The early 2000s ushered in the anti-VEGF era for wet age-related macular degeneration (AMD), which completely changed the approach to treating patients with wet AMD. Ophthalmology is on the verge of a similar advance that could provide a therapy for patients with advanced dry AMD. Age-related macular degeneration is the leading cause of irreversible blindness in developed countries and is expected to affect 288 million people globally by 2040.1 Geographic atrophy (GA), characterized by loss of the choriocapillaris, retinal pigment epithelium (RPE), and photoreceptors, represents the most advanced form of dry AMD and has been the target of numerous avenues of therapeutic approach, the most promising thus far proving to be the complement pathway inhibition. With the first 2 drugs in this class being evaluated by the FDA, it is an apt time to review the history of complement inhibition in GA treatment.
THE COMPLEMENT SYSTEM
The complement system forms a core component of the innate immune response and can be classified into 3 pathways that differ by their mode of activation: the classical pathway, the lectin pathway, and the alternative pathway. The classical pathway is activated by antigen-antibody complexes, resulting in activation of C1 by C1q. The C1 complex cleaves C2 and C4, allowing formation of the classical C3 convertase, C4b2a.2,3
The lectin pathway is activated by recognition of bacterial mannose-containing polysaccharides by mannose-binding lectin (MBL).3 Activated MBL binds to MBL serine peptidases (MASPs), cleaving C2 and C4 and again producing C4b2a.
The alternative pathway is initiated by spontaneous C3 hydrolysis into C3a and C3b. C3b binds to complement factor B (CFB) and complement factor D (CFD) to form the alternative C3 convertase, C3bBb. Regulation of this self-amplifying pathway is achieved through inhibitory complement factors H (CFH) and I (CFI). Alternative pathway activation accounts for the majority of complement activity (80%) and is likely the most significant pathway in AMD pathogenesis.2,4
The 3 pathways converge on C3, which cleaves C5 into C5a and C5b, the latter of which initiates the terminal complement pathway. Terminal pathway activation results in phagocytosis, inflammation, and formation of the membrane attack complex.5
RATIONALE FOR COMPLEMENT INHIBITION IN AMD
The role of complement dysfunction in AMD pathogenesis was suggested by genome-wide association studies (GWAS) that identified polymorphisms in complement proteins, including CFH, CFI, C2 and C3, in AMD patients.4,6,7 Particular polymorphisms, such as CFH Y402H, are associated with 2.45 to 5.57-fold increased risk of AMD.4 In vitro, many of these polymorphisms result in either increased complement activation or impaired complement regulation.2,8 Patients with AMD also demonstrate increased serum concentrations of complement activation products, which have also been identified within drusen and in drusen-adjacent RPE and choriocapillaris.3,9,10 Lastly, murine models of complement dysregulation reproduce the clinical features of AMD, including sub-RPE lipofuscin deposition, subretinal drusen-like deposits, and RPE and photoreceptor atrophy.11,12
EARLY TRIALS
Most agents targeting complement in GA have focused on alternative and common pathway proteins, such as C3 and C5. The earliest trials provided largely discouraging results but yielded important lessons that would inform subsequent trials. A summary of previous and ongoing clinical trials in complement inhibition for GA are provided in Table 1.13-29
