estimated Study Completion Date:December 2011
Estimated Primary Completion Date:September 2011 (Final data collection date for primary outcome measure)

Contact information

Fundación Mexicana de Retina

Notes

Sponsor: Especialistas en Retina Medica y Quirurgica Grupo de Investigacion

NCT01131585

Trial name or title

Safety and Efficacy of Ranibizumab in Diabetic Macular Edema (RELATION)

Methods

Allocation: Randomised
Intervention Model: Parallel Assignment
Masking: Double Masked (Patient, Investigator)

Participants

128, country: Germany

Interventions

Experimental: Active laser photocoagulation and ranibizumab
Active laser treatment applied at baseline and reapplied if needed at intervals no shorter than 3 months from the last treatment.
Ranibizumab intravitreal injection given at baseline, at 30, 60 and 90 days and if needed, reapplied at intervals no shorter than 28 days from last treatment.

Active Comparator: Active laser photocoagulation and sham injection
Active laser treatment applied at baseline and reapplied if needed at intervals no shorter than 3 months from the last treatment.
Sham intravitreal injection given at baseline, at 30, 60 and 90 days and if needed, reapplied at intervals no shorter than 28 days from last treatment.

Outcomes

Primary Outcome Measures:
Change in BCVA from baseline to month 12
Mean change in BCVA letters at 12 months compared to baseline was measured using VA. VA accounts for the number of letters a participant can see using EDTRS‐like VA testing charts, from a sitting position at a testing distance of 4 metres. BCVA means that the participant's refraction is already taken into account when VA is determined. A higher BCVA number at 12 months in reference to baseline indicates improved BCVA.

Starting date

Study Start Date: June 2010
Study Completion Date: July 2011

Contact information

Novartis (Novartis Pharmaceuticals)

Notes

Sponsor: Novartis Pharmaceuticals

NCT01171976

Trial name or title

Efficacy and Safety of Ranibizumab in Two "Treat and Extend" Treatment Algorithms Versus Ranibizumab As Needed in Patients With Macular Edema and Visual Impairment Secondary to Diabetes Mellitus (RETAIN)

Methods

Allocation: Randomised
Intervention Model: Parallel Assignment
Masking: Single Masked (Investigator)

Participants

374, 52 centres in Europe

Interventions

Experimental: 0.5 mg ranibizumab "Treat and Extend" + laser
Experimental: 0.5 mg ranibizumab "Treat and Extend" alone
Active Comparator: 0.5 mg ranibizumab alone given PRN

Outcomes

Primary Outcome Measures:
Mean average change from baseline inBCVA over a 12‐month treatment period.

Secondary Outcome Measures:
Evaluate whether the mean average change from baseline in BCVA obtained with either a 0.5 mg ranibizumab TE with adjunctive laser, or with 0.5 mg ranibizumab TE is non‐inferior to 0.5 mg ranibizumab PRN
Investigate within the TE dosing concepts the impact of laser treatment on the number of retreatments.
Investigate the efficacy of 0.5 mg ranibizumab TE with adjunctive laser, 0.5 mg ranibizumab TE and 0.5 mg ranibizumab PRN measured by the overall score assessed by VFQ‐25 and EQ‐5D.
Time course of mean BCVA change from baseline to month 12, and up to month 24 obtained with either a 0.5 mg ranibizumab TE with adjunctive laser, or with 0.5 mg ranibizumab TE and with 0.5 mg ranibizumab PRN.
To compare the changes in development of CSFT of 0.5 mg ranibizumab TE with adjunctive laser, 0.5 mg ranibizumab TE and 0.5 mg ranibizumab PRN over time.

Starting date

Study Start Date:September 2010
Estimated Study Completion Date:May 2013

Contact information

Novartis Pharmaceuticals

Notes

Sponsor: Novartis Pharmaceuticals

NCT01223612

Trial name or title

Lucentis (Ranibizumab) in Diabetic Macular Oedema: a Treatment Evaluation (LUCIDATE)

Methods

Study Start Date:October 2010
Estimated Study Completion Date:July 2012

Participants

36, country: UK

Interventions

Drug: Ranibizumab
Intravitreal injection of 0.5 mg in 0.05 mL. One injection at baseline, 4 and 8 weeks then four‐weekly as required to 44 weeks.

Procedure: Modified ETDRS laser
Argon laser therapy to the macula in accordance with the modified ETDRS protocol at baseline, 12, 24 and 36 weeks.

Outcomes

Patients will undergo detailed baseline evaluation which will include:
vision testing
optical coherence tomography scanning
fundus fluorescein angiography
microperimetry
colour contrast sensitivity testing
electrophysiological testing

Starting date

Study Start Date:October 2010
Estimated Study Completion Date:July 2012

Contact information

Mrs Sue Lydeard, Moorfields Eye Hospital NHS Foundation Trust

Notes

Sponsor: Moorfields Eye Hospital NHS Foundation Trust

NCT01445899

Trial name or title

An Open‐Label Dose Escalation Study of PF‐04523655 (Stratum I) Combined With a Prospective, Randomized, Double‐Masked, Multi‐Center, Controlled Study (Stratum II) Evaluating the Efficacy and Safety of PF‐04523655 Alone and in Combination With Ranibizumab Versus Ranibizumab Alone in Diabetic Macular Edema (MATISSE STUDY)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Double Masked (Subject, Caregiver, Investigator)

Participants

264, countries:USA, Israel

Interventions

Drug: PF‐04523655 (Stratum I)
Drug: PF‐04523655 and ranibizumab
Drug: ranibizumab
Drug: PF‐04523655 (Stratum II)

Outcomes

Primary Outcome Measures:
Safety and dose‐limiting toxicities (Stratum I)

‐ To determine the safety and dose‐limiting toxicities of a single intravitreal (IVT) injection of PF‐04523655 in people with low vision

Pharmacokinetics (Stratum I)
‐ To determine the pharmacokinetics (PK) of a single IVT injection of PF‐04523655 in people with low vision

Safety and tolerability (Stratum II)
‐ To evaluate the safety and tolerability of PF‐04523655 alone and in combination with ranibizumab in patients with DMO

Efficacy (Stratum II)
‐ To evaluate the ability of PF‐04523655 alone and in combination with ranibizumab to improve visual acuity compared to ranibizumab alone in people with DMO

Starting date

Study Start Date:February 2012
Estimated Study Completion Date:July 2014

Contact information

Quark Pharmaceuticals

Notes

Sponsor: Quark Pharmaceuticals

NCT01476449

Trial name or title

Monthly Ranibizumab Versus Treat and Extend Ranibizumab for Diabetic Macular Edema

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label

Participants

20, country:USA

Interventions

Active Comparator: Monthly ranibizumab
Patients randomised to the monthly ranibizumab arm of the study will be administered intravitreal injections each month for their diabetic MO for the duration of the study.

Experimental: Treat and Extend Ranibizumab
Patients randomised to this arm of the study will receive intravitreal injections of ranibizumab until their maculae are anatomically "dry," at which point the evaluation and injection interval will be extended.

Outcomes

NA

Starting date

Study Start Date:November 2011
Estimated Study Completion Date:June 2013

Contact information

Retina Vitreous Associates of Florida

Notes

NCT01487629

Trial name or title

Bevacizumab Versus Ranibizumab for the Treatment of Diabetic Macular Edema (IBERA‐DME)

Methods

Allocation: Randomised
Endpoint Classification: Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label

Participants

53, country: Brasil

Interventions

Drug: Bevacizumab
Bevacizumab, 1.5 mg, intravitreal, throughout the study

Drug: Ranibizumab
Ranibizumab, 0.5 mg, intravitreal, throughout the study

Outcomes

Primary Outcome Measures:
CSFT change
CSFT measured with spectral‐domain OCT

Secondary Outcome Measures:
BCVA change
BCVA using ETDRS charts

Starting date

Study Start Date:April 2010
Estimated Study Completion Date:September 2012

Contact information

Rodrigo Jorge, Principal investigator, University of Sao Paulo

Notes

Sponsor: University of Sao Paulo

NCT01552408

Trial name or title

A Phase I/II, Randomized, Study for Diabetic Macular Edema Using 0.5mg Ranibizumab Combined With Targeted PRP Monthly for 4 Months,Then PRN vs. 0.5mg Ranibizumab 4 Months Monotherapy, Then as Needed(DME‐AntiVEgf) DAVE

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Single Group Assignment
Masking: Open Label

Participants

40. country: USA

Interventions

Active Comparator: 0.50mg ranibizumab
4 mandatory monthly injections of 0.50mg ranibizumab, retreatment will be as needed

Experimental: Targeted PRP with 0.50mg ranibizumab
4 mandatory monthly injections of 0.50mg ranibizumab, and at V3 (day7) will receive Targeted PRP, then treatment with ranibizumab will be PRN

Outcomes

NA

Starting date

Study Start Date:March 2012
Estimated Study Completion Date:March 2013

Contact information

David M. Brown, M.D., Director Greater Houston Retina Research, Greater Houston Retina Research

Notes

Sponsor: David M. Brown, M.D.
Collaborator: Genentech

NCT01565148

Trial name or title

A Randomized, Multi‐center, Phase II Study of the Safety, Tolerability and Bioactivity of Repeated Intravitreal Injections of iCo‐007 as Monotherapy or in Combination With Ranibizumab or Laser Photocoagulation in the Treatment of Diabetic Macular Edema (the iDEAL Study)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Factorial Assignment
Masking: Open Label

Participants

208, country: USA

Interventions

Experimental: Group 1 Drug: iCo‐007 350 mcg
iCo‐007 (350 μg) as an intravitreal injection at baseline followed by another iCo‐007 dose (350 μg) at month 4

Experimental: Group 2 Drug: iCo‐007 700 mcg
iCo‐007 (700 μg) as an intravitreal injection at baseline followed by another iCo‐007 dose (700 μg) at month 4

Experimental: Group 3 Drug: iCo‐007 350 mcg plus laser
iCo‐007 (350 μg) as an intravitreal injection at baseline followed 7 days later by laser photocoagulation. At M4, intravitreal injection of iCo‐007 (350 μg) will be given as mandatory treatment. If the eye also meets retreatment criteria, it will also receive the second laser photocoagulation

Experimental: Group 4 Drug: ranibizumab Plus iCo‐007 350 mcg
Ranibizumab (0.5 mg) intravitreal injection at baseline followed by iCo‐007 (350 μg) intravitreal injection 2 weeks later; re‐treatment with ranibizumab (0.5 mg) mandatory at M4 followed by iCo‐007 (350 μg) 2 weeks later

Outcomes

Primary Outcome Measures:
Change in VA from baseline to month 8

Secondary Outcome Measures:
Number of participants in a given study arm experiencing the same drug‐related serious adverse event as a measure of safety and tolerability
Safety of repeated iCo‐007 intravitreal injections in treatment of people with DMO as monotherapy and in combination with ranibizumab or laser photocoagulation. Serious consideration will be given if 2 or more patients in a particular treatment arm experience the same drug‐related serious adverse event;

Change in VA from baseline to month 12
Change in retinal thickness measured by OCT from baseline to month 8
Change in retinal thickness measured by OCT from baseline to month 12
Duration of iCo‐007 treatment effect during the 12 month follow‐up period as measured by VA and OCt thickness
Peak plasma concentration (Cmax) of iCo‐007 after multiple injections

Starting date

Study Start Date:February 2012
Estimated Study Completion Date:December 2013

Contact information

Quan Dong Nguyen, MD, Johns Hopkins University

Notes

Sponsors and Collaborators
Quan Dong Nguyen
Juvenile Diabetes Research Foundation
iCo Therapeutics Inc.

NCT01572350

Trial name or title

Safety and Efficacy of Triamcinolone Acetonide Combined With Laser, Bevacizumab Combined With Laser Versus Laser Alone for the Treatment of Diffuse Non‐tractional Diabetic Macular Edema (ALBA)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Open Label

Participants

105, country: Spain

Interventions

Other: Grid laser
Drug: Triamcinolone Acetonide
Drug: Bevacizumab

Outcomes

Primary Outcome Measures:
Best‐Corrected Visual AcuiBCVA

Secondary Outcome Measures:
To assess the safety of intravitreal Triesence (r)
To measure average change in mean CMT in each group
To measure average change in mean CMT (in microns) obtained by OCT at each follow‐up visits compared to the baseline visit in each of the three groups.

To assess the safety of intravitreal Avastin (r)
Type of adverse events, severity and number of Participants with Adverse Events as a Measure of Safety and Tolerability
To assess the safety of intravitreal grid photocoagulation
Type of adverse events, severity and number of Participants with Adverse Events as a Measure of Safety and Tolerability

Starting date

Study Start Date:October 2010
Estimated Primary Completion Date:October 2012 (Final data collection date for primary outcome measure)

Contact information

Alicia Pareja, MD, Hospital Universitario de Canarias

Notes

Sponsor: Hospital Universitario de Canarias

NCT01610557

Trial name or title

A Phase II Randomized Study to Compare Anti‐VEGF Agents in the Treatment of Diabetic Macular Edema (CADME)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Cross‐over assignment
Masking: Double‐Masked

Participants

60, country: USA

Interventions

Drug: Ranibizumab and Bevacizumab
Eyes are randomly assigned to receive a set sequence of monthly eye injections; all eyes receive ranibizumab at some time points and bevacizumab at others during the cross‐over study.
Drug: Bevacizumab

Outcomes

Primary Outcome Measures:
The primary outcome measure is the mean change in BCVA.

