Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.
The REMoDL-B phase III adaptive trial compared rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone (R-CHOP) versus R-CHOP + bortezomib (RB-CHOP) in patients with diffuse large B-cell lymphoma (DLBCL), stratified by molecular subtype. Primary analysis at a median follow-up of 30 months found no effect of bortezomib on progression-free survival (PFS) or overall survival (OS). Retrospective analysis using a gene expression–based classifier identified a molecular high-grade (MHG) group with worse outcomes. We present an updated analysis for patients successfully classified by the gene expression profile (GEP). Eligible patients were age older than 18 years with untreated DLBCL, fit enough for full-dose chemotherapy, and with adequate biopsies for GEP. Of 1,077 patients registered, 801 were identified with Activated B-Cell (ABC), Germinal Center B-cell, or MHG lymphoma. At a median follow-up of 64 months, there was no overall benefit of bortezomib on PFS or OS (5-year PFS hazard ratio [HR], 0.81; P = .085; OS HR, 0.86; P = .32). However, improved PFS and OS were seen in ABC lymphomas after RB-CHOP: 5-year OS 67% with R-CHOP versus 80% with RB-CHOP (HR, 0.58; 95% CI, 0.35 to 0.95; P = .032). Five-year PFS was higher in MHG lymphomas: 29% versus 55% (HR, 0.46; 95% CI, 0.26 to 0.84). Patients with ABC and MHG DLBCL may benefit from the addition of bortezomib to R-CHOP in initial therapy.
INTRODUCTION
Molecular heterogeneity is a recognized feature of diffuse large B-cell lymphoma (DLBCL), with varying outcomes among karyotypic, genomic, and transcriptomic subtypes.1-4 Clinical trials have tested whether additional targeted therapies might improve outcomes, by modulating aberrant intracellular pathways in malignant B cells.5-7 The REMoDL-B trial compared rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone (R-CHOP) versus R-CHOP + bortezomib (RB-CHOP) in patients with newly diagnosed DLBCL. Primary analysis at a median follow-up of 30 months found no difference in progression-free survival (PFS) between the two treatment arms (hazard ratio [HR], 0.86; 95% CI, 0.65 to 1.13).8 Subsequent analysis of biopsies using a gene expression–based classifier identified a more aggressive subtype (molecular high-grade [MHG]) characterized by a proliferative phenotype closely related to centroblasts, which showed a trend toward therapeutic benefit from bortezomib (PFS HR, 0.58; 95% CI, 0.31 to 1.07).9
This article updates the trial results after the full 5-year follow-up of all patients whose lymphomas were successfully classified by gene expression profile (GEP).
METHODS
Details of the REMoDL-B design and primary analysis have been published.8 REMoDL-B was an open-label randomized phase III adaptive trial, which recruited from centers in the United Kingdom and Switzerland. Participants had DLBCL with sufficient diagnostic material from initial biopsies for GEP and pathology review; were age 18 years or older; had an Eastern Cooperative Oncology Group performance status of ≤2; bulky stage I or stage II–IV disease; measurable disease, and cardiac, lung, renal, and liver function sufficient to tolerate full-dose chemotherapy.
All patients received R-CHOP for one cycle and were randomly assigned to R-CHOP or R-CHOP + bortezomib for cycles 2-6, stratified by International Prognostic Index (IPI) and cell of origin (COO), determined by GEP. Biopsies were analyzed by fluorescence in situ hybridization for MYC, BCL2, and BCL6 rearrangements and by targeted genomic analysis using enrichment of 70 genes recurrently mutated in lymphomas by customized HaloPlex HS probe library (Agilent Technologies, Santa Clara, CA).
