Advertisement
Research Article|Articles in Press, 100286

Alternative Complement Pathway Inhibition by Lampalizumab: Analysis of Data from Chroma and Spectri Phase 3 Clinical Trials

Open AccessPublished:February 13, 2023DOI:https://doi.org/10.1016/j.xops.2023.100286

      Abstract

      Objective

      Lampalizumab, an antigen-binding fragment of a humanized monoclonal antibody directed against complement factor D (CFD), is designed to treat geographic atrophy (GA) secondary to age-related macular degeneration. Given the lack of clinical efficacy observed in patients with GA in phase 3 Chroma/Spectri trials, we investigated the impact of lampalizumab on the complement system in vivo. We developed 6 novel assays to measure changes in complement pathway activities in aqueous humor samples collected from patients enrolled in these trials.

      Design

      Chroma/Spectri were double-masked, sham-controlled, 96-week trials.

      Participants

      Aqueous humor samples from 97 patients with bilateral GA across all groups (ie, intravitreous lampalizumab 10 mg every 6 weeks, every 4 weeks, or corresponding sham procedures) were tested.

      Methods

      Novel antibody capture assays were developed on the Simoa platform for complement factor B (CFB), the Bb fragment of CFB, intact complement component 3 (C3), processed C3, intact complement component 4 (C4), and processed C4.

      Main Outcome Measures

      The ratio of processed versus intact complement factors (ie, complement activity) in aqueous humor were assessed.

      Results

      Patients treated with either of the lampalizumab regimens demonstrated an increase in CFD level at week 24 compared with baseline, along with a corresponding median reduction in the Bb:CFB ratio of 41% to 43%. There were no strong correlations between lampalizumab concentrations in aqueous humor and change in CFD levels or Bb:CFB ratio over time. No change in downstream C3 processing was observed with lampalizumab treatment. Additionally, there was no change in C4 processing.

      Conclusions

      The collection of aqueous humor samples from patients in Chroma and Spectri trials provided key insights on the effects of lampalizumab, a novel complement inhibitor, on local ocular complement activation. Lampalizumab inhibited the alternative complement pathway in the eyes of patients with GA; however, this did not translate into a measurable reduction in either classical or total complement activity, based on absence of changes in C4 and C3 processing, respectively.

      Keywords

      Abbreviations and Acronyms:

      Ab (antibody), AMD (age-related macular degeneration), BLLOQ (below lower limit of quantification), C2 (complement component 2), C3 (complement component 3), C4 (complement component 4), C5 (complement component 5), CFB (complement factor B), CFD (complement factor D), CFI (complement factor I), CV% (percent coefficient of variation), Fab (antigen-binding fragment), FAF (fundus autofluorescence), GA (geographic atrophy), gD (glycoprotein D), Q4W (every 4 weeks), Q6W (every 6 weeks)
      Geographic atrophy (GA), an advanced form of age-related macular degeneration (AMD), is a major cause of visual function loss.
      • Sunness J.S.
      • Rubin G.S.
      • Applegate C.A.
      • et al.
      Visual function abnormalities and prognosis in eyes with age-related geographic atrophy of the macula and good visual acuity.
      ,
      • Flaxman S.R.
      • Bourne R.R.A.
      • Resnikoff S.
      • et al.
      Global causes of blindness and distance vision impairment 1990–2020: a systematic review and meta-analysis.
      Unlike the neovascular presentation of advanced AMD, GA has no approved treatment to halt progressive vision loss associated with this disease.
      • Holz F.G.
      • Strauss E.C.
      • Schmitz-Valckenberg S.
      • van Lookeren Campagne M.
      Geographic atrophy: clinical features and potential therapeutic approaches.
      Several studies have shown that polymorphisms in genes encoding proteins in the alternative complement pathway are associated with the risk of developing AMD, including complement component 3 (C3) and complement factors B (CFB), H, and I (CFI).
      • Fritsche L.G.
      • Chen W.
      • Schu M.
      • et al.
      Seven new loci associated with age-related macular degeneration.
      ,
      • Gorin M.B.
      Genetic insights into age-related macular degeneration: controversies addressing risk, causality, and therapeutics.
      Dysregulation of the alternative complement pathway has also been implicated in the pathogenesis of AMD;
      • Loyet K.M.
      • Deforge L.E.
      • Katschke Jr., K.J.
      • et al.
      Activation of the alternative complement pathway in vitreous is controlled by genetics in age-related macular degeneration.
      studies have shown increased levels of complement proteins, including complement factor D (CFD), in the serum of patients with AMD compared with controls.
      • Scholl H.P.
      • Charbel Issa P.
      • Walier M.
      • et al.
      Systemic complement activation in age-related macular degeneration.
      ,
      • Stanton C.M.
      • Yates J.R.
      • den Hollander A.I.
      • et al.
      Complement factor D in age-related macular degeneration.
      A recent study demonstrated a significant association between AMD stage and complement activation in the serum.
      • Heesterbeek T.J.
      • Lechanteur Y.T.E.
      • Lores-Motta L.
      • et al.
      Complement activation levels are related to disease stage in AMD.
      CFD is a pivotal regulator of the alternative complement pathway that binds to the C3b/CFB complex and cleaves CFB into Ba and Bb. Ba is then released, but Bb remains attached to C3b; this C3bBb complex forms the C3 convertase, which catalyzes additional C3 cleavage (Fig 1).
      • Boyer D.S.
      • Schmidt-Erfurth U.
      • van Lookeren Campagne M.
      • et al.
      The pathophysiology of geographic atrophy secondary to age-related macular degeneration and the complement pathway as a therapeutic target.
      CFD is the rate-limiting enzyme in the alternative complement pathway and is present in the plasma at low concentrations relative to other complement proteins.
      • Volanakis J.E.
      • Narayana S.V.
      Complement factor D, a novel serine protease.
      ,
      • Do D.V.
      • Pieramici D.J.
      • van Lookeren Campagne M.
      • et al.
      A Phase Ia dose-escalation study of the anti-factor D monoclonal antibody fragment FCFD4514S in patients with geographic atrophy.
      For these reasons, CFD was considered a potential therapeutic target for inhibition of the alternative complement pathway, and consequently, treatment of GA.
      • Do D.V.
      • Pieramici D.J.
      • van Lookeren Campagne M.
      • et al.
      A Phase Ia dose-escalation study of the anti-factor D monoclonal antibody fragment FCFD4514S in patients with geographic atrophy.
      ,
      • Katschke Jr., K.J.
      • Wu P.
      • Ganesan R.
      • et al.
      Inhibiting alternative pathway complement activation by targeting the factor D exosite.
      Figure thumbnail gr1
      Figure 1Overview of the complement pathways. C2 = complement component 2; C3 = complement component 3; C4 = complement component 4; C5 = complement component 5; CFB = complement factor B; CFD = complement factor D.
      Lampalizumab, an antigen-binding fragment (Fab) of a humanized monoclonal antibody directed against CFD, was developed as a therapeutic agent for GA.
      • Do D.V.
      • Pieramici D.J.
      • van Lookeren Campagne M.
      • et al.
      A Phase Ia dose-escalation study of the anti-factor D monoclonal antibody fragment FCFD4514S in patients with geographic atrophy.
      • Katschke Jr., K.J.
      • Wu P.
      • Ganesan R.
      • et al.
      Inhibiting alternative pathway complement activation by targeting the factor D exosite.
      • Loyet K.M.
      • Good J.
      • Davancaze T.
      • et al.
      Complement inhibition in cynomolgus monkeys by anti–factor D antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration.
      In in vitro and in vivo studies, lampalizumab inhibited the alternative complement pathway but had no direct effect on the classical or lectin pathways of the complement system.
      • Katschke Jr., K.J.
      • Wu P.
      • Ganesan R.
      • et al.
      Inhibiting alternative pathway complement activation by targeting the factor D exosite.
      ,
      • Loyet K.M.
      • Good J.
      • Davancaze T.
      • et al.
      Complement inhibition in cynomolgus monkeys by anti–factor D antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration.
      Lampalizumab was investigated in the phase 2 MAHALO trial in 123 patients with GA secondary to AMD, and met the primary endpoint of reducing GA lesion area progression compared with sham treatment.
      • Yaspan B.L.
      • Williams D.F.
      • Holz F.G.
      • et al.
      Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration.
      Lampalizumab showed particular benefit in the subgroup of patients who carried the CFI risk allele.
      • Yaspan B.L.
      • Williams D.F.
      • Holz F.G.
      • et al.
      Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration.
      The efficacy and safety of lampalizumab were further investigated in the phase 3 Chroma (906 patients) and Spectri (975 patients) trials.
      • Holz F.G.
      • Sadda S.R.
      • 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.
      In these identically designed trials, patients with bilateral GA and no prior active choroidal neovascularization in either eye were randomized to receive lampalizumab administered intravitreally (every 6 weeks [Q6W] or every 4 weeks [Q4W]) or sham injections. Among 1881 patients, the adjusted mean increase in GA lesion area from baseline to week 48 was between 1.93 and 2.09 mm2 across all groups in both trials. There were no significant reductions in GA lesion area growth with lampalizumab compared with sham treatment in any group in either study. There was also no observed benefit from lampalizumab treatment in any subgroup, including those with the CFI risk allele.
      To better understand why a clinical effect of lampalizumab might not have been observed in the Chroma and Spectri trials, we investigated the effect of lampalizumab on complement pathway activities. To this end, we developed 6 novel assays using procured aqueous humor samples. Thereafter, aqueous humor samples collected from patients in the Chroma and Spectri trials were analyzed using the final assays.