Secondary Outcome Measures:
Retinal thickness on Optical Coherence Tomography

Starting date

Study Start Date:May 2012
Estimated Study Completion Date:August 2014

Contact information

Henry E. Wiley IV, M.D./National Eye Institute, National Institutes of Health

Notes

Sponsor: National Eye Institute (NEI)

NCT01627249

Trial name or title

Comparative Effectiveness Study of Intravitreal Aflibercept, Bevacizumab, and Ranibizumab for DME (Protocol T)

Methods

Allocation: Randomised
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Single Masked (Patient)

Participants

660

Interventions

Drug: 0.5 mg intravitreal ranibizumab
Intravitreal injection of 0.5 mg ranibizumab (Lucentis™) at baseline and up to every 4 weeks using defined retreatment criteria.

Experimental: Aflibercept Drug: 2.0 mg intravitreal aflibercept
Intravitreal injection of 2.0 mg aflibercept at baseline and up to every 4 weeks using defined retreatment criteria.

Experimental: Bevacizumab Drug: 1.25 mg intravitreal bevacizumab
Intravitreal injection of 1.25 mg bevacizumab at baseline and up to every 4 weeks using defined retreatment criteria.

Outcomes

Allocation: Randomized
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Single Masked (Patient)

Starting date

Study Start Date: August 2012
Estimated Study Completion Date: January 2016

Contact information

Diabetic Retinopathy Clinical Research Network

Notes

Sponsor:
Diabetic Retinopathy Clinical Research Network

BCVA: best‐corrected visual acuity
CMT: central macular thickness
CRT: central retinal thicknessCSMO: clinically significant macular oedema
DMO: diabetic macular oedema (DME:US spelling edema)
ETDRS: Early Treatment Diabetic Retinopathy Study
IOP: intraocular pressure
OCT: optical coherence tomography
VA: visual acuity

Data and analyses

Open in table viewer
Comparison 1. Anti‐VEGF versus laser

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 1 year Show forest plot

5

556

Risk Ratio (M‐H, Fixed, 95% CI)

2.76 [2.02, 3.76]

Analysis 1.1

Comparison 1 Anti‐VEGF versus laser, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

1.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

3.06 [1.54, 6.05]

1.2 Ranibizumab

2

300

Risk Ratio (M‐H, Fixed, 95% CI)

3.06 [1.93, 4.87]

1.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.26, 3.51]

2 Gain 3+ lines of visual acuity at 1 year Show forest plot

5

556

Risk Ratio (M‐H, Fixed, 95% CI)

3.20 [2.07, 4.95]

Analysis 1.2

Comparison 1 Anti‐VEGF versus laser, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

2.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

2.52 [1.20, 5.29]

2.2 Ranibizumab

2

300

Risk Ratio (M‐H, Fixed, 95% CI)

3.51 [1.78, 6.92]

2.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

3.72 [1.52, 9.08]

3 Loss 2+ lines of visual acuity at 1 year Show forest plot

4

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.23 [0.12, 0.44]

Analysis 1.3

Comparison 1 Anti‐VEGF versus laser, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

3.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.07, 0.52]

3.2 Ranibizumab

1

225

Risk Ratio (M‐H, Fixed, 95% CI)

0.27 [0.09, 0.80]

3.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.05, 1.09]

4 Loss 3+ lines of visual acuity at 1 year Show forest plot

4

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.13 [0.05, 0.34]

Analysis 1.4

Comparison 1 Anti‐VEGF versus laser, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

4.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.05, 0.51]

4.2 Ranibizumab

1

225

Risk Ratio (M‐H, Fixed, 95% CI)

0.11 [0.01, 0.83]

4.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

0.08 [0.00, 1.30]

5 Mean difference in logMAR visual acuity at 1 year Show forest plot

5

554

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.16, ‐0.10]

Analysis 1.5

Comparison 1 Anti‐VEGF versus laser, Outcome 5 Mean difference in logMAR visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 5 Mean difference in logMAR visual acuity at 1 year.

5.1 Bevacizumab

2

165

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.28, ‐0.12]

5.2 Ranibizumab

2

300

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.14, ‐0.07]

5.3 Aflibercept

1

89

Mean Difference (IV, Fixed, 95% CI)

‐0.27 [‐0.40, ‐0.13]

6 Mean difference in (change of) OCT central macular thickness at 1 year Show forest plot

4

477

Mean Difference (IV, Fixed, 95% CI)

‐60.71 [‐83.87, ‐37.54]

Analysis 1.6

Comparison 1 Anti‐VEGF versus laser, Outcome 6 Mean difference in (change of) OCT central macular thickness at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 6 Mean difference in (change of) OCT central macular thickness at 1 year.

6.1 Bevacizumab

2

165

Mean Difference (IV, Fixed, 95% CI)

‐43.61 [‐82.11, ‐5.11]

6.2 Ranibizumab

1

225

Mean Difference (IV, Fixed, 95% CI)

‐57.40 [‐89.86, ‐24.94]

6.3 Aflibercept

1

87

Mean Difference (IV, Fixed, 95% CI)

‐121.9 [‐186.47, ‐57.33]

7 Mean difference in logMAR visual acuity at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.7

Comparison 1 Anti‐VEGF versus laser, Outcome 7 Mean difference in logMAR visual acuity at 2 years.

Comparison 1 Anti‐VEGF versus laser, Outcome 7 Mean difference in logMAR visual acuity at 2 years.

7.1 Bevacizumab

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Mean difference in (change of) OCT Central Macular Thickness at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.8

Comparison 1 Anti‐VEGF versus laser, Outcome 8 Mean difference in (change of) OCT Central Macular Thickness at 2 years.

Comparison 1 Anti‐VEGF versus laser, Outcome 8 Mean difference in (change of) OCT Central Macular Thickness at 2 years.

8.1 Bevacizumab

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+ Show forest plot

3

380

Risk Ratio (M‐H, Fixed, 95% CI)

3.29 [1.77, 6.13]

Analysis 1.9

Comparison 1 Anti‐VEGF versus laser, Outcome 9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+.

Comparison 1 Anti‐VEGF versus laser, Outcome 9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+.

10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+ Show forest plot

2

305

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.02, 0.41]

Analysis 1.10

Comparison 1 Anti‐VEGF versus laser, Outcome 10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+.

Comparison 1 Anti‐VEGF versus laser, Outcome 10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+.

Open in table viewer
Comparison 2. Anti‐VEGF versus sham

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

2.46 [1.77, 3.40]

Analysis 2.1

Comparison 2 Anti‐VEGF versus sham, Outcome 1 Gain 2+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 1 Gain 2+ lines of visual acuity at 8 to 12 months.

1.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.43, 3.09]

1.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

3.31 [1.80, 6.09]

2 Gain 3+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

2.19 [1.36, 3.53]

Analysis 2.2

Comparison 2 Anti‐VEGF versus sham, Outcome 2 Gain 3+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 2 Gain 3+ lines of visual acuity at 8 to 12 months.

2.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

1.79 [1.01, 3.16]

2.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

3.17 [1.32, 7.62]

3 Loss 2+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.19, 0.60]

Analysis 2.3

Comparison 2 Anti‐VEGF versus sham, Outcome 3 Loss 2+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 3 Loss 2+ lines of visual acuity at 8 to 12 months.

3.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

0.44 [0.21, 0.92]

3.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

0.20 [0.07, 0.54]

4 Loss 3+ lines of visual acuity at 8 to 12 months Show forest plot

2

411

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.13, 0.59]

Analysis 2.4

Comparison 2 Anti‐VEGF versus sham, Outcome 4 Loss 3+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 4 Loss 3+ lines of visual acuity at 8 to 12 months.

4.1 Pegaptanib

1

260

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.16, 1.21]

4.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

0.14 [0.04, 0.50]

5 Mean change of visual acuity at 6 to 12 months Show forest plot

4

575

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.17, ‐0.08]

Analysis 2.5

Comparison 2 Anti‐VEGF versus sham, Outcome 5 Mean change of visual acuity at 6 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 5 Mean change of visual acuity at 6 to 12 months.

5.1 Bevacizumab

1

78

Mean Difference (IV, Fixed, 95% CI)

‐0.15 [‐0.26, ‐0.04]

5.2 Pegaptanib

2

346

Mean Difference (IV, Fixed, 95% CI)

‐0.08 [‐0.13, ‐0.03]

5.3 Ranibizumab

1

151

Mean Difference (IV, Fixed, 95% CI)

‐0.23 [‐0.32, ‐0.15]

6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months Show forest plot

3

315

Mean Difference (IV, Fixed, 95% CI)

‐126.38 [‐160.27, ‐92.49]

Analysis 2.6

Comparison 2 Anti‐VEGF versus sham, Outcome 6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months.

6.1 Bevacizumab

1

78

Mean Difference (IV, Fixed, 95% CI)

‐130.6 [‐187.27, ‐73.93]

6.2 Pegaptanib

1

86

Mean Difference (IV, Fixed, 95% CI)

‐71.7 [‐149.71, 6.31]

6.3 Ranibizumab

1

151

Mean Difference (IV, Fixed, 95% CI)

‐145.80 [‐196.12, ‐95.48]

7 Gain 2+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

1.99 [1.64, 2.40]

Analysis 2.7

Comparison 2 Anti‐VEGF versus sham, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

7.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.87, 1.88]

7.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

2.30 [1.85, 2.86]

8 Gain 3+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.85, 3.23]

Analysis 2.8

Comparison 2 Anti‐VEGF versus sham, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

8.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

1.56 [0.87, 2.78]

8.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

2.80 [2.03, 3.86]

9 Loss 3+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.17, 0.64]

Analysis 2.9

Comparison 2 Anti‐VEGF versus sham, Outcome 9 Loss 3+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 9 Loss 3+ lines of visual acuity at 2 years.

9.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

0.42 [0.13, 1.31]

9.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

0.30 [0.13, 0.68]

10 Loss 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.10

Comparison 2 Anti‐VEGF versus sham, Outcome 10 Loss 2+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 10 Loss 2+ lines of visual acuity at 2 years.

10.1 Ranibizumab

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Mean change of visual acuity at 2 years Show forest plot

2

716

Mean Difference (IV, Fixed, 95% CI)

‐0.16 [‐0.20, ‐0.12]

Analysis 2.11

Comparison 2 Anti‐VEGF versus sham, Outcome 11 Mean change of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 11 Mean change of visual acuity at 2 years.

11.1 Pegaptanib

1

207

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.17, ‐0.02]

11.2 Ranibizumab

1

509

Mean Difference (IV, Fixed, 95% CI)

‐0.19 [‐0.23, ‐0.14]

12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 2.12

Comparison 2 Anti‐VEGF versus sham, Outcome 12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks).

Comparison 2 Anti‐VEGF versus sham, Outcome 12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks).

13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 2.13

Comparison 2 Anti‐VEGF versus sham, Outcome 13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks).

Comparison 2 Anti‐VEGF versus sham, Outcome 13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks).

Open in table viewer
Comparison 3. Anti‐VEGF plus laser versus laser alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 1 year Show forest plot

3

1267

Risk Ratio (M‐H, Fixed, 95% CI)

1.94 [1.66, 2.28]

Analysis 3.1

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

1.1 Prompt photocoagulation

3

786

Risk Ratio (M‐H, Fixed, 95% CI)

2.14 [1.73, 2.64]

1.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.33, 2.15]

2 Gain 3+ lines of visual acuity at 1 year Show forest plot

3

1267

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.67, 2.67]

Analysis 3.2

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

2.1 Prompt photocoagulation

3

786

Risk Ratio (M‐H, Fixed, 95% CI)

2.29 [1.67, 3.13]

2.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

1.88 [1.31, 2.70]

3 Loss 2+ lines of visual acuity at 1 year Show forest plot

2

1189

Risk Ratio (M‐H, Fixed, 95% CI)

0.26 [0.15, 0.43]

Analysis 3.3

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

3.1 Prompt photocoagulation

2

708

Risk Ratio (M‐H, Fixed, 95% CI)

0.27 [0.14, 0.51]

3.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.10, 0.56]

4 Loss 3+ lines of visual acuity at 1 year Show forest plot

2

1189

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.15, 0.55]

Analysis 3.4

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

4.1 Prompt photocoagulation

2

708

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.13, 0.67]

4.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.10, 0.77]

5 Mean difference in change of logMAR visual acuity at 6 to 12 months Show forest plot

3

1266

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.13, ‐0.08]

Analysis 3.5

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 5 Mean difference in change of logMAR visual acuity at 6 to 12 months.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 5 Mean difference in change of logMAR visual acuity at 6 to 12 months.

5.1 Prompt photocoagulation

3

785

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.13, ‐0.07]

5.2 Deferred photocoagulation

1

481

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.17, ‐0.07]

6 Mean difference in change of OCT central macular thickness at 6 to 12 months Show forest plot

2

1116

Mean Difference (IV, Fixed, 95% CI)

‐40.90 [‐57.19, ‐24.62]

Analysis 3.6

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 6 Mean difference in change of OCT central macular thickness at 6 to 12 months.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 6 Mean difference in change of OCT central macular thickness at 6 to 12 months.

6.1 Prompt photocoagulation

2

670

Mean Difference (IV, Fixed, 95% CI)

‐44.28 [‐64.69, ‐23.86]

6.2 Deferred photocoagulation

1

446

Mean Difference (IV, Fixed, 95% CI)

‐35.0 [‐62.00, ‐6.00]

7 Gain 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.7

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

7.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Gain 3+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.8

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

8.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Loss 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.9

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 9 Loss 2+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 9 Loss 2+ lines of visual acuity at 2 years.

