Abstract
Introduction:
Diabetic retinopathy (DR) is one of the most frequent microvascular complications of diabetes that can lead to blindness. Laser treatment has been the gold standard treatment for diabetic macular edema (DME) and proliferative diabetic retinopathy (PDR) for many years. Recently, the role of vascular endothelial growth factor (VEGF) has been established in the pathogenesis of DR, and the use of intravitreal anti-VEGF therapy has gained popularity for the management of DR.
Areas covered:
This review includes a brief overview of the efficacy and safety of currently available (bevacizumab, ranibizumab, and aflibercept) and potential future (brolucizumab, faricimab, and KSI-301) anti-VEGF agents in patients with DR based mainly on publicly available data from phase 1, 2 and 3 clinical trials.
Expert opinion:
Clinical trials investigating the efficacy of intravitreal bevacizumab, ranibizumab, and aflibercept injections demonstrated favorable functional and anatomical outcomes in patients with DME. Moreover, the use of these anti-VEGF agents showed a significant improvement in the severity of DR. Recent clinical research for future anti-VEGF molecules aims to provide higher target-protein binding affinity and prolonged therapeutic effect. Brolucizumab, faricimab, and KSI-301 are three novel anti-VEGF agents that demonstrate promising data for the management of DME and potentially DR.
Keywords: Aflibercept, bevacizumab, diabetic macular edema, diabetic retinopathy, ranibizumab, vascular endothelial growth factor, anti-vascular endothelial growth factor
1. Introduction
Diabetic retinopathy (DR) is among the most common complications of diabetes and is the leading cause of visual impairment and blindness among the working-age population (aged 15–64) worldwide [1–4]. Diabetic macular edema (DME) by far is the most frequent vision-threatening complication of DR that can develop at any stage of DR and therefore creates a major challenge for the health-care system. Complications of proliferative DR (PDR) include vitreous hemorrhage, tractional retinal detachment, and neovascular glaucoma, all of which are other significant reasons for vision loss in DR [5,6].
Although the complex pathophysiology of DR is not entirely understood, vascular endothelial growth factor (VEGF), particularly VEGF-A, is known to be a key mediator in disease progression and subsequent development of vision threatening complications. In DR, chronic hyperglycemia is thought to damage endothelial cells and basement membrane proteins in retinal vasculature. Such damage, accompanied by vascular hyperpermeability and production of proinflammatory cytokines, can lead to retinal ischemia that can result in a maladaptive increase in VEGF expression. The breakdown of the retinal-blood barrier and resulting angiogenesis from increased VEGF is thought to play a major role in visual loss from DR [7,8]. This understanding has led physicians to use intravitreal anti-VEGF therapy to reduce DR-associated complications and improve vision in affected patients. To date, several VEGF inhibitors, such as bevacizumab (BVZ), ranibizumab (RBZ) and aflibercept (AFB), have been administered by intravitreal injection and have been studied in prospective, randomized phase 2 and phase 3 clinical trials with favorable results in patients with DME and DR.
In this review, we aim to conduct a comprehensive overview summarizing the role, efficacy and safety of several intravitreal anti-VEGF therapies and promising therapeutic targets in patients with DME and DR.
2. Agents in clinical usage
2.1. Bevacizumab
2.1.1. Structure and mechanism of action
BVZ is a 148 kilodalton (kDa) recombinant whole humanized monoclonal immunoglobulin G1κ antibody produced in Chinese hamster ovary (CHO) mammalian cells [9]. It can generally be described as consisting of two main regions, the fragment antigen-binding (Fab) and fragment crystallizable (Fc) regions. The antibodies consist of two light chains (VL-CL) and two complete heavy chains (VH-CH1-CH2-CH3) (kappa subclass) which provide two binding sites for VEGF [10]. Each heavy chain contains an N-linked glycan at a consensus site in the Fc region, which is the site for glycosylation (Figure 1).
Figure 1.

The structure of a full-length IgG1 bevacizumab antibody. Yellow stars represent glycosylation sites.
Fc, fragment crystallizable; Fab, fragment antigen-binding; Variable domains, VL and VH; Constant domains, CL, CH1, CH2, CH3.
The mechanism of action of BVZ is its binding to all active VEGF-A isoforms. Such binding prevents the interaction of VEGF-A with its receptors (VEGFR1 and VEGFR2) on the surface of endothelial cells, reducing endothelial cell proliferation, vascular leakage, and angiogenesis [9]. Table 1 summarizes the structural features and pharmacodynamics of BVZ.
Table 1.
Comparison of structural properties, pharmacodynamics, advantages and disadvantages, and status of different anti-VEGFs used in the clinical trials.
| Drug Name | Structure | Size | IV dose (mg) | Biological Target | Advantages | Disadvantages | Status |
|---|---|---|---|---|---|---|---|
| Bevacizumab | Humanized monoclonal G1ΰ antibody | 148kDa | 1.25 | All isoforms of VEGF-A | Cost-effective | No preservative free vials for intravitreal administration | Off label use |
| Ranibizumab | Humanized IgG1 kappa isotype monoclonal antibody fragment | 48kDa | 0.5 | All isoforms of VEGF-A | Small molecular weight allows for excellent retinal penetration,Less systemic absorption | One binging site for VEGF, Shorter systemic half-life | FDA approved |
| Aflibercept | Human recombinant fusion protein: VEGFR-1 and VEGFR-1 extracellular domains fused with IgG1 Fc | 115kDa | 2 | All isoforms of VEGF-A, VEGF-B, PIGF-1 and PIGF-2 | Great affinity for VEGF-A, VEGF-B, PIGF-1 and PIGF-2, Less systemic absorption, Allows for less frequent intravitreal injections | Incidence of intraocular inflammation | FDA approved |
| Brolicizumab | Humanized single-chain antibody fragment | 26kDa | 6 mg | All isoforms of VEGF-A | Low molecular weight allows for a higher molecular dosing and more durability | Concerns over inflammatory profile, particularly occlusive retinal vasculitis | FDA approved |
| Faricimab | Humanized bispecific antibody | 150kDa | 6 mg | All isoforms of VEGF-A and Ang-2 | Dual pathway inhibition allows extended dosing intervals | Incidence of intraocular inflammation greater numerically compared to Aflibecept | FDA approved |
| KSI-301 | Anti-VEGF antibody-biopolymer conjugate | 950kDa | 5 mg | All isoforms of VEGF-A | Large molar dose of drug is designed for more bioavailability and durability | Efficacy and safety are still under investigation | Not-FDA approved |
Ang-2, Angiopoietin-2, Fc, fragment crystallizable, FDA, Food and Drug Administration, IV, intravitreal, kDa, kilodalton, mg, milligram, PIGF, placental growth factor, VEFG, vascular endothelial growth factor, VEGFR, vascular endothelial growth factor receptor.