      Methods

      Study Design

      This was a post hoc study analyzing aqueous humor samples collected from patients included in the Chroma (NCT02247479) and Spectri (NCT02247531) trials. A detailed description of the trial designs has been published previously.
      • Holz F.G.
      • Sadda S.R.
      • 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.
      Briefly, Chroma and Spectri were 2 identically designed, phase 3, double-masked, sham-controlled trials conducted at 275 sites in 23 countries. Nine-hundred and six patients were enrolled in Chroma and 975 patients were enrolled in Spectri. Patients were randomized in a 2:1:2:1 ratio to receive intravitreous lampalizumab 10 mg Q6W, sham procedure Q6W, lampalizumab 10 mg Q4W, or sham procedure Q4W for 96 weeks. All sites received institutional review board or ethics committee approval before study initiation and all participants provided written informed consent.
      • Holz F.G.
      • Sadda S.R.
      • 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.

      Development of Complement Assays

      CFD concentrations in aqueous humor were determined by enzyme-linked immunosorbent assay as previously described.
      • Le K.N.
      • Gibiansky L.
      • Good J.
      • et al.
      A mechanistic pharmacokinetic/pharmacodynamic model of factor D inhibition in cynomolgus monkeys by lampalizumab for the treatment of geographic atrophy.
      Six novel assays were developed on the Simoa (Quanterix, Billerica, MA, USA) platform to assess complement activity in the eye. The assays were designed to measure levels of full-length CFB; the Bb fragment; full-length C3 (intact C3); the C3c, iC3b, and C3b fragments of C3 (processed C3); full-length complement component 4 (C4; intact C4); and the C4c and C4b fragments of C4 (processed C4).
      Fig S2 (available at www.aaojournal.org) shows the configuration of each of the 6 assays. For all assays, the capture antibody was conjugated to carboxylated beads and the detection antibody was biotinylated. Per the standard Simoa assay setup, all assays used streptavidin-conjugated beta-galactosidase and resorufin galactopyranoside from Quanterix to allow for final readout. The CFB assay used a monoclonal capture antibody specific to Ba from AbCam (Cambridge, MA, USA) coupled with a monoclonal detection antibody, GNE Ab 4674, specific to Bb from Genentech, Inc. (South San Francisco, CA, USA). The Bb assay used GNE Ab 4674 as the capture antibody and a Bb neoepitope monoclonal antibody from Quidel (San Diego, CA, USA) as the detection antibody. The intact C3 assay used a polyclonal antibody specific for C3a from R&D Systems (Minneapolis, MN, USA) as the capture antibody and a CappelTM polyclonal antibody against C3 from MP Biomedicals (Solon, OH, USA) as the detection antibody. The processed C3 assay used a monoclonal capture antibody against iC3b/C3b/C3c from Kerafast (Boston, MA, USA) and a polyclonal detection antibody against C3 from Protos Immunoresearch (Burlingame, CA, USA) for detection. The intact C4 assay used a polyclonal capture antibody against C4 from Abbexa (Houston, TX, USA) and a monoclonal detection antibody against C4c from Quidel (San Diego, CA, USA). The processed C4 assay used a monoclonal capture antibody against C4c from United States Biological (Salem, MA, USA) and a monoclonal detection antibody against C4 from LSBio (Seattle, WA, USA).
      To demonstrate that the 6 assays could capture dynamic activation of the complement pathways, 25 micrograms of lampalizumab or glycoprotein D Fab (control Fab) were added to normal human serum aliquots (0.5 ml each) and either the alternative or classical complement pathway was activated with zymosan (0.5 mg/ml final concentration) or heat-aggregated gamma globulin (0.5 mg/ml final concentration), respectively, followed by incubation at 37°C. At regular time points from 0 to 30 or 60 minutes, portions of the samples were collected, and the activation was stopped by adding ethylenediaminetetraacetic acid and storing on ice. After each time course was complete, the resulting samples were measured using microtiter plate assays.
      Overall, all 6 novel assays were found to have good sensitivity and specificity (Table S1, available at www.aaojournal.org) for measuring the activation of the alternative and classical complement pathways (Appendix and Fig S3A–C, available at www.aaojournal.org).

      Aqueous Humor Sample Collection and Testing

      Optional aqueous humor samples were collected from patients who consented to the procedure and sample acquisition. The aqueous humor sample was collected in the clinic by the treating physician before any treatment was administered. The collection consisted of an anterior chamber paracentesis whereby a drop of topical anesthetic was placed on the cornea and a 30-gauge needle was passed through the limbus into the anterior chamber and approximately 0.1 mL of aqueous fluid was removed. All samples were subsequently frozen, shipped on dry ice, and stored at < -70˚C at a central laboratory prior to analysis.
      Longitudinal baseline and week 24 aqueous humor samples were selected for analysis in a blinded manner across all treatment (lampalizumab Q6W and lampalizumab Q4W) and sham groups (sham Q6W and sham Q8W) in the Chroma and Spectri trials. Each sample was tested in duplicate with each of the 6 complement assays developed for this study.

      Statistical Analysis

      We calculated the percent coefficient of variation (CV%) for each analyte in every sample by dividing the standard deviation by the mean and multiplying by 100. Samples with CV% greater than 30% were excluded from the analysis (n = 2). For the remaining samples, their mean values were analyzed. Results below the limit of quantification were assigned the value of the assay lower limit of quantification.
      For each complement pathway, we calculated the ratio of the processed form to the intact form (ie, complement activity). Percentage changes from baseline were calculated by subtracting the baseline value from the week 24 value and dividing the result by the baseline value. Correlations were calculated using Spearman’s rho, a nonparametric measure for the strength of association. A Wilcoxon rank-sum test with continuity correction was used to assess differences between treatment groups.