9.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

10 Loss 3+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.10

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 10 Loss 3+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 10 Loss 3+ lines of visual acuity at 2 years.

10.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Mean difference in change of logMAR visual acuity at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.11

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 11 Mean difference in change of logMAR visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 11 Mean difference in change of logMAR visual acuity at 2 years.

11.1 Prompt photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Deferred photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Mean difference in change of OCT central macular thickness at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.12

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 12 Mean difference in change of OCT central macular thickness at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 12 Mean difference in change of OCT central macular thickness at 2 years.

12.1 Prompt photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Deferred photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 4. Adverse events: Anti‐VEGF versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total ATC thromboembolic events at 6 to 24 months Show forest plot

9

2159

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.56, 1.28]

Analysis 4.1

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 1 Total ATC thromboembolic events at 6 to 24 months.

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 1 Total ATC thromboembolic events at 6 to 24 months.

1.1 Follow‐up 6 to 12 months

6

868

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.49, 4.06]

1.2 Follow‐up 24 months

3

1291

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.42, 1.53]

2 Death Show forest plot

9

2159

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.52, 1.74]

Analysis 4.2

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 2 Death.

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 2 Death.

2.1 Follow‐up 6 to 12 months

6

868

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.24, 2.83]

2.2 Follow‐up 24 months

3

1291

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.36, 3.45]

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.
Figuras y tablas -
Figure 1

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.

Forest plot of comparison: 2 AntiVEGF versus Laser, outcome: 2.2 Gain 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Figure 2

Forest plot of comparison: 2 AntiVEGF versus Laser, outcome: 2.2 Gain 3+ lines of visual acuity at 1 year.

Forest plot of comparison: 2 AntiVEGF versus laser, outcome: 2.4 Loss 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Figure 3

Forest plot of comparison: 2 AntiVEGF versus laser, outcome: 2.4 Loss 3+ lines of visual acuity at 1 year.

Forest plot of comparison: 1 AntiVEGF versus sham, outcome: 1.2 Gain 3+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 AntiVEGF versus sham, outcome: 1.2 Gain 3+ lines of visual acuity at 8 to 12 months.

Forest plot of comparison: 1 AntiVEGF versus sham, outcome: 1.4 Loss 3+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 AntiVEGF versus sham, outcome: 1.4 Loss 3+ lines of visual acuity at 8 to 12 months.

Forest plot of comparison: 4 Adverse events: antiVEGF versus control at 6 to 24 months, outcome: 4.2 Death.
Figuras y tablas -
Figure 6

Forest plot of comparison: 4 Adverse events: antiVEGF versus control at 6 to 24 months, outcome: 4.2 Death.

Comparison 1 Anti‐VEGF versus laser, Outcome 1 Gain 2+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 1.1

Comparison 1 Anti‐VEGF versus laser, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 2 Gain 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 1.2

Comparison 1 Anti‐VEGF versus laser, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 3 Loss 2+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 1.3

Comparison 1 Anti‐VEGF versus laser, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 4 Loss 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 1.4

Comparison 1 Anti‐VEGF versus laser, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 5 Mean difference in logMAR visual acuity at 1 year.
Figuras y tablas -
Analysis 1.5

Comparison 1 Anti‐VEGF versus laser, Outcome 5 Mean difference in logMAR visual acuity at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 6 Mean difference in (change of) OCT central macular thickness at 1 year.
Figuras y tablas -
Analysis 1.6

Comparison 1 Anti‐VEGF versus laser, Outcome 6 Mean difference in (change of) OCT central macular thickness at 1 year.

Comparison 1 Anti‐VEGF versus laser, Outcome 7 Mean difference in logMAR visual acuity at 2 years.
Figuras y tablas -
Analysis 1.7

Comparison 1 Anti‐VEGF versus laser, Outcome 7 Mean difference in logMAR visual acuity at 2 years.

Comparison 1 Anti‐VEGF versus laser, Outcome 8 Mean difference in (change of) OCT Central Macular Thickness at 2 years.
Figuras y tablas -
Analysis 1.8

Comparison 1 Anti‐VEGF versus laser, Outcome 8 Mean difference in (change of) OCT Central Macular Thickness at 2 years.

Comparison 1 Anti‐VEGF versus laser, Outcome 9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+.
Figuras y tablas -
Analysis 1.9

Comparison 1 Anti‐VEGF versus laser, Outcome 9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+.

Comparison 1 Anti‐VEGF versus laser, Outcome 10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+.
Figuras y tablas -
Analysis 1.10

Comparison 1 Anti‐VEGF versus laser, Outcome 10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+.

Comparison 2 Anti‐VEGF versus sham, Outcome 1 Gain 2+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Analysis 2.1

Comparison 2 Anti‐VEGF versus sham, Outcome 1 Gain 2+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 2 Gain 3+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Analysis 2.2

Comparison 2 Anti‐VEGF versus sham, Outcome 2 Gain 3+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 3 Loss 2+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Analysis 2.3

Comparison 2 Anti‐VEGF versus sham, Outcome 3 Loss 2+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 4 Loss 3+ lines of visual acuity at 8 to 12 months.
Figuras y tablas -
Analysis 2.4

Comparison 2 Anti‐VEGF versus sham, Outcome 4 Loss 3+ lines of visual acuity at 8 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 5 Mean change of visual acuity at 6 to 12 months.
Figuras y tablas -
Analysis 2.5

Comparison 2 Anti‐VEGF versus sham, Outcome 5 Mean change of visual acuity at 6 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months.
Figuras y tablas -
Analysis 2.6

Comparison 2 Anti‐VEGF versus sham, Outcome 6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months.

Comparison 2 Anti‐VEGF versus sham, Outcome 7 Gain 2+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 2.7

Comparison 2 Anti‐VEGF versus sham, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 8 Gain 3+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 2.8

Comparison 2 Anti‐VEGF versus sham, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 9 Loss 3+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 2.9

Comparison 2 Anti‐VEGF versus sham, Outcome 9 Loss 3+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 10 Loss 2+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 2.10

Comparison 2 Anti‐VEGF versus sham, Outcome 10 Loss 2+ lines of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 11 Mean change of visual acuity at 2 years.
Figuras y tablas -
Analysis 2.11

Comparison 2 Anti‐VEGF versus sham, Outcome 11 Mean change of visual acuity at 2 years.

Comparison 2 Anti‐VEGF versus sham, Outcome 12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks).
Figuras y tablas -
Analysis 2.12

Comparison 2 Anti‐VEGF versus sham, Outcome 12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks).

Comparison 2 Anti‐VEGF versus sham, Outcome 13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks).
Figuras y tablas -
Analysis 2.13

Comparison 2 Anti‐VEGF versus sham, Outcome 13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks).

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 1 Gain 2+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 3.1

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 1 Gain 2+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 2 Gain 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 3.2

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 2 Gain 3+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 3 Loss 2+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 3.3

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 3 Loss 2+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 4 Loss 3+ lines of visual acuity at 1 year.
Figuras y tablas -
Analysis 3.4

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 4 Loss 3+ lines of visual acuity at 1 year.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 5 Mean difference in change of logMAR visual acuity at 6 to 12 months.
Figuras y tablas -
Analysis 3.5

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 5 Mean difference in change of logMAR visual acuity at 6 to 12 months.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 6 Mean difference in change of OCT central macular thickness at 6 to 12 months.
Figuras y tablas -
Analysis 3.6

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 6 Mean difference in change of OCT central macular thickness at 6 to 12 months.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 7 Gain 2+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 3.7

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 7 Gain 2+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 8 Gain 3+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 3.8

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 8 Gain 3+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 9 Loss 2+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 3.9

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 9 Loss 2+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 10 Loss 3+ lines of visual acuity at 2 years.
Figuras y tablas -
Analysis 3.10

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 10 Loss 3+ lines of visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 11 Mean difference in change of logMAR visual acuity at 2 years.
Figuras y tablas -
Analysis 3.11

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 11 Mean difference in change of logMAR visual acuity at 2 years.

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 12 Mean difference in change of OCT central macular thickness at 2 years.
Figuras y tablas -
Analysis 3.12

Comparison 3 Anti‐VEGF plus laser versus laser alone, Outcome 12 Mean difference in change of OCT central macular thickness at 2 years.

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 1 Total ATC thromboembolic events at 6 to 24 months.
Figuras y tablas -
Analysis 4.1

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 1 Total ATC thromboembolic events at 6 to 24 months.

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 2 Death.
Figuras y tablas -
Analysis 4.2

Comparison 4 Adverse events: Anti‐VEGF versus control, Outcome 2 Death.

Summary of findings for the main comparison. Anti‐VEGF versus laser for diabetic macular oedema

Anti‐VEGF versus laser for diabetic macular oedema

Patient or population: patients with diabetic macular oedema
Settings:
Intervention: anti‐VEGF

Comparison: laser

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Anti‐VEGF versus laser

Gain 3+ lines of visual acuity at 1 year
Follow‐up: mean 12 months

81 per 1000

258 per 1000
(167 to 399)

RR 3.2
(2.07 to 4.95)

556
(5 studies)

⊕⊕⊕⊝
moderate1,2,3,4,5

BOLT; DA VINCI; READ2; RESTORE; Soheilian 2007

no overall or drug subgroup heterogeneity (I2 = 0%)

Loss 3+ lines of visual acuity at 1 year
Follow‐up: 12 months

140 per 1000

18 per 1000
(7 to 48)

RR 0.13
(0.05 to 0.34)

481
(4 studies)

⊕⊕⊕⊝
moderate1,2,3,4,5

BOLT; DA VINCI; RESTORE; Soheilian 2007

no overall or drug subgroup heterogeneity (I2 = 0%)

Adverse events: seesummary of findings Table 4

Cost‐effectiveness studies, UK setting

Incremental cost

Incremental QALY

ICER (cost per QALY gained)

Base‐case

Main sensitivity analyses

Criticism

Comments

NICE 2011

not available

not available

£30,277

1) 35% of people treated in both eyes: £44,400

2) Utilities source Brazier 20086: £23,664

2) Utilities source Lloyd 2008: £24,779                

Bilateral treatment not considered.

Assumption that no treatment is needed beyond year 3.

Manufacturer's submission to NICE. Several assumptions were criticised by the NICE Appraisal Committee that estimated higher ICER under different assumptions.

Mitchell 2012

£4191

0.17

£24,028

1) 14 total ranibizumab injections: £38,836

2) Utilities source Lloyd 2008: £19,238

Bilateral treatment not considered.

Assumption that no treatment is needed beyond year 2.

Manufacturer's sponsored study.

Main difference versus NICE 2011 likely related to lower number of injections (10 in years 1 to 2 for both NICE 2011 and Mitchell 2012, but 3 versus 0 injections in year 3, respectively (total 13 versus 10).

*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).
CI: Confidence interval; RR: Risk ratio.

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.

1 One small study had problems of baseline imbalance and loss to follow‐up, but we did not downgrade quality because the consequences are not known.
2 Heterogeneity difficult to estimate with few trials in the analysis, some of which are relatively small; differences between drugs hard to investigate (‐1).
3 Optimal Information Size criterion not met according to Guyatt 2011 (‐1).

4 Large effect measured precisely (+1).
5 Unclear quality for most items in READ2, which did not provide loss data; however, no overall quality penalty was applied since READ‐2 received little weight in the meta‐analysis.

6Referenced in NICE 2011 but could not find the reference.

Figuras y tablas -
Summary of findings for the main comparison. Anti‐VEGF versus laser for diabetic macular oedema
Summary of findings 2. Anti‐VEGF compared with sham for diabetic macular oedema

Anti‐VEGF compared with sham for diabetic macular oedema

Patient or population: patients with diabetic macular oedema
Settings:
Intervention: anti‐VEGF
Comparison: sham

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Sham

Anti‐VEGF

Gain 3+ lines of visual acuity at 6 to 12 months

96 per 1000

203 per 1000
(131 to 339)

RR 2.19
(1.36 to 3.53)

497
(3 studies)

⊕⊕⊕⊝
moderate1,2,3

Macugen 2005; Macugen 2011; RESOLVE

heterogeneity: overall I2 = 0%

drug subgroup I2 = 12.9% (P = 0.28)

Loss 3+ lines of visual acuity at 6 to 12 months

119 per 1000

33 per 1000
(15 to 70)

RR 0.28
(0.13 to 0.59)

411
(2 studies)

⊕⊕⊕⊝
moderate1,2,3

Macugen 2011; RESOLVE

heterogeneity: overall I2 = 44%

drug subgroup I2 = 44.2% (P = 0.18)

Adverse events: seesummary of findings Table 4

*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).
CI: Confidence interval; RR: Risk ratio.

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.

1 Heterogeneity difficult to estimate with few trials in the analysis, two of which are relatively small. Also, data collected at variable follow‐up (‐1).
2 Optimal Information Size criterion not met according to Guyatt 2011 (‐1).
3 Large effect measured precisely (+1).

