Scolaris Content Display Scolaris Content Display

تاثیر بورتزومیب (bortezomib) در درمان مالتیپل میلوما

Contraer todo Desplegar todo

Referencias

All India Institute Study {published data only}

Sahai S, Kumar L, Raina V, Sharma A, Gupta R, Mahajan S, et al. Multiple myeloma with light chain‐induced acute renal failure ‐ role of bortezomib‐dexamethasone & cyclophosphamide‐thalidomide & dexamethasone: a prospective randomized study (Abstract 3648). Proceedings of the European Cancer Congress. Amsterdam, 27 Sep ‐ 01 Oct 2013.

APEX Study {published data only}

Chanan‐Khan A, Sonneveld P, Schuster MW, Stadtmauer EA, Facon T, Harousseau JL, et al. Analysis of herpes zoster events among bortezomib‐treated patients in the phase III APEX study. Journal of Clinical Oncology 2008;26(29):4784‐90.
Lee SJ, Richardson PG, Sonneveld P, Schuster MW, Irwin D, San Miguel JF, et al. Bortezomib is associated with better health‐related quality of life than high‐dose dexamethasone in patients with relapsed multiple myeloma: results from the APEX study. British Journal of Haematology 2008;143(4):511‐9.
Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer EA, Facon T, et al. Bortezomib or high‐dose dexamethasone for relapsed multiple myeloma. New England Journal of Medicine 2005;352(24):2487‐98.
Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer EA, Facon T, et al. Extended follow‐up of a phase 3 trial in relapsed multiple myeloma: final time‐to‐event results of the APEX trial. Blood 2007;110(10):3557‐60.

CREST Study {published data only}

Jagannath S, Barlogie B, Berenson J, Siegel D, Irwin D, Richardson PG, et al. A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. British Journal of Haematology 2004;127(2):165‐72.
Jagannath S, Barlogie B, Berenson JR, Siegel DS, Irwin D, Richardson PG, et al. Updated survival analyses after prolonged follow‐up of the phase 2, multicenter CREST study of bortezomib in relapsed or refractory multiple myeloma. British Journal of Haematology 2008;143(4):537‐40.

GEM05MENOS65 Study {published data only}

Rosinol L, Oriol A, Teruel AI, Hernandez D, Lopez‐Jiminez J, de la Rubia J, et al. Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood 2012;120(8):1589‐96.
Rosinol L, Oriol A, Teruel AL, Hernandez D, Lopez‐Jimenez J, De La Rubia J, et al. Maintenance therapy after stem‐cell transplantation for multiple myeloma with bortezomib/thalidomide vs.thalidomide vs. alfa2b‐interferon: final results of a phase iii pethema/gem randomized trial. Blood (ASH Annual Meeting Abstracts) Abstract 334. 2012; Vol. 120, issue 21.

GEM2010MAS65 Study {published data only}

Mateos MV, Martinez‐Lopez J, Hernandez MT, Martinez R, Rosinnol L, Ocio EM, et al. Comparison of sequential vs alternating administration of bortezomib, melphalan and prednisone (VMP) and lenalidomide plus dexamethasone (Rd) in elderly patients with newly diagnosed multiple myeloma (MM) patients: GEM2010MAS65 trial. Blood (ASH Annual Meeting Abstracts) Abstract 178. 2014; Vol. 124.
Mateos MV, Martinez‐Lopez J, Hernandez MT, Martinez R, Rosinnol L, Ocio EM, et al. Comparison of sequential vs alternating administration of bortezomib, melphalan and prednisone (VMP) and lenalidomide plus dexamethasone (Rd) in elderly patients with newly diagnosed multiple myeloma (MM) patients: GEM2010MAS65 trial. Blood (ASH Annual Meeting Abstracts) Abstract 403. 2013; Vol. 122, issue 21.

GIMEMA‐MM‐03‐05 Study {published data only}

Palumbo A, Bringhen S, De Paoli L, Cotzia E, Ria R, Gentili S, et al. Bortezomib‐melphalan‐prednisone‐thalidomide followed by continuous bortezomib thalidomide (VMPT‐VT) improved survival. Clinical Lymphoma, Myeloma and Leukemia 2013;13:S106‐107.
Palumbo A, Bringhen S, Larocca A, Rossi D, Di Raimondo F, Magarotto V, et al. Bortezomib‐melphalan‐prednisone‐thalidomide followed by maintenance with bortezomib‐thalidomide compared with bortezomib‐melphalan‐prednisone for initial treatment of multiple myeloma: updated follow‐up and improved survival. Journal of Clinical Oncology 2014;32(7):634‐40.
Palumbo A, Bringhen S, Rossi D, Cavalli M, Larocca A, Ria R, et al. Bortezomib‐melphalan‐prednisone‐thalidomide followed by maintenance with bortezomib‐thalidomide compared with bortezomib‐melphalan‐prednisone for initial treatment of multiple myeloma: a randomized controlled trial. Journal of Clinical Oncology 2010;28(34):5101‐9.

GIMEMA‐MMY‐3006 Study {published data only}

Cavo M, Galli M, Pantani L, Di Raimondo F, Crippa C, Offidani M, et al. Bortezomib‐thalidomide‐dexamethasone incorporated into autotransplantation is associated with more favorable outcomes after relapse in comparison with thalidomide‐dexamethasone plus autotransplantation in multiple myeloma. Blood (ASH Annual Meeting Abstracts) Abstract 4210. 2012; Vol. 120.
Cavo M, Galli M, Pezzi A, Di Raimondo F, Crippa C, Offidani M, et al. Persistent improvement in clinical outcomes with bortezomib‐thalidomide‐dexamethasone vs thalidomide‐dexamethasone incorporated into double autologous transplantation for multiple myeloma: an updated analysis of phase 3 Gimema‐MMY‐3006 study. Blood (ASH Annual Meeting Abstracts) Abstract 2090. 2013; Vol. 122, issue 21.
Cavo M, Pantani L, Petrucci MT, Patriarca F, Zamagni E, Donnarumma D, et al. Bortezomib‐thalidomide‐dexamethasone is superior to thalidomide‐dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma. Blood 2012;120(1):9‐19.
Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem‐cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet 2010;376(9758):2075‐85.

HOVON‐65/GMMG‐HD4 Study {published data only}

Sonneveld P, Scheid C, van der Holt B, el Jarari L, Bertsch U, Salwender H, et al. Bortezomib induction and maintenance treatment improves survival in patients with newly diagnosed multiple myeloma: extended follow‐up of the HOVON‐65/GMMG‐HD4 trial. Blood (ASH Annual Meeting Abstracts) Abstract 404. 2013; Vol. 122, issue 21.
Sonneveld P, Schmidt‐Wolf IGH, van der Holt B, El Jarari L, Bertsch U, Salwender H, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON‐65/ GMMG‐HD4 trial. Journal of Clinical Oncology 2012;30(24):2946‐55.

IFM 2005‐01 Study {published data only}

Harousseau JL, Attal M, Avet‐Loiseau H, Marit G, Caillot D, Mohty M, et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem‐cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005‐01 phase III trial. Journal of Clinical Oncology 2010;28(30):4621‐9.

IFM 2007‐02 Study {published data only}

Moreau P, Avet‐Loiseau H, Facon T, Attal M, Tiab M, Hulin C, et al. Bortezomib plus dexamethasone versus reduced‐dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood 2011;118(22):5752‐8.

MD Anderson Study {published data only}

Sharma M, Khan H, Thall PF, Orlowski RZ, Bassett Jr RL, Shah N, et al. A randomized phase 2 trial of a preparative regimen of bortezomib, high‐dose melphalan, arsenic trioxide, and ascorbic acid. Cancer 2012;118(9):2507‐15.

MMVAR/IFM 2005‐04 Study {published data only}

Garderet L, Iacobelli S, Moreau P, Mamoun D, Lafon I, Niederwieser D, et al. Superiority of the triple combination of bortezomib‐thalidomide‐dexamethasone over the dual combination of thalidomide‐dexamethasone in patients with multiple myeloma progressing or relapsing after autologous transplantation: the MMVAR/IFM 2005‐04 Randomized Phase III Trial from the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Journal of Clinical Oncology 2012;30(20):2475‐82.

MMY‐3021 Study {published data only}

Arnulf B, Pylypenko H, Grosicki S, Karamanesht I, Leleu X, van de Velde H, et al. Updated survival analysis of a randomized phase III study of subcutaneous versus intravenous bortezomib in patients with relapsed multiple myeloma. Haematologica 2012;97(12):1925‐8.
Moreau P, Pylypenko H, Grosicki S, Karamanesht I, Leleu X, Grishunina M, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non‐inferiority study. Lancet Oncology 2011;12(5):431‐40.

NMSG 15/05 Study {published data only}

Mellqvist UH, Gimsing P, Hjertner O, Lenhoff S, Laane E, Remes K, et al. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood 2013;121(23):4647‐54.

NMSG 17/07 Study {published data only}

Hjorth M, Hjertner O, Knudsen LM, Gulbrandsen N, Homberg E, Pedersen PT, et al. Thalidomide and dexamethasone vs. bortezomib and dexamethasone for melphalan refractory myeloma: a randomized study. European Journal of Haematology 2012;88(6):485‐96.

VISTA Study {published data only}

Delforge M, Dhawan R, Robinson D, Meunier J, Regnault A, Esseltine DL, et al. Health‐related quality of life in elderly, newly diagnosed multiple myeloma patients treated with VMP vs. MP: results from the VISTA trial. European Journal of Haematology 2012;89(1):16‐27.
Mateos MV, Richardson PG, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, et al. Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow‐up and impact of subsequent therapy in the phase III VISTA trial. Journal of Clinical Oncology 2010;28(13):2259‐66.
San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. New England Journal of Medicine 2008;359(9):906‐17.
San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Persistent overall survival benefit and no increased risk of second malignancies with bortezomib‐melphalan‐prednisone versus melphalan‐prednisone in patients with previously untreated multiple myeloma. Journal of Clinical Oncology 2013;31(4):448‐55.
Spicka I, Mateos MV, Redman K, Dimopoulos MA, Richardson PG. An overview of the VISTA trial: newly diagnosed, untreated patients with multiple myeloma ineligible for stem cell transplantation. Immunotherapy 2011;3(9):1033‐40.

Chen 2011b {published data only}

Chen RA, Tu Y, Cao Y, Liu L, Liang Y. Bortezomib‐dexamethasone or vincristine‐doxorubicin‐dexamethasone as induction therapy followed by thalidomide as maintenance therapy in untreated multiple myeloma patients. Journal of International Medical Research 2011;39(5):1975‐84.

Goldschmidt 2012 {published data only}

Goldschmidt H, Salwender H, Bertsch U, et al. GMMG MM5 trial in newly diagnosed multiple myeloma to evaluate PAD vs VCD induction prior to HDT followed by lenalidomide consolidation and maintenance ‐ first interim analysis on induction. Haematologica 2012;97(115):Abstract 285.

Kumar 2012 {published data only}

Kumar S, Flinn I, Richardson PG, Hari P, Callander N, Noga SJ, et al. Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood 2012;119(19):4375‐82.

Mateos 2010 {published data only}

Mateos MV, Oriol A, Martínez‐López J, Gutiérrez N, Teruel AI, De Paz R, et al. Bortezomib, melphalan, and prednisone versus bortezomib, thalidomide, and prednisone as induction therapy followed by maintenance treatment with bortezomib and thalidomide versus bortezomib and prednisone in elderly patients with untreated multiple myeloma: a randomised trial. Lancet Oncology 2010;11(10):934‐41.

Niesvisky 2010 {published data only}

Niesvizky R, Flinn IW, Rifkin RM, Gabrail NY, Charu V, et al. Phase 3b UPFRONT study: safety and efficacy of weekly bortezomib maintenance therapy after bortezomib‐based induction regimens in elderly, newly diagnosed multiple myeloma patients. American Society of Hematology. 2010.

Orlowski 2007 {published data only}

Orlowski R, Nagler A, Sonneveld P, Bladé J, Hajek R, Spencer A, et al. Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression. Journal of Clinical Oncology 2007;25(25):3892‐901.

Mayo Clinic Study {unpublished data only}

 

Velcade Consolidation Bone Study {unpublished data only}

 

CLARION Study {unpublished data only}

CLARION Study. Ongoing study2013.

Consolidation (61‐75 years) Study {unpublished data only}

CR006127 Study. Ongoing study2006.

Consolidation (less than 60 years) Study {unpublished data only}

CR006124 Study. Ongoing study2006.

E1A11 Study {unpublished data only}

ECOG E1A11 Study. Ongoing study2013.

ENDEAVOR Study {unpublished data only}

ENDEAVOR Study. Ongoing study2012.

Hackensack University Study {unpublished data only}

PRO# 1307 Study. Ongoing study2010.

HOVON 95 Study {unpublished data only}

HOVON 95 Study. Ongoing study2011.