2.1.2. Bevacizumab for diabetic macular edema
A survey to evaluate the safety and efficacy of BVZ for DME was first conducted as phase 2, multicenter, randomized controlled trial of the diabetic retinopathy clinical research (DRCR) network investigators. In this study, subjects with DME were randomized into five arms: 1. focal laser group, 2. 1.25 mg BVZ at baseline and week 6, 3. 2.5 mg BVZ at baseline and week 6, 4. 1.25 mg BVZ at baseline and sham at week 6 and 5. 1.25 mg BVZ with photocoagulation at week 3. Patients treated with both 1.25 mg and 2.5 mg BVZ had a greater reduction in central macular thickness (CMT) at 3 weeks than those with focal laser. However, no statistical difference in CMT was found between patients treated with BVZ and focal laser in long-term follow-up. Patients treated regularly with BVZ had greater improvement in best-corrected visual acuity (BCVA) over one line compared to baseline over the 12 weeks [11]. Another interventional retrospective multicenter study by the Pan-American Collaborative Retina Study Group (PACORES) showed significant improvement in BCVA and CMT at 12 months in both two BVZ regimens (1.25 and 2.5 mg) in patients with DME (101 eyes in 82 patients) [12]. No differences were seen between two different BVZ doses in both studies [11,12]. This positive data have been supported by a prospective single-center, randomized 2-year trial, intravitreal BVZ (1.25 mg, q6) study (BOLT) showing significant improvement in VA (mean BCVA improvement of 8.6 letters) in BVZ arm compared to macular laser treatment arm (mean BCVA loss of 0.5 letters) over 24 months [13]. Table 2 summarizes the clinical trials using intravitreal BVZ therapy in the management of DME.°Results for pooled RISE and RIDE study *Result for the subset of patients with a VA score of 20/50 or worse
Table 2.
List of clinical trials summarizing intravitreal anti-VEGF therapy in the management of diabetic macular edema
| Study | No. of Patients (Study Eyes) | Duration, mo | Inclusion Criteria | Intervention | BCVA Change, letters, (Mean) | CMT Change, (Mean) | No of injections, (Mean) |
|---|---|---|---|---|---|---|---|
| DRCR.net [Citation11] | 121 | 12 | BCVA 20/32–20/320 CMT ≥275 μm |
1. Focal laser 2. BVZ (1.25 mg, q6) 3. BVZ (2.5 mg, q6) 4. BVZ at baseline (1.25 mg) and sham at 6 weeks 5. BVZ (1.25 mg, q6) with photocoagulation at 3 weeks |
−1†, (−6,+5) +5†, (+1. +12) +7†, (+4, +11) +4†, (−3, +7) 0†, (−5, +8) |
−40 μm†, (−146, +85) −56 μm†, (−120, −6) −47 μm†, (−125, −16) −5 μm†, (−4,+53) −40 μm†, (−103, +33) |
N/A |
| BOLT [Citation13] | 80 | 24 | BCVA 20/40–20/320 CMT ≥270 μm |
1. BVZ (1.25 mg, q6) 2. Laser therapy |
8.6 ± 9.1 –0.5 ± 10.6 |
−146 ± 171 –118 ± 112 |
12†, (11,15) |
| READ 2 [Citation27] | 74 | 36 | BCVA 20/40–20/320 CMT ≥250 μm |
1. RBZ (0.5 mg, q8, after initial 2q4 doses) 2. Laser alone 3. RBZ (0.5 mg, q12) + laser |
10.3 1.4 8.9 |
−132 −193 −243 |
5.4 2.3 3.3 |
| READ 3 [Citation28] | 152 | 24 | BCVA 20/40–20/320 CMT ≥250 μm |
1. RBZ (0.5 mg, PRN, after 6 initial q4 doses) 2. RBZ (2 mg, PRN, after 6 initial q4 doses) |
11.06 6.78 |
−192.53 –170.64 |
18.4 17.3 |
| RESOLVE [Citation29] | 151 | 12 | BCVA 20/40–20/160 CMT ≥300 μm |
1. RBZ (pooled 0.3 and 0.5 mg, q4) 2. Sham |
7.8 ± 7.7 –0.1 ± 9.8 |
−194.2 ± 135.1 –48.4 ± 153.4 |
10.2 ± 2.5 8.9 ± 3.5 |
| RISE [Citation37] | 377 | 36 | BCVA 20/40–20/320 CMT ≥275 μm |
1. RZB (0.3 mg, q4) 2. RZB (0.5 mg, q4) 3. Sham |
11.0 ± 12.9 14.2 ± 12.8 4.3 ± 14.9 |
−261.2 ± 196.2 –269.1 ± 178.9 –200.1 ± 215.6 |
10.6° ±2.6 10.9° ± 2.2 10.0° ± 2.0 |
| RIDE [Citation37] | 382 | 36 | BCVA 20/40–20/320 CMT ≥275 μm |
1. RZB (0.3 mg, q4) 2. RZB (0.5 mg, q4) 3. Sham |
11.4 ± 16.3 10.6 ± 12.9 4.7 ± 13.3 |
−261.8 ± 180.8 –266.7 ± 207.8 –213.2 ± 193.5 |
10.6° ±2.6 10.9° ± 2.2 10.0° ± 2.0 |
| Protocol I [Citation30] | 235 | 60 | BCVA 20/32–20/320 | 1. RZB (0.5 mg, q4) +prompt laser 2. RZB (0.5 mg, q4) +deferred laser |
8 ± 13 10 ± 13 |
−167 ± 168 –165 ± 165 |
13† (9,24) 17† (11, 27) |
| RESTORE [Citation34] | 345 | 12 | BCVA 20/32–20/160 | 1. RZB (0.5 mg, q4) +sham laser 2. RZB (0.5 mg, q4) + laser 3. Sham injection + laser |
6.1 ± 6.43 5.9 ± 7.92 0.8 ± 8.56 |
−118.7 ± 115.07 –128.3 ± 114.43 –61.3 ± 132.29 |
7.0 ± 2.81 6.8 ± 2.95 7.3 ± 3.22 |
| REVEAL [Citation35] | 396 | 12 | BCVA 20/32–20/160 | 1. RZB (0.5 mg, q4) +sham laser 2. RZB (0.5 mg, q4) + laser 3. Sham injection + laser |
6.6 ± 7.68 6.4 ± 10.67 1.8 ± 8.27 |
−134.6 –171.8 −57.2 |
7.8 ± 2.94 7.0 ± 3.07 7.4 ± 3.14 |
| DA VINCI [Citation55] | 176 | 12 | BCVA 20/40–20/320 CMT ≥250 μm |
1. AFB (0.5 mg, q4) 2. AFB (2 mg, q4) 3. AFB (2 mg, q8) 4. AFB (2 mg, PRN) 5. Laser |
11.0 13.1 9.7 12.0 –1.3 |
−165.4 –227.4 −187.8 –180.3 −58.4 |
11.7 ± 2.49 10.8 ± 2.87 7.2 ± 1.74 7.4 ± 3.19 N/A |