Year | Trial | Phase | Drug | Target | Design | N | Primary Outcome | Result | |
2014 | COMPLETE (NCT00935883) | 2 | Eculizumab (intravenous) (Soliris; Alexion) | C5 | 2:1:1 randomization to low-dose, high-dose, sham weekly for 4 weeks, then Q2W until week 24 | 30 | Growth of geographic atrophy (GA); decrease in drusen volume at 26 weeks | No significant reduction in GA growth rate at 26 weeks (P=.96) or 52 weeks (P=.93) | |
2017 | MAHALO (NCT01229215) | 2 | Lampalizumab (intravitreal) (Genentech) | CFD | 1:1:1 randomization to monthly or every other month (EOM) vs sham | 123 | Growth rate of GA area from baseline at 18 months | 20% reduction in GA growth rate vs sham (P=.117). CFI risk-allele carriers showed 44% reduction (P=.0037) | |
2018 | NCT01527500 | 2 | LFG316 (intravitreal) (Novartis) | C5 | 2:1 randomization to monthly dosing vs sham | 158 | GA growth from baseline at 505 days; safety | Well tolerated. No significant difference in GA growth vs sham | |
2018 | CHROMA (NCT02247479) | 3 | Lampalizumab (intravitreal) (Genentech) | CFD | 2:1:2:1 randomization to Q4W or Q6W vs sham | 906 | Change in GA area from baseline at 48 weeks | No significant reduction in GA growth with Q4W (P=.80) or Q6W dosing (P=.59) | |
2018 | SPECTRI (NCT02247531) | 3 | Lampalizumab (intravitreal) (Genentech) | CFD | 2:1:2:1 randomization to Q4W or Q6W vs sham | 975 | Change in GA area from baseline at 48 weeks | No significant reduction in GA growth with Q4W (P=.048, favoring sham) or Q6W dosing (P=.27) | |
2020 | FILLY (NCT02503332) | 2 | Pegcetacoplan (intravitreal) (Apellis Pharmaceuticals) | C3 | 2:2:1:1 randomization to monthly or EOM vs sham | 246 | Change in GA area from baseline at 12 months ; Safety | Well tolerated. Reduction in GA growth of 29% (P=.008) with monthly and 20% (P=.067) with EOM vs sham | |
2020 | GATHER1 (NCT02686658) | 2/3 | Avacincaptad pegol (Zimura; Iveric Bio) | C5 | Part 1: 1:1:1 randomization to 1 mg, 2 mg, sham dosed monthly. Part 2: 1:1:1 randomization to 2 mg, 4 mg, sham dosed monthly | 286 | Mean rate of change in GA area at 12 months | 27.4% reduction in GA growth rate over 12 months with 2 mg dosed monthly (P=.0072) and 27.8% with 4 mg dosed monthly (P=.0051) vs sham | |
2021 | DERBY (NCT03525600) | 3 | Pegcetacoplan (intravitreal) (Apellis Pharmaceuticals) | C3 | 2:2:1:1 randomization to monthly, EOM or sham | 621 | Change in GA area from baseline at 12 months | Nonsignificant reduction in GA growth of 12% with monthly dosing (P=.0528) and 11% with EOM dosing (P=.0750) vs sham | |
2021 | OAKS (NCT03525613) | 3 | Pegcetacoplan (intravitreal) (Apellis Pharmaceuticals) | C3 | 2:2:1:1 randomization to monthly, EOM or sham | 637 | Change in GA area from baseline at 12 months | Reduction in GA growth of 22% over 12 months with monthly dosing (P=.0003) and 16% with EOM dosing (P=.0052) vs sham | |
2022 | GATHER2 (NCT04435366) | 3 | Avacincaptad pegol (Zimura; Iveric Bio) | C5 | Part 1 (12 months): 1:1 randomization to 2 mg vs sham monthly. Part 2 (12 months): 2 mg monthly arm re-randomized to 2 mg monthly or EOM, sham continues monthly. | 448 | Mean rate of change in GA over 12 months | Reduction in GA growth rate of 14.3% over 12 months (P=.0064) using square root transformation, or of 17.7% using observed GA area | |
2022 | NCT03144999 | 1 | JNJ-81201887 (Janssen) (intravitreal gene therapy) | CD59 | Dose escalation study | 17 | Safety | No SAEs or systemic SAEs over 24 months | |
2022 | ReGAtta (NCT04643886) | 2a | GEM103 (Gemini) | CFH | Open-label, multiple dose study | 62 | Safety over 18 months | Study terminated by sponsor | |