King Fasail Hospital Study {unpublished data only}

2081‐113 Study. Ongoing study2009.

Optimized Retreatment Study {unpublished data only}

CR018796 Study. Ongoing study2013.

Subcutaneous Bortezomib Maintenance Study {unpublished data only}

Subcutaneous bortezomib maintenance Study. Ongoing study2013.

SWOG‐S0777 Study {unpublished data only}

SWOG‐S0777 Study. Ongoing study2008.

VCAT Study {unpublished data only}

CR018751 Study. Ongoing study2012.

Wuerzburg University Hospital Study {unpublished data only}

DSMM XIV Study. Ongoing study2012.

Bertolotti 2008

Bertolotti P, Bilotti E, Colson K, Curran K, Doss D, Faiman B, et al. Management of side effects of novel therapies for multiple myeloma: consensus statements developed by the International Myeloma Foundation's Nurse Leadership Board. Clinical Journal of Oncology Nursing 2008;12(3):9‐12.

Chen 2011a

Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP. Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Current Cancer Drug Targets 2011;11(3):239‐53.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG. Chapter 9: Analysing data and undertaking meta‐analyses. Cochrane Handbook for Systematic Reviews of Interventions. Vol. 5.1.0, http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

EndNote 2012 [Computer program]

Thomson Reuters. EndNote. Version X6. Thomson Reuters, 2012.

Ferlay 2010

Ferlay J, Shin H, Bray F, Forman D, Mathers CD, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. International Journal of Cancer 2010;127:2893‐917.

Higgins 2011a

Higgins JPT, Altman DG, Sterne JAC. Chapter 16: Special topics in statistics. Cochrane Handbook for Systematic Reviews of Interventions. Vol. 5.1.0, Available from http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

Higgins 2011b

Higgins JPT, Altman DG, Sterne JAC. Chapter 8: Assessing risk of bias in included studies. Cochrane Handbook for Systematic Reviews of Interventions. Vol. 5.1.0, Available from http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

Kyle 2009

Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia 2009;23(1):3‐9.

Lawasut 2012

Lawasut P, Chauhan D, Laubach J, Hayes C, Fabre C, Maglio M, et al. New proteasome inhibitors in myeloma. Current Haematologic Malignancy Reports 2012;7:258‐66.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for Studies. Cochrane Handbook for Systematic Review of Interventions. Vol. 5.1.0, Available from http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG. The PRISMA Group Preferred Reporting Items for Systematic Reviews and Meta‐Analyses: The PRISMA Statement. PLOS Medicine 2009;6(7):e1000097.

Moreau 2012

Moreau P, Richardson PG, Cavo M. Proteasome inhibitors in multiple myeloma: 10 years later. Blood 2012;120(5):947‐59.

Nooka 2013

Nooka AK, Kaufman JL, Behera M, Langston A, Waller EK, Flowers CR, et al. Bortezomib‐containing induction regimens in transplant‐eligible myeloma patients: a meta‐analysis of phase 3 randomized clinical trials. Cancer 2013;119(23):4119‐28.

Palumbo 2011

Palumbo A, Anderson KC. Multiple myeloma. New England Journal of Medicine 2011;364:1046‐60.

Parmar 1998

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

Picot 2011

Picot J, Cooper K, Bryant J, Clegg AJ. The clinical effectiveness and cost‐effectiveness of bortezomib and thalidomide in combination regimens with an alkylating agent and a corticosteroid for the first‐line treatment of multiple myeloma: a systematic review and economic evaluation. Health Technology Assessment 2011;15(41):1‐204.

Pulte 2011

Pulte D, Gondos A, Brenner H. Improvement in survival of older adults with multiple myeloma: results of an updated period of analysis of SEER data. Oncologist 2011;16:1600‐03.

Raab 2009

Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma. Lancet 2009;374:324‐39.

RevMan 5.3 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Richardson 2003

Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D. A phase 2 study of bortezomib in relapsed, refractory myeloma. New England Journal of Medicine 2003;348:2609‐17.

Richardson 2005

Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadmauer EA, Facon T, et al. Bortezomib or high‐dose dexamethasone for relapsed multiple myeloma. New England Journal of Medicine 2005;352(24):2487‐98.

Richardson 2007

Richardson PG, Sonneveld P, Schuster P, Irwin D, Stadmauer EA, Facon T, et al. Extended follow‐up of a phase 3 trial in relapsed multiple myeloma: final time‐to‐event results of the APEX trial. Blood 2007;110(10):3557‐60.

Schünemann 2011

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and 'Summary of findings' tables. Cochrane Handbook for Systematic Reviews of Interventions. 5.1.0. http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

Shah 2009

Shah JJ, Orlowski RZ. Proteasome inhibitors in the treatment of multiple myeloma. Leukemia 2009;23:1964‐79.

Siegel 2012

Siegel SG, Martin T, Wang M, Vij R, Jakubowiak AJ, Lonial S, et al. A phase 2 study of single‐agent carfilzomib (PX‐171‐003‐A1) in patients with relapsed and refractory multiple myeloma. Blood 2012;120(14):2817‐25.

Sonneveld 2013

Sonneveld P, Goldschmidt H, Rosinol L, Blade J, Lahuerta JJ, Cavo M, et al. Bortezomib‐based versus nonbortezomib‐based induction treatment before autologous stem‐cell transplantation in patients with previously untreated multiple myeloma: a meta‐analysis of phase III randomized, controlled trials. Journal of Clinical Oncology 2013;31(26):3279‐87.

Sterne 2011

Sterne JAC, Egger M, Moher D. Chapter 10: Addressing reporting biases. Cochrane Handbook for Systematic Reviews of Interventions. Vol. 5.1.0, http://www.cochrane‐handbook.org: The Cochrane Collaboration, 2011.

Tierney 2007

Tierney J, Stewart L, Ghersi D, Burdett S, Sydes M. Practical methods for incorporating summary time‐to‐event data into meta‐analysis. Trials 2007;8:16. [DOI: 10.1186/1745‐6215‐8‐16]

Zeng 2013

Zeng Z, Lin J, Chen J. Bortezomib for patients with previously untreated multiple myeloma: a systematic review and meta‐analysis of randomized controlled trials. Annals of Hematology 2013;92:935‐43.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

All India Institute Study

Methods

  • Design: Randomised open‐label trial conducted at the All India Institute of Medical Science, Delhi, India from Feb 2011 to Sep 2012.

  • Sample size: N = 43 patients. Experimental arm (BD) = 22 patients. Control arm (CTD) = 21 patients.

Participants

  • Patient Population: Multiple myeloma with light‐chain induced acute renal failure.

  • Inclusion/Exclusion Criteria: Not reported.

  • Baseline Characteristics: Not reported.

Interventions

  • Experimental Arm (BD): IV bortezomib 1.3 mg/m2, oral dexamethasone 40 mg both weekly, q 4 weekly.

  • Control Arm (CTD): Oral cyclophosphamide 100 mg/m2 for 7 days, oral thalidomide 100 mg daily and oral dexamethasone 40 mg/week, q 4 weekly.

  • Additional Treatments: Not reported.

Outcomes

  • Renal response and myeloma response according to IMWG Criteria.

Notes

  • Sponsor/Funding: Not reported.

  • Type of Publication (Full text or abstract only): Abstract only.

  • Linked to other reports: None.

  • IMWG = International Myeloma Working Group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study design described as 'randomised' in title however randomisation methods not reported.

Allocation concealment (selection bias)

Unclear risk

Not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective outcomes measured (e.g. response rate and progression according to IMWG guidelines, number of patients alive). Independent blinded outcomes assessment not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Patient status reported at median follow‐up of 7.5 months.

Selective reporting (reporting bias)

High risk

Key eligibility criteria not reported. Baseline characteristics per arm not reported. Selected adverse events reported only.

Other bias

Unclear risk

Not reported.

APEX Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 93 centres in the US, Canada, Europe and Israel from Jun 2002 to Oct 2003.

  • Sample size: N = 669 patients. Experimental arm (bortezomib) = 333 patients. Control arm (high‐dose dexamethasone) = 336 patients.

  • Cross‐over: Patients on the high‐dose dexamethasone were permitted to cross‐over to the bortezomib arm at disease progression.

Participants

  • Patient Population: Multiple myeloma patients with measurable progressive disease after 1 to 3 previous treatments.

  • Inclusion Criteria: KPS) ≥ 60%; platelets ≥ 50,000/mL3; haemoglobin ≥ 7.5g/dL; ANC ≥750/mL3; creatinine clearance 20 mL/min.

  • Exclusion Criteria: Disease refractory to high‐dose dexamethasone; ≥ grade 2 peripheral neuropathy.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Experimental arm: IV bortezomib 1.3 mg/m2 on days 1, 4, 8, 11 of cycles 1 to 8 (21‐day cycle) and days 1,8, 15, 22 of cycles 9 to 11 (35‐day cycle). Maximum treatment period = 273 days.

  • Control arm: Oral dexamethasone 40 mg on days 1 to 4, 9 to 12, 17 to 20 of cycles 1 to 4 (35‐day cycle) and days 1 to 4 of cycles 5 to 9 (28‐day cycles). Maximum treatment period = 280 days.

  • Additional Treatments: Platelet and red‐cell transfusions, neutrophil growth factors and epoetin alfa. IV bisphosphonates unless clinically contraindicated.

Outcomes

  • Primary: Time to disease progression.

  • Secondary: Overall survival; 1‐year survival; response rate (complete plus partial response); duration of response; time to first evidence of confirmed response; time to first infection ≥ grade 3; incidence of ≥ grade 3 infection; time to first skeletal event; adverse events; health‐related quality of life.

Notes

  • Sponsor/Funding: Millennium Pharmaceuticals.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Richardson, 2007 (extended follow‐up data).

  • KPS = Karnofsky Performance Status.

  • ANC = Absolute Neutrophil Count.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not specified. Central randomisation probably performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression by the European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Response data based on central laboratory analysis.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data for the final analyses of the time to disease progression and the response were censored. Full details of censoring reported.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

Not reported.

CREST Study

Methods

  • Design: Exploratory, randomised, open‐label, phase II trial conducted at 10 centres in the US from May 2001 to Jan 2002.

  • Sample size: N = 54 patients. Arm A (1.0 mg/m2 bortezomib) = 28 patients. Arm B (1.3 mg/m2 bortezomib) = 26 patients.

  • Trial not designed to conduct formal statistical comparisons between groups.

Participants

  • Patient Population: Multiple myeloma patients with relapsed/refractory disease who have received front‐line therapy only.

  • Eligibility Criteria: Age ≥ 18 years; KPS) ≥ 60%; life expectancy > 3 months; platelets ≥ 30 x 109/L; haemoglobin ≥ 8 g/dL; ANC ≥ 0.5 x 109/L; creatinine clearance ≥ 30 mL/min; bilirubin ≤ 2 x ULN; AST or ALT ≤ 3 x ULN.

  • Baseline Characteristics: Some imbalances noted. More females, patients with IgG myeloma and patients with abnormal cytogenetics in 1.3 mg/m2 group. More patients with platelets < 75 x 109/L at baseline in 1.0 mg/m2 group.

Interventions

  • Arm A: Bortezomib 1.0 mg/m2 IV days 1, 4, 8, 11 of 21‐day cycle up to 8 cycles.

  • Arm B: Bortezomib 1.3 mg/m2 IV days 1, 4, 8, 11 of 21‐day cycle up to 8 cycles.

  • Additional Treatments: Patients with progressive disease after 2 cycles, or stable disease after 4 cycles were eligible to receive 20 mg oral dexamethasone on the day of, and day following bortezomib treatment.

Outcomes

  • Primary: Overall response rate (ORR): sum of complete response (CR), partial response (PR) and minimal response (MR).

  • Secondary: Response rate to bortezomib in combination with dexamethasone; time to progression on bortezomib alone and in combination with dexamethasone; overall survival; safety.

Notes

  • Sponsor/Funding: Millennium Pharmaceuticals.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Jagannath, 2008 (extended follow‐up data).

  • KPS = Karnofsky Performance Status.

  • ANC = Absolute Neutrophil Count.

  • AST = Aspartate aminotransferase.

  • ALT = Alanine aminotransferase.

  • IgG = Immunoglobulin G.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Randomisation envelopes at each centre selected based on stage of disease and front‐line chemo‐therapeutic regimen. Type of envelope used e.g. opaque and who had access to the envelopes not adequately reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression by the European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Response data assessed by independent review committee of 3 myeloma experts independent of trial.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Patient status at > 5‐year median follow‐up reported.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

Not reported.

GEM05MENOS65 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 66 centres in Spain from Apr 2006 to Aug 2009.

