YOSEMITE and RHINE: Phase 3 Randomized Clinical Trials of Faricimab for Diabetic Macular Edema: Study Design and Rationale

Nicole Eter, Rishi P Singh, Francis Abreu, Kemal Asik, Karen Basu, Caroline Baumal, Andrew Chang, Karl G Csaky, Zdenka Haskova, Hugh Lin, Carlos Quezada Ruiz, Paisan Ruamviboonsuk, David Silverman, Charles C Wykoff, Jeffrey R Willis, Nicole Eter, Rishi P Singh, Francis Abreu, Kemal Asik, Karen Basu, Caroline Baumal, Andrew Chang, Karl G Csaky, Zdenka Haskova, Hugh Lin, Carlos Quezada Ruiz, Paisan Ruamviboonsuk, David Silverman, Charles C Wykoff, Jeffrey R Willis

Abstract

Purpose: Faricimab is a novel anti-angiopoietin-2 and anti-vascular endothelial growth factor (VEGF) bispecific antibody with high affinities and specificities for both VEGF and angiopoietin-2. It is postulated that targeting angiogenic factors and inflammatory pathways in addition to the VEGF pathway will increase treatment durability and improve outcomes. The phase 3 YOSEMITE (ClinicalTrials.gov identifier, NCT03622580) and RHINE (ClinicalTrials.gov identifier, NCT03622593) trials are designed to assess efficacy, safety, and durability of faricimab compared with aflibercept in patients with diabetic macular edema (DME). The trials evaluate a personalized treatment interval (PTI) approach to address heterogeneity in treatment response among patients with DME.

Design: Two identically designed, global, double-masked, randomized, controlled phase 3 trials (YOSEMITE and RHINE).

Participants: Adults with center-involving DME secondary to type 1 or 2 diabetes mellitus.

Methods: These studies were designed to evaluate 3 treatment groups: faricimab 6.0 mg dosed either at fixed dosing every 8 weeks after initial treatment with 6 intravitreal doses at 4-week intervals, or faricimab 6.0 mg dosed according to PTI after initial treatment with 4 every-4-week doses, compared with aflibercept 2.0 mg dosed every 8 weeks after 5 initial every-4-week doses. The primary end point of the studies was change from baseline in best-corrected visual acuity at 1 year, averaged over weeks 48, 52, and 56. Secondary end points included anatomic, durability, and patient-reported outcomes. Safety outcomes included incidence and severity of ocular and nonocular adverse events. The PTI is a protocol-defined flexible regimen based on the treat-and-extend concept, which allowed up to every-16-week adjustable dosing based on objective and standardized criteria. The PTI design aimed to maximize therapeutic results while minimizing treatment burden.

Main outcome measures: We describe the rationale for the study design and the novel PTI (up to every-16-week adjustable dosing) approach for treatment with faricimab.

Results: YOSEMITE and RHINE enrolled 940 and 951 patients, respectively. Results from each study will be reported separately.

Conclusions: YOSEMITE and RHINE were the first registrational trials in retinal disease to incorporate an objective PTI regimen, allowing for up to every-16-week adjustable dosing with a dual angiopoietin-2 and VEGF-A inhibitor, faricimab 6.0 mg, for treatment of DME.

Keywords: AE, adverse event; Adjustable dosing; Angiopoietin-2; Anti–vascular endothelial growth factor; BCVA, best-corrected visual acuity; Bispecific antibody; CFP, color fundus photography; CRC, central reading center; CST, central subfield thickness; DME, diabetic macular edema; DR, diabetic retinopathy; Diabetic macular edema; ETDRS, Early Treatment Diabetic Retinopathy Study; FFA, fundus fluorescein angiography; Faricimab; ITT, intention-to-treat; IxRS, interactive voice or web-based response system; PRN, pro re nata; PTI, personalized treatment interval; Personalized treatment interval; Phase 3 clinical trial design; SD, spectral-domain; T&E, treat-and-extend; VEGF, vascular endothelial growth factor.

© 2021 by the American Academy of Ophthalmology.