Ongoing | GALE (NCT04770545) | 3 | Pegcetacoplan (intravitreal) (Apellis Pharmaceuticals) | C3 | Open label extension | ~1,200 | Safety over 36 months | Pending | |
Ongoing | ARCHER (NCT04656561) | 2 | ANX007 (intravitreal) (Annexon) | C1q | 1:1:1 randomization to monthly or EOM vs sham | 270 | GA lesion growth rate at 12 months | Pending | |
Ongoing | GOLDEN (NCT03815825) | 2 | IONIS-FB-LRx (subcutaneous) (Ionis) | CFB | Randomization to 3 dose levels vs placebo | ~330 | Change in GA area at week 49 | Pending | |
Ongoing | NCT05019521 | 2 | Danicopan (oral) (Alexion) | CFD | Randomization to 100 mg BID, 200 mg BID, 400 mg QD, or placebo | ~332 | Change in GA area at week 52 | Pending | |
Ongoing | FOCUS (NCT03846193) | 1/2 | GT011 (subretinal gene therapy) (Gyroscope) | CFI | Nonrandomized dose escalation study via transvitreal subretinal injection or suprachoroidal cannulation approach | 56 | Safety | Pending | |
Ongoing | EXPLORE (NCT04437368) | 2 | GT011 (subretinal gene therapy) (Gyroscope) | CFI | 1:1:1 randomization to low-dose or high-dose vs untreated control in patients with rare CFI allele | ~75 | Change in GA area at week 48 | Pending | |
Ongoing | HORIZON (NCT04566445) | 2 | GT011 (subretinal gene therapy) (Gyroscope) | CFI | 1:1:1 randomization to medium-dose or high-dose vs untreated control | ~250 | Change in GA area at week 48 | Pending |
COMPLETE (NCT00935883) was a small, phase 2 trial evaluating intravenous eculizumab, an anti-C5 monoclonal antibody (Soliris; Alexion Pharmaceuticals) that failed to demonstrate reduction in mean GA growth rate at 26 weeks.13
MAHALO (NCT01229215) was a small, phase 2 trial evaluating lampalizumab, an anti-CFD monoclonal antibody fragment (Genentech). MAHALO met a prespecified alpha of 0.20 for reduction of GA growth at 18 months (P=.117). However, it provided intriguing results in a subset of patients with high-risk CFI alleles, in whom a more substantial GA growth reduction of 44% was observed.14 Based on these results, lampalizumab was evaluated in the larger CHROMA (NCT02247479) and SPECTRI (NCT02247531) trials. These identical phase 3 trials were the largest studies not only of complement inhibition in AMD but also of GA more generally, at the time they were conducted (n=1,881).16 Both failed to show a benefit of lampalizumab in reducing GA growth rate, including in the high-risk CFI allele subgroup. Around the same time, a phase 2 trial (NCT01527500) of LFG316 (Novartis), an anti-C5 monoclonal antibody, also failed to demonstrate a significant reduction in GA growth compared to sham.15,30
The failure of COMPLETE raised fundamental questions about the promise of the complement system as a therapeutic target, about the association between the complement polymorphisms identified in GWAS and GA progression, and whether an optimal therapeutic target within the complement cascade may exist.13 These concerns were only amplified following the failure of CHROMA and SPECTRI. However, these trials made important contributions. They established GA area growth rate assessed by fundus autofluorescence (FAF) as a standard endpoint and demonstrated good tolerability of a 0.1 mL injection volume. CHROMA and SPECTRI also provided useful information about GA natural history by identifying an average annual lesion growth rate of ~2 mm2 and by identifying baseline lesion size, multifocality, and nonfoveal location as risk factors for rapid enlargement, consistent with AREDS2 findings also released in 2018.16,31 They also highlighted problems with post hoc subset analyses.