Whole-genome GEP was performed on mRNA extracted from diagnostic tissue by Illumina DASL array (Cambridge, UK). Classification in the trial was by DLBCL automatic classifier10 in real time. Biopsies were categorized as activated B-cell (ABC), germinal center B-cell (GCB), or unclassifiable—the MHG group was not characterized at initial implementation. The COO classification was also analyzed retrospectively, with improved data normalization over the complete trial data set to classify patients as ABC, GCB, MHG, or unclassifiable. The analysis of results with extended follow-up prespecified these subgroups, for consistency with the previous paper identifying the MHG group.9 All patients were followed for 5 years after treatment ended. This trial is registered with ClinicalTrials.gov identifier: NCT01324596.
The primary end point was PFS. The sample size calculation of the trial has been previously reported.1 All analyses followed a prespecified statistical analysis plan. Time-to-event analyses by trial arm were summarized using the Kaplan-Meier method and Cox regression modeling (including IPI and where applicable COO), both overall and within the subgroups. 95% confidence intervals for HR for the treatment effect were estimated from the Cox model and a two-sided P value of <.05 used to define statistical significance. To assess for differences in treatment effect by subgroups, heterogeneity and interaction tests were performed. No adjustment for multiple testing was performed. We used Stata statistical software v17 and SAS v9.4 (College Station, TX) for all analyses.
RESULTS
Survival Outcomes According to Molecular Subtype
In the trial, 1,129 patients were registered between June 2011 and June 2015. Of these, 1,077 underwent GEP, with 801 retrospectively classified as ABC, GCB, or MHG. 407 were randomly assigned to R-CHOP, and 394 to RB-CHOP, an increase of 82 from the 719 classified prospectively for random assignment as ABC or GCB. Seventy-eight of 199 biopsies assessed as unclassifiable at random assignment could be allocated to ABC, GCB, or MHG. Five of 719 allocated to ABC or GCB prospectively could not be classified retrospectively. Eight of 15 who had initially unsuccessful GEP could be allocated as ABC, GCB, or MHG. One GCB patient randomly assigned to RB-CHOP had not provided data for primary analysis but provided data for the long-term follow-up. Full details of prospective and retrospective classifications are provided in the Data Supplement (online only).9
Baseline characteristics of the patients were well-balanced (Table 1).
TABLE 1.
At a median follow-up of 64 months for survivors, there was no overall benefit of bortezomib in PFS or OS among all the successfully classified patients (Data Supplement).
Improved PFS was seen with bortezomib in MHG and ABC lymphomas. With additional follow-up, 95 patients with ABC lymphoma have progressed or died, an increase of 22 (13 after R-CHOP and nine after RB-CHOP) from the previous report, resulting in a 60-month PFS of 54.4% (95% CI, 45.1 to 62.8) after R-CHOP versus 69.4% (95% CI, 60.2 to 76.9) after RB-CHOP (HR, 0.65; 95% CI, 0.43 to 0.98; Fig 1A). Six patients with MHG lymphomas have progressed since the previous analysis (five after R-CHOP and one after RB-CHOP), leading to a 60-month PFS of 29.3% (95% CI, 16.4 to 43.5) after R-CHOP versus 54.9% (95% CI, 38.3 to 68.7) after RB-CHOP (HR, 0.46; 95% CI, 0.26 to 0.84; Fig 1C).
A 60-month OS advantage was seen in the ABC group. Sixty-four of 249 patients with ABC lymphomas have died (21 more than the previous analysis: 15 after R-CHOP and six after RB-CHOP, Data Supplement), leading to a 60-month OS of 67.4% (95% CI, 58.2 to 75.0) after R-CHOP versus 80.4% (95% CI, 72.0 to 86.5) after RB-CHOP (HR, 0.58; 95% CI, 0.35 to 0.95; Fig 1D). Thirty-seven of 83 patients with MHG lymphomas have died (four more, two on each arm), leading to a 60-month OS of 47.5% (95% CI, 31.5 to 61.9) after R-CHOP versus 60.0% (95% CI, 43.2 to 73.3) after RB-CHOP (HR, 0.62; 95% CI, 0.32 to 1.20; Fig 1F).