  • Randomisation to one of 3 induction therapy arms (N = 386 patients):

  1. Arm A = VTD (bortezomib, thalidomide, dexamethasone) = 127 patients

  2. Arm B = TD (thalidomide, dexamethasone) = 130 patients

  3. Arm C = VBMCP/VBAD/B (vincristine, BCNU, melphalan, cyclophosphamide, prednisone/vincristine, BCNU, doxorubicin, dexamethasone/bortezomib) = 129 patients

  • Post ASCT, randomisation to one of 3 maintenance therapy arms (N = 266 patients):

  1. Arm A = TV (thalidomide, bortezomib) = 89 patients

  2. Arm B = T (thalidomide only) = 87 patients

  3. Arm C = alfa2‐IFN (Interferon alpha‐2b) = 90 patients

Participants

  • Patient Population: Patients with newly diagnosed and untreated symptomatic multiple myeloma.

  • Inclusion Criteria: Age ≤ 65 years; PS < 3; Platelets ≥ 50x109/L; Haemoglobin ≥ 8 g/dL; ANC ≥ 1x109/L; serum creatinine ≤ 2 mg/dL.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Induction Therapy (24 weeks):

  1. Arm A: VTD: TD as per Arm B and IV bortezomib 1.3 mg/m2 on D1, 4, 8 and 11 of each cycle.

  2. Arm B: TD: Oral thalidomide 200 mg daily (escalating dose first cycle), oral dexamethasone 40 mg D1‐4 and 9‐12 for 6 cycles.

  3. Arm C: VBMCP/VBAD/B: VBMCP and VBAD chemotherapy plus bortezomib for 4 cycles alternating VBMCP and VBAD.

  • Maintenance Therapy (3 years):

  1. Arm A: TV: Oral thalidomide 100 mg per day plus 1 cycle of bortezomib on D1,4,8 and 11 every 3 months.

  2. Arm B: T: Oral thalidomide 100 mg per day.

  3. Arm C: alfa2‐IFN: Interferon alpha‐2b SC 3MU 3 times per week.

  • Additional Treatments: LMW heparin/aspirin for thromboprophylaxis for patients receiving thalidomide, zoledronic acid up to 2 years.

Outcomes

  • Primary: Complete response rate after induction and after ASCT.

  • Secondary: Progression‐free survival, overall survival and safety.

Notes

  • Sponsor/Funding: PETHEMA Foundation supported by 2 grants from Janssen‐Cilag and Pharmion.

  • Type of Publication (Full Text or abstract only): Full text.

  • Linked to other reports: Rosinol, 2012 (maintenance data).

  • PS = Performance Status.

  • ANC = Absolute Neutrophil Count.

  • LMW = Low molecular weight.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Patients were 'centrally randomised'.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate according to the European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Response data and toxicity monitored by an external contract research organisation and centrally reviewed by the principal investigators.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Acceptable rates of withdrawal during induction therapy reported.

Selective reporting (reporting bias)

Unclear risk

Benefits and harms reported, however selected adverse events reported only.

Other bias

Unclear risk

Not reported.

GEM2010MAS65 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted in Spain.

  • Overall sample size: N = 241 patients. Experimental arm (alternating scheme) = 120 patients. Control arm (sequential scheme) = 121 patients.

Participants

  • Patient Population: Elderly patients with newly diagnosed multiple myeloma.

  • Inclusion Criteria: Age > 65 years; ECOG PS ≤2; platelets ≥ 75 x 109/L; haemoglobin ≥ 8 g/dL; ANC ≥ 1.0 x 109/L; serum bilirubin ≤ 1.5 mg/dL; alkaline phosphatase, AST, ALT ≤ 2.5 x ULN; serum creatinine ≤ 2.5 mg/dL.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; prior bortezomib or lenalidomide.

  • Baseline Characteristics: Not known.

Interventions

  • Sequential scheme: 9 cycles of VMP (IV bortezomib 1.3 mg/m2 twice weekly for 1 six‐week cycle followed by once weekly for 8 four‐weeks cycles, oral melphalan 9 mg/m2, prednisone 60 mg/m2 once daily on days 1‐4 of each cycle) followed by 9 cycles of Rd (oral lenalidomide 25 mg daily on days 1‐21, dexamethasone 40 mg weekly).

  • Alternating scheme: One cycle of VMP alternating with one cycle of Rd (half of patients started VMP and half by Rd) for up to 18 cycles.

Outcomes

  • Primary: Time to progression; toxicity (safety and tolerability).

  • Secondary: Response; genomics analysis; duration of response; progression‐free survival, time to next therapy; overall survival.

Notes

  • Sponsor/Funding: PETHEMA Foundation.

  • Type of Publication (Full text or abstract only): Abstract only.

  • Linked to other reports: None.

  • ECOG PS = Eastern Coooperative Oncology Group Performance Status.

  • ANC = Absolute Neutrophil Count.

  • AST = Aspartate aminotransferase.

  • ALT = Alanine aminotransferase.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not known. Central randomisation probably performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective endpoints measured (e.g. response rate according to the European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Not known if blinded/independent outcomes assessment conducted.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not clear if any patients lost to follow‐up, withdrawn etc (abstract only).

Selective reporting (reporting bias)

Unclear risk

Not clear (abstract only). Benefits and harms reported.

Other bias

Unclear risk

Not reported.

GIMEMA‐MM‐03‐05 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 61 centres in Italy from May 2006 to Jan 2009.

  • Overall sample size: N = 511 patients. Experimental arm (VMPT‐VT) = 254 patients. Control arm (VMP) = 257 patients.

Participants

  • Patient Population: Patients with newly diagnosed transplant ineligible multiple myeloma.

  • Inclusion Criteria: Age ≥ 65 years; not candidates for high‐dose therapy plus stem cell transplantation due to age or co‐existing co‐morbidities; KPS ≥ 60%.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; renal insufficiency (creatinine ≥ 25 mg/mL); psychiatric disease; uncontrolled/severe cardiovascular disease; other malignancy within 5 years.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Experimental arm (VMPT‐VT): Induction therapy with nine 6‐weekly cycle of IV bortezomib 1.3 mg/m2 on days 1, 4, 8, 11, 22, 25, 29, 32 of cycles 1to 4 and on days 1, 8, 22, 29 of cycles 5‐9, oral melphalan 9 mg/m2 on days 1 to 4, oral prednisone 60 mg/m2 on days 1 to 4, oral thalidomide 50 mg/day continuously. Maintenance therapy with IV bortezomib 1.3 mg/m2 every 14 days and oral thalidomide 50mg/day for 2 years or until progression.

  • Control arm (VMP): Standard induction therapy with nine 6‐weekly cycles of VMP at same doses and no maintenance therapy.

  • Post safety analysis of 139 patients, induction changed to nine 5‐week cycles and weekly bortezomib cycles 1 to 9 to reduce neuropathy.

  • Additional Treatments: Thromboprophylaxis for VMPT‐VT patients.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rate, time to first response, overall survival, incidence of ≥ grade 3 adverse events.

Notes

  • Sponsor/Funding: Fondazione Neoplasie Sangue Onlus.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Palumbo 2014 (updated follow‐up).

  • KPS = Karnofsky Performance Status.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not reported. Multi‐centre trial, therefore probably centrally randomised.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective endpoints measured (e.g. response rate and progression according to International Uniform Response Criteria, overall survival) however independent blinded outcomes assessment not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart accounting for all patients. Acceptable rates of withdrawal/lost to follow‐up.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

Not reported.

GIMEMA‐MMY‐3006 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 73 centres in Italy from May 2006 to Apr 2008.

  • Overall sample size: N = 480 patients. Experimental arm (VTD: bortezomib, thalidomide, dexamethasone) = 241 patients. Control Arm (TD: thalidomide, dexamethasone) = 239 patients.

Participants

  • Patient Population: Patients with newly diagnosed and untreated symptomatic multiple myeloma.

  • Inclusion Criteria: Age 18‐65 years; KPS ≥ 60%; Platelets ≥ 70 x 109/L; ANC ≥ 1 x 109/L; serum creatinine ≤ 176 µmol/L.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; history of venous thromboembolism; diagnosis of thrombophylic alterations.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Experimental Arm (VTD): Induction: IV bortezomib 1.3 mg/m2 on days 1, 4, 8, 11 with TD for 3 x 21‐day cycles. Consolidation: IV bortezomib 1.3mg/m2 on Days 1, 8, 15 and 22 with TD for 2 x 35‐day cycles.

  • Control Arm (TD): Induction: oral thalidomide 100 mg daily first 14 days, 200 mg daily thereafter and dexamethasone 40 mg on days 1, 2, 4, 5, 8, 9, 11 and 12. Consolidation: oral thalidomide 100 mg daily and oral dexamethasone 40 mg on days 1, 2, 8, 9, 15, 16, 22 and 23.

  • Additional Treatments: Double ASCT 3‐6 months apart following induction therapy and stem cell mobilisation.

Outcomes

  • Primary: Complete response rate plus near complete response after induction.

  • Secondary: Complete response rate plus near complete response rate to double transplantation and consolidation therapy, time to progression or relapse, progression‐free survival, overall survival and safety.

Notes

  • Sponsor/Funding: Seràgnoli Institute of Haematology at the University of Bologna. Janssen‐Cilag provided bortezomib.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Cavo 2012 (median follow‐up 43 months); Cavo 2012 (abstract only, median follow‐up 52 months).

  • KPS = Karnofsky Performance Status.

  • ANC = Absolute Neutrophil Count.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Patients were randomised via 'web‐based system' at central coordinating centre.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression according to the European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Responses monitored by external contract research organisation and centrally reassessed by central coordinating team.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart accounting for all patients. Acceptable rates of withdrawal/lost to follow‐up.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

Not reported.

HOVON‐65/GMMG‐HD4 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted in Belgium, Netherlands and Germany from May 2005 to May 2008.

  • Overall sample size: N = 827 patients. Experimental arm (PAD ) = 413 patients. Control arm (VAD) = 414 patients.

Participants

  • Patient Population: Patients with newly diagnosed symptomatic multiple myeloma.

  • Inclusion Criteria: Age 18‐65 years; Performance Status 0‐2.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; serum bilirubin ≥ 30 µmol/L; amino transferases ≥ 2.5 normal level.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Experimental arm (PAD): Induction chemotherapy with bortezomib, Adriamycin and dexamethasone (PAD) followed by intensive chemotherapy with melphalan 200 mg/m2 and ASCT, followed by maintenance therapy with bortezomib for 2 years. PAD = 3 cycles of IV bortezomib 1.3 mg/m2 on days 1, 4, 8 and 11, IV doxorubicin 9 mg/m2/day on days 1 to 4, oral dexamethasone 40 mg/day on days 1 to 4, 9 to 12, 17 to 20 every 28 days. Maintenance IV bortezomib = 1.3 mg/m2 every 2 weeks for 2 years.

  • Control arm (VAD): Induction chemotherapy with vincristine, Adriamycin and dexamethasone (VAD) followed by intensive chemotherapy with melphalan 200 mg/m2 and ASCT, followed by maintenance therapy with thalidomide for 2 years. VAD = 3 cycles of IV vincristine 0.4 mg/day on days 1 to 4, IV doxorubicin 9 mg/m2/day on days 1 to 4, oral dexamethasone 40 mg/day on days 1 to 4, 9 to 12, 17 to 20 every 28 days. Maintenance oral thalidomide = 50 mg/day for 2 years.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rate; progression‐free survival without censoring patients with ASCT; progression‐free survival from last high‐dose melphalan; overall survival; safety; toxicity.

Notes

  • Sponsor/Funding: Dutch‐Belgian Hemato‐Oncology Cooperative Group and the German Multicenter Myeloma Group (GMMG). Supported by Dutch Cancer Foundation, German Federal Ministry of Education and Research, Janssen‐Cilag‐Ortho Biotech, Novartis, Amgen, Chugai, and Roche.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Sonneveld 2013 (abstract only, median follow‐up 67 months).

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Patients were randomised via 'web‐based' system.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective endpoints measured (e.g. response rate and progression according to modified European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival), however, independent/blinded outcomes assessment not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart accounting for all patients included.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

IFM 2005‐01 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 89 centres in France, Belgium and Switzerland from Aug 2005 to Jan 2008.

  • Overall sample size: N = 482 patients:

  1. Arm A1 (VAD induction + no consolidation) = 121 patients.

  2. Arm A2 (VAD induction + DCEP consolidation) = 121 patients.

  3. Arm B1 (BD induction + no consolidation) = 121 patients.

  4. Arm B2 (BD induction + DCEP consolidation) = 119 patients.

  • VAD induction (A1 + A2) = 242 patients.

  • BD induction (B1 + B2) = 240 patients.

Participants

  • Patient Population: Patients with newly diagnosed untreated symptomatic multiple myeloma.

  • Inclusion Criteria: Age ≤ 65 years; Performance Status ≤ 2; adequate renal, haematologic, and hepatic function.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; HIV positive; uncontrolled diabetes.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • VAD: 4 cycles of vincristine 0.4 mg/day on days 1 to 4, doxorubicin 9 mg/m2/day by continuous infusion on days 1 to 4, oral dexamethasone 40 mg/day on days 1 to 4 (all cycles), 9 to 12, 17 to 20 (cycles 1 and 2) every 28 days.