FIRST SIGNS OF SUCCESS
Initial success came in 2020 with the phase 2 FILLY trial (NCT02503332) evaluating intravitreal pegcetacoplan, a pegylated anti-C3 and C3b peptide (APL-2; Apellis Pharmaceuticals).17 FILLY demonstrated a significant reduction of 29% in GA growth rate at 12 months with monthly dosing. Twelve-month efficacy data from the DERBY (NCT03525600) and OAKS (NCT03525613) phase 3 trials evaluating pegcetacoplan was released the following year. Pegcetacoplan significantly reduced GA growth at 12 months in OAKS, but narrowly missed this endpoint in DERBY.19
Also in 2020, the GATHER1 trial (NCT02686658) evaluating intravitreal avacincaptad pegol (Zimura; Iveric Bio), a pegylated RNA aptamer targeting C5, demonstrated significant reductions of 27.4% and 27.8% in GA growth at 12 months with 2 mg and 4 mg dosed monthly.18 The phase 3 GATHER2 trial (NCT04435366) (n=448) met its primary endpoint and demonstrated a 14.3% to 17.7% reduction in GA growth rate over 12 months vs sham.20 Post hoc analysis of GATHER1 data found a strong correlation between GA as measured by FAF and as measured by OCT, and identified OCT ellipsoid zone integrity as a possible biomarker for GA. This has important implications for clinical practice, as OCT-based assessment of GA is likely to be more widely utilized by practicing retina specialists compared to FAF.32 Iveric Bio completed its NDA submission in December 2022.20
RECENT DEVELOPMENTS
Pegcetacoplan continues to show promise with 24-month data from DERBY and OAKS. At 24 months, pegcetacoplan reduced GA growth by 19% when dosed monthly (P=.0004) and by 16% when dosed every other month (EOM) (P=.0030) in DERBY, and by 22% (monthly) (P<.0001) and 18% (EOM) (P=.0002) in OAKS.33 Notably, DERBY met its primary endpoint at 24 months after narrowly missing its 12-month primary endpoint. Pegcetacoplan also showed positive trends toward preventing perilesional photoreceptor functional loss as assessed by microperimetry, which met significance in the EOM dosing group in OAKS. This represents some of the first human functional data demonstrating a benefit to complement inhibition in GA. An open-label extension study of pegcetacoplan (GALE; NCT04770545) is ongoing with approximately 1,200 patients.23 Pegcetacoplan will be the first drug for GA to be evaluated by the FDA, with a PDUFA date of February 26, 2023.34
An interesting trend to emerge from DERBY and OAKS is an acceleration of treatment effect over time. In DERBY and OAKS, the effect of pegcetacoplan treatment increased over months 18 to 24, with a reduction of GA growth of 36% with monthly dosing (P<.0001) and 29% with EOM dosing (P=.0002) in DERBY and 24% (P=.0080) and 25% (P=.0007), respectively, in OAKS.33 Earlier data from FILLY also demonstrated reductions of 45% and 33% in GA growth rate with monthly and EOM dosing during the second 6 months of the trial, compared to 29% and 20% overall.17 The reason for this treatment acceleration remains unclear. The concept of “dead retina walking” has been proposed as a possible explanation, in which terminally damaged perilesional retina leads to an inevitable, initial progression of GA with or without treatment, followed by stabilization of GA growth once complement inhibition arrests further retinal damage.35
A concerning theme also emerged from these studies: increased rates of choroidal neovascularization (CNV) in the setting of complement inhibition. Both FILLY treatment arms demonstrated increased rates of CNV vs sham, accounting in part for the high rate of treatment dropout.17 DERBY and OAKS demonstrated rates of new CNV of 6.0%, 4.1%, and 2.4% at 12 months and 12.2%, 6.7%, and 3.1% at 24 months in the monthly, EOM, and sham arms, respectively.36,37 Retrospective analysis of FILLY found OCT double-layer sign and contralateral eye CNV to correlate with new CNV, suggesting the possible role of complement inhibition in unmasking pre-existing subclinical CNV.38 While pre-existing anatomic features may influence risk of CNV with complement inhibition, more research is needed to fully elucidate the role of complement in vascular homeostasis and CNV in the context of AMD.
NEW AGENTS ON THE HORIZON
Numerous promising agents are in early development for complement inhibition in patients with GA. Ongoing phase 2 trials include ARCHER (NCT04656561) (n=270) evaluating intravitreal ANX007 anti-C1q monoclonal antibody fragment (Annexon); GOLDEN (NCT03815825) (n=~330) evaluating IONIS-FB-LRx antisense oligonucleotide targeting CFB (Ionis Pharmaceuticals); and NCT05019521 (n=~332) evaluating danicopan small-molecule inhibitor of CFD (ALXN2040; Alexion Pharmaceuticals).24,25,26,39-43 Other agents, such as CB2782-PEG (Catalyst Biosciences, Inc.) a C3-inactivating protease, are in preclinical development.44,45
Recent results were less encouraging for other agents. The phase 2a ReGAtta trial (NCT04643886) (n=62) evaluating GEM103 recombinant human CFH (Gemini Therapeutics) demonstrated good tolerability and positive CFH biomarkers, but was not designed to detect statistically significant treatment effect and was terminated in Q1 2022.22 The phase 2 CATALINA trial (NCT04465955) (n=320) evaluating NGM621 anti-C3 monoclonal antibody (NGM Bio) failed to yield a significant reduction in GA growth compared to sham after showing positive phase 1 results.46,47 However, CATALINA post hoc analyses have highlighted a variety of challenges related to grading methodology and FAF assessment of GA lesions, particularly large or complex lesions.48 These challenges may have complicated the interpretation of CATALINA results, and also provide useful lessons for future trials.