| VISTA [Citation56] | 461 | 36 | BCVA 20/40–20/320 | 1. AFB (2 mg, q4) 2. AFB (2 mg, q8) 3. Laser |
10.4 10.5 1.4 |
−200.4 –190.1 −109.8 |
29.6 ± 9.8 18.1 ± 4.8 |
| VIVID [Citation57] | 404 | 36 | BCVA 20/40–20/320 | 1. AFB (2 mg, q4) 2. AFB (2 mg, q8) 3. Laser |
10.3 11.7 1.6 |
−215.2 –202.8 −122.6 |
32.0 ± 9.7 18.1 ± 11.9 |
| Protocol V [Citation58] | 702 | 24 | BCVA >20/25 CMT ≥250 μm |
1. AFB (2 mg, q4) 2. Laser 3.Observation |
0.9 ± 6.4 0.1 ± 6.3 –0.4 ± 6.4 |
−48 ± 65 –41 ± 75 –42 ± 75 |
8† (6–11) 7† (5–9) 9† (6–11) |
| Protocol T [Citation59] | 660 | 24 | BCVA 20/40–20/320 | 1. AFB (2 mg, 4q) 2. BVZ (1.25 mg, q4) 3. RBZ (0.3 mg. q4) |
18.1 ± 13.8* 13.3 ± 13.4* 16.1 ± 12.1* |
−211 ± 155* −185 ± 158* −174 ± 159* |
15† (11, 17) 16† (12–20) 15† (11–19) |
Median.
AFB, aflibercept; BCVA, best corrected visual acuity; BVZ, bevacizumab; CMT, central macular thickness; mo: month; PRN, pro re nata; q, every; RBZ, ranibizumab; VEGF, vascular endothelial growth factor.
2.1.3. Bevacizumab for diabetic retinopathy
Panretinal photocoagulation (PRP) is an established treatment for PDR and severe non-proliferative DR (NPDR) described by the Early Treatment Diabetic Retinopathy Study (ETDRS) Research Group [14]. However, since the introduction of intravitreal VEGF inhibitors, numerous studies have sought to further elucidate the most effective treatments for DR.
One prospective study (IBeHi) of patients with high-risk PDR compared patients who were randomized to PRP (PRP group, n = 15) to those randomized to PRP plus intravitreal injection (PRP plus, n = 15). This study showed that the adjunctive use of intravitreal BVZ with PRP in patients with high-risk PDR was associated with a greater reduction in active neovascularization (NV) than PRP alone, but showed no significant difference in BCVA compared to PRP alone [15]. In a separate retrospective review of 60 consecutive eyes (30 patients) with severe PDR, PRP was performed in two sessions in all eyes in both the control group and the interventional group, and intravitreal BVZ 1.25 mg was performed at 1 week before initial PRP in only the interventional group. Though seven eyes (23.3%) in the control group had worse vision by 2 lines and increased foveal thickness by over 50 μm at 24 weeks, none in the interventional group with BVZ had either worse vision or a significant increase in foveal thickness (P = 0.011) [15]. As a result, it was concluded that intravitreal BVZ before PRP might be able to prevent PRP-induced ocular complications.
A prospective, paired-eye, randomized pilot study was conducted for 12 months to investigate intravitreal BVZ monotherapy without prior PRP for symmetric untreated patients with severe NPDR (n = 5) and patients with PDR (n = 5). The right eye was randomly assigned to treatment with PRP or intravitreal BVZ 2.5 mg every 2 months, and the left eye received the other treatment. Final CMT was significantly decreased in eyes treated with BVZ (197 ± 17 μm) compared to those treated with PRP (243 ± 49 μm), respectively (p = 0.012). There was no difference in BCVA. Moreover, none of 10 eyes treated with BVZ injections every 2 months developed a complication, though two eyes treated with PRP developed vitreous hemorrhage (VH) and three of 10 eyes treated with PRP developed macular edema [16].
A subsequent representative retrospective multicenter interventional case series study was conducted by the PACORES group to evaluate the effects of intravitreal BVZ on retinal NV in patients with PDR [17]. Eighty-one (81) consecutive patients (97 eyes) with retinal NV due to PDR who received at least 1 intravitreal BVZ injection were reviewed in this study. Reinjections of BVZ were performed if there had been a recurrence of NV on ophthalmic examination on fluorescein angiography (FA). In the evaluation of leakage on FA, complete NV regression occurred in 58 (59.7%) eyes, partial resolution in 17 (17.7%) eyes, and no regression in 22 (22.6%) eyes at the end of follow-up. There was a statistically significant change in mean BCVA from 20/125 (0.8 ± 0.4 logMAR) at baseline to 20/60 (0.5 ± 0.4 logMAR) at the last follow-up visit (p < 0.0005). Final BCVA remained stable in 49 (50.5%) eyes, improved by 2 or more Snellen lines in 42 (43.3%) eyes, and decreased by 2 or more Snellen lines in 6 (6.2%) eyes. In particular, it was shown that 8 (42.1%) eyes out of 19 eyes with only intravitreal BVZ treatment without laser therapy or surgery achieved a complete regression of PDR during the 24 months of follow-up. Due to limitations from study design, this study could not answer the question of whether BVZ or PRP was a superior management approach for DR. However, the authors recommended that intravitreal BVZ alone provides a reasonable chance of control of retinal NV in a patient with prior PRP.