  • BD: 4 cycles of IV bortezomib 1.3 mg/m2 on days 1, 4, 8 and 11, oral dexamethasone 40 mg/day on days 1 to 4 (all cycles), days 9 to 12 (cycles 1 and 2) every 21 days.

  • DCEP: 2 cycles of dexamethasone 40 mg/day on days 1 to 4, cyclophosphamide 400 mg/m2, etoposide 40 mg/m2, cisplatin 15 mg/m2/day by continuous infusion on days 1 to 4 every 28 days.

  • Additional treatments: Bisphosphonates, antibiotics, anti‐fungal and anti‐viral therapies according to local practice.

Outcomes

  • Primary: Post‐induction Complete Response/near Complete Response (CR/nCR) rate.

  • Secondary: Post‐induction overall response rate; CR/nCR rate with and without DCEP consolidation; CR/nCR and at least VGPR rates post first transplantation; proportion of patients requiring second transplantation; safety and toxicity.

Notes

  • Sponsor/Funding: Investigator‐initiated trial.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: None.

  • HIV = human immunodeficiency virus.

  • CR/nCR = Complete Response/near Complete Response.

  • VGPR = Very Good Partial Response.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Patients were 'centrally randomly assigned'.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression according to modified European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Responses confirmed by independent review committee.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart accounting for all patients included. Acceptable rates of withdrawal.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

IFM 2007‐02 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 50 centres in France from Mar 2008 to Jan 2009.

  • Overall sample size: N = 199 patients. Experimental arm (vtD) = 100 patients. Control arm (VD) = 99 patients.

Participants

  • Patient Population: Patients with newly diagnosed untreated symptomatic multiple myeloma.

  • Inclusion Criteria: Age ≤ 65 years; Performance Status ≤ 2; adequate renal function.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; HIV positive; uncontrolled diabetes; amyloidosis; history of other cancer (except basal cell carcinoma and cervix cancer in situ).

  • Baseline Characteristics: No significant imbalance apparent or reported except difference in proportion of patients with t(4;14) and/or del (17p) higher in vtD arm than VD arm.

Interventions

  • vtD: 4 cycles of IV bortezomib 1.0 mg/m2 on days 1, 4, 8 and 11, oral thalidomide 100 mg/day and oral dexamethasone 40 mg/day on days 1 to 4 (all cycles), days 9 to 12 (cycles 1 and 2) every 21 days. In case of < PR after cycle 2, bortezomib increased to 1.3 mg/m2 and thalidomide to 200 mg/day.

  • VD: 4 cycles of IV bortezomib 1.3 mg/m2 on days 1, 4, 8 and 11, oral dexamethasone 40 mg/day on days 1 to 4 (all cycles), days 9 to 12 (cycles 1 and 2) every 21 days.

  • Additional treatments: Bisphosphonates, antibiotics and anti‐viral therapies according to local practice.

Outcomes

  • Primary: Post‐induction CR rate.

  • Secondary: CR plus VGPR rates after cycle 2, after induction and after ASCT; overall response rates (≥ PR) after cycle 2, after induction and after ASCT; safety and toxicity.

Notes

  • Sponsor/Funding: Investigator‐initiated trial by the Intergroupe Francophone du Myelome (IFM).

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: None.

  • CR = Complete Response.

  • VGPR = Very Good Partial Response.

  • PR = Partial Response.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Patients were 'centrally randomised'.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression according to International Myeloma Working Group Uniform Criteria, overall survival). Laboratory samples to evaluate response data were centrally evaluated.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart accounting for all patients included. Acceptable rates of withdrawal.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

MD Anderson Study

Methods

  • Design: Randomised, open‐label, phase II trial conducted at 1 centre in US from October 2006 to September 2007.

  • Overall sample size: N = 60 patients. Randomised to 1 of 3 groups (N = 20 per group).

  • Group 1 Control Arm: No bortezomib + Melphalan (Mel) + Ascorbic Acid (AA) +Arsenic Trioxide (ATO).

  • Group 2 Treatment Arm: Bortezomib 1.0 mg/m2 + Mel + AA +ATO.

  • Group 3 Treatment Arm: Bortezomib 1.5 mg/m2+ Mel + AA +ATO.

Participants

  • Patient Population: Patients with newly diagnosed multiple myeloma undergoing ASCT.

  • Inclusion Criteria: Age ≤ 75 years; PS < 2; serum bilirubin < 2 x ULN; SGPT < 4 x ULN.

  • Baseline Characteristics: No significant imbalances apparent or reported.

Interventions

  • Group 1 Control Arm: Mel (100 mg/m2 IV days ‐4, ‐3), AA (100 mg/m2 IV days ‐9 to ‐3), ATO (0.25 mg/kg IV days ‐9 to ‐3).

  • Group 2 Treatment Arm: Bortezomib 1.0 mg/m2 IV days ‐9, ‐6, ‐3 + Mel + AA +ATO.

  • Group 3 Treatment Arm: Bortezomib 1.5 mg/m2 IV days ‐9, ‐6, ‐3 + Mel + AA +ATO.

  • Additional treatments: Supportive care according to established guidelines.

Outcomes

  • Primary: Complete response, time to grade IV toxicity and death.

  • Secondary: Response rate, progression‐free survival, overall survival and safety.

Notes

  • Sponsor/Funding: Sponsored by M.D. Anderson Cancer Center. Part supported by grant from Cephalon Oncology.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: None.

  • PS = Performance Status.

  • ULN = Upper Normal Limit.

  • SGPT = Serum glutamic pyruvic transaminase.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not specified.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate, overall survival).

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 patients withdrew early (from group 1 and group 3). No lost to follow‐up patients reported.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

MMVAR/IFM 2005‐04 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 60 centres in 9 countries in Europe and Israel from Jan 2006 to Jul 2010.

  • Overall sample size: N = 269 patients. Experimental Arm A (VTD: bortezomib + thalidomide + dexamethasone) = 135 patients. Control Arm (TD: thalidomide + dexamethasone Alone) = 134 patients.

Participants

  • Patient Population: Patients with relapsed multiple myeloma following ASCT.

  • Inclusion Criteria: KPS > 50%; platelets ≥ 40,000/µL; ANC ≥ 1,000/µL; creatinine clearance ≥ 30 mL/min.

  • Exclusion Criteria: ≥ grade 2 peripheral neuropathy; prior allogeneic SCT.

  • Baseline Characteristics: No significant imbalances apparent or reported.

Interventions

  • Experimental Arm (VTD): IV bortezomib 1.3 mg/m2 on days 1, 4, 8, 11 of 21 day cycle for 8 cycles (6 months), followed by IV bortezomib 1.3 mg/m2 on days 1, 8, 15 and 22 of 42 day cycle for 4 cycles (6 months) with TD. Total duration = 1 year.

  • Control Arm (TD): Oral thalidomide 200 mg daily and oral dexamethasone 40 mg on 4 days every 3 weeks. Total duration = 1 year.

Outcomes

  • Primary: Time to disease progression.

  • Secondary: Progression‐free survival; overall survival; overall response rate (complete plus partial); adverse events.

Notes

  • Sponsor/Funding: European Group for Blood and Marrow Transplantation.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: None.

  • KPS = Karnofksy Performance Status.

  • ANC = Absolute Neutrophil Count.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not specified. Central randomisation most probably performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective endpoints measured (e.g. response rate and progression according to European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Independent outcomes assessment not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Flow chart with all randomised patients accounted for. Low rate of lost to follow‐up. All patients included in intention‐to‐treat Analysis.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

MMY‐3021 Study

Methods

  • Design: Randomised, open‐label, phase III non‐inferiority trial conducted at 53 centres in 10 countries in Europe, Asia and South America from Jul 2008 to Feb 2010.

  • Overall sample size: N = 222 patients. 2 : 1 allocation. Experimental arm (SC bortezomib) = 148 patients. Control arm (IV bortezomib) = 74 patients.

Participants

  • Patient Population: Multiple myeloma patients with measurable progressive disease.

  • Inclusion Criteria: Age ≥ 18 years; KPS ≥ 70%; 1 to 3 prior lines of therapy; adequate haematologic, hepatic and renal function.

  • Exclusion Criteria: Prior bortezomib therapy; ≥ grade 2 peripheral neuropathy or neuropathic pain; anti‐neoplastic, experimental, corticosteroid (> 10 mg/day prednisone or equivalent) therapy within 3 weeks of randomisation.

  • Baseline Characteristics: No significant imbalance apparent or reported except: higher number of patients in SC group with KPS < 80%, creatinine clearance < 60 mL/min and from Eastern Europe; higher number of male patients and patients with high‐risk cytogenetics in IV group.

Interventions

  • Experimental Arm: Bortezomib 1.3 mg/m2 by SC injection on days 1, 4, 8, 11 of 21‐day cycle up to 8 cycles.

  • Control Arm: Bortezomib 1.3 mg/m2 by IV infusion on days 1, 4, 8, 11 of 21‐day cycle up to 8 cycles.

  • Patients with suboptimal response (< CR, without disease progression) at end of cycle 4 could additionally receive oral dexamethasone from cycle 5 onwards. Patients with stable disease or PR as best response at end of cycle 8, evolving steadily to late PR or CR, respectively, could receive two additional cycles.

  • Additional Treatments: Concomitant supportive care therapies allowed except systemic steroids and anti‐neoplastic therapies with anti‐myeloma effects. Bisphosphonates permitted.

Outcomes

  • Primary: Overall response rate (complete plus partial response) after 4 cycles.

  • Secondary: CR, nCR and VGPR rates after 4 cycles; ORR after 8 cycles; time to response; duration of response; time to progression; progression‐free survival; 1‐year overall survival; safety and tolerability; pharmacokinetics and pharmacodynamics.

Notes

  • Sponsor/Funding: Millennium Pharmaceuticals.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Arnulf, 2012 (extended follow‐up data).

  • KPS = Karnofksy Performance Status.

  • CR/nCR = Complete Response/near Complete Response.

  • VGPR = Very Good Partial Response.

  • ORR = Overall Response Rate.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised', employing a 'computer‐generated randomisation schedule' based on 'permuted blocks'.

Allocation concealment (selection bias)

Low risk

Central randomisation using an 'interactive voice response system' (IVRS).

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression by European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Response data based on central laboratory analysis and blinded response evaluation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart with all randomised patients accounted for. No lost to follow‐up patients reported.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

NMSG 15/05 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 23 centres in Denmark, Estonia, Finland, Iceland, Norway and Sweden from Oct 2005 to Apr 2009.

  • Overall sample size: N = 370 patients. Experimental arm (bortezomib consolidation therapy) = 187 patients. Control arm (No consolidation therapy) = 183 patients.

Participants

  • Patient Population: Patients with newly diagnosed multiple myeloma following ASCT.

  • Exclusion Criteria: > grade 2 peripheral neuropathy; severe heart disease or heart failure; history of hypotension; prior exposure to bortezomib.

  • Baseline Characteristics: No significant imbalances apparent or reported.

Interventions

  • Experimental Arm: Consolidation therapy with IV bortezomib 1.3 mg/m2 on days 1, 4, 8, 11 of 21‐day cycle for 2 cycles, followed by IV bortezomib 1.3 mg/m2 on Days 1, 8, 15 of 28‐day cycle for 4 cycles.

  • Control Arm: No consolidation therapy.

  • Additional Treatments: No steroids permitted except < 50 mg prednisone for 1 week for other medical conditions. Bisphosphonates allowed.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rate; overall survival; health‐related quality of life; tolerability.

Notes

  • Sponsor/Funding: Nordic Myeloma Study Group.

  • Type of Publication (Full Text or abstract only): Full Text.

  • Linked to other reports: None.

  • ASCT = Autologous Stem Cell Transplant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Central computer randomisation performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. response rate and progression according to European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Independent outcomes assessment not reported but data monitored by independent contract research organization.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Clear flow chart with all randomised patients accounted for. No lost to follow‐up reported. All patients included in intention‐to‐treat analysis.

Selective reporting (reporting bias)

Unclear risk

Benefits and harms reported however selected adverse events reported (peripheral neuropathy and neuropathic pain) only. 2 secondary malignancies reported (1 on treatment arm; 1 on control arm).

Other bias

Unclear risk

None reported.

NMSG 17/07 Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 29 centres in Norway, Sweden and Denmark from Oct 2007 to Sep 2010.

  • Overall sample size: N = 131 patients. Trial prematurely closed due to low accrual.

  • Experimental arm (bortezomib + dexamethasone) = 64 patients. Control arm (thalidomide + dexamethasone) = 67 patients.

  • Cross‐over: Patients offered cross‐over to other arm at disease progression: 39 patients received bort‐dex after failure on thal‐dex, 33 received thal‐dex after failure on bort‐dex.

Participants

  • Patient Population: Patients with refractory multiple myeloma.

  • Inclusion Criteria: Any age; refractory to prior melphalan treatment.