GENE THERAPY
One of the biggest concerns with respect to current phase 3 agents under investigation is the significant treatment burden incurred by monthly or EOM injections. Long-term, durable treatment for GA may be on the horizon with gene therapy finally approaching late-stage clinical development.
Interim safety results of the phase 1/2 FOCUS trial (NCT03846193) evaluating GT005 AAV2-based gene therapy for CFI (Gyroscope Therapeutics) demonstrated good tolerability of subretinal, transvitreal delivery of GT005 (n=31).27,49 Results of GT005 delivered by the Orbit subretinal delivery system are pending. Two concurrent phase 2 trials evaluating GT005, EXPLORE (NCT04437368) and HORIZON (NCT04566445), are ongoing and will assess patients with (EXPLORE) or without (HORIZON) a rare CFI allele.28,29
JNJ-81201887 (also JNJ-1887, formerly AAVCAGsCD59 or HMR59) (Janssen Research & Development, LLC), is a gene therapy expressing CD59, thereby inhibiting C9 insertion and membrane attack complex formation. Phase 1 results (NCT03144999) demonstrated that JNJ-81201887 was well tolerated at 3 escalating doses given by intravitreal injection (n=17).21,50,51
CONCLUSIONS
Complement inhibition has shown remarkable and rapid progress as a viable treatment for GA following the uncertain beginnings and trial failures that marked its inception. Several drugs are in late-stage development and many more are in early phases of development. Complement inhibition may soon begin to address one of the largest unmet needs currently facing the practicing retinal physician. RP
Editor’s note: This article is part of a special edition of Retinal Physician that was supported by Apellis Pharmaceuticals. Authors and editors maintained editorial control for all articles in this special edition.
REFERENCES
- Wong WL, Su X, Li X, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health. 2014;2(2):e106-e116. doi:10.1016/S2214-109X(13)70145-1
- Boyer DS, Schmidt-Erfurth U, van Lookeren Campagne M, Henry EC, Brittain C. The pathophysiology of geographic atrophy secondary to age-related macular degeneration and the complement pathway as a therapeutic target. Retina. 2017;37(5):819-835. doi:10.1097/IAE.0000000000001392
- Whitmore SS, Sohn EH, Chirco KR, et al. Complement activation and choriocapillaris loss in early AMD: implications for pathophysiology and therapy. Prog Retin Eye Res. 2015;45:1-29. doi:10.1016/j.preteyeres.2014.11.005
- Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant increases the risk of age-related macular degeneration. Science. 2005;308(5720):419-421. doi:10.1126/science.1110359
- Lin JB, Halawa OA, Miller JW, Vavvas DG. Complement inhibition for geographic atrophy: a tempting target with mixed results. J Clin Med. 2021;10(13):2890. Published 2021 Jun 29. doi:10.3390/jcm10132890
- Fritsche LG, Igl W, Bailey JN, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2016;48(2):134-143. doi:10.1038/ng.3448
- Fritsche LG, Chen W, Schu M, et al. Seven new loci associated with age-related macular degeneration. Nat Genet. 2013;45(4):433-439e4392. doi:10.1038/ng.2578
- Seddon JM, Yu Y, Miller EC, et al. Rare variants in CFI, C3 and C9 are associated with high risk of advanced age-related macular degeneration. Nat Genet. 2013;45(11):1366-1370. doi:10.1038/ng.2741
- Scholl HP, Charbel Issa P, Walier M, et al. Systemic complement activation in age-related macular degeneration [published correction appears in PLoS ONE. 2008;3(7). doi: 10.1371/annotation/32b9bc31-ed6d-4d31-9ce0-480407017bad] [published correction appears in PLoS ONE. 2008;3(7). doi: 10.1371/annotation/511a1029-bc43-4510-a4ca-c1db31810acc]. PLoS One. 2008;3(7):e2593. doi:10.1371/journal.pone.0002593