Figuras y tablas -
Summary of findings 2. Anti‐VEGF compared with sham for diabetic macular oedema
Summary of findings 3. Ranibizumab plus laser compared with laser alone for diabetic macular oedema

Ranibizumab plus laser compared with laser alone for diabetic macular oedema

Patient or population: patients with diabetic macular oedema
Settings:
Intervention: ranibizumab plus laser
Comparison: laser alone

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Laser alone

Ranibizumab plus laser

Gain 3+ lines of visual acuity at 1 year ‐ Prompt photocoagulation
Follow‐up: 12 months

118 per 1000

249 per 1000
(197 to 369)

RR 2.11
(1.67 to 3.13)

786
(3 studies)

⊕⊕⊕⊝
moderate1,2,3,4

DRCRnet; READ2; RESTORE; heterogeneity: I2 = 0%

Gain 3+ lines of visual acuity at 1 year ‐ Deferred photocoagulation
Follow‐up: 12 months

147 per 1000

276 per 1000
(193 to 397)

RR 1.88
(1.31 to 2.7)

481
(1 study)

⊕⊕⊕⊝
moderate1,2,3,4

DRCRnet

Loss 3+ lines of visual acuity at 1 year ‐ Prompt photocoagulation
Follow‐up: 12 months

79 per 1000

23 per 1000
(10 to 53)

RR 0.29
(0.13 to 0.67)

708
(2 studies)

⊕⊕⊕⊝
moderate1,2,3,4

DRCRnet; READ2; RESTORE;

no heterogeneity: I2 = 0%

Loss 3+ lines of visual acuity at 1 year ‐ Deferred photocoagulation
Follow‐up: 12 months

78 per 1000

21 per 1000
(8 to 60)

RR 0.27
(0.1 to 0.77)

481
(1 study)

⊕⊕⊕⊝
moderate1,2,3,4

DRCRnet

Adverse events: seesummary of findings Table 4

*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).
CI: Confidence interval; RR: Risk ratio.

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.

1 Unclear quality for most items in READ‐2, which did not provide loss data; however, no overall quality penalty was applied since READ‐2 received little weight in the meta‐analysis.
2 Heterogeneity impossible or difficult to estimate with one or few trials in the analysis.
3 Optimal Information Soize criterion not met according to Guyatt 2011 (‐1).
4 Large effect measured precisely (+1).

Figuras y tablas -
Summary of findings 3. Ranibizumab plus laser compared with laser alone for diabetic macular oedema
Summary of findings 4. Adverse events: Anti‐VEGF compared with control for diabetic macular oedema

Adverse events: Anti‐VEGF compared with control for diabetic macular oedema

Patient or population: patients with diabetic macular oedema
Settings:
Intervention: adverse events: anti‐VEGF
Comparison: control

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Adverse events: anti‐VEGF

Total ATC thromboembolic events
Follow‐up: 6 to 24 months

50 per 1000

43 per 1000
(28 to 64)

RR 0.85
(0.56 to 1.28)

2159
(9 studies)

⊕⊕⊝⊝
low1,2,3

heterogeneity: I2 = 6%

Death
Follow‐up: 6 to 24 months

22 per 1000

21 per 1000
(12 to 39)

RR 0.95

[0.52 to 1.74]

2159
(9 studies)

⊕⊕⊝⊝
low1,2,3

heterogeneity: I2 = 8%

*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).
CI: Confidence interval; RR: Risk ratio.

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.

1 Variable follow‐up (6 to 24 months) (‐1).

2 Large 95% confidence intervals, including effects that are clinically relevant (‐1).

3 Antiplatelet Trialists' Collaboration (ATC) events definition not fully met for studies Macugen 2011; RISE‐RIDE and problem with extracting data in many studies (‐1).

Figuras y tablas -
Summary of findings 4. Adverse events: Anti‐VEGF compared with control for diabetic macular oedema
Table 1. Outcome reporting grid: visual acuity

Outcome 

Gain 3+ lines

Loss 3+ lines

Gain 3+ lines

Loss 3+ lines

Study

antiangiogenic drug

6 to 12 months

2 years

Soheilian 2007

bevacizumab

yes

yes

yes

yes

Ahmadieh 2008

bevacizumab

E

E

E

E

BOLT

bevacizumab

yes

yes

NA

NA

Macugen 2005

pegaptanib

yes

E

NA

NA

Macugen 2011

pegaptanib

yes

yes

yes

yes

DRCRnet

ranibizumab

yes

yes

yes

yes

READ2

ranibizumab

yes

E

NA

NA

RESOLVE

ranibizumab

yes

yes

NA

NA

RESTORE

ranibizumab

yes

yes

NA

NA

RISE‐RIDE

ranibizumab

E

E

yes

yes

DA VINCI

aflibercept

yes

yes

NA

NA

yes: outcome analysed and fully reported allowing its inclusion in the meta‐analysis.

E: clear that outcome was measured (for example, includes structurally related outcomes) but not necessarily analysed.

(adapted from list provided by Paula Williamson at Cochrane training workshop on selective outcome reporting bias, Edinburgh March 2009).

NA: not applicable since follow‐up shorter than 2 years

Figuras y tablas -
Table 1. Outcome reporting grid: visual acuity
Table 2. Mean or median (*) number of intravitreal injections

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

antiVEGF

+ laser (prompt)

ranibizumab

+ laser (deferred)

aflibercept 2 mg PRN§

Macugen 2005

36 weeks (reported at 30 weeks)

4.5 (1.5)

5 (1.2)

Soheilian 2007

2 years

 

 

3.1 (1.6)

 

1 (0.1)

Ahmadieh 2008

24 weeks

3

3

DRCRnet

1 year

 

 

 

 

 

8 (7,11)*

9 (7,11)*

DRCRnet

year 2 only

 

 

 

 

 

2 (0,4)*

3 (1,7)*

RESTORE

1 year

 

7 (2.81)

 

 

7.3 (3.22)

6.8 (2.95)

 

RESOLVE

1 year

8.9 (3.5)

10.2 (2.5)

 

 

 

 

READ2

1.5 years

 

5.3

 

 

4.4

2.9

 

BOLT

1 year

 

 

9 (8,9)*

 

3 (2, 4)*

Macugen 2011

1 year

8.4 (1.4)

 

 

8.3 (1.7) 

Macugen 2011

2 years

12.9 (4.4)

 

 

12.7 (4.6)

RISE‐RIDE (two studies)

2 years

20 (7.5)

20.8 (7.1) 

20.9 (6.3) 

21.9 (5.8)

 

DA VINCI

1 year

7.4 (3.19)

(*): median (interquartile range) number of injection; mean otherwise

(§): only regimen 2q8 included: loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

Figuras y tablas -
Table 2. Mean or median (*) number of intravitreal injections
Table 3. Presentation of absolute risk and number‐needed‐to‐treat (NNT)

 Cut‐off

Studies

Patients

Control

Absolute effect

RR

NNT

pegaptanib versus sham (1 year)

Gain 3+

2

346

9%

+7% (0, +20%)

1.79 (1.01, 3.16)

13 (5, 1056)

Gain 2+

2

346

17%

+19% (+7%, +36%)

2.11 (1.43, 3.09)

5 (3, 14)

Stable

2

346

78%

 

 

 

Loss 2+

2

346

12%

‐7% (‐1%, ‐9%)

0.44 (0.21, 0.92)

15 (11, 106)

Loss 3+

1

260

9%

‐5% (+2%, ‐7%)

0.43 (0.16, 1.21)

20 (14, 55)

ranibizumab versus sham (1 year)

Gain 3+

1

151

10%

+22% (3%, +68%)

3.17 (1.32, 7.62)

5 (1, 31)

Gain 2+

1

151

18%

+42% (+15%, +93%)

3.31 (1.80, 6.09)

2 (1, 7)

Stable

1

151

57%

 

 

 

Loss 2+

1

151

24%

‐20% (‐11%, ‐23%)

0.20 (0.07, 0.54)

5 (4, 9)

Loss 3+

1

151

20%

‐18% (‐10%, ‐20%)

0.14 (0.04, 0.50)

6 (5, 10)

bevacizumab versus laser (1 year)

Gain 3+

2

167

10%

+15% (2%, +42%)

2.52 (1.20, 5.29)

7 (2, 51)

Gain 2+

2

167

11%

+22% (+6%, +56%)

3.06 (1.54, 6.05)

4 (2, 17)

Stable

2

167

64%

 

 

 

Loss 2+

2

167

25%

‐20% (‐11%, ‐23%)

0.19 (0.07, 0.52)

5 (4, 9)

Loss 3+

2

167

22%

‐17% (‐11%, ‐21%)

0.16 (0.05, 0.51)

5 (5, 9)

ranibizumab versus laser (1 year)

Gain 3+

1

225

8%

+21% (6%, +48%)

3.51 (1.78, 6.92)

7 (3, 34)

Gain 2+

1

225

15%

+32% (+14%, +60%)

3.06 (1.93, 4.87)

5 (2, 14)

Stable

1

225

72%

 

 

 

Loss 2+

1

225

13%

‐9% (‐3%, ‐12%)

0.27 (0.09, 0.80)

11 (9, 39)

Loss 3+

1

225

8%

‐7% (‐1%, ‐8%)

0.11 (0.01, 0.83)

14 (12, 72)

ranibizumab plus prompt laser versus laser (1 year)

Gain 3+

3

786

12%

+15% (8%, +25%)

2.29 [1.67, 3.13)

7 (4, 13)

Gain 2+

3

786

23%

+26% (+17%, +37%)

2.14 [1.73, 2.64)

4 (3, 6)

Stable

2

708

63%

 

 

 

Loss 2+

2

708

13%

‐10% (‐6%, ‐11%)

0.27 (0.14, 0.51)

10 (9, 16)

Loss 3+

2

708

8%

‐6% (‐3%, ‐7%)

0.29 [0.13, 0.67)

17 (14, 38)

aflibercept (2PRN§) versus laser (1 year)

Gain 3+

1

89

11%

31% (6%, 92%)

3.72 (1.52, 9.08)

3 (1, 17)

Gain 2+

1

89

30%

33% (8%, 74%)

2.11 (1.26, 3.51)

3 (1, 13)

Stable

1

89

52%

 

 

 

Loss 2+

1

89

18%

‐14% (‐17%, 2%)

0.24 (0.05, 1.09)

7 (6, 61)

Loss 3+

1

89

14%

‐13% (‐14%,4%)

0.08 (0.00, 1.30)

8 (7, 24)

(*) NNT not presented because difference not statistically significant and both benefit and harm are possible according to 95% confidence limits.

(§): data extracted from only 1 of 4 active drug comparison groups (loading dose of 3 initial 2 mg injection, then as needed; PRN: Pro Re Nata)

Figuras y tablas -
Table 3. Presentation of absolute risk and number‐needed‐to‐treat (NNT)
Table 4. Dataset used in indirect comparisons among antiangiogenic drugs.

study

treatment

gain 2+

gain 3+

total

BOLT

laser

3

2

38

BOLT

bevacizumab

13

5

42

DRCRnet

laser

81

43

293

DRCRnet

ranibizumab/laser

95

57

187

Macugen 2005

sham

4

3

42

Macugen 2005

pegaptanib

15

8

44

Macugen 2011

sham

25

13

127

Macugen 2011

pegaptanib

49

22

133

READ2

laser

2

0

38

READ2

ranibizumab

17

8

37

READ2

ranibizumab/laser

12

3

40

RESOLVE

sham

9

5

49

RESOLVE

ranibizumab

62

33

102

RESTORE

laser

17

9

110

RESTORE

ranibizumab

43

26

115

RESTORE

ranibizumab/laser

51

27

118

Soheilian 2007

laser

6

6

43

Soheilian 2007

bevacizumab

16

16

44

DA VINCI

laser

28

5

44

DA VINCI

aflibercept*

13

19

45

(*): only 1 of 4 aflibercept regimens was selected, based on similarity to current clinical practice

Figuras y tablas -
Table 4. Dataset used in indirect comparisons among antiangiogenic drugs.
Table 5. Ocular adverse events: endophthalmitis

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

ranibizumab + laser (prompt)

ranibizumab + laser (deferred)

aflibercept 2 mg PRN§

Macugen 2005

36 weeks

0/42

1/44

Soheilian 2007 *

2 years

 

 

0/48

 

0/48

Ahmadieh 2008 (#)

24 weeks

0

0

DRCRnet

2 years

 

 

 

 

1/293

2/187

2/188

RESTORE

1 year

 

0/115

 

 

0/110

0/120

 

RESOLVE

1 year

0/49

2/102

 

 

 

 

 

READ2

2 years

 

 

 

 

 

 

BOLT

1 year

 

 

0/42

 

0/40

 

 

Macugen 2011

2 years

0/127

 

 

0/133

 

 

 

RISE‐RIDE

2 years

0/250

3/250*

DA VINCI

1 year

0/44

1/45

(*): denominator is total number of patients at mean follow‐up

(#): no cases mentioned but number of eyes, not patients, given for each group

(§): loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

Figuras y tablas -
Table 5. Ocular adverse events: endophthalmitis
Table 6. Ocular adverse events: retinal detachment

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

ranibizumab + laser (prompt)

ranibizumab + laser (deferred)

aflibercept 2 mg PRN§

Macugen 2005

36 weeks

0/42

0/44

Soheilian 2007

36 weeks

0/48

 

0/48

Ahmadieh 2008 (#)

24 weeks

0

0

DRCRnet

2 years

 

 

 

 

1/293

0/187

1/188

RESTORE

1 year

 

0/115

 

 

0/110

0/120

 

RESOLVE

1 year

1/49

0/102

 

 

 

 

 

READ2

2 years

 

 

 

 

 

 

BOLT

1 year

 

 

0/42

 

0/40

 

 

Macugen 2011

2 years

0/127

 

 

0/133

 

 

 

RISE‐RIDE

2 years

1/250

1/250*

DA VINCI

0/44

0/45

(#): no cases mentioned but number of eyes, not patients, given for each group

(§): loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

Figuras y tablas -
Table 6. Ocular adverse events: retinal detachment
Table 7. Total non‐ocular adverse events

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

ranibizumab + laser (prompt)

ranibizumab + laser (deferred)

aflibercept 2 mg PRN§

Macugen 2005

36 weeks

0/42

0/44

Soheilian 2007 (*)

36 weeks

 

 

0/48

0/48

Ahmadieh 2008 (*)

24 weeks

1

0

DRCRnet

2 years

 

 NA

 

 

 NA

 NA

 NA

RESTORE

1 year

 

23/115

 

 

15/110

17/120

 

RESOLVE

1 year

8/49

14/102

 

 

 

 

 

READ2

2 years

 

 

 

 

BOLT

1 year

 

 

3/42

 

7/38

 

 

Macugen 2011

2 years

 27/142

 

 

 28/144

 

 

 

RISE‐RIDE (#)

2 years

25/250

22/250*

DA VINCI

1 year

10/44

6/45

(*): eyes, not patients, as unit of analysis

(§): loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

(#): only 0.5 mg dose

NA: not available in the manuscript

Figuras y tablas -
Table 7. Total non‐ocular adverse events
Table 8. Total arterial thromboembolic events or APTC (Antiplatelet Trialists` Collaboration) events

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

ranibizumab + laser

aflibercept 2 mg PRN§

Macugen 2005

36 weeks

0/42

0/44

Soheilian 2007 (*)

36 weeks

0/44

2/43

Ahmadieh 2008 (*)

24 weeks

1/?