No PFS or OS difference was observed in the GCB group according to the treatment arm (Figs 1B and 1E). There was a small increase in lymphoma-related deaths in the group treated with RB-CHOP (27 [21.8%] v 18 [14.4%]; Data Supplement).
Subgroups characterized by double-hit cytogenetics, with MYC and BCL2 translocation, and by high levels of MYC and BCL2 mRNA showed similar PFS results to those previously reported (Fig 2). Those with high MYC and BCL2 mRNA show an increase in PFS with RB-CHOP (HR, 0.61; 95% CI, 0.39 to 0.96). Four hundred patients could be retrospectively analyzed using the LymphGen algorithm11 although 179 (45%) of these could not be classified (Data Supplement). There was no clear difference in outcomes by treatment arm in any LymphGen subgroup (Data Supplement).
Adverse Events and Second Cancers
Adverse events were similar to those reported previously.8 The addition of bortezomib was well-tolerated (Data Supplement). RB-CHOP was not associated with increased hematologic toxicity, and 398 (87.1%) of 459 participants assigned to RB-CHOP completed six cycles of treatment. Neuropathy of any grade was more common after RB-CHOP, but grade 3 or worse neuropathy was reported in 17 (3.8%) patients who were given RB-CHOP versus 10 (2.2%) who were given R-CHOP. Serious adverse events occurred in 190 (42.5%) patients who were given R-CHOP, including five treatment-related deaths, and 225 (50.7%) who were given RB-CHOP, including four treatment-related deaths.
Of 1,041 patients included in safety analyses, there were 20 second cancers reported, 14 after R-CHOP (2.3%) and six after RB-CHOP (1.4%; see the Data Supplement).
DISCUSSION
This study confirms that different molecular subtypes of DLBCL exhibit different responses to bortezomib when given in combination with R-CHOP and specifically that the ABC and MHG subtypes show improvements in PFS after the addition of bortezomib, which is also reflected in improved OS for the ABC group (60-month PFS difference 15%; OS difference 13%). Conversely, no such effect was seen in the GCB group. Targeting of constitutive NFkB pathway activation provides a potential explanation for the effect of bortezomib in ABC-DLBCL. However, this mechanism is unlikely to explain the effect in MHG. An alternate explanation for the therapeutic effect across these disparate subtypes may lie in MYC-driven proteotoxic stress, rendering cells sensitive to bortezomib-mediated proteasome inhibition.
This is one of the largest studies conducted in DLBCL using GEP in real time for stratified random assignment. However, the proportion of MHG lymphoma in DLBCL is relatively small (approximately 10%), with the result that even in this trial, only 83 cases were identified, limiting the ability to detect important differences in survival.
With mature follow-up, this study suggests a benefit from the addition of bortezomib to R-CHOP for ABC and MHG subtypes of DLBCL. Ideally, this should be confirmed in a prospective trial, with a more potent proteasome inhibitor carrying less risk of additive neurotoxicity.
ACKNOWLEDGMENT
Bortezomib was supplied free of charge by Janssen-Cilag. C.M. and U.N. are members of the Swiss Group for Clinical Cancer Research, Switzerland.