- Johnson LV, Ozaki S, Staples MK, Erickson PA, Anderson DH. A potential role for immune complex pathogenesis in drusen formation. Exp Eye Res. 2000;70(4):441-449. doi:10.1006/exer.1999.0798
- Ufret-Vincenty RL, Aredo B, Liu X, et al. Transgenic mice expressing variants of complement factor H develop AMD-like retinal findings. Invest Ophthalmol Vis Sci. 2010;51(11):5878-5887. doi:10.1167/iovs.09-4457
- Cashman SM, Desai A, Ramo K, Kumar-Singh R. Expression of complement component 3 (C3) from an adenovirus leads to pathology in the murine retina. Invest Ophthalmol Vis Sci. 2011;52(6):3436-3445. doi:10.1167/iovs.10-6002
- Yehoshua Z, de Amorim Garcia Filho CA, Nunes RP, et al. Systemic complement inhibition with eculizumab for geographic atrophy in age-related macular degeneration: the COMPLETE study. Ophthalmology. 2014;121(3):693-701. doi:10.1016/j.ophtha.2013.09.044
- Yaspan BL, Williams DF, Holz FG, et al. Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration. Sci Transl Med. 2017;9(395):eaaf1443. doi:10.1126/scitranslmed.aaf1443
- Intravitreal LFG316 in Patients With Age-Related Macular Degeneration (AMD). ClinicalTrials.gov identifier: NCT01527500. Updated January 5, 2021. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT01527500
- Holz FG, Sadda SR, Busbee B, et al. Efficacy and safety of lampalizumab for geographic atrophy due to age-related macular degeneration: Chroma and Spectri phase 3 randomized clinical trials. JAMA Ophthalmol. 2018;136(6):666-677. doi:10.1001/jamaophthalmol.2018.1544
- Liao DS, Grossi FV, El Mehdi D, et al. Complement C3 inhibitor pegcetacoplan for geographic atrophy secondary to age-related macular degeneration: a randomized phase 2 trial. Ophthalmology. 2020;127(2):186-195. doi:10.1016/j.ophtha.2019.07.011
- Jaffe GJ, Westby K, Csaky KG, et al. C5 inhibitor avacincaptad pegol for geographic atrophy due to age-related macular degeneration: a randomized pivotal phase 2/3 trial. Ophthalmology. 2021;128(4):576-586. doi:10.1016/j.ophtha.2020.08.027
- Heier JS. Efficacy of intravitreal pegcetacoplan in geographic atrophy: results from two phase 3 trials. Presented at: Retina Society Annual Meeting 2021, Chicago, IL.
- Iveric Bio announces completion of rolling NDA submission to FDA for avacincaptad pegol for the treatment of geographic atrophy. News release. December 20, 2022. Accessed December 20, 2022. https://www.businesswire.com/news/home/20221219005815/en/Iveric-Bio-Announces-Completion-of-Rolling-NDA-Submission-to-FDA-for-Avacincaptad-Pegol-for-the-Treatment-of-Geographic-Atrophy
- Treatment of advanced dry age related macular degeneration with AAVCAGsCD59. ClinicalTrials.gov identifier: NCT03144999. Updated April 2, 2021. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT03144999
- A multiple dose study of repeat intravitreal injections of gem103 in dry age-related macular degeneration. ClinicalTrials.gov identifier: NCT04643886. Updated September 23, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT04643886
- An extension study to evaluate the long-term safety and efficacy of pegcetacoplan (APL-2) in subjects with geographic atrophy secondary to AMD (GALE). ClinicalTrials.gov identifier: NCT04770545. https://clinicaltrials.gov/ct2/show/NCT04770545 . Updated August 30, 2022. Accessed November 1, 2022.