  • Exclusion Criteria: Former treatment with thalidomide, bortezomib, lenalidomide; ≥ grade 3 sensory neuropathy; ≥ grade 2 neuropathic pain; platelets < 25 x 109/L; severe co‐morbidity.

  • Baseline Characteristics: Signficantly more females on control arm (thal‐dex); no prognostic importance on univariate analysis. All other characteristics balanced.

Interventions

  • Experimental Arm (bort‐dex); Bortezomib 1.3 mg/m2 IV on days 1, 4, 8, 11 of 21‐day cycle. Dexamethasone 20 mg oral on days 1‐2, 4‐5, 8‐9, 11‐12 of 21‐day cycle.

  • Control Arm (thal‐dex): Thalidomide 50 mg oral daily, escalated by 50 mg every 3 weeks to a maximum of 200 mg. Dexamethasone 40 mg oral on days 1 to 4 every 3 weeks.

  • Additional Treatments: Anti‐thrombotic prophylaxis and acyclovir prophylaxis used routinely.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rate; duration of response; toxicity; health‐related quality of life; time to next treatment; overall survival.

Notes

  • Sponsor/Funding: Nordic Myeloma Study Group.

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: None.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Low risk

Central web‐based randomisation performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Objective endpoints measured (e.g. response rate and progression according to International Myeloma Working Group (IMWG) guidelines, overall survival). Independent outcomes assessment not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart for randomised patients included. Acceptable rates of withdrawal, no lost to follow‐up patients reported. All patients included in intention‐to‐treat analysis.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

VISTA Study

Methods

  • Design: Randomised, open‐label, phase III trial conducted at 151 centres in 22 countries in Europe, North and South America and Asia from Dec 2004 to Sep 2006.

  • Overall sample size: N = 682 patients. Experimental arm A (VMP: melphalan + prednisone + bortezomib) = 344 patients. Control arm (MP: melphalan + prednisone alone) = 338 patients.

Participants

  • Patient Population: Newly diagnosed, untreated, symptomatic, measurable multiple myeloma patients not candidates for high‐dose therapy plus stem‐cell transplantation.

  • Inclusion Criteria: Age ≥ 65 years; measurable disease.

  • Exclusion Criteria: Serum creatinine > 2 mg/dL; ≥ grade 2 peripheral neuropathy or neuropathic pain.

  • Baseline Characteristics: No significant imbalance apparent or reported.

Interventions

  • Experimental Arm: VMP: Nine 6‐weekly cycles of melphalan 9 mg/m2and prednisone at 60 mg/m2on days 1 to 4 of each cycle with IV bortezomib. 1.3 mg/m2 on Days 1, 4, 8, 11, 22, 25, 29 and 32 during cycles 1 to 4 and on days 1, 8, 22 and 29 during cycles 5 to 9.

  • Control Arm: MP: Nine 6‐weekly cycles of melphalan 9 mg/m2and prednisone at 60 mg/m2on days 1 to 4 alone.

  • Additional Treatments: Bisphosphonates for patients with myeloma‐associated bone disease.

Outcomes

  • Primary: Time to disease progression.

  • Secondary: Progression‐free survival, complete plus partial response rate, complete response rate, duration of response; time to subsequent myeloma therapy, overall survival, adverse events, health‐related quality of life.

Notes

  • Sponsor/Funding: Millennium Pharmaceuticals/Johnson & Johnson Pharmaceutical Research & Development

  • Type of Publication (Full text or abstract only): Full text.

  • Linked to other reports: Mateos 2010, San Miguel 2013, Delforge 2013, Spicka 2011.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Study design described as 'randomised'.

Allocation concealment (selection bias)

Unclear risk

Not specified. Central randomisation most probably performed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias) for OS

Low risk

Low risk of bias for OS.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Objective endpoints measured (e.g. e.g. response rate and progression according to European Group for Blood and Bone Marrow Transplant (EBMT) criteria, overall survival). Response data based on central laboratory analysis of blood and urine samples.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Detailed flow chart. Low numbers lost to follow‐up/excluded from analysis.

Selective reporting (reporting bias)

Low risk

Benefits and harms reported.

Other bias

Unclear risk

None reported.

IV: intravenous
SC: subcutaneous

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Chen 2011b

Described as a 'retrospective randomised' study.  Small study of 46 patients randomised according to date of hospitalisation.

Goldschmidt 2012

Same dose of bortezomib on same days on both arms (PAD regimen = 28 day cycle and VCD regimen = 21‐day cycle).

Kumar 2012

Same dose/schedule of bortezomib on each arm.

Mateos 2010

Same dose/schedule of bortezomib on each arm.

Niesvisky 2010

Same dose/schedule of bortezomib on each arm.

Orlowski 2007

Same dose/schedule of bortezomib on each arm.

PAD:
VCD:

Characteristics of studies awaiting assessment [ordered by study ID]

Mayo Clinic Study

Methods

Design: Randomised, open‐label, phase II trial.

Overall sample size: N = 150 patients.

Participants

Patients with newly diagnosed multiple myeloma who have completed stem cell transplant.

Interventions

Arm A (bortezomib). Patients receive bortezomib subcutaneously (SC) on days 1 and 15 of courses 1 to 12 and day 1 of courses 13 to 24.

Arm B (bortezomib, cyclophosphamide, dexamethasone). Patients receive bortezomib SC as in Arm A, cyclophosphamide orally on days 1 and 15 of courses 1 to12 and day 1 of courses 13 to 24, and dexamethasone orally on days 1 and 15 of courses 1 to 12 and day 1 of courses 13 to 24.

Arm C (bortezomib, lenalidomide). Patients receive bortezomib SC as in Arm A and lenalidomide orally on days 1 to 28.

Outcomes

Primary: Proportion of patients with stringent complete response.

Secondary: Survival time, progression‐free survival, adverse events.

Notes

Sponsor: Mayo Clinic, US. This study is not eligible for inclusion (same dose/schedule of bortezomib on each arm). To be verified post‐publication for addition to next update of this review.

Velcade Consolidation Bone Study

Methods

Design: Randomised, open‐label, phase II trial.

Overall sample size: N = 106 patients.

Participants

Patients with multiple myeloma who have received high‐dose chemotherapy and autologous stem cell transplantation.

Interventions

Experimental arm: Bortezomib 1.6 mg/m² bolus injection on Days 1, 8, 15 and 22 every 5 weeks for 4 cycles.

Control arm: Observation only.

Outcomes

Primary: Change From baseline in Bone Mineral Density (BMD).

Secondary: Progression‐free survival, bone markers, skeletal events, appearance of new bone lesions, Karnosfsky performance status, overall survival.

Notes

Sponsor: Janssen‐Cilag International NV. BMD data published by abstract only (European Haematology Association (EHA) Congress, June 2014). To consider eligibility of study for inclusion in future update of review if progression‐free survival and/or overall survival data are published.

Characteristics of ongoing studies [ordered by study ID]

CLARION Study

Trial name or title

CLARION Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 882 patients.

Participants

Transplant‐ineligible patients with multiple myeloma.

Interventions

Carfilzomib, Melphalan, and Prednisone (CMP) versus Bortezomib, Melphalan, and Prednisone (VMP).

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Overall survival, response rates, health‐related quality of life.

Starting date

2013

Contact information

Onyx Pharmaceuticals

Notes

Study in recruitment phase. November 2017 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov April 2015.

Consolidation (61‐75 years) Study

Trial name or title

CR006127 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 154 patients.

Participants

Patients with multiple myeloma aged 61 to 75.

Interventions

Bortezomib 1.6 mg/m2 IV on days 1, 8, 15 and 22 of 35‐day cycle for 4 cycles as consolidation therapy versus observation.

Outcomes

  • Primary: Event‐free survival.

  • Secondary: Best response, response rates.

Starting date

2006

Contact information

Janssen‐Cilag G.m.b.H

Notes

May 2013 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015. Contact with company confirmed study not yet published.

Consolidation (less than 60 years) Study

Trial name or title

CR006124 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 217 patients.

Participants

Patients with multiple myeloma aged less than 60 years.

Interventions

Bortezomib 1.6 mg/m2 IV on days 1, 8, 15 and 22 of 35‐day cycle for 4 cycles as consolidation therapy versus observation.

Outcomes

  • Primary: Event‐free survival.

  • Secondary: Best response, response rates.

Starting date

2006

Contact information

Janssen‐Cilag G.m.b.H

Notes

May 2013 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015. Contact with company confirmed study not yet published.

E1A11 Study

Trial name or title

ECOG E1A11 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 525 patients.

Participants

Patients with newly diagnosed symptomatic multiple myeloma.

Interventions

Bortezomib, Lenalidomide and Dexamethasone (VRd) versus Carfilzomib, Lenalidomide, Dexamethasone (CRd) followed by limited or indefinite Lenalidomide maintenance.

Outcomes

  • Primary: Overall survival for maintenance analysis.

  • Secondary: Progression‐free survival for maintenance analysis, overall survival for induction analysis, response rates, time to progression, duration of response, adverse events, patient‐reported outcomes.

Starting date

2013

Contact information

Dr SK Kumar, Eastern Cooperative Oncology Group (ECOG)

Notes

May 2016 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov September 2014.

ENDEAVOR Study

Trial name or title

ENDEAVOR Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 898 patients.

Participants

Patients with relapsed multiple myeloma

Interventions

Carfilzomib and Dexamethasone versus Bortezomib and Dexamethasone

Outcomes

Primary: Progression‐free survival

Starting date

2012

Contact information

Onyx Pharmaceuticals

Notes

January 2016 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015.

Hackensack University Study

Trial name or title

PRO# 1307 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 398 patients.

Participants

Patients with multiple myeloma 65 years or older.

Interventions

Autologous Stem Cell Transplantation with high‐dose Melphalan versus high‐dose Melphalan and Bortezomib.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Overall survival, toxicity, response rates.

Starting date

2010

Contact information

Dr M Donato, John Theurer Cancer Center, Hackensack University Medical Center, New Jersey, US.

Notes

Study open to recruitment. November 2014 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov Aug 2014.

HOVON 95 Study

Trial name or title

HOVON 95 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 1500 patients.

Participants

Patients with newly diagnosed multiple myeloma.

Interventions

Bortezomib, Melphalan, Prednisone (VMP) With high‐dose Melphalan followed by Bortezomib, Lenalidomide, Dexamethasone (VRD) Consolidation and Lenalidomide Maintenance.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Overall survival, toxicity, response rates.

Starting date

2011

Contact information

Prof. P Sonneveld, Stichting Hemato‐Oncologie voor Volwassenen Nederland

Notes

April 2021 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015.

King Fasail Hospital Study

Trial name or title

2081‐113 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 440 patients.

Participants

Patients with previously untreated multiple myeloma without an intent for immediate autologous stem cell transplant.

Interventions

Lenalidomide and low dose dexamethasone (LLD) versus bortezomib, lenalidomide and low dose dexamethasone (BLLD) as induction therapy.

Outcomes

Progression‐free survival.

Starting date

2009

Contact information

Dr N Chaudri, King Faisal Specialist Hospital & Reseach Center, Riyadh, Saudi Arabia

Notes

January 2013 (Final data collection date for primary outcome measure). Recruitment status unknown; last update on Clinicaltrials.gov Feb 2012.

Optimized Retreatment Study

Trial name or title

CR018796 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

Overall sample size: N = 80 patients.

Participants

Patients with multiple myeloma in first or second relapse.

Interventions

  • Experimental Arm: Retreatment with 6 cycles of bortezomib and dexamethasone (two 21‐day cycles followed by four 35‐day cycles) followed by a second randomisation in a 1:1 ratio to 1 of 2 prolonged therapy schedules with bortezomib alone (Group A1: once weekly for the first 4 weeks in 35‐day cycles; or Group A2: once every other week).

  • Control Arm: Patients will start retreatment with eight 21‐day bortezomib and dexamethasone cycles, followed by posttreatment follow‐up every 6 weeks.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rates, time to progression, duration of response, time to next myeloma therapy, performance status, health‐related quality of life.

Starting date

2013

Contact information

Janssen‐Cilag International NV

Notes

January 2016 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov April 2015.

Subcutaneous Bortezomib Maintenance Study

Trial name or title

Subcutaneous bortezomib maintenance Study

Methods

  • Design: Randomised, open‐label, phase II trial.

  • Overall sample size: N = 216 patients.

Participants

Patients with relapsed and refractory multiple myeloma after salvage with bortezomib‐based therapy.

Interventions

  • Experimental arm: SC bortezomib and oral dexamethasone every 2 weeks. Patients randomised in this group will be observed. At the occurrence of biochemical relapse, 4 cycles of SC bortezomib and oral dexamethasone weekly will be administered.

  • Control arm: Observation only.

Outcomes

  • Primary: Time to progression.

  • Secondary: Response rates, overall survival.

Starting date

2013

Contact information

Stichting Hemato‐Oncologie voor Volwassenen Nederland.