Most recently, at the 2021 American Academy of Ophthalmology Annual Congress (New Orleans, November 12–15), it was reported that investigative ophthalmic bevacizumab-vikg formulation (Outlook Therapeutics, ONS-5010) significantly improved visual acuity versus RBZ in patients with neovascular age-related macular degeneration (nv-AMD) over 12 months, according to the phase 3 NORSE findings and outcomes (NCT04516278) [18]. The phase 3 superiority pivotal trial assessed 12-month efficacy and safety of ONS-5010 1.25 mg monthly intravitreal injections (IVIs) versus RBZ IVIs in patients with nv-AMD [18]. Given the positive results, it is very likely that ONS-5010 will be evaluated in DR and DME in the near future to elucidate further the protean applications of intraocular BVZ. If approved, bevacizumab-vikg (Lytenaya) would be the first ophthalmic formulation of BVZ approved to treat retinal conditions. It is a full-length, humanized anti-VEGF recombinant monoclonal antibody that inhibits VEGF and associated angiogenic activity.
2.1.4. Safety of bevacizumab
Ocular and systemic side effects of BVZ have been addressed by a number of studies [19–21]. The most common ocular adverse events (AE) are subconjunctival hemorrhage, regurgitation of drug from the site of injection, transient rise of intraocular pressure, uveitis, lens injury, conjunctival chemosis, and iatrogenic VH [19]
Goldberg et al. reported an outbreak of infectious endophthalmitis in 12-patients after an intravitreal BVZ. The outbreak was attributed to contamination of the BVZ during syringe preparation by the compounding pharmacy [20]. However, a study on 1376 BVZ IVIs by Afarid et al. showed no ocular adverse effects after injections [22]. Sangroongruangsri et al. in the prospective observational study on 5975 patients who received intravitreal BVZ and 397 patients who received intravitreal RBZ, categorized serious systemic adverse events (SAE) into three subgroups due to low prevalence of the adverse systemic events. All-cause mortality, arterial thromboembolic events (ATE), and non-fatal heart failure (HF) were the three main subgroups, which included 66 (1.10%), 46 (0.77%), and 20 (0.33%) of patients, respectively. They reported that there were no significant differences between serious SAE of intravitreal BVZ and RBZ. Their data also showed that 6 treated eyes (0.08%) got endophthalmitis [23].
2.2. Ranibizumab
2.2.1. Structure and mechanism of action
RBZ, which is produced in prokaryotic Escherichia coli, is a 48 kDa recombinant humanized IgG1 kappa isotype monoclonal antibody fragment. It contains only Fab fragment compared to BVZ, which has both Fab and Fc fragments. The fragment consists of a light chain (VL-CL) covalently connected to one heavy chain (VH-CH1), which provides only one binding site for VEGF [10]. Because it lacks the Fc fragment, RBZ does not carry any glycosylation sites (Figure 2).
Figure 2.

The structure of a ranibizumab IgG1 antibody fragment composed of the affinity maturated Fab of bevacizumab.
Fab, fragment antigen-binding; Variable domains, VL; Constant domains, CL, CH1.
The mechanism of action for RBZ is binding with high affinity to the receptor-binding site of all active VEGF-A isoforms, including the biologically active, cleaved form of this molecule, VEGF110. This prevents the interaction of VEGF-A with its receptors (VEGFR1 and VEGFR2) on the surface of endothelial cells, reducing endothelial cell proliferation, vascular leakage, and angiogenesis [24]. Table 1 summarizes the structural features and pharmacodynamics of RBZ.
2.2.2. Ranibizumab for diabetic macular edema
RBZ has undergone significant evaluation in various clinical studies assessing its efficacy and safety. The Ranibizumab for Edema of the Macula in Diabetes – Study 2 (READ-2) was one of the earliest phase 2 clinical trials evaluating 0.5 mg RBZ against laser therapy alone and in combination with RBZ in 1:1:1. The outcomes of the study demonstrated superiority of RBZ therapy (BCVA improvement of 7.4 letters) over the laser therapy alone (BCVA improvement of 0.5 letters) at month 6 [25]. At the end of two years, however, the changes between the three groups were not statistically significant despite RBZ only group showing better improvement in BCVA [26]. Additionally, the combination of laser therapy with RBZ resulted in lower frequency of intravitreal injection at the end of two years. The three-year extension of the READ-2 study showed that aggressive treatment with RBZ therapy maintained the improvement in BCVA and CRT [27]. Following the READ-2 study, the READ-3 Study was conducted, which was a double masked, multicenter clinical trial that evaluated the role of higher dose of RBZ (2.0 mg vs 0.5 mg). The outcomes of the READ-3 clinical trial did not show any additional benefit of 2.0 mg dose of RBZ compared to 0.5 mg dose at the primary end point (ΔBCVA/ΔCRT of 7.01 letters/ 159.70 μm and 9.42 letters/168.58 μm for 2.0 mg and 0.5 mg, respectively). However, the improvement in BCVA at the end of month 24 in the READ-3 study was significantly more in the lower dose group (11.06 letters) compared to the high-dose group (6.78 letters) establishing 0.5 mg as more ideal dose for the management of DME [28]. The change in CRT between the two groups, however, was not significantly different highlighting the fact that the change in CRT does not always directly correlate with the visual improvement.
Safety and efficacy of RBZ in diabetic macular edema (RESOLVE) study was a randomized, sham-controlled, double-masked, multicenter phase 2 clinical trial that compared the two doses of RBZ (0.5 mg and 0.3 mg) against sham treatment [29]. The pooled analysis for the RBZ groups demonstrated significant improvement in BCVA (10.39 letters) compared to sham group (−1.4 letters). Protocol I by the DRCR.net was a phase 3 clinical trial that compared 0.5 mg dose of RBZ along with prompt or deferred laser (≥ 24 weeks) therapy, or triamcinolone plus prompt laser therapy to sham plus prompt laser therapy [30]. The study demonstrated superiority of RBZ plus prompt/deferred laser treatment at the end of 1 and 2-year compared to sham plus prompt laser treatment [30,31]. The study was extended to compare the RBZ plus prompt against RBZ plus deferred laser treatments at 3 and 5-year [32,33]. At both time-points the group with RBZ plus prompt laser treatment demonstrated lower improvement in BCVA compared to RBZ plus deferred treatment (difference in BCVA between RBZ plus deferred vs prompt was 2.9 and 2.6 at 3 and 5 years, respectively). However, the difference was only statistically significant at 3 years (p = 0.02). RESTORE and REVEAL studies were two phase 3 clinical trials that demonstrated the advantage of RBZ-only therapy over laser therapy (Δ BCVA in RESTORE (RBZ group = 6.1 vs laser group = 0.8) and in REVEAL (RBZ group = 5.9 vs laser group = 1.4) [34,35]. Both studies also showed that addition of laser therapy to the RBZ treatment did not provide any additional benefit.