0/?

READ2

6 months

 0/39

 

 

 0/40

 1/38

DRCRnet

2 years

 

 

 

 

17/130

25/375

RESTORE

1 year

 

6/115

 

 

1/110

1/120

RESOLVE

1 year

2/49

3/102

 

 

 

 

BOLT

1 year

 

 

0/42

 

1/38

 

Macugen 2011 (#)

2 years

 9/142

 

 

 7/144

 

 

RISE‐RIDE (#)

2 years

13/250

18/250

DA VINCI

1 year

1/44

1/45

(*): eyes, not patients, as unit of analysis

(§): loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

(#) approved dose only (0.5 mg ranibizumab, 0.3 mg pegaptanib)

Figuras y tablas -
Table 8. Total arterial thromboembolic events or APTC (Antiplatelet Trialists` Collaboration) events
Table 9. All cause mortality

study

follow‐up

sham

ranibizumab

bevacizumab

pegaptanib

laser

ranibizumab + laser

aflibercept 2 mg PRN§

Macugen 2005

36 weeks

0/42

0/44

Soheilian 2007 (*)

36 weeks

 

 

0/44

 

2/43

Ahmadieh 2008 (*)

24 weeks

1/?

0/?

READ2

6 months

 0/39

 

 

 0/40

 1/38

RESTORE

1 year

 

2/115

 

 

2/110

2/120

RESOLVE

1 year

0/49

1/102

 

 

 

 

BOLT

1 year

 

 

0/42

0/38

DRCRnet

2 years

 

 

 

 

8/130

13/375

Macugen 2011 (#)

2 years

5/142

 

 4/144

 

 

RISE‐RIDE (#)

2 years

3/250

11/250

 

DA VINCI

1 year

1/44

0/45

(*): eyes, not patients, as unit of analysis

(§): loading dose of 3 initial injection and then as needed (PRN: Pro Re Nata)

(#) approved dose only (0.5 mg ranibizumab, 0.3 mg pegaptanib)

Figuras y tablas -
Table 9. All cause mortality
Table 10. Economic evaluations: 1. Smiddy 2011

Form of economic analysis

 

Cost‐effectiveness analysis (cost per unit line of vision saved).

 

Population

 

Patients with macular oedema due to diabetic retinopathy enrolled in phase I/III RCTs

Interventions

 

Laser grid photocoagulation, intravitreal triamcinolone, dexamethasone implant, pegaptanib, bevacizumab, ranibizumab

 

Comparators

 

Not defined a priori

 

Perspective, time horizon

 

6 to 12 months in included studies, life‐long horizon.

 

Modelling used and Key assumptions

 

Quote: “Most index studies offered 1‐year follow‐up data, but resource use data were conservatively extrapolated to 1 year when shorter, assuming a durable result at 1 year, and until death for line‐year calculations. […] By design, costs were generally underestimated and benefits were overestimated for more expensive modalities; the converse was applied to less expensive modalities.

Quote: “By using the estimate of lines saved, cost/line saved was calculated. The mean age of each study cohort was used to calculate a life expectancy, by consulting the actuarial table of the Social Security Administration, and to calculate cost/line years saved.”

 

Effectiveness data

 

Single randomised trial  comparing any listed intervention to any control. Non comparative studies also used (natural course of all conditions, outcome of bevacizumab treatment for DMO).

 

Health state valuations (utilities)

 

Cost‐benefit analysis, but [quote] “A cost utility analysis was performed by using the lines saved estimate and ascribing 0.03 marginal quality‐adjusted life years (QALYs) for each line gained, a reasonable estimate from previously published data in the VA range of the index study cohorts, albeit for a “better eye,” which was not usually the case in the index study cohorts.”

 

Unit cost data, price year, resource use data

 

 

Direct cost only, including follow‐up visits, OCT, fluorescein angiography, treatment. Quote: “The Medicare allowable amounts for hospital‐based use (in South Florida as of June 1, 2010) were applied as costs (including hospital facility usage allowable) for the hypothetic 1 year of treatment.

All costs were calculated in US dollars; present value adjustments were not included in cost calculations.”

 

Discounting

 

Not mentioned.

 

Results and sensitivity analyses

 

Not reported because we judged study quality was insufficient.

 

DMO: diabetic macular oedema

Figuras y tablas -
Table 10. Economic evaluations: 1. Smiddy 2011
Table 11. Evers Checklist: Smiddy 201

 

yes

no

quote/comment

Item

 

 

1. Is the study population clearly described?

Y

 

 Quote: "Purpose: To relate costs and treatment benefits for diabetic macular edema, branch retinal vein occlusion, and central retinal vein occlusion."

Comment: very general statement including macular oedema due to several diseases.

2. Are competing alternatives clearly described?

 

 N

Quote: "For each of the 3 diagnostic groups, an index study (usually a collaborative, randomized, controlled study with 6‐month to 1‐year follow‐up information) was chosen for natural history, focal laser, intravitreal triamcinolone acetonide, intravitreal dexamethasone implant, pegaptanib, bevacizumab, and ranibizumab, and a surgical series for DMO only".

Comment: treatments listed, but reasons for this choice are not reported. A discussion of the appropriate comparator was not made.

3. Is a well‐defined research question posed in answerable form?

 Y

 

 Quote: "Purpose: To relate costs and treatment benefits for diabetic macular edema, branch retinal vein occlusion, and central retinal vein occlusion."

Comment: same quotation as above, very general, broad purpose economic study.

4. Is the economic study design appropriate to the stated objective?

 

 N

Comment: calculation of cost per line of vision saved for each treatment, as well as of QALYs, made with no probabilistic modelling of transition probabilities between health states, including benefits and adverse events; non probabilistic sensitivity analyses were performed.

5. Is the chosen time horizon appropriate to include relevant costs and consequences?

 Y

 

Quote: ""Most index studies offered 1‐year follow‐up data, but resource use data were conservatively extrapolated to 1 year when shorter, assuming a durable result at 1 year, and until death for line‐year calculations".

Comment: life‐long time horizon appropriate.

6. Is the actual perspective chosen appropriate?

 ?

 ?

Comment: perspective not declared. Given the measurement of benefits and direct costs, the perspective should be that of the public health care provider.

7. Are all important and relevant costs for each alternative identified?

 

 N

Comment: Costs of treatment, follow‐up visits, OCT and fluorescein angiography considered, but not cost of adverse events.

8. Are all costs measured appropriately in physical units?

 Y

 

Quote: "The Medicare allowable amounts for hospital‐based use (in South Florida as of June 1, 2010) were applied as costs (including hospital facility usage allowable) for the hypothetic 1 year of treatment".

9. Are costs valued appropriately?

N

Quote: "By design, costs were generally underestimated and benefits were overestimated for more expensive modalities; the converse was applied to less expensive modalities."

"All costs were calculated in US dollars; present value adjustments were not included in cost calculations." 

Comment: cost assumptions neither justified nor checked in sensitivity analyses; no discount rate applied.

10. Are all important and relevant outcomes for each alternative identified?

 

 N

Comment: adverse events not considered.

11. Are all outcomes measured appropriately?

 

 N

 See above.

12. Are outcomes valued appropriately?

 

 N

 See above.

13. Is an incremental analysis of costs and outcomes of alternatives performed?

 Y

 

Quote: "By using the estimate of lines saved, cost/line saved was calculated."

Comment: cost and outcomes were combined, however the comparator generating the incremental cost‐effectiveness was insufficiently reported.

14. Are all future costs and outcomes discounted appropriately?

 

 N

 Comment: no discount rate applied.

15. Are all important variables, whose values are uncertain, appropriately subjected to sensitivity analysis?

 

 N

 Comment: no sensitivity analysis.

16. Do the conclusions follow from the data reported?

 Y

 

 Comment: conclusions are coherent with results, but the methodology is inappropriate.

17. Does the study discuss the generalizability of the results to other settings and patient/ client groups?

 

 N

Quote: "Certainly, selection biases and application differences may apply not only to the index study patient cohorts but also, more important, to the patients actually treated in the clinic on the basis of the results of these studies. Thus, the “real world” differential may be even less. Furthermore, most index studies report up to 1‐year results; the durability and amount of additional costs necessary to maximize longer‐term results are even more conjectural."

Comment: the study acknowledges that included RCTs may not be pragmatic and reflect routine clinical practice.

18. Does the article indicate that there is no potential conflict of interest of study researcher(s) and funder(s)?

 N

 

Comment: no conflict of interest explicitly declared.

19. Are ethical and distributional issues discussed appropriately?

 

 N

Quote: "Although physicians have historically been committed to making treatment decisions independently of treatment costs, at least for distinctly important medical problems, the 15‐fold cost differences, the high prevalence of these conditions, and the ongoing nature of treatment make the issue of costs inescapable. It would seem good stewardship for the physician, the traditional gatekeeper of medical care, to be cognizant of the magnitude of the cost differential and to consider it in some manner in formulating treatment regimens. A problem is that there is no policy basis or even short‐term motivation for the physician to act on these findings. This difficult ethical problem is even more difficult for an individual patient, payor, or health policy maker."

Comment: the perspective taken is that of the physician, which does not completely overlap with the perspective of policy‐maker.

OCT: optical coherence tomography; RCT: randomised controlled trial

Figuras y tablas -
Table 11. Evers Checklist: Smiddy 201
Table 12. Economic evaluations: NICE 2011

Form of economic analysis

 

Cost‐utility analysis.  

 

Population

 

Patients with macular oedema due to diabetic retinopathy enrolled in phase III RCTs RESTORE and DRCRnet. READ2 was excluded because follow‐up was short and treatment schedule was inadequate.

Interventions

 

Ranibizumab alone or added to laser grid photocoagulation.

 

Comparators

 

Laser grid photocoagulation.

Perspective, time horizon

 

Perspective of health care provider; follow‐up 12‐24 months in included studies,15 years horizon in modelling.

 

Modelling used and Key assumptions

 

The key section of the original manufacturer's submission is presented here:

1) transition probabilities between three health states in year 1 were drew from changes in best corrected visual acuity (BCVA) in RESTORE for treated and control patients; treatment effect was modelled as stable in year 2 based on DRCRnet and no treatment effect was assumed thereafter, meaning that BCVA would remain constant and decline at the same rate in the two groups after year 1.

2) to estimate the HRQoL associated with each health state corresponding to vision, EQ‐5D data from RESTORE were transformed to utility values using standard social tariffs and then related to BCVA in the treated eye using linear regression.

The original manufacturer's submission was revised following the Evidence Review Group comments. Manufacturer's revised assumptions are presented here.

1) patients receiving ranibizumab would receive an average of two injections in a third year of treatment and one injection in a fourth year of treatment. In the laser photocoagulation arm, the model assumed once‐yearly treatments for years 3 and 4.

2) following criticisms, the manufacturer provided a scenario analysis, using its revised model, which simulated treatment in both eyes for 35% of people. This analysis assumed that, in people with bilateral disease, both eyes would be treated and monitored at the same visit, with ranibizumab drug and treatment costs doubled. The analysis applied reduced costs associated with severe visual impairment because fewer people would go blind in both eyes. The analysis assumed that treating the second eye would result in utility gains one quarter the magnitude of those achieved by treating the first eye; this is because the HRQoL of people who can see well with both eyes is only a little better than the HRQoL of people who can see well with one eye. The model calculated this figure by applying a 25% uplift to the QALYs generated by ranibizumab.

3) the revised model predicted that 43% of the cohort would remain alive after 15 whereas the original model had suggested that 65% of people would be alive at that time.

4) had concluded that, by assuming people whose BCVA rose to 76 letters or higher would stop receiving ranibizumab, the manufacturer's original model had not reflected likely clinical practice. Acknowledging this view, the manufacturer removed the stopping rule from the base case of its revised model.