- A study investigating the efficacy and safety of intravitreal injections of ANX007 in pa tients with geographic atrophy (ARCHER). ClinicalTrials.gov identifier: NCT04656561. Updated July 5, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT04656561
- GOLDEN study: a study to assess safety and efficacy of multiple doses of IONIS-FB-LRx in participants with geographic atrophy secondary to age-related macular degeneration (AMD). ClinicalTrials.gov identifier: NCT03815825. Updated August 22, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT03815825
- A study of danicopan in participants with geographic atrophy secondary to age-related macular degeneration. ClinicalTrials.gov identifier: NCT05019521. Updated October 20, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT05019521
- FOCUS: first in human study to evaluate the safety and efficacy of GT005 administered in subjects with dry AMD. ClinicalTrials.gov identifier: NCT03846193. Updated October 13, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT03846193
- EXPLORE: a phase II study to evaluate the safety and efficacy of two doses of GT005. ClinicalTrials.gov identifier: NCT04437368. Updated December 2, 2021. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT04437368
- HORIZON: a phase II study to evaluate the safety and efficacy of two doses of GT005. ClinicalTrials.gov identifier: NCT04566445. Updated August 2, 2022. Accessed November 1, 2022. https://clinicaltrials.gov/ct2/show/NCT04566445
- Zamiri P. Complement C5 inhibition in AMD. Paper presented at: Angiogenesis, Exudation, and Degeneration meeting. February 6, 2016, Miami, FL.
- Keenan TD, Agrón E, Domalpally A, et al. Progression of geographic atrophy in age-related macular degeneration: AREDS2 report number 16. Ophthalmology. 2018;125(12):1913-1928. doi:10.1016/j.ophtha.2018.05.028
- Ehlers JP, Yordi S, Cetin H, et al. Exploration of machine learning-enhanced compartmental retinal integrity assessment for progression risk and treatment response in the GATHER1 study. Presented at: The Macula Society Annual Meeting 2022, Berlin, Germany. https://investors.ivericbio.com/static-files/14eb43db-c394-49f4-8b4f-ec0289bd47bb
- Wykoff CC. Treatment of geographic atrophy secondary to AMD with pegcetacoplan: two-year outcomes from the randomized phase 3 OAKS and DERBY trials. Presented at: American Academy of Ophthalmology Annual Meeting 2022, Chicago, IL. https://apellismedicalhub.com/geographic-atrophy-ga/american-academy-of-ophthalmology-aao-2022/treatment-of-geographic-atrophy-secondary-to-amd-with-pegcetacoplan-two-year-outcomes-from-the-randomized-phase-3-oaks-and-derby-trials/
- Apellis announces FDA acceptance of NDA amendment and new PDUFA date of February 26, 2023 for pegcetacoplan for geographic atrophy (GA). News release. November 18, 2022. Accessed November 18, 2022. https://investors.apellis.com/news-releases/news-release-details/apellis-announces-fda-acceptance-nda-amendment-and-new-pdufa .
- Wykoff CC, Singer M. Pegcetacoplan combats geographic atrophy in DERBY and OAKS 2-year data. Retinal Physician. October 2022. Accessed November 1, 2022. https://www.retinalphysician.com/newsletter/retina-minute/october-2022
- Apellis announces 24-month phase 3 post hoc results showing treatment with pegcetacoplan led to preservation of function in high-risk areas of the retina in patients with geographic atrophy (GA) at AAO annual meeting. News release. September 30, 2022. Accessed November 1, 2022. https://investors.apellis.com/news-releases/news-release-details/apellis-announces-24-month-phase-3-post-hoc-results-showing
- Wykoff CC. Safety of intravitreal pegcetacoplan in geographic atrophy: results from two phase 3 trials. Presented at: the Retina Society Annual Meeting 2021, Chicago, IL.