Two landmark phase 3 multicenter clinical trials that led to approval of RBZ by the United States Food and Drug Administration (FDA) were RISE and RIDE. These studies compared two doses of monthly treatment with RBZ (0.3 mg and 0.5 mg) of RBZ to sham injections in patients with DME at the end of month 24. In both the studies, the percentage of patients achieving ≥15 letters improvement from the baseline was significantly higher for patients treated with RBZ (RISE (0.3 mg: 44.8% and 0.5 mg: 39.2%) and RIDE (0.3 mg: 33.6% and 0.5 mg: 45.7%)) compared to sham group (RISE: 18.1% and RIDE: 12.3%) [36]. The studies were extended to month 36 and the patients in the sham groups were allowed to cross over to receive 0.5 mg RBZ. The month 36 outcomes demonstrated sustenance in the efficacy of monthly treatment with RBZ. However, the group that received delayed treatment with RBZ (Crossover from sham group) gained fewer letters compared to the patients receiving RBZ treatment from the start of the study (Sham:2.8 vs. 0.3 mg: 10.6 and 0.5 mg: 11.1 letters) [37]. These results suggested that chronic macular edema may result in permanent vision loss which may not improve with delayed treatment with RBZ.
In various clinical trials, monthly dosing of RBZ has been utilized. However, despite being the optimal regimen, monthly dosing may not be very practical in real-world scenarios. Therefore, some trials have been conducted looking at the pro re nata (PRN) or treat-and-extend (T&E) dosing regimen of RBZ and this has also been studied in extension phases of trials with initial monthly treatment. In the Protocol I, the number of injections received by the patients progressively declined over the 5-year period despite maintenance of BCVA benefits in the RBZ + prompt laser and RBZ + deferred laser arms (8–9 injections in year 1, 2–3 injections in year 2, 1–2 injections in year 3, 0–1 injection in year 4, and no injections in year 5 in RBZ + prompt laser and RBZ + deferred laser arms, respectively). Thus, around 52% to 62% of the patients in the two arms received no injection during year 5. Similarly, year-3 extension of the RESTORE study also demonstrated efficacy of RBZ in maintaining BCVA while lowering the number of injections needed using PRN regimen [38]. The RELIGHT study was a phase 3b, open-label, single-arm study evaluating individualized re-treatment with 0.5 mg RBZ after initial three monthly doses, along with bimonthly follow-up from month 6 to month 18. The results of the study showed that BCVA gain achieved during initial 6 months with monthly follow-ups was maintained at the end of month 18 after a year of bimonthly follow-up [39]. The randomized trial of treat-and-extend (T&E) RBZ with and without navigated laser for DME: TREX-DME study was a multicenter, prospective, RCT with three groups comparing monthly dosing of 0.3 mg RBZ with T&E algorithm using 0.3 mg RBZ with and without angiography guided macular laser photocoagulation [40]. The primary endpoint results of the study showed that there was no significant difference in BCVA gains between the monthly (8.6 letters), T&E (9.6 letters), and T&E with angiography guided macular laser photocoagulation (9.5 letters) groups. In addition, both T&E groups needed significantly lower injections (10.7 and 10.1) compared to monthly treatment arm (13.1, p < 0.001). The addition of laser photocoagulation, however, did not infer any additional benefit to the T&E regimen. Table 2 summarizes the clinical trials using intravitreal RBZ therapy in the management of DME.
2.2.3. Ranibizumab for diabetic retinopathy
RBZ has been shown to not only slow the progression of DR but also improve the severity the DR in various studies. Monthly dosing of RBZ has recently been approved for the management of all forms of DR. In the post-hoc analysis of the RISE and RIDE clinical trial, 37.2% and 35.9% of the patients receiving 0.3 mg and 0.5 mg RBZ demonstrated ≥ 2-step improvement in the DR severity score compared to 5.4% in the sham group at the end of month 24 which persisted at the end of extension phase at 36 months [41,42]. The post-hoc analysis of the READ-3 study demonstrated that monthly dosing of RBZ has shown improvement DR severity similar to other clinical trials as early as month 6 and these changes persisted at the end of the month 24 [43,44]. The improvement in DR status by RBZ was also shown in the DRCR.net Protocol I [45]. Among patients with NPDR the improvement in DR severity was shown by 29–32% of the people along the time of study.
For the management of PDR, DRCR.net protocol I demonstrated efficacy of RBZ in improving the severity in eyes of these patients [45]. The improvement in DR severity among patients with PDR was 23–38% during the course of the study. Protocol S was a noninferiority trial by the DRCR.net which was structured to compare the standard PRP treatment for PDR against 0.5 mg [46,47]. The results of the study demonstrated non-inferiority of RBZ compared to PRP in terms of BCVA improvement in eyes with PDR both at 2- and 5-year end points. The mean BCVA improvement was +2.8 and +0.2 letters at 2 year in the RBZ and PRP group, respectively [46,47]. Surprisingly, the 5-year results showed that the difference in improvement in BCVA between the RBZ and PRP groups had resolved (+3.1 letters for the RBZ group and +3.0 letters for the PRP group, respectively) [47]. It is important to note that approximately 53% of the patients in the PRP group of the study received RBZ for DME during the course of the study that may have some contribution toward the VA changes in this group. Table 3 summarizes the Protocol S study.
Table 3.