Notes

Study open to recruitment. November 2016 (final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015.

SWOG‐S0777 Study

Trial name or title

SWOG‐S0777 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 756 patients.

Participants

Previously untreated multiple myeloma without intent for immediate Autologous Stem Cell Transplant.

Interventions

Lenalidomide and Low Dose Dexamethasone (LLD) versus Bortezomib, Lenalidomide and Low Dose Dexamethasone (BLLD) as induction therapy.

Outcomes

  • Primary: Progression‐free survival.

  • Secondary: Response rates, overall survival.

Starting date

2008

Contact information

Dr BG Durie, Southwest Oncology Group (SWOG).

Notes

Follow‐up continuing. Last update on Clinicaltrials.gov September 2014.

VCAT Study

Trial name or title

CR018751 Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 253 patients.

Participants

Patients with multiple myeloma after receiving Bortezomib, Cyclophosphamide, Dexamethasone (VCD) Induction and Autologous Stem Cell Transplant.

Interventions

Bortezomib Consolidation (With Thalidomide and Prednisolone) versus Thalidomide and Prednisolone Alone.

Outcomes

  • Primary: Response rates.

  • Secondary: Response rates, progression‐free survival, disease‐free survival, overall survival.

Starting date

2012

Contact information

Janssen Scientific Affairs, LLC

Notes

October 2015 (Final data collection date for primary outcome measure). Last update on Clinicaltrials.gov March 2015.

Wuerzburg University Hospital Study

Trial name or title

DSMM XIV Study

Methods

  • Design: Randomised, open‐label, phase III trial.

  • Overall sample size: N = 406 patients.

Participants

Patients with newly diagnosed multiple myeloma.

Interventions

Lenalidomide, Adriamycin, Dexamethasone (RAD) versus Lenalidomide, Bortezomib, Dexamethasone (VRD) as induction therapy followed by response‐adapted consolidation and lenalidomide maintenance.

Outcomes

  • Primary: Response rates, progression‐free survival.

  • Secondary: Response rates, overall survival, toxicity, number of hospital days/hospitalisations.

Starting date

2012

Contact information

Dr Stefan Knop, Wuerzburg University Hospital.

Notes

Study still recruiting. Last update on Clinicaltrials.gov September 2012.

Data and analyses

Open in table viewer
Comparison 1. All Studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

9

4118

Peto Odds Ratio (95% CI)

0.77 [0.69, 0.86]

Analysis 1.1

Comparison 1 All Studies, Outcome 1 Overall Survival.

Comparison 1 All Studies, Outcome 1 Overall Survival.

1.1 Bortezomib versus no bortezomib (same background therapy)

4

1586

Peto Odds Ratio (95% CI)

0.77 [0.65, 0.92]

1.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2532

Peto Odds Ratio (95% CI)

0.76 [0.67, 0.88]

2 Progression‐Free Survival Show forest plot

9

4344

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.72]

Analysis 1.2

Comparison 1 All Studies, Outcome 2 Progression‐Free Survival.

Comparison 1 All Studies, Outcome 2 Progression‐Free Survival.

2.1 Bortezomib versus no bortezomib (same background therapy)

5

1855

Peto Odds Ratio (95% CI)

0.65 [0.57, 0.74]

2.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2489

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.75]

3 Complete Response Rate Show forest plot

12

4630

Odds Ratio (M‐H, Fixed, 95% CI)

2.35 [2.02, 2.73]

Analysis 1.3

Comparison 1 All Studies, Outcome 3 Complete Response Rate.

Comparison 1 All Studies, Outcome 3 Complete Response Rate.

3.1 Bortezomib versus no bortezomib (same background therapy)

6

2064

Odds Ratio (M‐H, Fixed, 95% CI)

2.63 [2.13, 3.24]

3.2 Bortezomib versus no bortezomib (different background therapy or other agents)

6

2566

Odds Ratio (M‐H, Fixed, 95% CI)

2.08 [1.67, 2.58]

4 Overall Response Rate Show forest plot

12

4630

Odds Ratio (M‐H, Fixed, 95% CI)

2.62 [2.25, 3.05]

Analysis 1.4

Comparison 1 All Studies, Outcome 4 Overall Response Rate.

Comparison 1 All Studies, Outcome 4 Overall Response Rate.

4.1 Bortezomib versus no bortezomib (same background therapy)

6

2064

Odds Ratio (M‐H, Fixed, 95% CI)

3.45 [2.72, 4.37]

4.2 Bortezomib versus no bortezomib (different background therapy or other agents)

6

2566

Odds Ratio (M‐H, Fixed, 95% CI)

2.17 [1.78, 2.64]

5 Treatment‐related death Show forest plot

5

2389

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.43, 1.34]

Analysis 1.5

Comparison 1 All Studies, Outcome 5 Treatment‐related death.

Comparison 1 All Studies, Outcome 5 Treatment‐related death.

5.1 Bortezomib versus no bortezomib (same background therapy)

2

737

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.30, 2.16]

5.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1652

Odds Ratio (M‐H, Fixed, 95% CI)

0.73 [0.36, 1.48]

6 Adverse Events: Thrombocytopenia Show forest plot

8

3791

Odds Ratio (M‐H, Fixed, 95% CI)

2.05 [1.70, 2.48]

Analysis 1.6

Comparison 1 All Studies, Outcome 6 Adverse Events: Thrombocytopenia.

Comparison 1 All Studies, Outcome 6 Adverse Events: Thrombocytopenia.

6.1 Bortezomib versus no bortezomib (same background therapy)

3

1196

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [1.13, 2.00]

6.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2595

Odds Ratio (M‐H, Fixed, 95% CI)

2.60 [2.01, 3.35]

7 Adverse Events: Neutropenia Show forest plot

8

3791

Odds Ratio (M‐H, Fixed, 95% CI)

1.33 [1.10, 1.60]

Analysis 1.7

Comparison 1 All Studies, Outcome 7 Adverse Events: Neutropenia.

Comparison 1 All Studies, Outcome 7 Adverse Events: Neutropenia.

7.1 Bortezomib versus no bortezomib (same background therapy)

3

1196

Odds Ratio (M‐H, Fixed, 95% CI)

0.95 [0.73, 1.24]

7.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2595

Odds Ratio (M‐H, Fixed, 95% CI)

1.85 [1.41, 2.41]

8 Adverse Events: Anaemia Show forest plot

6

3404

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.65, 1.00]

Analysis 1.8

Comparison 1 All Studies, Outcome 8 Adverse Events: Anaemia.

Comparison 1 All Studies, Outcome 8 Adverse Events: Anaemia.

8.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

0.67 [0.48, 0.94]

8.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2465

Odds Ratio (M‐H, Fixed, 95% CI)

0.92 [0.69, 1.21]

9 Adverse Events: Nausea/Vomiting Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

2.37 [1.64, 3.42]

Analysis 1.9

Comparison 1 All Studies, Outcome 9 Adverse Events: Nausea/Vomiting.

Comparison 1 All Studies, Outcome 9 Adverse Events: Nausea/Vomiting.

9.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

4.55 [1.99, 10.42]

9.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [1.28, 2.93]

10 Adverse Events: Diarrhoea Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

2.44 [1.74, 3.43]

Analysis 1.10

Comparison 1 All Studies, Outcome 10 Adverse Events: Diarrhoea.

Comparison 1 All Studies, Outcome 10 Adverse Events: Diarrhoea.

10.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

6.24 [2.79, 13.98]

10.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.80 [1.22, 2.65]

11 Adverse Events: Constipation Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.14, 2.22]

Analysis 1.11

Comparison 1 All Studies, Outcome 11 Adverse Events: Constipation.

Comparison 1 All Studies, Outcome 11 Adverse Events: Constipation.

11.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

1.88 [1.04, 3.41]

11.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.47 [0.98, 2.20]

12 Adverse Events: Peripheral Neuropathy Show forest plot

10

4636

Odds Ratio (M‐H, Fixed, 95% CI)

3.71 [2.92, 4.70]

Analysis 1.12

Comparison 1 All Studies, Outcome 12 Adverse Events: Peripheral Neuropathy.

Comparison 1 All Studies, Outcome 12 Adverse Events: Peripheral Neuropathy.

12.1 Bortezomib versus no bortezomib (same background therapy)

5

2040

Odds Ratio (M‐H, Fixed, 95% CI)

5.10 [3.37, 7.72]

12.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2596

Odds Ratio (M‐H, Fixed, 95% CI)

3.09 [2.30, 4.14]

13 Adverse Events: Infections (All) Show forest plot

9

4266

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [1.27, 1.79]

Analysis 1.13

Comparison 1 All Studies, Outcome 13 Adverse Events: Infections (All).

Comparison 1 All Studies, Outcome 13 Adverse Events: Infections (All).

13.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

1.37 [0.97, 1.93]

13.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2596

Odds Ratio (M‐H, Fixed, 95% CI)

1.55 [1.27, 1.90]

14 Adverse Events: Herpes Zoster infection Show forest plot

4

1733

Odds Ratio (M‐H, Fixed, 95% CI)

1.83 [0.91, 3.67]

Analysis 1.14

Comparison 1 All Studies, Outcome 14 Adverse Events: Herpes Zoster infection.

Comparison 1 All Studies, Outcome 14 Adverse Events: Herpes Zoster infection.

14.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [0.74, 5.03]

14.2 Bortezomib versus no bortezomib (different background therapy or other agents)

2

794

Odds Ratio (M‐H, Fixed, 95% CI)

1.71 [0.62, 4.74]

15 Adverse Events: Cardiac Disorders Show forest plot

5

2191

Odds Ratio (M‐H, Fixed, 95% CI)

1.74 [1.17, 2.58]

Analysis 1.15

Comparison 1 All Studies, Outcome 15 Adverse Events: Cardiac Disorders.

Comparison 1 All Studies, Outcome 15 Adverse Events: Cardiac Disorders.

15.1 Bortezomib versus no bortezomib (same background therapy)

2

736

Odds Ratio (M‐H, Fixed, 95% CI)

1.17 [0.39, 3.52]

15.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1455

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.21, 2.81]

16 Adverse Events: Fatigue Show forest plot

5

2926

Odds Ratio (M‐H, Fixed, 95% CI)

1.96 [1.35, 2.84]

Analysis 1.16

Comparison 1 All Studies, Outcome 16 Adverse Events: Fatigue.

Comparison 1 All Studies, Outcome 16 Adverse Events: Fatigue.

16.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

3.30 [1.66, 6.58]

16.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1987

Odds Ratio (M‐H, Fixed, 95% CI)

1.52 [0.97, 2.38]

Open in table viewer
Comparison 2. Subgroup Analyses ‐ Disease Setting

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

8

4075

Peto Odds Ratio (95% CI)

0.77 [0.70, 0.86]

Analysis 2.1

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 1 Overall Survival.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 1 Overall Survival.

1.1 Relapsed / Refactory

1

669

Peto Odds Ratio (95% CI)

0.77 [0.61, 0.97]

1.2 Transplant Eligible

5

2213

Peto Odds Ratio (95% CI)

0.86 [0.73, 1.02]

1.3 Transplant Ineligible

2

1193

Peto Odds Ratio (95% CI)

0.70 [0.59, 0.82]

2 Progression Free Survival Show forest plot

11

4344

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.72]

Analysis 2.2

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 2 Progression Free Survival.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 2 Progression Free Survival.

2.1 Relapsed / Refactory

3

938

Peto Odds Ratio (95% CI)

0.57 [0.48, 0.68]

2.2 Transplant Eligible

6

2213

Peto Odds Ratio (95% CI)

0.76 [0.68, 0.85]

2.3 Transplant Ineligible

2

1193

Peto Odds Ratio (95% CI)

0.57 [0.49, 0.67]

3 Complete Response Rate Show forest plot

11

4593

Odds Ratio (M‐H, Fixed, 95% CI)

2.38 [2.05, 2.77]

Analysis 2.3

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 3 Complete Response Rate.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 3 Complete Response Rate.

3.1 Relapsed / Refactory

3

998

Odds Ratio (M‐H, Fixed, 95% CI)

3.35 [2.06, 5.43]

3.2 Transplant Eligible

6

2424

Odds Ratio (M‐H, Fixed, 95% CI)

1.88 [1.56, 2.27]

3.3 Transplant Ineligibile

2

1171

Odds Ratio (M‐H, Fixed, 95% CI)

3.69 [2.71, 5.02]

4 Overall Response Rate Show forest plot

11

4593

Odds Ratio (M‐H, Fixed, 95% CI)

2.61 [2.24, 3.05]

Analysis 2.4

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 4 Overall Response Rate.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 4 Overall Response Rate.