5) [quote] "a further series of scenario analyses adopted alternative estimates of utility drawn from various published sources. When utility values from the better seeing eye study by Lloyd et al. were used, the ICER was £24,779 per QALY gained. When utility was estimated according to an equation published by Sharma et al., associating visual acuity in the better‐seeing eye with HRQoL, the ICER was between £12,312 and £12,610 per QALY gained, depending on the version of the equation used. A final analysis adopted utility values estimated in a study by Brazier et al., in which members of the general public valued levels of visual impairment that were simulated by custom‐made contact lenses, using the time trade‐off method. Participants wore the same lenses in both eyes, so the resulting utility values reflected bilateral impairment of vision. This was the source of utility values the Committee had judged most accurately reflected the HRQoL associated with visual impairment in NICE technology appraisal guidance 155. When these values were used in the revised ranibizumab model, the ICER was £23,664 per QALY gained."

Conclusive comments by the Appraisal Commitee are presented here.

Quote: “In summary, the Committee considered that amendments to the manufacturer's model made during the consultation process had resulted in a more robust analysis. However, the Committee believed that the manufacturer's revised base‐case model still provided an inaccurate reflection of likely clinical practice in at least six respects:

  • By not accounting for the need to treat both eyes in a large proportion of people with diabetic macular oedema, the manufacturer's base‐case revised model underestimated the benefits and – to a greater degree – the costs of treatments. The manufacturer's scenario analysis simulating treatment in both eyes for 35% of people provided a more realistic reflection of likely clinical practice. This analysis generates an ICER that is almost 50% higher than the revised base case (see section 4.21).

  • The range of utility values used in the manufacturer's revised base case was broader than would be expected according to the assumptions of the model. The Committee preferred the manufacturer's scenario analysis adjusting for factors that may influence the relationship between BCVA and HRQoL. This analysis generates an ICER that is more than 10% higher than the revised base case (see sections 4.22 and 4.23).

  • The model underestimates the amount of ranibizumab that people with diabetic macular oedema are likely to need over time. Basing the number of injections for year 2 of the model's ranibizumab monotherapy arm on observed experience in DRCR.net overlooks the fact that the trial participants also received laser photocoagulation, which clinicians believe may have a ranibizumab‐sparing effect. The declining number of ranibizumab injections assumed in years 3 and 4 is not evidence‐based, and is unlikely to lead to stable vision during that period, as assumed. It may also be unrealistic to assume that ranibizumab treatment will not continue beyond 4 years. The full impact of these assumptions is uncertain, but a more realistic approach could be assumed to raise the ICER given in the revised base case (see section 4.25).

  • The model's assumption that the relative benefit achieved during the treatment phase lasts indefinitely is unrealistic. If NICE technology appraisal guidance 155 is considered a precedent for this approach, then it should be noted that the model in that appraisal had a shorter time horizon, which limited the Committee's uncertainty about extrapolating treatment effects into the future. Shortening the time horizon in the present appraisal to limit uncertainty similarly would result in an ICER that could be expected to be up to 50% higher than the ICER in the revised base case (see section 4.26).

  • The model applies unequal assumptions about treatment visits and monitoring visits for people treated with ranibizumab and those treated with laser photocoagulation. When this imbalance is corrected, the ICER in the revised base case rises by 10–25% (see section 4.27).

  • If glycaemic control in the modelled population reflected that in people treated in clinical practice, it is very likely that the ICER in the revised base case would rise (see section 4.28)."

Effectiveness data

 

RESTORE, DRCRnet

 

Health state valuations (utilities)

 

See above.

 

Unit cost data, price year, resource use data

 

 

Treatment costs:

  • Cost of ranibizumab: £761.20 per injection

  • Treatment with both ranibizumab and laser photocoagulation on an outpatient basis: costs £150 per visit.

  • Combination therapy: ranibizumab injections and laser photocoagulation at the same visit, cost £184 per visit.

  • Visits to monitor patients: £126 each. Patients receiving ranibizumab monotherapy had 12 visits in the first year and 10 visits in the second year; patients receiving combination therapy had 12 visits in the first year and eight visits in the second year; those receiving laser photocoagulation alone in the first and second years, and all patients from the third year onwards, had four visits per year. For the ranibizumab‐containing arms, a visit for treatment was assumed to include monitoring as well. The same assumption was not applied for patients receiving laser photocoagulation alone; that is, patients receiving laser photocoagulation required separate visits for treatment and monitoring.

Costs of low vision:

  • Costs associated  with severe vision loss for patients with the lowest BCVA in the treated eye (0–25 or 26–35 letters), regardless of vision in the nontreated eye: £6067 in the first year and  £5936 in subsequent years. These accounted for a range of tems including low‐vision aids, rehabilitation, residential care, district nursing, community care and the cost of treating complications including depression and falls. The manufacturer drew cost data largely from a published costing study of blindness in the UK that focused on people with age‐related macular degeneration (Meads and Hyde 2003), with costs updated or adjusted for inflation as appropriate.

Discounting

 

Not mentioned.

 

Results and sensitivity analyses

 

Results, description of ICER for base‐case scenario and sensitivity analyses:

  • Base‐case scenario on the comparison of ranibizumab monotherapy and laser photocoagulation: ICER £30,277 per QALY gained. Subgroup of subgroup of people with central foveal thickness greater than 400 micrometres: ICER £21,418 per QALY gained.

  • Treatment in both eyes for 35% of people: ICER £44,400 per QALY gained. Subgroup of people with central foveal thickness greater than 400 micrometres: ICER £35,719 per QALY gained

  • Utilities re‐estimated from RESTORE data using an extended model with additional covariates reflecting patient characteristics and risk factors for diabetic complications: ICER £33,857 per QALY gained.

  • Utility values from the better seeing eye study by other sources:ICER £12,312 ‐ £12,610 (Sharma), ICER £24,779 (Lloyd), ICER £23,664 (Brazier et al., recommended in NICE guidance 155)

Quote: “The Committee considered that the manufacturer's model, as revised during consultation, resulted in an ICER for ranibizumab monotherapy compared with laser photocoagulation of £30,277 per QALY gained, which was at the upper limit of the range it could consider to represent an effective use of NHS resources. However, the Committee concluded that this revised model underestimated the ICER because, despite improvements made in response to the Committee's comments in the appraisal consultation document, the model remained reliant on several implausible assumptions. Where the effect of using individual alternative assumptions was quantifiable, the ICER rose by an appreciable amount; for example the ICER increased to £44,400 per QALY gained when 35% of people were assumed to be treated in both eyes. The Committee concluded that a model that relied on a combined set of plausible assumptions would be certain to produce an ICER that substantially exceeded the acceptable range. Therefore, ranibizumab could not be recommended as a treatment for people with diabetic macular oedema.

[...]

The Committee heard conflicting evidence about the extent to which bevacizumab is currently used to treat diabetic macular oedema in England and Wales for this indication. It concluded that, although bevacizumab is not in routine use throughout the NHS, it is among the treatments adopted by some clinicians and is funded by some NHS trusts. The Committee was mindful that some consultees and commentators supported the evaluation of bevacizumab as a comparator and others opposed it. Because of this, the Committee agreed that, in order to evaluate bevacizumab as a comparator consider a cost‐effectiveness analysis of ranibizumab compared with bevacizumab. However, because it had not been provided with plausible evidence that ranibizumab represents an effective use of NHS resources when compared with laser photocoagulation, the Committee did not believe that considering evidence about the costs and effects of bevacizumab would alter its decision. For this reason, given the current model and its assumptions, the Committee concluded that it could make recommendations on the use of ranibizumab for diabetic macular oedema without requiring additional evidence comparing it with bevacizumab."

Figuras y tablas -
Table 12. Economic evaluations: NICE 2011
Table 13. Economic evaluations: Mitchell 2012

Form of economic analysis

 

Cost‐utility analysis.  

 

Population

 

Patients with macular oedema due to diabetic retinopathy enrolled in phase III RCTs RESTORE and DRCRnet.

Interventions

 

Ranibizumab alone or added to laser grid photocoagulation.

 

Comparators

 

Laser grid photocoagulation.

Perspective, time horizon

 

12‐24 months in included studies, 15‐year horizon.

 

Modelling used and Key assumptions

 

Quote: “The model also assumed the same baseline BCVA distribution [asRESTORE], but excluded patients with BCVA >75 letters, consistent with guidance from the ranibizumab summary of product characteristics that such patients may benefit less from treatment than those with baseline BCVA ≤75 letters. The model framework allocated eight linear health states defined by BCVA in the treated eye using a set of 10‐letter (two‐line) categories. Movement of patients from one health state to another was determined by transition probabilities that depended on the effectiveness of treatment and natural BCVA changes over time

Costs and outcomes were accrued over 3‐month cycles, applying half‐cycle corrections. The time horizon in the base case was 15 years; although a lifetime horizon could be justified given the chronic nature of the condition, we selected a more conservative approach for the base case because of the lack of evidence on long‐term prognosis.

[…] Patients were assumed to receive ranibizumab treatment in year 1 at the frequency observed in RESTORE. In year 2, patients were assumed to need fewer injections, as observed in the Diabetic Retinopathy Clinical Research Network (DRCR.net) protocol I study (which included patients with comparable baseline demographics to RESTORE).1 A proportionately smaller number of monitoring visits was therefore assumed in year 2. After year 2, laser therapy was assumed to be administered as required in all arms, with no further need for ranibizumab; the assumed number of monitoring visits was further reduced accordingly.

The average BCVA achieved in year 1 was assumed to be maintained during year 2, as was observed in the DRCR.net protocol I study.1 After year 2, all arms of the model followed natural disease history based on 4‐year health state transition outcomes modelled from the Wisconsin Epidemiologic Study of Diabetic Retinopathy (WESDR) reports, the Diabetes Control and Complications Trial and UK Prospective Diabetes Study. Transition probabilities were calibrated to adjust for the improvement in diabetes management since the WESDR reports, and predicted that around 30% of patients would be expected to exhibit a worsening in BCVA of at least 10 letters and 20% of patients would show an improvement of at least 10 letters over a 4‐year time horizon. […]

Mortality was estimated by adjusting general UK population death rates according to the increased RR of death in patients with DME. Mulnier et al estimated an increased mortality (HR 1.93) in a UK type 2 diabetes population relative to patients without diabetes, while Hirai et al estimated an HR of 1.27 for death in patients with clinically significant macular oedema (CSME) and diabetes relative to diabetic patients without CSME. We calculated a 2.45 RR of death in a DME population by multiplying these two ratios.

 

Utility scores were calculated based on patient‐reported outcomes data from RESTORE […], in which patients completed the EuroQoL (EQ‐5D) questionnaire at baseline and months 3, 6 and 12. Individual EQ‐5D health scores were converted into utility scores using preferences from a UK population survey; mean utility scores were calculated for each health state […]. As these states were defined by BCVA in the treated eye, of which 67.2% were the worse‐seeing eye at baseline, this method established an association between utility and BCVA changes in the treated eye.

 

Health state costs included the costs of treatment and monitoring (Supplementary tables 3–6), and the costs associated with blindness (Supplementary table 2). Treatment costs included the costs of ranibizumab (Novartis UK, personal communication) and its administration, laser therapy and investigative procedures. Monitoring costs, including consultation and procedure costs, were estimated from the UK National Health Service Reference Costs. Costs of blindness included those incurred by the UK National Health Service for items such as low‐vision aids, low‐vision rehabilitation, residential or home care, depression and hip fracture/replacement as listed in the costing study by Meads and Hyde. Where older cost estimates were used, these were inflated to 2010 prices using the Hospital and Community Health Services index.26 The cost of blindness would be incurred only in patients reaching health states with BCVA ≤35 letters (Snellen ≤6/60) in the better‐seeing eye. However, as the study assesses treatment response according to the enrolled eye, the proportion of patients reaching this level within the time horizon of the model is therefore uncertain. As such, the base case model adjusts for the cost of blindness on the basis of treated eyes reaching the BCVA ≤35‐letter threshold. As with other model parameters subject to uncertainty, deviations from this assumption were explored in sensitivity analyses (Supplementary table 7).

 

[…] An annual 3.5% discount rate was applied for future costs and utilities, consistent with the standard UK approach.

Effectiveness data

 

RESTORE, DRCRnet

 

Health state valuations (utilities)

 

See above.

 

Unit cost data, price year, resource use data

 

 

Cost of treatment

Ranibizumab injection (0.5 mg vial [x1]) ) £742.17, 7 injections year 1, 3 in year 2.  Novartis UK, personal communication.

Laser treatment per session (weighted average of day cases and outpatient procedures for vitreous retinal procedures category 1) £274.19, 2 treatments year1, 1 in year 2. NHS Reference Costs 2008–09 – NHS Trusts and PCTs  combined (unless otherwise stated).

Ophthalmologist visit (weighted first  attendance and follow‐up attendance)  £84.42, 12 visits ranibizumab, 5 visits laser both in year 1 and 2. NHS Reference Costs 2008–09 – NHS Trusts and PCTs  combined (unless otherwise stated)  Additional ophthalmologist visit £73.16.  

Pre‐injection VA and BCVA assessment  (first attendance for ophthalomology non‐consultant‐led, non‐admitted visit)   £83.97 NHS Reference Costs 2008–09 – NHS Trusts and PCTs  combined (unless otherwise stated)

Optometrist visit (follow‐up attendance for  ophthalomology non‐consultant‐led, non‐admitted visit) £60.92, 12 visits ranibizumab, 5 visits laser. NHS Reference Costs 2008–09 – NHS Trusts and PCTs  combined (unless otherwise stated).