List of clinical trials summarizing intravitreal anti-VEGF treatment in diabetic retinopathy.
| Study | No. of Patients (Study Eyes) | Duration, mo | Inclusion Criteria | Intervention | Regression of DRP (%) | BCVA Change, letters, (Mean) | CMT Change, (Mean) | No of Injections |
|---|---|---|---|---|---|---|---|---|
| Protocol S [Citation46] | 394 | 60 | PDR BCVA≥20/320 |
1. RBZ (0.5 mg, q4 PRN) 2. PRP |
46 N/A |
3.1 ± 14.3 3.0 ± 10.5 |
−139 ± 157 –59 ± 102 |
19.2 ± 10.9 5.4 ± 7.9 |
| CLARITY [Citation63] | 232 | 12 | PDR BCVA≥20/320 |
1. AFB (2 mg, q4 PRN, after 3 initial q4 doses) 2. PRP |
64 34 |
1.3 ± 0.6 –2.9 ± 0.7 |
−8.9 ± 2.3 24.0 ± 5.5 |
4.4 ± 1.7- |
| PANORAMA [Citation64] | 402 | 24 | DRSS score 47 and 53 BCVA ≥20/40 |
1. AFB (2 mg, q16, after 3 initial q4 doses and one q8 interval) 2. AFB (2 mg, q8, after 5 initial q4 doses) 3. Sham |
65.2 79.9 15.0 |
1.7 ± 3.5 1.3 ± 3.49 0.5 ± 3.01 |
N/A N/A N/A |
N/A N/A N/A |
AFB, aflibercept; BCVA, best corrected visual acuity; CMT, central macular thickness; DRP, diabetic retinopathy; DRSS, diabetic retinopathy severity scale; mo, month; PDR, Proliferative diabetic retinopathy; PRP, panretinal photocoagulation; q, every; RBZ, ranibizumab.
2.2.4. Safety of ranibizumab
2.2.4.1. Ocular adverse events.
Most recently, the REFINE study evaluated the safety of the RBZ versus laser photocoagulation in 384 DME patients [48]. The most common observed ocular AEs was increased IOP (RBZ 5.2%), followed by vitreous hemorrhage (RBZ 1.6%; laser 5.3%), conjunctival hemorrhage (RBZ 3.6%; laser 1.3%), and dry eye (RBZ 3.6%; laser 1.3%). Ocular serious AEs occurred in 0.3% of patients in RBZ arm due to cataract. The authors reported no cases with endophthalmitis [48]. These findings were consistent with what have been reported from the READ-3 study, which compared the safety outcomes between the 0.5 mg and 2.0 mg RBZ in 152 eyes. They found no ocular SAEs in either of the study groups [28].
In a 24-month follow-up of the RISE and RIDE studies, the most common SAE was vitreous hemorrhage, which occurred in 4 sham-treated and 2 RBZ-treated eyes in RISE and in 3 sham-treated eyes in RIDE. Serious intraocular inflammation (IOI) was uncommon among RBZ-treated patients, occurring only once. SAEs arising from the injection procedure were also uncommon; 1 case of endophthalmitis occurred in RISE and 3 in RIDE, along with three cases of traumatic cataract and one rhegmatogenous retinal detachment out of 10,584 IVIs [36]. The ocular AEs after a 36-month follow-up of the RISE and RIDE study were generally consistent with the results seen at month 24 [37].
2.2.4.2. Systemic adverse events.
Since the SAEs of RBZ are rare, studies with an extremely large study population are required to address the small difference between the systemic complications. The present evidence are not powered sufficiently to project these AEs due to relatively small sample sizes. However, in 2014 Yanagida et al. launched a meta-analysis of the six trials, including 2456 patients with DME, to evaluate the systemic safety of the RBZ. Risk ratio for cerebrovascular accident, myocardial infarction, vascular death, and overall mortality were 0.80 (95% confidence interval, 0.37 1.73; P = 0.57), 0.91 (95% confidence interval, 0.46–1.80; P = 0.78), 1.29 (95% confidence interval, 0.58–2.86; P = 0.53), and 1.92 (95% confidence interval, 0.78–4.73; P = 0.16), respectively. The meta-analysis showed that RBZ is systemically safe in patients with eligible systemic vascular conditions (history of cerebrovascular accident, myocardial infarction, uncontrolled blood pressure, uncontrolled diabetes mellitus, or renal failure were considered as exclusion criteria in the six included trials) [49]. In the REFINE study, hypertension (6.2%), nasopharyngitis (6.5%), upper respiratory tract infection (8.5%), and cough (5.9%) considered as the SAEs. The overall incidence of SAEs was 16.3% [48].
2.3. Aflibercept
2.3.1. Structure and mechanism of action
AFB is a 115 kDa human recombinant decoy receptor, consisting of portions of human VEGF receptor 1 and 2 extracellular domains fused to the Fc portion of human IgG1, produced in Chinese hamster ovary (CHO) K1 cells. It has five putative N-glycosylation sites on each polypeptide chain. The affinity of AFB for VEGF is higher than their natural receptors and is greater than that of RBZ and BVZ [50,51] (Figure 3).
Figure 3.

The structure of aflibercept composed of domains from VEGFR1 and 2 fused to an IgG1 Fc-region.
VEGFR1, vascular endothelial growth factor receptor 1; VEGFR2, vascular endothelial growth factor receptor 2; IgG1, immunoglobulin class G; Fc, fragment crystallization; Constant domains, CH2, CH3.
A major functional distinction between AFB and other anti-VEGF agents is that it blocks VEGF-B, PlGF1, and PlGF-2 in addition to VEGF-A isoforms. AFB antagonizes a broader spectrum of growth factors, and the potency of AFB for blocking VEGF-mediated signaling by VEGF121 and VEGF165 is greater than that for RBZ and BVZ by several orders of magnitude; in particular, the affinity of AFB for VEGF-A165 is 94 times greater than RBZ and approximately 120 times greater than BVZ [50]. VEGF normally binds its receptors as homo- or heterodimers [9]. AFB has a unique binding action and binds to both sides of the VEGF dimer, ‘like two hands on a football,’ forming an inert 1:1 complex, also termed a VEGF trap which minimizes the potential for VEGF to interact with more than one AFB molecule. Compared to anti-VEGF antibodies, such as BVZ and RBZ, AFB binds in a manner that allows the VEGF dimer to interact further with other molecules [51,52]. Table 1 summarizes the structural features and pharmacodynamics of AFB.
2.3.2. Aflibercept for diabetic macular edema
Do and colleagues published the first study worldwide of intravitreal AFB for DME [53]. Subsequently, the DA VINCI study is a randomized, double-masked, multicenter, phase 2 clinical trial that compared the efficacy and safety of intravitreal AFB at 4 different dosing regimens (0.5 mg every 4 weeks (0.5q4); 2 mg every 4 weeks (2q4); 2 mg for every 8 weeks (2q8) after 3 initial monthly doses; 2 mg on an as-needed basis after 3 initial monthly doses) with macular laser treatment [n = 44] in patients with DME [54,55]. The results showed significant improvement in BCVA and CRT (ΔBCVA/ΔCRT of 11.0 letters/ 165.4 μm, 13.1 letters/ 227.4 μm, 9.7 letters/ 187.8 μm, and 12.0 letters/ 180.3 μm for 0.5q4, 2q4, 2q8 and 2 PRN, respectively) from baseline to week 24 (primary endpoint) and week 52 for each AFB regimen compared to macular laser photocoagulation (ΔBCVA/ΔCRT of −1.3 letters/ 58.4 μm). Although all AFB dosing regimens showed significant treatment effects compared with laser treatment, 2q4 had the highest percentages of eyes with VA improvements. However, the differences between the 2q4 and 2q8 groups were attributed to the baseline differences between the two AFB groups instead of the dosing regimens.