4.1 Relapsed / Refactory

3

998

Odds Ratio (M‐H, Fixed, 95% CI)

2.39 [1.79, 3.20]

4.2 Transplant Eligible

6

2424

Odds Ratio (M‐H, Fixed, 95% CI)

2.24 [1.76, 2.85]

4.3 Transplant Ineligibile

2

1171

Odds Ratio (M‐H, Fixed, 95% CI)

3.45 [2.63, 4.53]

Open in table viewer
Comparison 3. Subgroup Analyses ‐ Therapy Setting

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

5

Peto Odds Ratio (95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 1 Overall Survival.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 1 Overall Survival.

1.1 Induction

3

1783

Peto Odds Ratio (95% CI)

0.81 [0.67, 0.96]

1.2 Consolidation

2

844

Peto Odds Ratio (95% CI)

1.02 [0.71, 1.47]

1.3 Maintenance

2

1338

Peto Odds Ratio (95% CI)

0.76 [0.64, 0.91]

2 Progression Free Survival Show forest plot

5

Peto Odds Ratio (95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 2 Progression Free Survival.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 2 Progression Free Survival.

2.1 Induction

3

1783

Peto Odds Ratio (95% CI)

0.75 [0.66, 0.85]

2.2 Consolidation

2

844

Peto Odds Ratio (95% CI)

0.70 [0.58, 0.84]

2.3 Maintenance

2

1338

Peto Odds Ratio (95% CI)

0.70 [0.61, 0.79]

3 Complete Response Rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 3 Complete Response Rate.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 3 Complete Response Rate.

3.1 Induction

4

1999

Odds Ratio (M‐H, Fixed, 95% CI)

2.04 [1.65, 2.53]

3.2 Consolidation

2

839

Odds Ratio (M‐H, Fixed, 95% CI)

1.77 [1.34, 2.33]

3.3 Maintenance

2

1330

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.44, 2.31]

4 Overall Response Rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.4

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 4 Overall Response Rate.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 4 Overall Response Rate.

4.1 Induction

4

1999

Odds Ratio (M‐H, Fixed, 95% CI)

2.33 [1.82, 2.99]

4.2 Consolidation

2

839

Odds Ratio (M‐H, Fixed, 95% CI)

2.21 [1.20, 4.06]

4.3 Maintenance

2

1330

Odds Ratio (M‐H, Fixed, 95% CI)

1.90 [1.38, 2.62]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

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

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

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

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

Forest plot of comparison: 3 All studies, outcome: 3.1 Overall Survival.
Figuras y tablas -
Figure 4

Forest plot of comparison: 3 All studies, outcome: 3.1 Overall Survival.

Forest plot of comparison: 3 All studies, outcome: 3.2 Progression Free Survival.
Figuras y tablas -
Figure 5

Forest plot of comparison: 3 All studies, outcome: 3.2 Progression Free Survival.

Forest plot of comparison: 3 All studies, outcome: 3.3 Complete Response Rate.
Figuras y tablas -
Figure 6

Forest plot of comparison: 3 All studies, outcome: 3.3 Complete Response Rate.

Forest plot of comparison: 3 All studies, outcome: 3.4 Overall Response Rate.
Figuras y tablas -
Figure 7

Forest plot of comparison: 3 All studies, outcome: 3.4 Overall Response Rate.

Comparison 1 All Studies, Outcome 1 Overall Survival.
Figuras y tablas -
Analysis 1.1

Comparison 1 All Studies, Outcome 1 Overall Survival.

Comparison 1 All Studies, Outcome 2 Progression‐Free Survival.
Figuras y tablas -
Analysis 1.2

Comparison 1 All Studies, Outcome 2 Progression‐Free Survival.

Comparison 1 All Studies, Outcome 3 Complete Response Rate.
Figuras y tablas -
Analysis 1.3

Comparison 1 All Studies, Outcome 3 Complete Response Rate.

Comparison 1 All Studies, Outcome 4 Overall Response Rate.
Figuras y tablas -
Analysis 1.4

Comparison 1 All Studies, Outcome 4 Overall Response Rate.

Comparison 1 All Studies, Outcome 5 Treatment‐related death.
Figuras y tablas -
Analysis 1.5

Comparison 1 All Studies, Outcome 5 Treatment‐related death.

Comparison 1 All Studies, Outcome 6 Adverse Events: Thrombocytopenia.
Figuras y tablas -
Analysis 1.6

Comparison 1 All Studies, Outcome 6 Adverse Events: Thrombocytopenia.

Comparison 1 All Studies, Outcome 7 Adverse Events: Neutropenia.
Figuras y tablas -
Analysis 1.7

Comparison 1 All Studies, Outcome 7 Adverse Events: Neutropenia.

Comparison 1 All Studies, Outcome 8 Adverse Events: Anaemia.
Figuras y tablas -
Analysis 1.8

Comparison 1 All Studies, Outcome 8 Adverse Events: Anaemia.

Comparison 1 All Studies, Outcome 9 Adverse Events: Nausea/Vomiting.
Figuras y tablas -
Analysis 1.9

Comparison 1 All Studies, Outcome 9 Adverse Events: Nausea/Vomiting.

Comparison 1 All Studies, Outcome 10 Adverse Events: Diarrhoea.
Figuras y tablas -
Analysis 1.10

Comparison 1 All Studies, Outcome 10 Adverse Events: Diarrhoea.

Comparison 1 All Studies, Outcome 11 Adverse Events: Constipation.
Figuras y tablas -
Analysis 1.11

Comparison 1 All Studies, Outcome 11 Adverse Events: Constipation.

Comparison 1 All Studies, Outcome 12 Adverse Events: Peripheral Neuropathy.
Figuras y tablas -
Analysis 1.12

Comparison 1 All Studies, Outcome 12 Adverse Events: Peripheral Neuropathy.

Comparison 1 All Studies, Outcome 13 Adverse Events: Infections (All).
Figuras y tablas -
Analysis 1.13

Comparison 1 All Studies, Outcome 13 Adverse Events: Infections (All).

Comparison 1 All Studies, Outcome 14 Adverse Events: Herpes Zoster infection.
Figuras y tablas -
Analysis 1.14

Comparison 1 All Studies, Outcome 14 Adverse Events: Herpes Zoster infection.

Comparison 1 All Studies, Outcome 15 Adverse Events: Cardiac Disorders.
Figuras y tablas -
Analysis 1.15

Comparison 1 All Studies, Outcome 15 Adverse Events: Cardiac Disorders.

Comparison 1 All Studies, Outcome 16 Adverse Events: Fatigue.
Figuras y tablas -
Analysis 1.16

Comparison 1 All Studies, Outcome 16 Adverse Events: Fatigue.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 1 Overall Survival.
Figuras y tablas -
Analysis 2.1

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 1 Overall Survival.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 2 Progression Free Survival.
Figuras y tablas -
Analysis 2.2

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 2 Progression Free Survival.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 3 Complete Response Rate.
Figuras y tablas -
Analysis 2.3

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 3 Complete Response Rate.

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 4 Overall Response Rate.
Figuras y tablas -
Analysis 2.4

Comparison 2 Subgroup Analyses ‐ Disease Setting, Outcome 4 Overall Response Rate.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 1 Overall Survival.
Figuras y tablas -
Analysis 3.1

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 1 Overall Survival.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 2 Progression Free Survival.
Figuras y tablas -
Analysis 3.2

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 2 Progression Free Survival.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 3 Complete Response Rate.
Figuras y tablas -
Analysis 3.3

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 3 Complete Response Rate.

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 4 Overall Response Rate.
Figuras y tablas -
Analysis 3.4

Comparison 3 Subgroup Analyses ‐ Therapy Setting, Outcome 4 Overall Response Rate.

Summary of findings for the main comparison. Bortezomib versus no bortezomib for the treatment of multiple myeloma

Bortezomib versus no bortezomib

Patient or population: All patients with a diagnosis of multiple myeloma
Setting: International multicentre studies
Intervention: Bortezomib
Comparison: Bortezomib versus no bortezomib (same or different background therapy or other agents)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk without bortezomib

Risk with bortezomib

Overall Survival

Study population

Peto Odds Ratio 0.77
(0.69 to 0.86)

4118
(9 RCTs)

⨁⨁⨁⨁
HIGH

215 per 1000

166 per 1000
(148 to 185)

Overall Survival ‐ Bortezomib versus no bortezomib (same background therapy)

Follow‐up 36 to 60 months

Study population

Peto Odds Ratio 0.77
(0.65 to 0.92)

1586
(4 RCTs)

⨁⨁⨁◯
MODERATE 1

354 per 1000

273 per 1000
(230 to 326)

Overall Survival ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Follow‐up 7.5 to 67 months

Study population

Peto Odds Ratio 0.76
(0.67 to 0.88)

2532
(5 RCTs)

⨁⨁⨁⨁
HIGH

129 per 1000

98 per 1000
(87 to 114)

Progression‐Free Survival

Study population

Peto Odds Ratio 0.67
(0.61 to 0.72)

4344
(9 RCTs)

⨁⨁◯◯
LOW 2 3

523 per 1000

350 per 1000
(319 to 377)

Progression‐Free Survival ‐ Bortezomib versus no bortezomib (same background therapy)

Follow‐up 30 to 60 months

Study population

Peto Odds Ratio 0.65
(0.57 to 0.74)

1855
(5 RCTs)

⨁⨁⨁◯
MODERATE 2

324 per 1000

211 per 1000
(185 to 240)

Progression‐Free Survival ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Follow‐up 22 to 67 months

Study population

Peto Odds Ratio 0.67
(0.61 to 0.75)

2489
(4 RCTs)

⨁⨁◯◯
LOW 2 4

669 per 1000

448 per 1000
(408 to 501)

Treatment‐related death

Study population

OR 0.76
(0.43 to 1.34)

2389
(5 RCTs)

⨁⨁◯◯
LOW 6

22 per 1000

17 per 1000
(10 to 29)

Moderate

27 per 1000

21 per 1000
(12 to 36)

Treatment‐related death ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 0.81
(0.30 to 2.16)

737
(2 RCTs)

⨁⨁◯◯
LOW 6

22 per 1000

18 per 1000
(7 to 47)

Moderate

35 per 1000

29 per 1000
(11 to 73)

Treatment‐related death ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 0.73
(0.36 to 1.48)

1652
(3 RCTs)

⨁◯◯◯
VERY LOW 6 7

22 per 1000

16 per 1000
(8 to 32)

Moderate

27 per 1000

20 per 1000
(10 to 40)

Health‐related quality of life

see comment

see comment

see comment

717

(4 RCTs)

see comment

Each trial used the same validated quality of life instrument (European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ‐C30), whereas the time points of administration of the questionnaire varied between the four trials.

Adverse Events: Thrombocytopenia

Study population

OR 2.05
(1.70 to 2.48)

3791
(8 RCTs)

⨁⨁◯◯
LOW 8

114 per 1000

209 per 1000
(180 to 242)

Moderate

48 per 1000

94 per 1000
(79 to 111)

Adverse Events: Thrombocytopenia ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 1.51
(1.13 to 2.00)

1196
(3 RCTs)

⨁⨁⨁⨁
HIGH

197 per 1000

271 per 1000
(217 to 330)

Moderate

47 per 1000

70 per 1000
(53 to 90)

Adverse Events: Thrombocytopenia ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 2.60
(2.01 to 3.35)

2595
(5 RCTs)

⨁⨁◯◯
LOW 9

76 per 1000

176 per 1000
(142 to 216)

Moderate

49 per 1000

118 per 1000
(93 to 147)

Adverse Events: Diarrhoea

Study population

OR 2.44
(1.74 to 3.43)

3788
(8 RCTs)

⨁⨁◯◯
LOW 5 10

26 per 1000

62 per 1000
(45 to 85)

Moderate

17 per 1000

39 per 1000
(28 to 54)

Adverse Events: Diarrhoea ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 6.24
(2.79 to 13.98)

1670
(4 RCTs)

⨁⨁⨁◯
MODERATE 5

8 per 1000

50 per 1000
(23 to 106)

Moderate

7 per 1000

41 per 1000
(19 to 87)

Adverse Events: Diarrhoea ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 1.80
(1.22 to 2.65)

2118
(4 RCTs)

⨁⨁⨁◯
MODERATE 10

40 per 1000

71 per 1000
(49 to 100)

Moderate

23 per 1000

40 per 1000
(28 to 58)

Adverse Events: Peripheral Neuropathy

Study population

OR 3.71
(2.92 to 4.70)

4636
(10 RCTs)

⨁⨁⨁◯
MODERATE 5

44 per 1000

145 per 1000
(118 to 176)

Moderate

80 per 1000

244 per 1000
(203 to 291)

Adverse Events: Peripheral Neuropathy ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 5.10
(3.37 to 7.72)

2040
(5 RCTs)

⨁⨁◯◯
LOW 5 10

31 per 1000

139 per 1000
(96 to 196)

Moderate

139 per 1000

453 per 1000
(353 to 556)

Adverse Events: Peripheral Neuropathy ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 3.09
(2.30 to 4.14)

2596
(5 RCTs)

⨁⨁⨁⨁
HIGH

54 per 1000

149 per 1000
(116 to 190)