- Wykoff CC, Rosenfeld PJ, Waheed NK, et al. Characterizing new-onset exudation in the randomized phase 2 FILLY trial of complement inhibitor pegcetacoplan for geographic atrophy. Ophthalmology. 2021;128(9):1325-1336. doi:10.1016/j.ophtha.2021.02.025
- Jiao H, Rutar M, Fernando N, et al. Subretinal macrophages produce classical complement activator C1q leading to the progression of focal retinal degeneration. Mol Neurodegener. 2018;13(1):45. doi:10.1186/s13024-018-0278-0
- Annexon Biosciences completes enrollment in ARCHER phase 2 trial of novel C1q inhibitor, ANX007, in patients with geographic atrophy. News release. April 7, 2022. Accessed November 1, 2022. https://ir.annexonbio.com/news-releases/news-release-details/annexon-biosciences-completes-enrollment-archer-phase-2-trial
- Jaffe JG, Sahni J, Fauser S, et al. Development of IONIS-FB-LRX to treat geographic atrophy associated with AMD. Presented at: ARVO Annual Meeting 2020, Denver, CO. https://iovs.arvojournals.org/article.aspx?articleid=2768483
- Ionis Pharmaceuticals, Inc. (IONS) CEO Brett Monia on Q2 2022 Results - Earnings Call Transcript. August 9, 2022. Accessed November 1, 2022. https://seekingalpha.com/article/4532145-ionis-pharmaceuticals-inc-ions-ceo-brett-monia-on-q2-2022-results-earnings-call-transcript
- Boyer D, Rivera J, Ya-Ping Ko, et al. Oral administration of the complement factor D inhibitor danicopan (ALXN2040) in preclinical studies demonstrates high and sustained drug concentrations in posterior ocular tissues for the potential treatment of geographic atrophy. Presented at: ARVO Annual Meeting 2021, San Francisco, CA. https://iovs.arvojournals.org/article.aspx?articleid=2773030
- Catalyst Biosciences & Mosaic Biosciences Present Preclinical Data on Pegylated CB 2782 for the Treatment of Dry Age-Related Macular Degeneration. April 29, 2019. Accessed November 1, 2022. Available at: https://ir.catalystbiosciences.com/news-releases/news-release-details/catalyst-biosciences-mosaic-biosciences-present-preclinical-data
- Catalyst Biosciences regains rights to CB 2782-PEG for the treatment of dry AMD expands CBIO’s complement portfolio in ophthalmology. News release. March 15, 2022. Accessed November 1, 2022. https://ir.catalystbiosciences.com/news-releases/news-release-details/catalyst-biosciences-regains-rights-cb-2782-peg-treatment-dry
- Wykoff CC. Inhibition of complement C3 in geographic atrophy with NGM621: phase 1 study results. Presented at: Angiogenesis, Exudation, and Degeneration 2021 Annual Meeting, Miami, FL. Accessed December 20, 2022. http://www.ngmbio.com/wp-content/uploads/2021/12/Angiogenesis-2021-NGM621-Phase-1-slides-FINAL.pdf
- NGM Bio announces topline results from the CATALINA phase 2 trial of NGM621 in patients with geographic atrophy (GA) secondary to age-related macular degeneration. October 17, 2022. Accessed November 1, 2022. https://ir.ngmbio.com/news-releases/news-release-details/ngm-bio-announces-topline-results-catalina-phase-2-trial-ngm621
- Wykoff, CC. The results of the CATALINA phase 2 study of NGM621 for geographic atrophy secondary to AMD. Presented at: The Retina Society Annual Meeting 2022, Pasadena, CA. Accessed November 15, 2022. https://www.ngmbio.com/wp-content/uploads/2022/11/Wykoff_CATALINA-Topline-Results_Retina-Society_vPosted.pdf
- Kiss, S. Preliminary results from a first-in-human phase I/II gene therapy study (FO CUS) of subretinally delivered GT005, an investigational AAV2 vector, in patients with geographic atrophy secondary to age-related macular degeneration. Presented at: ASRS 2022 Annual Meeting, New York, NY. Accessed December 20, 2022. https://www.gyroscopetx.com/wp-content/uploads/2022/07/KISS_ASRS-2022_PRESENTATION.pdf
- Cohen, MN. Phase 1 study of JNJ-81201887 gene therapy in geographic atrophy (GA) due to age-related macular degeneration (AMD). Presented at: AAO Annual Meeting 2022, Chicago, IL.
- Janssen announces late-breaking data from two gene therapy programs at the American Academy of Ophthalmology annual meeting 2022. News release. October 1, 2022. Accessed November 1, 2022. Available at: https://www.jnj.com/janssen-announces-late-breaking-data-from-two-gene-therapy-programs-at-the-american-academy-of-ophthalmology-2022-annual-meeting