Andrew J. Davies
Honoraria: Gilead Sciences, Kite, a Gilead company, AbbVie, Incyte, Roche, Celgene/Bristol Myers Squibb, Genmab, AstraZeneca
Consulting or Advisory Role: Gilead Sciences, Roche, Acerta Pharma, Kite, a Gilead company, Janssen, Celgene, Regeneron, AbbVie
Research Funding: Roche (Inst), Gilead Sciences (Inst), GlaxoSmithKline (Inst), Takeda (Inst), Pfizer (Inst), Celgene (Inst), Karyopharm Therapeutics (Inst), Acerta Pharma (Inst), MSD
Travel, Accommodations, Expenses: Roche
Urban Novak
Consulting or Advisory Role: Roche, Gilead Sciences, Celgene, Takeda, AstraZeneca
Travel, Accommodations, Expenses: Celgene, Takeda, Amgen, Roche
Andrew McMillan
Honoraria: Roche/Genentech, Amgen, Protherics, Takeda
Consulting or Advisory Role: Amgen
Speakers' Bureau: Roche/Genentech
Research Funding: Pfizer, Roche/Genentech (Inst)
Travel, Accommodations, Expenses: Takeda
Uncompensated Relationships: Kite/Gilead
Uncompensated Relationships: Novartis
Graham P. Collins
Honoraria: Roche, Takeda, Gilead Sciences, Pfizer, Novartis, Daiichi Sankyo, Incyte, Celleron Therapeutics, MSD Oncology, BeiGene, ADC Therapeutics, AstraZeneca
Consulting or Advisory Role: Roche, Takeda, Incyte, Pfizer, MSD, Celgene, BeiGene, Daiichi Sankyo, Celleron Therapeutics, ADC Therapeutics
Speakers' Bureau: Roche, Takeda, Novartis, Gilead Sciences
Research Funding: MSD Oncology (Inst), Celgene (Inst), BMS (Inst), Amgen (Inst), Pfizer (Inst)
Travel, Accommodations, Expenses: Roche, Takeda
Moniek van Hoppe
Employment: Leeds Teaching Hospitals NHS Trust
Reuben Tooze
Honoraria: Roche, Celgene
Consulting or Advisory Role: Medicxi
Research Funding: UCB
Travel, Accommodations, Expenses: Celgene
Gareth Griffiths
Honoraria: AstraZeneca, AbbVie
Research Funding: AstraZeneca (Inst), Astex Pharmaceuticals (Inst), Bristol Myers Squibb (Inst), Jansen-Cilag (Inst), Novartis (Inst), Roche (Inst), HeartFlow (Inst), BioNTech (Inst), GlaxoSmithKline (Inst), HeartFlow (Inst)
Anna Schuh
Stock and Other Ownership Interests: Illumina, SERENOx
Consulting or Advisory Role: AbbVie, Roche, Janssen, AstraZeneca
Research Funding: Johnson & Johnson, Illumina (Inst), Oxford Nanopore Technologies (Inst)
Travel, Accommodations, Expenses: AbbVie, Janssen Oncology
Catherine Burton
Honoraria: Takeda, Roche
Consulting or Advisory Role: Celgene, Bristol Myers Squibb
Speakers' Bureau: Roche, Celgene
Travel, Accommodations, Expenses: Takeda Science Foundation
Ming-Qing Du
Employment: GlaxoSmithKline
Peter W.M. Johnson
Honoraria: Epizyme, InCyte
Consulting or Advisory Role: Epizyme
Patents, Royalties, Other Intellectual Property: Combined use of Fc gamma RIIb (CD32b) and CD20 - specific antibodies. WO Patent, PCT/GB2011/051572; EU11760819.0
No other potential conflicts of interest were reported.
PRIOR PRESENTATION
Presented in part at the Annual Meeting of the American Society of Hematology, New Orleans, LA, December 10-13, 2022.
SUPPORT
Supported by a grant from Janssen-Cilag and endorsed by Cancer Research UK (C328/A12128). Translational work was supported by a Blood Cancer UK specialist program grant to the Precision Medicine for Aggressive Lymphoma consortium (15002).The Southampton Clinical Trials Unit receives core funding from Cancer Research UK. The study was supported by the National Institute for Healthcare Research and Cancer Research UK Experimental Cancer Medicine Centres at Southampton, Oxford, and Cambridge, UK.
CLINICAL TRIAL INFORMATION
NCT01324596 (REMoDL-B)
DATA SHARING STATEMENT
Individual participant data will be made available, including data dictionaries, for approved data sharing requests. Individual participant data will be shared that underlie the results reported in this article, after deidentification and normalization of information (text, tables, figures, and appendices). The statistical analysis plan will be provided upon request. Anonymized data will be available beginning 3 months after and ending 5 years after publication of this article to researchers who provide a completed Data Sharing Agreement that describes a methodologically sound proposal for the purpose of the approved proposal. Proposals should be directed to ctu@soton.ac.uk. Data will be shared once all relevant parties approve and sign the Data Sharing Agreement. Data sharing requests are available for 5 years via the Southampton Clinical Trials Unit website.