Subsequently, two similarly designed, randomized, double-masked, multicenter phase 3 trials, VISTA and VIVID, compared the efficacy of intravitreal AFB injections of 2q4 and 2q8 after 5 initial monthly doses with macular laser treatment in patients with DME [56,57]. Both VISTA and VIVID groups demonstrated significant improvements in mean BCVA gain, proportion of eyes that gained ≥ 15 letters, and diabetic retinopathy severity scale (DRSS) score (visual and anatomical outcomes) with similar overall efficacy in two intravitreal regimens (2q4 and 2q8 groups) compared to laser group at week 52 and 100. Mean CRT changes also were significantly greater in eyes treated in both AFB arms compared with laser group at week 52 and 100. These favorable results maintained at week 148. Although VISTA (ΔBCVA/ΔCRT of 10.4 letters/ 200.4 μm, and 10.5 letters/ 190.1 μm for 2q4 and 2q8, respectively) and VIVID (ΔBCVA/ΔCRT of 10.3 letters/ 215.2 μm, and 11.7 letters/ 202.8 μm for 2q4 and 2q8, respectively) studies showed similar efficacy between 2q4 and 2q8 regimens in terms of visual and anatomic outcomes through week 148, mean injections from baseline to week 148 for 2q8 arms were being noted fewer (18.1 for both VISTA and VIVID) compared to 2q4 arm (29.6 and 32.0 in VISTA and VIVID, respectively). Favorable results obtained from clinical trials have led to the approval of intravitreal AFB for the treatment of DME in July 2014 by the FDA [56,57].
Intravitreal anti-VEGF therapies has been shown to be effective in the treatment of center involved-DME (ci-DME) in patients with decreased visual acuity (VA) (20/32 or worse) in many clinical trials [11,13,27–29,35,37,55,57]. Recently Protocol V study, a randomized and multicenter clinical trial, compared visual loss in patients with good VA (20/25 or better) and ci-DME among eyes managed with monthly intravitreal AFB, laser photocoagulation and observation. No statistically significant difference was observed in mean VA changes and CRT among three groups at 2 years [58]. However, the study was not designed as a comparison of monotherapy of these approaches, and patients in the laser and observation groups received intravitreal AFB during follow-up if they meet the worsening criteria (VA worsened by 1 line at 2 consecutive visits or by 2 lines at 1 visit). Nevertheless, the majority of eyes in the observation group (two-thirds) and laser group (three-fourths) did not meet the worsening criteria for VA and did not receive AFB during 2 years of follow up. Therefore, observation without treatment only if VA worsened was considered as a reasonable approach in the management of ci-DME in eyes with good VA [58]. Table 2 summarizes the clinical trials using intravitreal AFB therapy in the management of DME.
The DRCR.net has also conducted a multicenter randomized, double-masked, multicenter, phase 3 study clinical trial, Protocol T, to compare the efficacy and safety of AFB (2.0 mg), BVZ (1.25 mg) and RBZ (0.3 mg) for ci-DME with vision impairment [59]. All three intravitreal groups showed improved BCVA and decreased the number of injections at 2 years. The subset of patients with better VA (20/32–20/40) had similar visual outcomes among three intravitreal groups (BCVA improvement of 7.8, 6.8, and 8.6 letters for AFB, BVZ, and RBZ, respectively). On the other hand, AFB demonstrated better visual outcomes compared to BVZ and RBZ in the subset of patients with worse VA (20/50–20/320) at year one. The significant difference between AFB (BCVA improvement of 18.1 letters) and BVZ (BCVA improvement of 13.3 letters) continued to be the same at year 2. However, AFB showed no superiority over RBZ in terms of VA (BCVA improvement of 16.1 letters for RBZ) at year 2 [59]. Recently, 5-year follow-up outcomes of Protocol T was published [60]. About two-thirds of the Protocol T study participants were eligible for the continuation of the study. However, outcomes among three anti-VEGF regimens were not comparable as half of the eyes were treated with a different anti-VEGF agent after the initial study ended at 2 years. Overall, 5-year outcomes showed a better mean BCVA than baseline but worse than 2 years (BCVA improvement of 12.1 and 7.4 letters for year 2 and 5, respectively), although there was no significant difference in the mean CRT between 2 and 5 years [60]. Table 2 summarizes the protocol T.
2.3.3. Aflibercept for diabetic retinopathy
AFB not only improves the DR severity but also slows down the progression of DR. In the DA VINCI study, eyes treated with each AFB regimen at week 52 demonstrated more significant improvement in the DR severity score (40%, 31%, 64%, and 32% improvement for 0.5q4, 2q4, 2q8, and 2PRN regimens, respectively) compared to the laser group (12%). Moreover, worsening of DRSS score was rare compared to the laser group (0%, 13%, 0%, and 14% in the 0.5q4, 2q4, 2q8, and 2PRN groups and 24% in the laser group) [55].
The improvement of DR by AFB was also shown in VISTA and VIVID trials. Intravitreal AFB injection has demonstrated a two-step or greater regression in DRSS score at week 100 both in VISTA (37%, 2q4 and 37.1%, 2q8) and VIVID (29.3%, 2q4 and 32.6%, 2q8) compared to laser treatment [56]. The number of patients treated with PRP through week 100 was smaller in two intravitreal AFB groups compared to laser group in both VIVID and VISTA [61]. Based on positive data in VIVID and VISTA, the use of intravitreal AFB in the treatment of DR with DME was approved by the FDA.
Similarly, AFB has been found to be associated with DR regression (31.2% at year 1 and 24.8% at year 2) in patients with NPDR in Protocol T study [59]. Additionally, a greater proportion of eyes with PDR had experienced improvement in DR after treated with AFB (75.9%) compared to RBZ (55.2%) or BVZ (31.4%) at each of the two annual visits [62].