VA and BCVA checks £55.59 NHS Reference Costs 2008–09 – NHS Trusts and PCTs.

combined (unless otherwise stated).

Cost of low vision

Low vision aids annual cost £194.16 £ in 33% of blind patients Inflated to base year 2008–09.

Low vision rehabilitation (occupational health therapist): annual cost £221.00 in 11% of blind patients. Section 7.2: NHS community  occupational therapist.

Residential care (homecare) – 30% private payer: annual cost £16,998.80 in 30% blind patients. Section 1.2: Private residential  care for older people: fees (A) only.

Community care £12,064.00 annual cost in 6% of blind patients. £723.84 Section 9.5: Local authority  home care worker.

Depression annual cost: £558.24in 39% blind patients. Inflated to base year 2008–09.

Hip replacement annual cost: £6952.93 in 5% blind patients. Weighted average of major hip.

procedures category – 12B and  12C TPCTEI.

 

Discounting

 

3.5%

 

Results and sensitivity analyses

 

Base Case: ICER £24,028.

Discount rate 0‐5% (vs 3.5%): ICER £17,051‐27,042.

Time horizon 10‐20 years (vs 15 years): ICER £33,139‐21,343.

Cost of blindness (base £6477) ‐25%‐+25%: ICER £27,907‐20,150.

Long term progression of visual acuity (vs declining) stable or improved: ICER £26,198‐28,4131.

Total n. ranibizumab injections (vs 10) 6‐14: ICER £12,446‐38,836.

Baseline age (vs 63 years) 58 years: ICER £19,259.

Source of utilities (vs RESTORE) Brazier: ICER £21,953.

 

 

Figuras y tablas -
Table 13. Economic evaluations: Mitchell 2012
Table 14. Economic evaluations: Philips 2004 checklist

QUALITY CRITERION: question

Study

Response

Comments

STRUCTURE: Statement of decision problem/objective

Is there a clear statement of the decision problem?

NICE 2011

Yes

Clinical effectiveness and cost effectiveness of ranibizumab monotherapy and ranibizumab in combination with laser photocoagulation compared with laser photocoagulation alone.

Mitchell 2012

Yes

Is the objective of the evaluation and model specified and consistent with the stated decision problem?

NICE 2011

Yes

See above.

Mitchell 2012

Yes

Is the primary decision‐maker specified?

NICE 2011

Yes

Not reported, but models submitted to NICE should take the perspective of the health service provider.

Mitchell 2012

Yes

UK healthcare payer perspective.

STRUCTURE: Statement of scope/perspective

Is the perspective of the model stated clearly?

 

NICE 2011

Yes

Ranibizumab manufacturer’s submission to NICE including an effectiveness review of two RCTs (RESTORE and DRCRnet), costs of treatment and visits and of some of its complications, as well as costs of low vision. Bevacizumab was not included in the submission but was considered a comparator by the Appraisal Committee.

Mitchell 2012

Yes

Manufacturer’s sponsored study on cost‐effectiveness of ranibizumab adopting similar model boundaries as NICE 2011. Bevacizumab not mentioned as a potential comparator.

Are the model inputs consistent with the stated perspective?

NICE 2011

Yes

See above.

Mitchell 2012

Yes

See above.

Has the scope of the model been stated and justified?

NICE 2011

Yes

See above.

Mitchell 2012

Yes

See above.

Are the outcomes of the model consistent with the perspective, scope and overall objective of the model?

NICE 2011

Yes

Utility‐based health‐related quality‐of‐life associated to visual acuity data from RCTs and costs of disease and treatment.

Mitchell 2012

Yes

STRUCTURE: Rationale for structure

Is the structure of the model consistent with a coherent theory of the health condition under evaluation?

NICE 2011

Yes

Health states and transition probabilities based on changes of best corrected visual acuity changes, although treatment of better‐seeing vs. worse eye or both eyes is a problem.

Mitchell 2012

Yes

Are the sources of data used to develop the structure of the model specified?

NICE 2011

Yes

EQ‐5D data from RESTORE related to visual acuity, NHS reference cost of treatment and visits. Cost of low vision from a published cost study.

Mitchell 2012

Yes

Same as above, plus Wisconsin study (WESDR) data for natural disease history.

Are the causal relationships described by the model structure justified appropriately?

NICE 2011

Unclear

Treatment effect and adverse events are based on RCTs, but costs, and not utility decrement, related to adverse events considered. Although adverse events associated with the treatments were low, unclear if they impact on cost‐effectiveness.

Mitchell 2012

No

Adverse events assumed to have negligible impact on cost‐effectiveness, but no demonstration given.

STRUCTURE: Structural assumptions

Are the structural assumptions transparent and justified?

NICE 2011

No

See Table 12. We accept the criticism of the Appraisal committee, including the fact that treatment in better‐seeing, worse‐seeing eye or both eyes s not considered.

Mitchell 2012

No

Same as for NICE 2011.

Are the structural assumptions reasonable given the overall objective, perspective and scope of the model?

 

NICE 2011

No

See above.

Mitchell 2012

No

STRUCTURE: Strategies/comparators

Is there a clear definition of the options under evaluation?

NICE 2011

Yes

The manufacturer considered bevacizumab as a potential comparator but concluded that significant methodological and clinical differences between studies precluded a valid analysis.

Mitchell 2012

No

Options under evaluation not discussed.

Have all feasible and practical options been evaluated?

NICE 2011

No

See above. Bevacizumab is used off‐label for treatment of DMO in several countries.

Mitchell 2012

No

See above.

Is there justification for the exclusion of feasible options?

NICE 2011

No

See above.

Mitchell 2012

No

See above.

STRUCTURE: Model type

Is the chosen model type appropriate given the decision problem and specified causal relationships within the model?

NICE 2011

Yes

Appropriate given what reported above.

Mitchell 2012

Yes

STRUCTURE: Time horizon

Is the time horizon of the model sufficient to reflect all important differences between options?

NICE 2011

Yes

15‐year horizon.

Mitchell 2012

Yes

Are the time horizon of the model, the duration of treatment and  the duration of treatment effect described and justified?

NICE 2011

No

The model’s assumption that the relative benefit achieved during the treatment phase lasts indefinitely is unrealistic.

Mitchell 2012

No

STRUCTURE: Disease states/pathways

Do the disease states (state transition model) or the pathways (decision tree model) reflect the underlying biological process of the disease in question and the impact of interventions?

NICE 2011

Yes

Course of visual acuity change after treatment for diabetic macular oedema.

Mitchell 2012

Yes

STRUCTURE: Cycle length

Is the cycle length defined and justified in terms of the natural history of disease?

NICE 2011

Yes

3‐monthly cycles.

Mitchell 2012

Yes

DATA: Data identification

Are the data identification methods transparent and appropriate given the objectives of the model?

NICE 2011

Unclear

A discussion of the choices of the sources is given but a systematic search is not mentioned.

Mitchell 2012

No

No discussion of the choices of the sources.

Where choices have been made between data sources, are these justified appropriately?

 

NICE 2011

Yes

RESTORE and DRCRnet data used. The choice of excluding RESOLVE and READ2 is discussed.

Mitchell 2012

No

No discussion of the choices of the sources.

Has particular attention been paid to identifying data for the important parameters in the model?

NICE 2011

Yes

Model revised following ERG comments considered all relevant parameters.

Mitchell 2012

Yes

We believe all the important parameters have been considered.

Has the quality of the data been assessed appropriately?

 

NICE 2011

Yes

Unclear, but NICE comment, quoted above, uses standard quality assessments methods, and assessments correspond to our review.

Mitchell 2012

No

No discussion of data quality.

Where expert opinion has been used, are the methods described and justified?

 

NICE 2011

NA

 

Mitchell 2012

NA

 

DATA: Data modelling

Is the data modelling methodology based on justifiable statistical and epidemiological techniques?

NICE 2011

Yes

Markov model using RCT data.

 

Mitchell 2012

 Yes

DATA: Data modelling ‐ Baseline data

Is the choice of baseline data described and justified?

NICE 2011

Yes

See above: data from two RCTs.

Mitchell 2012

Yes

Are transition probabilities calculated appropriately?

NICE 2011

Yes

Extraction of transition probabilities were revised and agreed with NICE, see Table 12.

Mitchell 2012

Unclear

Insufficient details.

Has a half‐cycle correction been applied to both cost and outcome?

NICE 2011

Unclear

Not specified.

Mitchell 2012

Yes

If not, has this omission been justified?

NICE 2011

NA

 

Mitchell 2012

NA

 

DATA: Data modelling ‐ Treatment effect

If relative treatment effects have been derived from trial data, have they been synthesised using appropriate techniques? 

NICE 2011

NA

Effects from individual studies.

Mitchell 2012

NA

Have the methods and assumptions used to extrapolate short‐term results to final outcomes been documented and justified?

NICE 2011

Yes

Explanation given: no change in treatment effect during year 2, according to DRCRnet, and from year 2 to 15. Mortality data from UK‐based literature (2.45 RR for diabetics with DMO).

Mitchell 2012

Yes

Same as NICE 2011, plus decline of visual acuity after year 2 modelled according to Wisconsin study (WESDR), the Diabetes Control and Complications Trial and UK Prospective Diabetes Study.

Have alternative assumptions been explored through sensitivity analysis?

NICE 2011

Yes

The manufacturer presented a series of deterministic sensitivity analyses in which parameters were varied across plausible ranges, including: number of ranibizumab injections during year 3 and 4;  retreatment need and treatment of both eyes in 35% of patients.

Mitchell 2012

No

Sensitivity analyses on number of injections performed. Sensitivity analysis regarding treatment need after 2 years and treatment in better vs. worse seeing eye or both eyes not presented.

Have assumptions regarding the continuing effect of treatment once treatment is complete been documented and justified? Have alternative assumptions been explored through sensitivity analysis?

NICE 2011

No

See previous comments and Table 12.

Mitchell 2012

No

See previous comments and Table 12.

DATA: Data modelling ‐ Costs

Are the costs incorporated into the model justified?

NICE 2011

Yes

All sources of costs considered, UK setting. Cost of adverse events not considered but suggested to be minimal due to the very low rate.

Mitchell 2012

Yes

Has the source for all costs been described?

NICE 2011

Yes

See Table 12.

Mitchell 2012

Yes

See Table 10.

Have discount rates been described and justified given the target decision‐maker?

NICE 2011

Unclear

Cannot find it in the report.

Mitchell 2012

Yes

3.5% discount rate.

DATA: Data modelling ‐ Quality of life weights (utilities)

Are the utilities incorporated into the model appropriate?

NICE 2011

No

Utility gain for treatment in the better‐seeing vs. worse‐seeing eye or in both eyes still a problem in the revised model.

Mitchell 2012

No

Utility gain for treatment in the better‐seeing vs. worse‐seeing eye or in both eyes still not considered.

Is the source for the utility weights referenced?

NICE 2011

Yes

EQ‐5D data from RESTORE were transformed to utility values using standard social tariffs and then related to BCVA in the treated eye using linear regression. Covariate adjustment used in revised submission. sensitivity analyses based on other sources available.

Mitchell 2012

Yes

EQ‐5D data from RESTORE were transformed to utility values using standard social tariffs and then related to BCVA in the treated eye using linear regression.

Are the methods of derivation for the utility weights justified?

NICE 2011

No

See above.

Mitchell 2012

No

See above.

DATA: Data incorporation

Have all data incorporated into the model been described and referenced in sufficient detail?

NICE 2011

Yes

 

Mitchell 2012

Yes

 

Has the use of mutually inconsistent data been justified (i.e. are assumptions and choices appropriate)?

NICE 2011

NA

Sources were specific to each type of data and no calibration was possible.

Mitchell 2012

NA

Is the process of data incorporation transparent?

NICE 2011

Yes

Use of data from RCT described.

Mitchell 2012

Yes

Use of data from RCT  and population‐based study described.

If data have been incorporated as distributions, has the choice of distribution for each parameter been described and justified?

NICE 2011

NA

Individual data from RCTs used to estimate parameters.

Mitchell 2012

NA

If data have been incorporated as distributions, is it clear that second order uncertainty is reflected?

 

NICE 2011

NA

 

Mitchell 2012

NA

 

DATA: Assessment of uncertainty

Have the four principal types of uncertainty been addressed? 

NICE 2011

No

See below

 

Mitchell 2012

No

If not, has the omission of particular forms of uncertainty been justified?

NICE 2011

No

 

Mitchell 2012

No

 

DATA: Assessment of uncertainty: Methodological

Have methodological uncertainties been addressed by running alternative versions of the model with different methodological assumptions?

NICE 2011

No

Single model structure as far as reported in NICE document

Mitchell 2012

No

Single model structure.

DATA: Assessment of uncertainty: Structural

Is there evidence that structural uncertainties have been addressed via sensitivity analysis?

NICE 2011

Yes

Sensitivity analyses presented.

Mitchell 2012

Yes

DATA: Assessment of uncertainty: Heterogeneity

Has heterogeneity been dealt with by running the model separately for different subgroups?

NICE 2011

Yes/Unclear

The manufacturer’s subgroup analysis suggesting that ranibizumab has a favourable cost‐effectiveness profile in people with a thicker retina (central foveal thickness greater than 400 micrometres), but The Appraisal Committee concluded that it could not consider this subgroup analysis sufficiently robust to support separate recommendations to the NHS because of small sample sizes.