AUTHOR CONTRIBUTIONS
Conception and design: Andrew J. Davies, Louise Stanton, Richard Stephens, Peter W.M. Johnson
Financial support: Ming-Qing Du
Administrative support: Josh Caddy
Provision of study materials or patients: Andrew J. Davies, Urban Novak, Graham P. Collins, Peter W.M. Johnson
Collection and assembly of data: Andrew J. Davies, Sharon Barrans, Louise Stanton, Josh Caddy, Christoph Mamot, Andrew McMillan, Graham P. Collins, Moniek van Hoppe, Catherine Burton, Ming-Qing Du, Peter W.M. Johnson
Data analysis and interpretation: Andrew J. Davies, Louise Stanton, Sam Wilding, Geoff Saunders, Urban Novak, Andrew McMillan, Paul Fields, Graham P. Collins, Francesco Cucco, Chulin Sha, Reuben Tooze, John R. Davies, Gareth Griffiths, Anna Schuh, David R. Westhead, Ming-Qing Du, Peter W.M. Johnson
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Differential Efficacy From the Addition of Bortezomib to R-CHOP in Diffuse Large B-Cell Lymphoma According to the Molecular Subgroup in the REMoDL-B Study With a 5-Year Follow-Up
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Andrew J. Davies
Honoraria: Gilead Sciences, Kite, a Gilead company, AbbVie, Incyte, Roche, Celgene/Bristol Myers Squibb, Genmab, AstraZeneca
Consulting or Advisory Role: Gilead Sciences, Roche, Acerta Pharma, Kite, a Gilead company, Janssen, Celgene, Regeneron, AbbVie
Research Funding: Roche (Inst), Gilead Sciences (Inst), GlaxoSmithKline (Inst), Takeda (Inst), Pfizer (Inst), Celgene (Inst), Karyopharm Therapeutics (Inst), Acerta Pharma (Inst), MSD
Travel, Accommodations, Expenses: Roche
Urban Novak
Consulting or Advisory Role: Roche, Gilead Sciences, Celgene, Takeda, AstraZeneca
Travel, Accommodations, Expenses: Celgene, Takeda, Amgen, Roche
Andrew McMillan
Honoraria: Roche/Genentech, Amgen, Protherics, Takeda
Consulting or Advisory Role: Amgen
Speakers' Bureau: Roche/Genentech
Research Funding: Pfizer, Roche/Genentech (Inst)
Travel, Accommodations, Expenses: Takeda
Uncompensated Relationships: Kite/Gilead
Uncompensated Relationships: Novartis
Graham P. Collins
Honoraria: Roche, Takeda, Gilead Sciences, Pfizer, Novartis, Daiichi Sankyo, Incyte, Celleron Therapeutics, MSD Oncology, BeiGene, ADC Therapeutics, AstraZeneca
Consulting or Advisory Role: Roche, Takeda, Incyte, Pfizer, MSD, Celgene, BeiGene, Daiichi Sankyo, Celleron Therapeutics, ADC Therapeutics
Speakers' Bureau: Roche, Takeda, Novartis, Gilead Sciences
Research Funding: MSD Oncology (Inst), Celgene (Inst), BMS (Inst), Amgen (Inst), Pfizer (Inst)
Travel, Accommodations, Expenses: Roche, Takeda
Moniek van Hoppe
Employment: Leeds Teaching Hospitals NHS Trust
Reuben Tooze
Honoraria: Roche, Celgene
Consulting or Advisory Role: Medicxi
Research Funding: UCB
Travel, Accommodations, Expenses: Celgene
Gareth Griffiths
Honoraria: AstraZeneca, AbbVie
Research Funding: AstraZeneca (Inst), Astex Pharmaceuticals (Inst), Bristol Myers Squibb (Inst), Jansen-Cilag (Inst), Novartis (Inst), Roche (Inst), HeartFlow (Inst), BioNTech (Inst), GlaxoSmithKline (Inst), HeartFlow (Inst)
Anna Schuh
Stock and Other Ownership Interests: Illumina, SERENOx
Consulting or Advisory Role: AbbVie, Roche, Janssen, AstraZeneca
Research Funding: Johnson & Johnson, Illumina (Inst), Oxford Nanopore Technologies (Inst)
Travel, Accommodations, Expenses: AbbVie, Janssen Oncology
Catherine Burton
Honoraria: Takeda, Roche
Consulting or Advisory Role: Celgene, Bristol Myers Squibb
Speakers' Bureau: Roche, Celgene
Travel, Accommodations, Expenses: Takeda Science Foundation
Ming-Qing Du