      Results

      Pharmacodynamic Effect of Lampalizumab

      Aqueous humor samples of 100 patients from Chroma and Spectri trials balanced by treatment groups (lampalizumab Q6W, and lampalizumab Q4W) and sham group for two time points (baseline and week 24) were collected. After excluding patients with duplicate CV% values greater than 30% (n = 2) and those without paired baseline/week 24 detectable samples for all complement assays (n = 1) , 97 patients were included in the analysis population. Table 2 shows that demographic and baseline characteristics were balanced between the 3 groups of patients.
      Table 2Demographic and Baseline Characteristics of Patients Providing Aqueous Humor Samples
      CharacteristicSham (n = 30)LQ6W (n = 32)LQ4W (n = 35)Overall (N = 97)
      Sex, n (%)
      Female16 (53.3)21 (65.6)25 (71.4)62 (63.9)
      Male14 (46.7)11 (34.4)10 (28.6)35 (36.1)
      Age at parent reference date, years
      Mean79.7 (8.04)79.1 (8.61)80.1 (7.96)79.6 (8.13)
      Median [min, max]80.5 [60.0, 93.0]81.5 [61.0, 92.0]81.0 [63.0, 93.0]81.0 [60.0, 93.0]
      Tobacco use, n (%)
      Current3 (10.0)1 (3.1)4 (11.4)8 (8.2)
      Never10 (33.3)19 (59.4)9 (25.7)38 (39.2)
      Previous17 (56.7)12 (37.5)22 (62.9)51 (52.6)
      Baseline study eye GA area by FAF, mm2
      Mean6.33 (2.87)9.54 (4.92)8.63 (3.33)8.22 (4.00)
      Median [min, max]5.54 [2.94, 13.9]8.57 [2.63, 17.7]7.82 [4.25, 17.3]7.82 [2.63, 17.7]
      Baseline study eye GA lesion contiguity, n (%)
      Multifocal24 (80.0)27 (84.4)30 (85.7)81 (83.5)
      Not multifocal6 (20.0)5 (15.6)5 (14.3)16 (16.5)
      Baseline study eye location of GA lesion, n (%)
      Nonsubfoveal14 (46.7)16 (50.0)19 (54.3)49 (50.5)
      Subfoveal16 (53.3)16 (50.0)16 (45.7)48 (49.5)
      Baseline aqueous humor total CFD, ng/ml
      Mean47.6 (24.8)51.0 (33.9)54.5 (26.7)51.2 (28.6)
      Median [min, max]44.6 [17.2, 128]42.4 [15.0, 148]51.7 [20.4, 128]47.5 [15.0, 148]
      Change in GA area from baseline to week 24, mm2
      Mean1.13 (0.999)1.04 (0.718)1.00 (0.689)1.05 (0.798)
      Median [min, max]0.950 [0.140, 3.93]0.910 [0.0200, 2.66]0.920 [0.110, 2.52]0.940 [0.0200, 3.93]
      Missing1 (3.3)01 (2.9)2 (2.1)
      Change in GA area from baseline to week 48, mm2
      Mean2.15 (1.45)2.06 (1.26)2.16 (1.32)2.13 (1.33)
      Median [min, max]1.72 [0.500, 6.20]1.71 [0.220, 5.25]1.79 [0.520, 5.11]1.73 [0.220, 6.20]
      Intact C3, ng/ml
      Mean4260 (3600)4930 (3390)5160 (2940)4810 (3290)
      Median [min, max]3260 [763, 20 200]4010 [751, 13 700]4700 [1350, 12 700]3970 [751, 20 200]
      Processed C3, ng/ml
      Mean2230 (1740)2660 (2030)2530 (1370)2480 (1710)
      Median [min, max]1880 [666, 9970]2100 [309, 7740]2220 [729, 7120]2060 [309, 9970]
      Intact C4, ng/ml
      Mean844 (685)937 (988)890 (634)891 (775)
      Median [min, max]584 [160, 2930]790 [64.7, 4750]748 [195, 2990]697 [64.7, 4750]
      Processed C4, ng/ml
      Mean335 (194)373 (289)367 (203)359 (231)
      Median [min, max]309 [93.5, 866]298 [40.4, 1100]340 [112, 1060]317 [40.4, 1100]
      CFB, ng/ml
      Mean723 (425)758 (445)858 (447)784 (439)
      Median [min, max]600 [239, 2400]736 [128, 1670]729 [249, 2070]668 [128, 2400]
      Bb, ng/ml
      Mean136 (96.8)174 (149)168 (95.8)160 (116)
      Median [min, max]114 [40.1, 502]125 [22.8, 520]150 [38.5, 443]125 [22.8, 520]
      C3 = complement component 3; C4 = complement component 4; CFB, complement factor B; CFD, complement factor D; FAF = fundus autofluorescence; GA = geographic atrophy; LQ4W = lampalizumab every 4 weeks; LQ6W = lampalizumab every 6 weeks; max = maximum; min = minimum; SD = standard deviation.
      Proof of target engagement was evident by similar, marked percentage increases in CFD levels in the aqueous humor at week 24 compared with baseline in the lampalizumab Q6W group (median, +91.6%; mean, +97.0%) and Q4W group (median, +124.0%; mean, +144.0%), consistent with previous preclinical observations
      • Loyet K.M.
      • Good J.
      • Davancaze T.
      • et al.
      Complement inhibition in cynomolgus monkeys by anti–factor D antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration.
      ; this effect was not observed in the sham group (median, –2.6%; mean, +7.5%; Fig 4A). Only a moderate correlation was observed between the percentage change from baseline to week 24 in CFD levels and lampalizumab concentration in aqueous humor (Spearman’s rho = 0.583; Fig S5A, available at www.aaojournal.org).
      Figure thumbnail gr2
      Figure 4(A) Total complement factor D (CFD) levels (ng/ml) and (B) Bb:complement factor B (CFB) ratio. Plots on left show values at baseline and week 24; plots on right show percentage change from baseline to week 24. LQ4W = lampalizumab every 4 weeks; LQ6W = lampalizumab every 6 weeks.
      Complement factor B and Bb levels in the aqueous humor at baseline and week 24 by treatment and sham groups are shown in Fig S6 (available at www.aaojournal.org). Fig 4B show that there was a reduction in the Bb:CFB ratio from baseline to week 24 in the lampalizumab Q6W group (median, –41.3%; mean, –40.5%) and Q4W group (median, –43.4%; mean, –40.3%). A similar reduction was also observed in the combined Q6W and Q4W group (median, –42.4%; mean, –40.4%). Conversely, there was a small increase in the Bb:CFB ratio from baseline to week 24 in the sham group (median, +0.26%; mean, +9.04%; P < 0.001 for lampalizumab versus sham groups). These results confirmed that lampalizumab engaged and inhibited the target alternative complement pathway. Similar reductions in the Bb:CFB ratio in the lampalizumab Q6W and Q4W groups as well as no meaningful correlation between Bb:CFB ratio and percent changes from baseline to week 24 in either CFD (Fig S7A, available at www.aaojournal.org) or lampalizumab (Fig S5B, available at www.aaojournal.org; Spearman’s rho = 0.272) levels observed in aqueous humor suggested that maximum target inhibition had been achieved and dosing with higher concentrations or more frequent dosing would be unlikely to result in greater inhibition of the alternative complement pathway.