Mitchell 2012

No

No subgroup analyses reported.

DATA: Assessment of uncertainty: Parameter

Are the methods of assessment of parameter uncertainty appropriate?

NICE 2011

Yes

One‐way sensitivity analyses of point estimates of parameters.

Mitchell 2012

Yes

If data are incorporated as point estimates, are the ranges used for sensitivity analysis stated clearly and justified?

 

NICE 2011

Yes

 

Mitchell 2012

Yes

 

CONSISTENCY: Internal consistency

Is there evidence that the mathematical logic of the model has been tested thoroughly before use?

NICE 2011

Unclear

Not reported.

Mitchell 2012

Unclear

Not reported.

CONSISTENCY: External consistency

Are any counterintuitive results from the model explained and justified?

NICE 2011

Unclear

No such results.

Mitchell 2012

Unclear

If the model has been calibrated against independent data, have any differences been explained and justified?

 

NICE 2011

NA

No such calibration.

Mitchell 2012

NA

Have the results of the model been compared with those of previous models and any differences in results explained?

NICE 2011

NA

No previous models.

Mitchell 2012

No

No comparison with NICE 2011, published in July 2011 and available to manufacturer.

BCVA: best‐corrected visual acuity

Figuras y tablas -
Table 14. Economic evaluations: Philips 2004 checklist
Table 15. Comparison of NICE 2011 and Mitchell 2012

NICE 2011

 Mitchell 2012

Cost of resources

 

 

Ranibizumab– drug cost

£761.20

£742.17

Ranibizumab injection – procedure cost

£150

 

Injections year 1‐2

10

10

Injections after year 2

3

0

Laser photocoagulation – procedure cost

£150

£274.19

Procedures year 1‐2

3

3

Procedures after year 2

2

0

Ophthalmologist visit for ranibizumab plus any  injection

£126

£84.42

Visits year 1‐2

22

22

Ophthalmologist visit for laser

£126

£84.42

Visits year 1‐2

8

8

Visits after year 2

4

4

Cost of disease consequences

 

 

Low vision total cost ‐  1st year

£6067

 

Low vision total cost ‐ 2nd year

£5936

 

Low‐vision aids (annual, 33% blind patients)

 

£194.16

Residential care (private, 30% blind patients)

 

£16,998.80

Community Care (annual, 6% blind patients)

 

£12,064

Depression (annual, 39% blind patients)

 

£558.24

Hip replacement (5% blind patients)

 

£6952.93

Mortality of patients with DMO (vs. age‐sex matched)

RR: 2.45

RR: 2.45

Utility source

RESTORE

RESTORE

Time horizon

15 years

15 years

Discount rate

unclear

3.5%

ICER (cost per QALY) ‐ base‐case and main sensitivity analyses

 

 

Base‐case

£30,277

£24,028

Bilateral treatment 35%

£44,400

 

Covariate adjusted utility estimation

£33,857

 

Utility source: Lloyd

£24,779

£19,238

Uitlity source: Brazier

£23,664

 

Utility source: Brown

 

£21,953

Discount rate 0‐5% (vs 3.5%): ICER £17,051‐27,042

 

 

Total n. ranibizumab injections (vs 10) 14 ‐ 6

 

£38,836 ‐12,446

Cost of blindness (base £6477) ‐25%‐+25%: ICER

 

£27,907‐20,150

Long term progression of visual acuity (vs declining) stable or improved

 

£26,198‐28,4131

Baseline age (vs 63 years) 58 years

 

£19,259

Number of visits with laser equal to ranibizumab

£37,673

 

Number of visits with ranibizumab equal to laser

£33,074

 

*ICER values are presented in cost per QALY units

Figuras y tablas -
Table 15. Comparison of NICE 2011 and Mitchell 2012
Comparison 1. Anti‐VEGF versus laser

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 1 year Show forest plot

5

556

Risk Ratio (M‐H, Fixed, 95% CI)

2.76 [2.02, 3.76]

1.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

3.06 [1.54, 6.05]

1.2 Ranibizumab

2

300

Risk Ratio (M‐H, Fixed, 95% CI)

3.06 [1.93, 4.87]

1.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.26, 3.51]

2 Gain 3+ lines of visual acuity at 1 year Show forest plot

5

556

Risk Ratio (M‐H, Fixed, 95% CI)

3.20 [2.07, 4.95]

2.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

2.52 [1.20, 5.29]

2.2 Ranibizumab

2

300

Risk Ratio (M‐H, Fixed, 95% CI)

3.51 [1.78, 6.92]

2.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

3.72 [1.52, 9.08]

3 Loss 2+ lines of visual acuity at 1 year Show forest plot

4

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.23 [0.12, 0.44]

3.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.07, 0.52]

3.2 Ranibizumab

1

225

Risk Ratio (M‐H, Fixed, 95% CI)

0.27 [0.09, 0.80]

3.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.05, 1.09]

4 Loss 3+ lines of visual acuity at 1 year Show forest plot

4

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.13 [0.05, 0.34]

4.1 Bevacizumab

2

167

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.05, 0.51]

4.2 Ranibizumab

1

225

Risk Ratio (M‐H, Fixed, 95% CI)

0.11 [0.01, 0.83]

4.3 Aflibercept

1

89

Risk Ratio (M‐H, Fixed, 95% CI)

0.08 [0.00, 1.30]

5 Mean difference in logMAR visual acuity at 1 year Show forest plot

5

554

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.16, ‐0.10]

5.1 Bevacizumab

2

165

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.28, ‐0.12]

5.2 Ranibizumab

2

300

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.14, ‐0.07]

5.3 Aflibercept

1

89

Mean Difference (IV, Fixed, 95% CI)

‐0.27 [‐0.40, ‐0.13]

6 Mean difference in (change of) OCT central macular thickness at 1 year Show forest plot

4

477

Mean Difference (IV, Fixed, 95% CI)

‐60.71 [‐83.87, ‐37.54]

6.1 Bevacizumab

2

165

Mean Difference (IV, Fixed, 95% CI)

‐43.61 [‐82.11, ‐5.11]

6.2 Ranibizumab

1

225

Mean Difference (IV, Fixed, 95% CI)

‐57.40 [‐89.86, ‐24.94]

6.3 Aflibercept

1

87

Mean Difference (IV, Fixed, 95% CI)

‐121.9 [‐186.47, ‐57.33]

7 Mean difference in logMAR visual acuity at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

7.1 Bevacizumab

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Mean difference in (change of) OCT Central Macular Thickness at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

8.1 Bevacizumab

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): gain 3+ Show forest plot

3

380

Risk Ratio (M‐H, Fixed, 95% CI)

3.29 [1.77, 6.13]

10 Sensitivity analysis excluding studies with eyes as unit (Soheillian 2007): loss 3+ Show forest plot

2

305

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.02, 0.41]

Figuras y tablas -
Comparison 1. Anti‐VEGF versus laser
Comparison 2. Anti‐VEGF versus sham

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

2.46 [1.77, 3.40]

1.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.43, 3.09]

1.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

3.31 [1.80, 6.09]

2 Gain 3+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

2.19 [1.36, 3.53]

2.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

1.79 [1.01, 3.16]

2.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

3.17 [1.32, 7.62]

3 Loss 2+ lines of visual acuity at 8 to 12 months Show forest plot

3

497

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.19, 0.60]

3.1 Pegaptanib

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

0.44 [0.21, 0.92]

3.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

0.20 [0.07, 0.54]

4 Loss 3+ lines of visual acuity at 8 to 12 months Show forest plot

2

411

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.13, 0.59]

4.1 Pegaptanib

1

260

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.16, 1.21]

4.2 Ranibizumab

1

151

Risk Ratio (M‐H, Fixed, 95% CI)

0.14 [0.04, 0.50]

5 Mean change of visual acuity at 6 to 12 months Show forest plot

4

575

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.17, ‐0.08]

5.1 Bevacizumab

1

78

Mean Difference (IV, Fixed, 95% CI)

‐0.15 [‐0.26, ‐0.04]

5.2 Pegaptanib

2

346

Mean Difference (IV, Fixed, 95% CI)

‐0.08 [‐0.13, ‐0.03]

5.3 Ranibizumab

1

151

Mean Difference (IV, Fixed, 95% CI)

‐0.23 [‐0.32, ‐0.15]

6 Mean difference in (change of) OCT central macular thickness at 6 to 12 months Show forest plot

3

315

Mean Difference (IV, Fixed, 95% CI)

‐126.38 [‐160.27, ‐92.49]

6.1 Bevacizumab

1

78

Mean Difference (IV, Fixed, 95% CI)

‐130.6 [‐187.27, ‐73.93]

6.2 Pegaptanib

1

86

Mean Difference (IV, Fixed, 95% CI)

‐71.7 [‐149.71, 6.31]

6.3 Ranibizumab

1

151

Mean Difference (IV, Fixed, 95% CI)

‐145.80 [‐196.12, ‐95.48]

7 Gain 2+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

1.99 [1.64, 2.40]

7.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.87, 1.88]

7.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

2.30 [1.85, 2.86]

8 Gain 3+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.85, 3.23]

8.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

1.56 [0.87, 2.78]

8.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

2.80 [2.03, 3.86]

9 Loss 3+ lines of visual acuity at 2 years Show forest plot

2

716

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.17, 0.64]

9.1 Pegaptanib

1

207

Risk Ratio (M‐H, Fixed, 95% CI)

0.42 [0.13, 1.31]

9.2 Ranibizumab

1

509

Risk Ratio (M‐H, Fixed, 95% CI)

0.30 [0.13, 0.68]

10 Loss 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 Ranibizumab

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Mean change of visual acuity at 2 years Show forest plot

2

716

Mean Difference (IV, Fixed, 95% CI)

‐0.16 [‐0.20, ‐0.12]

11.1 Pegaptanib

1

207

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.17, ‐0.02]

11.2 Ranibizumab

1

509

Mean Difference (IV, Fixed, 95% CI)

‐0.19 [‐0.23, ‐0.14]

12 Quality of life (difference change in NEI‐VFQ 25 composite score at 54 weeks) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

13 Quality of life (difference change in NEI‐VFQ 25 composite score at 102 weeks) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. Anti‐VEGF versus sham
Comparison 3. Anti‐VEGF plus laser versus laser alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gain 2+ lines of visual acuity at 1 year Show forest plot

3

1267

Risk Ratio (M‐H, Fixed, 95% CI)

1.94 [1.66, 2.28]

1.1 Prompt photocoagulation

3

786

Risk Ratio (M‐H, Fixed, 95% CI)

2.14 [1.73, 2.64]

1.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.33, 2.15]

2 Gain 3+ lines of visual acuity at 1 year Show forest plot

3

1267

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.67, 2.67]

2.1 Prompt photocoagulation

3

786

Risk Ratio (M‐H, Fixed, 95% CI)

2.29 [1.67, 3.13]

2.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

1.88 [1.31, 2.70]

3 Loss 2+ lines of visual acuity at 1 year Show forest plot

2

1189

Risk Ratio (M‐H, Fixed, 95% CI)

0.26 [0.15, 0.43]

3.1 Prompt photocoagulation

2

708

Risk Ratio (M‐H, Fixed, 95% CI)

0.27 [0.14, 0.51]

3.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.10, 0.56]

4 Loss 3+ lines of visual acuity at 1 year Show forest plot

2

1189

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.15, 0.55]

4.1 Prompt photocoagulation

2

708

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.13, 0.67]

4.2 Deferred photocoagulation

1

481

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.10, 0.77]

5 Mean difference in change of logMAR visual acuity at 6 to 12 months Show forest plot

3

1266

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.13, ‐0.08]

5.1 Prompt photocoagulation

3

785

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.13, ‐0.07]

5.2 Deferred photocoagulation

1

481

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.17, ‐0.07]

6 Mean difference in change of OCT central macular thickness at 6 to 12 months Show forest plot

2

1116

Mean Difference (IV, Fixed, 95% CI)

‐40.90 [‐57.19, ‐24.62]

6.1 Prompt photocoagulation

2

670

Mean Difference (IV, Fixed, 95% CI)

‐44.28 [‐64.69, ‐23.86]

6.2 Deferred photocoagulation

1

446

Mean Difference (IV, Fixed, 95% CI)

‐35.0 [‐62.00, ‐6.00]

7 Gain 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Gain 3+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Loss 2+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

9.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

10 Loss 3+ lines of visual acuity at 2 years Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 Prompt photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Deferred photocoagulation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Mean difference in change of logMAR visual acuity at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

11.1 Prompt photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Deferred photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Mean difference in change of OCT central macular thickness at 2 years Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

12.1 Prompt photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Deferred photocoagulation

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 3. Anti‐VEGF plus laser versus laser alone
Comparison 4. Adverse events: Anti‐VEGF versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total ATC thromboembolic events at 6 to 24 months Show forest plot

9

2159

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.56, 1.28]

1.1 Follow‐up 6 to 12 months

6

868

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.49, 4.06]

1.2 Follow‐up 24 months

3

1291

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.42, 1.53]

2 Death Show forest plot

9

2159

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.52, 1.74]

2.1 Follow‐up 6 to 12 months

6

868

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.24, 2.83]

2.2 Follow‐up 24 months

3

1291

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.36, 3.45]

Figuras y tablas -
Comparison 4. Adverse events: Anti‐VEGF versus control