Figures

Figure 1
Figure 1
Diagram showing study design overview. ∗The personalized treatment interval (PTI) is a protocol-driven regimen based on the treat-and-extend concept. †Change from baseline in best-corrected visual acuity (BCVA), as measured on the ETDRS chart at a starting distance of 4 m at 1 year, is the average of the week 48, 52, and 56 visits. Q8W = every 8 weeks; R = randomization.
Figure 2
Figure 2
Decision tree for interactive voice or web-based response system–determined personalized treatment interval arm dosing intervals. ∗The first central subfield thickness (CST) value that is †The mean of the 3 best best-corrected visual acuity (BCVA) scores obtained at any previous active dosing visit. Q4W = every 4 weeks; Q16W = every 16 weeks.
Figure 3
Figure 3
Graphs showing personalized treatment interval scenario examples. Horizontal line represents central subfield thickness (CST) threshold of 325 μm. Weeks in boldface indicate when active treatment was administered. A, Central subfield thickness threshold of < 325 μm (represented by solid horizontal line) not met: patient continues every-4-week (Q4W) treatment. B, Week 12: CST < 325 μm, extend to every 8 weeks (Q8W); week 20: CST within ±10% of reference CST∗ (with no associated ≥10-letter best-corrected visual acuity [BCVA] decrease from reference BCVA†), extend to every 12 weeks (Q12W); week 32: within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), extend to every 16 weeks (Q16W); week 48: CST within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), maintain at Q16W. C, Week 12: CST < 325 μm, extend to Q8W; week 20: CST within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), extend to Q12W; week 32: CST within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), extend to Q16W; week 48: CST increased by more than 10%, but no more than 20% (with an associated ≥5-letter to <10-letter BCVA decrease), reduce to Q12W. D, Week 12: CST < 325 μm, extend to Q8W; week 20: CST within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), extend to Q12W; week 32: CST within ±10% of reference CST (with no associated ≥10-letter BCVA decrease from reference BCVA), extend to Q16W; week 48: CST increased by more than 10% (with an associated ≥10-letter BCVA decrease from reference), reduce to Q8W. ∗The first CST value that is < 325 μm (defined as the central 1-mm thickness from the internal limiting membrane to Bruch’s membrane), starting at week 12. Reference CST is adjusted if CST decreases by >10% from the previous reference CST for 2 consecutive study drug dosing visits and the values obtained are within 30 μm. The CST value obtained at the latter visit serves as the new reference CST. †The mean of the 3 best BCVA scores obtained at any previous active dosing visit. ETDRS = Early Treatment Diabetic Retinopathy Study.
Figure 4
Figure 4
Graphs showing that low anti–vascular endothelial growth factor (VEGF) injection frequency in clinical practice versus clinical trials correlates with suboptimal outcomes. Adapted from Ciulla TA, Pollack JS, Williams DF. Br J Ophthalmol. 2021;105:216–221. Adapted under the terms of the CC BY-NC 4.0 license. DME = diabetic macular edema.