      Effect of Lampalizumab on Downstream and Classical Complement Pathway Activity

      No change in C3 levels (P = 0.23; Fig S8A, available at www.aaojournal.org) or C3 processing (Fig 9A) was observed in lampalizumab-treated patients compared with the sham group despite the observed reduction in Bb:CFB ratio. There was also no correlation between magnitude of changes in processed C3:intact C3 and either Bb:CFB ratios or CFD levels in patients treated with lampalizumab (Fig S7B, available at www.aaojournal.org and Fig S8B, available at www.aaojournal.org; Spearman’s rho = 0.28 for lampalizumab Q6W and 0.06 for lampalizumab Q4W), illustrating that no discernable inhibition of intact C3 processing was evident, even in those patients who had the strongest inhibition of CFB processing. Similarly, no meaningful change in C4 levels (P = 0.75; Fig S10A, available at www.aaojournal.org) or C4 processing (Fig 9B) was observed in lampalizumab-treated patients compared with sham group, and there was no consistent relationship between the magnitude of changes in processed C4:intact C4 and the Bb:CFB ratio (Fig S10B, available at www.aaojournal.org; Spearman’s rho = –0.27 for lampalizumab Q6W and +0.28 for lampalizumab Q4W). These results suggest that the lack of observed inhibition of C3 processing in lampalizumab-treated patients was not due to compensatory activation of the classical pathway.
      Figure thumbnail gr3
      Figure 9(A) Processed complement component 3 (C3):intact C3 ratio and (B) processed complement component 4 (C4):intact C4 ratio. Plots on left show values at baseline and week 24; plots on right show percentage change from baseline to week 24. LQ4W = lampalizumab every 4 weeks; LQ6W = lampalizumab every 6 weeks.