Moderate

21 per 1000

62 per 1000
(47 to 81)

Adverse Events: Infections (All)

Study population

OR 1.51
(1.27 to 1.79)

4266
(9 RCTs)

⨁⨁⨁◯
MODERATE 11

128 per 1000

181 per 1000
(157 to 207)

Moderate

254 per 1000

339 per 1000
(302 to 378)

Adverse Events: Infections (All) ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 1.37
(0.97 to 1.93)

1670
(4 RCTs)

⨁⨁⨁⨁
HIGH

77 per 1000

103 per 1000
(75 to 139)

Moderate

57 per 1000

77 per 1000
(56 to 105)

Adverse Events: Infections (All) ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 1.55
(1.27 to 1.90)

2596
(5 RCTs)

⨁⨁◯◯
LOW 9

160 per 1000

228 per 1000
(194 to 265)

Moderate

209 per 1000

291 per 1000
(251 to 335)

Adverse Events: Cardiac Disorders

Study population

OR 1.74
(1.17 to 2.58)

2191
(5 RCTs)

⨁⨁⨁⨁
HIGH

38 per 1000

65 per 1000
(44 to 93)

Moderate

30 per 1000

51 per 1000
(35 to 74)

Adverse Events: Cardiac Disorders ‐ Bortezomib versus no bortezomib (same background therapy)

Study population

OR 1.17
(0.39 to 3.52)

736
(2 RCTs)

⨁⨁⨁◯
MODERATE 5

16 per 1000

19 per 1000
(6 to 55)

Moderate

14 per 1000

17 per 1000
(6 to 49)

Adverse Events: Cardiac Disorders ‐ Bortezomib versus no bortezomib (different background therapy or other agents)

Study population

OR 1.84
(1.21 to 2.81)

1455
(3 RCTs)

⨁⨁⨁⨁
HIGH

49 per 1000

87 per 1000
(59 to 127)

Moderate

49 per 1000

86 per 1000
(58 to 126)

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

CI: Confidence interval; RR: Risk ratio; OR: Odds ratio;

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

1 Downgraded one level due to heterogeneity 55%

2 Downgraded one level because TTP was analysed instead of PFS in one trial.

3 Downgraded one level due to heterogeneity 56%.

4 Downgraded one level due to heterogeneity 70% .

5 Downgraded one level due to low number of events, wide CI.

6 Downgraded two levels due to very low number of events, very wide CI.

7 Downgraded one level due to heterogeneity 53%.

8 Downgraded one level due to heterogeneity 81%.

9 Downgraded one level due to heterogeneity 86%.

10 Downgraded one level due to heterogeneity 57%.

11 Downgraded one level due to heterogeneity 76%.

Figuras y tablas -
Summary of findings for the main comparison. Bortezomib versus no bortezomib for the treatment of multiple myeloma
Comparison 1. All Studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

9

4118

Peto Odds Ratio (95% CI)

0.77 [0.69, 0.86]

1.1 Bortezomib versus no bortezomib (same background therapy)

4

1586

Peto Odds Ratio (95% CI)

0.77 [0.65, 0.92]

1.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2532

Peto Odds Ratio (95% CI)

0.76 [0.67, 0.88]

2 Progression‐Free Survival Show forest plot

9

4344

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.72]

2.1 Bortezomib versus no bortezomib (same background therapy)

5

1855

Peto Odds Ratio (95% CI)

0.65 [0.57, 0.74]

2.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2489

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.75]

3 Complete Response Rate Show forest plot

12

4630

Odds Ratio (M‐H, Fixed, 95% CI)

2.35 [2.02, 2.73]

3.1 Bortezomib versus no bortezomib (same background therapy)

6

2064

Odds Ratio (M‐H, Fixed, 95% CI)

2.63 [2.13, 3.24]

3.2 Bortezomib versus no bortezomib (different background therapy or other agents)

6

2566

Odds Ratio (M‐H, Fixed, 95% CI)

2.08 [1.67, 2.58]

4 Overall Response Rate Show forest plot

12

4630

Odds Ratio (M‐H, Fixed, 95% CI)

2.62 [2.25, 3.05]

4.1 Bortezomib versus no bortezomib (same background therapy)

6

2064

Odds Ratio (M‐H, Fixed, 95% CI)

3.45 [2.72, 4.37]

4.2 Bortezomib versus no bortezomib (different background therapy or other agents)

6

2566

Odds Ratio (M‐H, Fixed, 95% CI)

2.17 [1.78, 2.64]

5 Treatment‐related death Show forest plot

5

2389

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.43, 1.34]

5.1 Bortezomib versus no bortezomib (same background therapy)

2

737

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.30, 2.16]

5.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1652

Odds Ratio (M‐H, Fixed, 95% CI)

0.73 [0.36, 1.48]

6 Adverse Events: Thrombocytopenia Show forest plot

8

3791

Odds Ratio (M‐H, Fixed, 95% CI)

2.05 [1.70, 2.48]

6.1 Bortezomib versus no bortezomib (same background therapy)

3

1196

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [1.13, 2.00]

6.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2595

Odds Ratio (M‐H, Fixed, 95% CI)

2.60 [2.01, 3.35]

7 Adverse Events: Neutropenia Show forest plot

8

3791

Odds Ratio (M‐H, Fixed, 95% CI)

1.33 [1.10, 1.60]

7.1 Bortezomib versus no bortezomib (same background therapy)

3

1196

Odds Ratio (M‐H, Fixed, 95% CI)

0.95 [0.73, 1.24]

7.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2595

Odds Ratio (M‐H, Fixed, 95% CI)

1.85 [1.41, 2.41]

8 Adverse Events: Anaemia Show forest plot

6

3404

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.65, 1.00]

8.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

0.67 [0.48, 0.94]

8.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2465

Odds Ratio (M‐H, Fixed, 95% CI)

0.92 [0.69, 1.21]

9 Adverse Events: Nausea/Vomiting Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

2.37 [1.64, 3.42]

9.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

4.55 [1.99, 10.42]

9.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [1.28, 2.93]

10 Adverse Events: Diarrhoea Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

2.44 [1.74, 3.43]

10.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

6.24 [2.79, 13.98]

10.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.80 [1.22, 2.65]

11 Adverse Events: Constipation Show forest plot

8

3788

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.14, 2.22]

11.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

1.88 [1.04, 3.41]

11.2 Bortezomib versus no bortezomib (different background therapy or other agents)

4

2118

Odds Ratio (M‐H, Fixed, 95% CI)

1.47 [0.98, 2.20]

12 Adverse Events: Peripheral Neuropathy Show forest plot

10

4636

Odds Ratio (M‐H, Fixed, 95% CI)

3.71 [2.92, 4.70]

12.1 Bortezomib versus no bortezomib (same background therapy)

5

2040

Odds Ratio (M‐H, Fixed, 95% CI)

5.10 [3.37, 7.72]

12.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2596

Odds Ratio (M‐H, Fixed, 95% CI)

3.09 [2.30, 4.14]

13 Adverse Events: Infections (All) Show forest plot

9

4266

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [1.27, 1.79]

13.1 Bortezomib versus no bortezomib (same background therapy)

4

1670

Odds Ratio (M‐H, Fixed, 95% CI)

1.37 [0.97, 1.93]

13.2 Bortezomib versus no bortezomib (different background therapy or other agents)

5

2596

Odds Ratio (M‐H, Fixed, 95% CI)

1.55 [1.27, 1.90]

14 Adverse Events: Herpes Zoster infection Show forest plot

4

1733

Odds Ratio (M‐H, Fixed, 95% CI)

1.83 [0.91, 3.67]

14.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [0.74, 5.03]

14.2 Bortezomib versus no bortezomib (different background therapy or other agents)

2

794

Odds Ratio (M‐H, Fixed, 95% CI)

1.71 [0.62, 4.74]

15 Adverse Events: Cardiac Disorders Show forest plot

5

2191

Odds Ratio (M‐H, Fixed, 95% CI)

1.74 [1.17, 2.58]

15.1 Bortezomib versus no bortezomib (same background therapy)

2

736

Odds Ratio (M‐H, Fixed, 95% CI)

1.17 [0.39, 3.52]

15.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1455

Odds Ratio (M‐H, Fixed, 95% CI)

1.84 [1.21, 2.81]

16 Adverse Events: Fatigue Show forest plot

5

2926

Odds Ratio (M‐H, Fixed, 95% CI)

1.96 [1.35, 2.84]

16.1 Bortezomib versus no bortezomib (same background therapy)

2

939

Odds Ratio (M‐H, Fixed, 95% CI)

3.30 [1.66, 6.58]

16.2 Bortezomib versus no bortezomib (different background therapy or other agents)

3

1987

Odds Ratio (M‐H, Fixed, 95% CI)

1.52 [0.97, 2.38]

Figuras y tablas -
Comparison 1. All Studies
Comparison 2. Subgroup Analyses ‐ Disease Setting

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

8

4075

Peto Odds Ratio (95% CI)

0.77 [0.70, 0.86]

1.1 Relapsed / Refactory

1

669

Peto Odds Ratio (95% CI)

0.77 [0.61, 0.97]

1.2 Transplant Eligible

5

2213

Peto Odds Ratio (95% CI)

0.86 [0.73, 1.02]

1.3 Transplant Ineligible

2

1193

Peto Odds Ratio (95% CI)

0.70 [0.59, 0.82]

2 Progression Free Survival Show forest plot

11

4344

Peto Odds Ratio (95% CI)

0.67 [0.61, 0.72]

2.1 Relapsed / Refactory

3

938

Peto Odds Ratio (95% CI)

0.57 [0.48, 0.68]

2.2 Transplant Eligible

6

2213

Peto Odds Ratio (95% CI)

0.76 [0.68, 0.85]

2.3 Transplant Ineligible

2

1193

Peto Odds Ratio (95% CI)

0.57 [0.49, 0.67]

3 Complete Response Rate Show forest plot

11

4593

Odds Ratio (M‐H, Fixed, 95% CI)

2.38 [2.05, 2.77]

3.1 Relapsed / Refactory

3

998

Odds Ratio (M‐H, Fixed, 95% CI)

3.35 [2.06, 5.43]

3.2 Transplant Eligible

6

2424

Odds Ratio (M‐H, Fixed, 95% CI)

1.88 [1.56, 2.27]

3.3 Transplant Ineligibile

2

1171

Odds Ratio (M‐H, Fixed, 95% CI)

3.69 [2.71, 5.02]

4 Overall Response Rate Show forest plot

11

4593

Odds Ratio (M‐H, Fixed, 95% CI)

2.61 [2.24, 3.05]

4.1 Relapsed / Refactory

3

998

Odds Ratio (M‐H, Fixed, 95% CI)

2.39 [1.79, 3.20]

4.2 Transplant Eligible

6

2424

Odds Ratio (M‐H, Fixed, 95% CI)

2.24 [1.76, 2.85]

4.3 Transplant Ineligibile

2

1171

Odds Ratio (M‐H, Fixed, 95% CI)

3.45 [2.63, 4.53]

Figuras y tablas -
Comparison 2. Subgroup Analyses ‐ Disease Setting
Comparison 3. Subgroup Analyses ‐ Therapy Setting

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall Survival Show forest plot

5

Peto Odds Ratio (95% CI)

Subtotals only

1.1 Induction

3

1783

Peto Odds Ratio (95% CI)

0.81 [0.67, 0.96]

1.2 Consolidation

2

844

Peto Odds Ratio (95% CI)

1.02 [0.71, 1.47]

1.3 Maintenance

2

1338

Peto Odds Ratio (95% CI)

0.76 [0.64, 0.91]

2 Progression Free Survival Show forest plot

5

Peto Odds Ratio (95% CI)

Subtotals only

2.1 Induction

3

1783

Peto Odds Ratio (95% CI)

0.75 [0.66, 0.85]

2.2 Consolidation

2

844

Peto Odds Ratio (95% CI)

0.70 [0.58, 0.84]

2.3 Maintenance

2

1338

Peto Odds Ratio (95% CI)

0.70 [0.61, 0.79]

3 Complete Response Rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Induction

4

1999

Odds Ratio (M‐H, Fixed, 95% CI)

2.04 [1.65, 2.53]

3.2 Consolidation

2

839

Odds Ratio (M‐H, Fixed, 95% CI)

1.77 [1.34, 2.33]

3.3 Maintenance

2

1330

Odds Ratio (M‐H, Fixed, 95% CI)

1.82 [1.44, 2.31]

4 Overall Response Rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 Induction

4

1999

Odds Ratio (M‐H, Fixed, 95% CI)

2.33 [1.82, 2.99]

4.2 Consolidation

2

839

Odds Ratio (M‐H, Fixed, 95% CI)

2.21 [1.20, 4.06]

4.3 Maintenance

2

1330

Odds Ratio (M‐H, Fixed, 95% CI)

1.90 [1.38, 2.62]

Figuras y tablas -
Comparison 3. Subgroup Analyses ‐ Therapy Setting