References

    1. Brown D.M., Schmidt-Erfurth U., Do D.V., et al. Intravitreal aflibercept for diabetic macular edema: 100-week results from the VISTA and VIVID studies. Ophthalmology. 2015;122:2044–2052.
    1. Flaxel C.J., Adelman R.A., Bailey S.T., et al. Diabetic retinopathy Preferred Practice Pattern®. Ophthalmology. 2020;127:P66–P145.
    1. Korobelnik J.-F., Do D.V., Schmidt-Erfurth U., et al. RESTORE study group Intravitreal aflibercept for diabetic macular edema. Ophthalmology. 2014;121:2247–2254.
    1. Mitchell P., Bandello F., Schmidt-Erfurth U., et al. The RESTORE study: ranibizumab monotherapy or combined with laser versus laser monotherapy for diabetic macular edema. Ophthalmology. 2011;118:615–625.
    1. Schmidt-Erfurth U., Garcia-Arumi J., Bandello F., et al. Guidelines for the management of diabetic macular edema by the European Society of Retina Specialists (EURETINA) Ophthalmologica. 2017;237:185–222.
    1. Wells J.A., Glassman A.R., Ayala A.R., et al. Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema. N Engl J Med. 2015;372:1193–1203.
    1. Cantrell R.A., Lum F., Chia Y., et al. Treatment patterns for diabetic macular edema: an Intelligent Research in Sight (IRIS®) Registry analysis. Ophthalmology. 2020;127:427–429.
    1. Ciulla T.A., Bracha P., Pollack J., Williams D.F. Real-world outcomes of anti–vascular endothelial growth factor therapy in diabetic macular edema in the United States. Ophthalmol Retina. 2018;2:1179–1187.
    1. Curry B.A., Sanfilippo P.G., Chan S., et al. Clinical outcomes of a treat and extend regimen with intravitreal aflibercept injections in patients with diabetic macular edema: experience in clinical practice. Ophthalmol Ther. 2020;9:87–101.
    1. Freund K.B., Korobelnik J.-F., Devenyi R., et al. Treat-and-extend regimens with anti-VEGF agents in retinal diseases: a literature review and consensus recommendations. Retina. 2015;35:1489–1506.
    1. Payne J.F., Wykoff C.C., Clark W.L., et al. TREX-DME Study Group Randomized trial of treat and extend ranibizumab with and without navigated laser for diabetic macular edema: TREX-DME 1 year outcomes. Ophthalmology. 2017;124:74–81.
    1. Prünte C., Fajnkuchen F., Mahmood S., et al. RETAIN Study Group Ranibizumab 0.5 mg treat-and-extend regimen for diabetic macular oedema: the RETAIN study. Br J Ophthalmol. 2016;100:787–795.
    1. Regeneron Pharmaceuticals, Inc.; Tarrytown, NY: 2021. Eylea [prescribing information]
    1. Eylea 40mg/ml solution for injection in a vial. Summary of product characteristics. 2012. Available at.
    1. Bayer Yakuhin; Osaka, Japan: 2015. Eylea [prescribing information]
    1. American Society of Retina Specialists 2020 Global Trends in Retina Survey. 2020. Available at: Accessed 29.04.21.
    1. Sahni J., Patel S.S., Dugel P.U., et al. Simultaneous inhibition of angiopoietin-2 and vascular endothelial growth factor-A with faricimab in diabetic macular edema: BOULEVARD phase 2 randomized trial. Ophthalmology. 2019;126:1155–1170.
    1. Sahni J., Dugel P.U., Patel S.S., et al. Safety and efficacy of different doses and regimens of faricimab vs ranibizumab in neovascular age-related macular degeneration: the AVENUE phase 2 randomized clinical trial. JAMA Ophthalmol. 2020;138:955–963.
    1. Ashraf M., Souka A., Adelman R. Predicting outcomes to anti-vascular endothelial growth factor (VEGF) therapy in diabetic macular oedema: a review of the literature. Br J Ophthalmol. 2016;100:1596–1604.
    1. Jyothi S.B., Leung R., Richardson M., et al. Intersession repeatability of visual acuity, colour vision, contrast sensitivity, retinal sensitivity thresholds and retinal thickness in mild diabetic retinopathy. Invest Ophthalmol Vis Sci. 2011;52:1291.
    1. Patel P.J., Chen F.K., Rubin G.S., Tufail A. Intersession repeatability of visual acuity scores in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2008;49:4347–4352.
    1. Shah N., Laidlaw D.A., Shah S.P., et al. Computerized repeating and averaging improve the test-retest variability of ETDRS visual acuity measurements: implications for sensitivity and specificity. Invest Ophthalmol Vis Sci. 2011;52:9397–9402.
    1. Bretz F., Posch M., Glimm E., et al. Graphical approaches for multiple comparison procedures using weighted Bonferroni, Simes, or parametric tests. Biom J. 2011;53:894–913.
    1. Regula J.T., Lundh von Leithner P., Foxton R., et al. Targeting key angiogenic pathways with a bispecific CrossMAb optimized for neovascular eye diseases. EMBO Mol Med. 2016;8:1265–1288.
    1. Csaky K., Cheik D., Foxton R., et al. Presented at: American Academy of Ophthalmology Subspecialty Day; San Francisco, CA, USA: October 11–12, 2019. Data supporting the sustained efficacy of faricimab, a bispecific antibody neutralizing both angiopoietin-2 and VEGF-A.
    1. Khanani A.M., Patel S.S., Ferrone P.J., et al. Efficacy of every four monthly and quarterly dosing of faricimab vs ranibizumab in neovascular age-related macular degeneration: the STAIRWAY phase 2 randomized clinical trial. JAMA Ophthalmol. 2020;138:964–972.
    1. Sivaprasad S., Oyetunde S. Impact of injection therapy on retinal patients with diabetic macular edema or retinal vein occlusion. Clin Ophthalmol. 2016;10:939–946.
    1. Spooner K.L., Guinan G., Koller S., et al. Burden of treatment among patients undergoing intravitreal injections for diabetic macular oedema in Australia. Diabetes Metab Syndr Obes. 2019;12:1913–1921.
    1. Ciulla T.A., Pollack J.S., Williams D.F. Visual acuity outcomes and anti-VEGF therapy intensity in diabetic macular oedema: a real-world analysis of 28 658 patient eyes. Br J Ophthalmol. 2021;105:216–221.
    1. Pak K.Y., Shin J.P., Kim H.W., et al. One-year results of treatment of diabetic macular edema with aflibercept using the treat-and-extend dosing regimen: the VIBIM study. Ophthalmologica. 2020;243:255–262.
    1. Kim Y.C., Shin J.P., Pak K.Y., et al. Two-year outcomes of the treat-and-extend regimen using aflibercept for treating diabetic macular oedema. Sci Rep. 2020;10:22030.
    1. Holekamp N.M., Campbell J., Almony A., et al. Vision outcomes following anti–vascular endothelial growth factor treatment of diabetic macular edema in clinical practice. Am J Ophthalmol. 2018;191:83–91.
    1. Kiss S., Campbell J., Almony A., et al. Management and outcomes for neovascular age-related macular degeneration: analysis of United States electronic health records. Ophthalmology. 2020;127:1179–1188.

Source: PubMed

3
订阅