      Discussion

      Six novel assays were developed specifically to measure complement protein concentrations in aqueous humor, assess target engagement and pathway inhibition by lampalizumab, and characterize the resulting effect on the complement system. The aim was to gain better mechanistic insight into the effect of lampalizumab on the complement system in the context of a negative readout from the phase 3 Chroma and Spectri trials in patients with GA.
      • Holz F.G.
      • Sadda S.R.
      • 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.
      The collection of aqueous humor samples was important for studying ocular complement activation and the local effects of lampalizumab. Previous research has provided evidence for systemic, rather than local, complement activation in AMD, thereby supporting the analysis of aqueous humor samples.
      • Schick T.
      • Steinhauer M.
      • Aslanidis A.
      • et al.
      Local complement activation in aqueous humor in patients with age-related macular degeneration.
      In the samples from patients in the Chroma and Spectri trials, a 92% to 124% median increase in CFD levels, and a resulting median reduction in the Bb:CFB ratio of 41% to 43%, was observed, confirming that lampalizumab was effectively binding its target and inhibiting the alternative complement pathway. This increase in CFD is attributed to slower clearance of the lampalizumab–CFD complex relative to unbound CFD, and is consistent with previous preclinical studies
      • Loyet K.M.
      • Good J.
      • Davancaze T.
      • et al.
      Complement inhibition in cynomolgus monkeys by anti–factor D antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration.
      and limited observations in patients from the phase 1 and 2 lampalizumab trials.
      • Le K.N.
      • Gibiansky L.
      • van Lookeren Campagne M.
      • et al.
      Population Pharmacokinetics and Pharmacodynamics of Lampalizumab Administered Intravitreally to Patients With Geographic Atrophy.
      Similar changes in both CFD levels and the Bb:CFB ratio were observed between the 2 lampalizumab dosing schedules. While a modest correlation was observed between the aqueous humor concentration of lampalizumab and changes in CFD levels, no meaningful correlation was observed between lampalizumab concentrations and changes in the Bb:CFB ratio, suggesting that maximum pathway engagement with lampalizumab was achieved. Thus, there remains the question as to why a greater reduction in the Bb:CFB ratio was not observed. One explanation is that while previous free target suppression models estimate that only 0.001% to 0.01% of the CFD relative to baseline levels may be unbound and active at the time of aqueous humor sampling,
      • Le K.N.
      • Gibiansky L.
      • Good J.
      • et al.
      A mechanistic pharmacokinetic/pharmacodynamic model of factor D inhibition in cynomolgus monkeys by lampalizumab for the treatment of geographic atrophy.
      any residual CFD activity remaining in the presence of these saturating levels of lampalizumab may have driven the generation of Bb. Alternatively, it is worth noting that the vast majority of complement protein, including CFB, is produced in the liver and enters the circulation before reaching target organs such as the eye.
      • Lubbers R.
      • van Essen M.F.
      • van Kooten C.
      • Trouw L.A.
      Production of complement components by cells of the immune system.
      It is therefore possible that the residual Bb detected after lampalizumab treatment may largely have been generated in the periphery rather than within the eye, a notion supported by the fact that plasma levels of Ba have been reported to be much higher than in aqueous humor.
      • Schick T.
      • Steinhauer M.
      • Aslanidis A.
      • et al.
      Local complement activation in aqueous humor in patients with age-related macular degeneration.
      A limitation of our assays is that these are unable to distinguish between the disease-relevant Bb that was generated within the eye versus the inactive Bb that was produced elsewhere in the periphery. Finally, while care was taken to minimize ex vivo complement activation during testing, unintended activation during sample handling and laboratory processing cannot be ruled out entirely.
      Despite the significant reduction in alternative pathway activation, we observed no change in downstream C3 processing using our assays; this finding was surprising and may help explain the lack of clinical efficacy observed in the Chroma and Spectri trials. One explanation for this result is that the classical pathway plays a larger primary or compensatory role in C3 activation in the eye than previously assumed. However, it is worth noting that we also observed no change in C4 processing in the presence of lampalizumab and thus, believe it unlikely for classical pathway compensation to be the sole explanation for this result. Another possibility worth considering is that given the nature of the alternative pathway as a positive amplification loop, it is possible that some residual alternative pathway activity remains within the eye in the presence of lampalizumab; this in turn may be sufficient to fully activate downstream C3 processing (Fig 11). Finally, similar to the theory that peripherally generated Bb may be entering the eye, we also consider it possible that a significant amount of peripherally generated processed C3 is entering the eye, thus, preventing our ability to detect more subtle changes in C3 processing that occurred within the eye (Fig 11). Plasma levels of processed C3 are 15-fold higher than in the aqueous humor, while intact C3 levels are 353-fold higher.
      • Edmonds R.
      • Steffen V.
      • Honigberg L.A.
      • Chang M.C.
      The Role of the Complement Pathway in Clinical Progression of Geographic Atrophy: Analysis of the Phase 3 Chroma and Spectri Trials.
      However, higher complement activity measured in aqueous humor versus that in the plasma
      • Edmonds R.
      • Steffen V.
      • Honigberg L.A.
      • Chang M.C.
      The Role of the Complement Pathway in Clinical Progression of Geographic Atrophy: Analysis of the Phase 3 Chroma and Spectri Trials.
      along with the fact that we can detect changes in CFB processing despite similar differences in plasma versus aqueous humor concentrations would suggest that this hypothesis is unlikely .
      Figure thumbnail gr4
      Figure 11Schematic showing potential hypotheses for lower-than-expected reduction in CFB cleavage with CFD inhibition by lampalizumab
      Other complement pathway inhibitors are currently in development for GA. One example is pegcetacoplan (APL-2; Apellis Pharmaceuticals, Waltham, MA, USA), which directly targets and inhibits C3, and has shown promising phase 2 and 3 results for reducing the growth rate of GA lesions and slowing disease progression.