Employment: GlaxoSmithKline
Peter W.M. Johnson
Honoraria: Epizyme, InCyte
Consulting or Advisory Role: Epizyme
Patents, Royalties, Other Intellectual Property: Combined use of Fc gamma RIIb (CD32b) and CD20 - specific antibodies. WO Patent, PCT/GB2011/051572; EU11760819.0
No other potential conflicts of interest were reported.
REFERENCES
- 1.Alizadeh AA, Eisen MB, Davis RE, et al. : Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503-511, 2000 [DOI] [PubMed] [Google Scholar]
- 2.Chapuy B, Stewart C, Dunford AJ, et al. : Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med 24:679-690, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Morin RD, Arthur SE, Hodson DJ: Molecular profiling in diffuse large B-cell lymphoma: Why so many types of subtypes? Br J Haematol 196:814-829, 2022 [DOI] [PubMed] [Google Scholar]
- 4.Schmitz R, Wright GW, Huang DW, et al. : Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med 378:1396-1407, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Offner F, Samoilova O, Osmanov E, et al. : Frontline rituximab, cyclophosphamide, doxorubicin, and prednisone with bortezomib (VR-CAP) or vincristine (R-CHOP) for non-GCB DLBCL. Blood 126:1893-1901, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nowakowski GS, Chiappella A, Gascoyne RD, et al. : Robust: A phase III study of lenalidomide plus R-CHOP versus placebo plus R-CHOP in previously untreated patients with ABC-type diffuse large B-cell lymphoma. J Clin Oncol 39:1317-1328, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Younes A, Sehn LH, Johnson P, et al. : Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol 37:1285-1295, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Davies A, Cummin TE, Barrans S, et al. : Gene-expression profiling of bortezomib added to standard chemoimmunotherapy for diffuse large B-cell lymphoma (REMoDL-B): An open-label, randomised, phase 3 trial. Lancet Oncol 20:649-662, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sha C, Barrans S, Cucco F, et al. : Molecular high-grade B-cell lymphoma: Defining a poor-risk group that requires different approaches to therapy. J Clin Oncol 37:202-212, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Care MA, Barrans S, Worrillow L, et al. : A microarray platform-independent classification tool for cell of origin class allows comparative analysis of gene expression in diffuse large B-cell lymphoma. PLoS One 8:e55895, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wright GW, Huang DW, Phelan JD, et al. : A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications. Cancer Cell 37:551-568, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Individual participant data will be made available, including data dictionaries, for approved data sharing requests. Individual participant data will be shared that underlie the results reported in this article, after deidentification and normalization of information (text, tables, figures, and appendices). The statistical analysis plan will be provided upon request. Anonymized data will be available beginning 3 months after and ending 5 years after publication of this article to researchers who provide a completed Data Sharing Agreement that describes a methodologically sound proposal for the purpose of the approved proposal. Proposals should be directed to ctu@soton.ac.uk. Data will be shared once all relevant parties approve and sign the Data Sharing Agreement. Data sharing requests are available for 5 years via the Southampton Clinical Trials Unit website.