      Apellis Announces Top-Line Results from Phase 3 DERBY and OAKS Studies in Geographic Atrophy (GA) and Plans to Submit NDA to FDA in the First Half of 2022. Apellis Pharmaceuticals Inc., 2021. Available at: https://investors.apellis.com/news-releases/news-release-details/apellis-announces-top-line-results-phase-3-derby-and-oaks. Accessed August 9, 2022.

      ,
      • Liao D.S.
      • Grossi F.V.
      • El Mehdi D.
      • et al.
      Complement C3 Inhibitor Pegcetacoplan for Geographic Atrophy Secondary to Age-Related Macular Degeneration: A Randomized Phase 2 Trial.
      Similarly, avacincaptad pegol (Iveric Bio, New York, NY, USA), a C5 inhibitor, significantly reduced the GA growth rate in eyes with AMD over a 12-month period in a randomized, controlled phase 2/3 clinical trial.
      • Jaffe G.J.
      • Westby K.
      • Csaky K.G.
      • et al.
      C5 Inhibitor Avacincaptad Pegol for Geographic Atrophy Due to Age-Related Macular Degeneration: A Randomized Pivotal Phase 2/3 Trial.
      Another approach being investigated is targeting the alternative pathway using systemic CFB antisense oligonucleotides; the aim is to reduce complement activation in the eye by inhibiting the primary source of plasma CFB in the liver, which in turn reduces ocular CFB.
      • Grossman T.R.
      • Carrer M.
      • Shen L.
      • et al.
      Reduction in ocular complement factor B protein in mice and monkeys by systemic administration of factor B antisense oligonucleotide.
      Although confirmatory clinical data are needed, these strategies could potentially be more effective at inhibiting the C3 complement pathway than the targeting of CFD by lampalizumab.
      We showed that lampalizumab inhibited the cleavage of intact CFB into Bb in the eyes of patients with GA, as expected; however, this did not translate into a downstream reduction in complement pathway activation. Further assessment is required to clarify the role of the complement pathways in GA.
      Table S1Sensitivity and Specificity Results for Each of the 6 Assays
      Assay SensitivityAssay Specificity
      Assay (Range of Lower Limit of Quantification–Upper Limit of Quantification) (ng/ml)Tested Protein Tested Levels (ng/mL)Reactivity (%)
      CFB (0.012–2)Ba 2.5 - 50 (BLLOQ)
      Bb 0.22 - 25.0160 (BLLOQ)
      Bb (0.221–25.00)CFB 20- 605
      Ba 10 - 200 (BLLOQ)
      Intact C3 (0.028–6.9)C3a 20 - 400 (BLLOQ)
      C3b 3.33 - 100.4
      C3c 3.01 - 6.027.3
      C3d 20 - 40 iC3b 3 - 60 (BLLOQ)

      0 (BLLOQ)
      Processed C3 (0.014–10)C3 2.9 - 6.914
      C3a 20 - 400 (BLLOQ)
      iC3b 3 - 659
      C3c 3.01 - 6.02117
      C3d 20 - 400 (BLLOQ)
      Intact C4 (0.049–12)C4a lacking C-terminal arginine50 0.1
      C4b 502.0
      C4c 5

      C3d 50
      10–15

      0 (BLLOQ)
      Processed C4 (0.021–5)C4 1232
      C4a lackingC-terminal arginine 505.5
      C4b 267
      C4d 500

      Acknowledgments

      We acknowledge Teresa Davancaze for development of the CFD assay and sample testing support, Patrick Zoder for sample testing project management support, Eric Wakshul for assay development technical support, Janis Allen for assay development project management support, and Kelly Loyet for provision of assay protocols and reagents. We also acknowledge the contributions of all the Chroma and Spectri patients and investigators.
      Data Sharing
      Qualified researchers may request access to individual patient level data through the clinical study data request platform (https://vivli.org/). Further details on Roche's criteria for eligible studies are available here: https://vivli.org/members/ourmembers/. For further details on Roche's Global Policy on the Sharing of Clinical Information and how to request access to related clinical study documents, see here: https://www.roche.com/research_and_development/who_we_are_how_we_work/clinical_trials/our_commitment_to_data_sharing.htm.

      References

        • Sunness J.S.
        • Rubin G.S.
        • Applegate C.A.
        • et al.
        Visual function abnormalities and prognosis in eyes with age-related geographic atrophy of the macula and good visual acuity.
        Ophthalmology. 1997; 104: 1677-1691
        • Flaxman S.R.
        • Bourne R.R.A.
        • Resnikoff S.
        • et al.
        Global causes of blindness and distance vision impairment 1990–2020: a systematic review and meta-analysis.
        Lancet Glob Health. 2017; 5: e1221-e1234
        • Holz F.G.
        • Strauss E.C.
        • Schmitz-Valckenberg S.
        • van Lookeren Campagne M.
        Geographic atrophy: clinical features and potential therapeutic approaches.
        Ophthalmology. 2014; 121: 1079-1091
        • Fritsche L.G.
        • Chen W.
        • Schu M.
        • et al.
        Seven new loci associated with age-related macular degeneration.
        Nat Genet. 2013; 45: 433-439
        • Gorin M.B.
        Genetic insights into age-related macular degeneration: controversies addressing risk, causality, and therapeutics.
        Mol Aspects Med. 2012; 33: 467-486
        • Loyet K.M.
        • Deforge L.E.
        • Katschke Jr., K.J.
        • et al.
        Activation of the alternative complement pathway in vitreous is controlled by genetics in age-related macular degeneration.
        Invest Ophthalmol Vis Sci. 2012; 53: 6628-6637
        • Scholl H.P.
        • Charbel Issa P.
        • Walier M.
        • et al.
        Systemic complement activation in age-related macular degeneration.
        PLoS One. 2008; 3e2593
        • Stanton C.M.
        • Yates J.R.
        • den Hollander A.I.
        • et al.
        Complement factor D in age-related macular degeneration.
        Invest Ophthalmol Vis Sci. 2011; 52: 8828-8834
        • Heesterbeek T.J.
        • Lechanteur Y.T.E.
        • Lores-Motta L.
        • et al.
        Complement activation levels are related to disease stage in AMD.
        Invest Ophthalmol Vis Sci. 2020; 61: 18
        • Boyer D.S.
        • Schmidt-Erfurth U.
        • van Lookeren Campagne M.
        • et al.
        The pathophysiology of geographic atrophy secondary to age-related macular degeneration and the complement pathway as a therapeutic target.
        Retina. 2017; 37: 819-835
        • Volanakis J.E.
        • Narayana S.V.
        Complement factor D, a novel serine protease.
        Protein Sci. 1996; 5: 553-564
        • Do D.V.
        • Pieramici D.J.
        • van Lookeren Campagne M.
        • et al.
        A Phase Ia dose-escalation study of the anti-factor D monoclonal antibody fragment FCFD4514S in patients with geographic atrophy.
        Retina. 2014; 34: 313-320
        • Katschke Jr., K.J.
        • Wu P.
        • Ganesan R.
        • et al.
        Inhibiting alternative pathway complement activation by targeting the factor D exosite.
        J Biol Chem. 2012; 287: 12886-12892
        • Loyet K.M.
        • Good J.
        • Davancaze T.
        • et al.
        Complement inhibition in cynomolgus monkeys by anti–factor D antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration.
        J Pharmacol Exp Ther. 2014; 351: 527-537
        • Yaspan B.L.
        • Williams D.F.
        • Holz F.G.
        • et al.
        Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration.
        Sci Transl Med. 2017; 9eaaf1443
        • Holz F.G.
        • Sadda S.R.
        • 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: 666-677
        • Le K.N.
        • Gibiansky L.
        • Good J.
        • et al.
        A mechanistic pharmacokinetic/pharmacodynamic model of factor D inhibition in cynomolgus monkeys by lampalizumab for the treatment of geographic atrophy.
        J Pharmacol Exp Ther. 2015; 355: 288-296
        • Schick T.
        • Steinhauer M.
        • Aslanidis A.
        • et al.
        Local complement activation in aqueous humor in patients with age-related macular degeneration.
        Eye (Lond). 2017; 31: 810-813
        • Le K.N.
        • Gibiansky L.
        • van Lookeren Campagne M.
        • et al.
        Population Pharmacokinetics and Pharmacodynamics of Lampalizumab Administered Intravitreally to Patients With Geographic Atrophy.
        CPT Pharmacometrics Syst Pharmacol. 2015; 4: 595-604
        • Lubbers R.
        • van Essen M.F.
        • van Kooten C.
        • Trouw L.A.
        Production of complement components by cells of the immune system.
        Clin Exp Immunol. 2017; 188: 183-194
        • Edmonds R.
        • Steffen V.
        • Honigberg L.A.
        • Chang M.C.
        The Role of the Complement Pathway in Clinical Progression of Geographic Atrophy: Analysis of the Phase 3 Chroma and Spectri Trials.
        Ophthalmology. 2022; ([In Preparation])
      1. Apellis Announces Top-Line Results from Phase 3 DERBY and OAKS Studies in Geographic Atrophy (GA) and Plans to Submit NDA to FDA in the First Half of 2022. Apellis Pharmaceuticals Inc., 2021. Available at: https://investors.apellis.com/news-releases/news-release-details/apellis-announces-top-line-results-phase-3-derby-and-oaks. Accessed August 9, 2022.

        • Liao D.S.
        • Grossi F.V.
        • 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: 186-195
        • Jaffe G.J.
        • Westby K.
        • Csaky K.G.
        • 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: 576-586
        • Grossman T.R.
        • Carrer M.
        • Shen L.
        • et al.
        Reduction in ocular complement factor B protein in mice and monkeys by systemic administration of factor B antisense oligonucleotide.
        Mol Vis. 2017; 23: 561-571