CLARITY is a randomized, single-blinded, multicenter, controlled, phase 2b, non-inferiority trial comparing intravitreal AFB (2 mg on an as-needed basis after 3 initial monthly doses) to PRP in patients with PDR. The study has shown that AFB was non-inferior to PRP, showing a difference of 30% regression in DR ([95% CI: 16–42], p < 0.0001) and 3.9 letters in mean BCVA ([95% CI: 2.3–5.6], p < 0.0001) favoring AFB at week 52. Moreover, vitreous hemorrhage was found to be twice as high in the PRP group compared to AFB treatment group (p = 0.034) [63]. These successful outcomes of AFB may be attributed to its increased affinity for VEGF binding and blockade of other angiogenic pathways such as placental growth factor and galectin-1 [63]. Table 3 summarizes the CLARITY study.
Most recently, PANORAMA (NCT02718326), a randomized multicenter phase 3 clinical trial, evaluated the improvement of diabetic retinopathy in patients with moderately severe and severe NPDR in the absence of ci-DME among two intravitreal AFB 2q8 and 2q16 regimens and sham group. The result of the study showed a 65% and 80% ≥ 2-step improvement in DRSS scores for 2q16 and 2q8 groups, respectively, versus 15% in the sham group at two years (P < 0.0001 for both). The risk of developing vision-threatening complications such as PDR or anterior segment neovascularization significantly reduced by 85% and 88% in both 2q16 and 2q8 groups, respectively, compared to sham. In addition to the findings mentioned above, the proportion of ci-DME was lower in the 2q16 (7%) and 2q8 (8%) groups compared to the sham group (26%), (P < 0.0001 for both) [64]. Table 3 summarizes the PANORAMA study.
2.3.4. Safety of aflibercept
2.3.4.1. Ocular adverse events.
VISTA and VIVID are the two pivotal clinical trials that evaluated the safety and efficacy of the intravitreal AFB injection (IAI) in DME during a 2-year follow-up [56]. In 2016, Ziemssen et al. carried out a post hoc pooled analysis of the VISTA and VIVID trials [65]. There was no significant difference between the ocular and non-ocular AEs across treatment groups. The most common ocular AEs were conjunctival hemorrhage (31.1%), cataract (11.6%), and eye pain (10.7%). No endophthalmitis was reported in the treated eyes. Also, the ocular AEs after 148-week follow-up were consisted with the previous findings [57]. Well et al. performed a multi-center clinical trial on 660 adults with DME to compare the safety and efficacy of AFB, BVZ, and RBZ during a 1-year follow-up. Contrary to VIVID and VISTA studies, they reported 1 (1%) eye with endophthalmitis and 3 (2%) eyes with inflammation after intravitreal AFB injection. Other ocular AEs were increased IOP (12%), vitreous hemorrhage (4%), and cataract (1%) [66].
In 2016, Andrade et al. reported a short-term, prospective clinical trial assessing the efficacy and safety of intravitreal injection of ziv-AFB in DME therapy [67]. Seven patients with DME received intravitreal ziv-AFB every 4 weeks for 6 months. They concluded that the intravitreal injection of the ziv-AFB is safe with no systemic or ocular complication. These findings were supported by the very first pilot study conducted by Do et al. which reported no ocular toxicity after the intravitreal AFB injection [53]. Despite the satisfactory safety outcomes of the two previous studies, it should be considered that these studies were primarily investigation with small sample size, short-term follow-up, and had low internal validity due to the lack of the control group.
2.3.4.2. Systemic adverse events.
According to the VIVID and VISTA study, the most common SAEs were hypertension, nasopharyngitis, and urinary tract infection [56]. Overall rates of arterial thromboembolic events, including vascular deaths, which were defined by Anti-Platelet Trialists’ Collaboration criteria, were low and comparable across the treatment groups. The incidence of death in the intravitreal AFB injection after 4 and 8 weeks, and in the control groups was 5.2, 2.6, and 1.9%, respectively, in VISTA, and 2.9, 4.4, and 0.8% in VIVID [56].
In 2011, the DA VINCI study compared the efficacy and safety of AFB at different dosing with traditional laser photocoagulation in eyes with DME [54]. The most common SAEs in the intravitreal AFB groups were hypertension (9.7%), myocardial infarction (1.1%), cerebrovascular accident (1.1%) and death (1.7%). However, these results should be interpreted with caution, since the SAEs maybe attributed to the underlying disease in these patients rather than the drug itself.
3. Selected VEGF antagonists with strong promise for diabetic retinopathy
3.1. Brolucizumab
Brolucizumab (Novartis Pharmaceuticals, Basel, Switzerland) is a humanized single-chain variable fragment that binds to and inhibits activity of VEGF-A on the VEGF receptor 1 and 2 [68]. Since it is only a single-chain variable fragment, the molecular mass of brolucizumab is only 26kDa, which makes it the smallest of the anti-VEGF agents relative to BVZ (148kDa), RBZ (48kDa) and AFB (97–115 kDa). Table 1 summarizes the structural features and pharmacodynamics of brolucizumab. Because of its small size and therefore high solubility, brolucizumab can be concentrated easily, resulting in delivery of 6 mg of drug delivered via a single 0.05 ml intravitreal injection. The effective dose delivered is about 22 and 11 times more than RBZ and AFB, respectively [68,69]. Such property can potentially translate to longer duration of therapeutic effects, thereby increasing that time period needed between injections. HAWK and HARRIER were two double masked, randomized, clinical trials that indeed demonstrated non-inferiority of brolucizumab administered q8 or even q12 weekly after a loading doze compared to AFB and resulted in its FDA approval for the treatment of neovascular-AMD in 2019 [70,71].
Based on the success of brolucizumab in patients with nv-AMD, KITE (NCT03481660) and KESTERL (NCT03481634) were two phase-3, double masked, multicenter, randomized controlled trials that were launched to compared the efficacy and safety of 6 mg brolucizumab (KITE and KESTERL) and 3 mg brolucizumab (KESTREL) compared to AFB in treatment naïve patients with DME [72]. The 52-week outcomes of the KITE and KESTERL trials were recently reported at the 2021 virtual Stanford Retina Innovation Summit (SRIS, 17 July 2021), among other congresses. Brolucizumab 6 mg was non-inferior to AFB in mean change in BCVA at week 52, with fewer injections. Significant improvements in central subfield thickness (CST) from baseline were achieved with brolucizumab 6 mg. There was a higher proportion of patients with fluid resolution (IRF/SRF) on brolucizumab 6 mg at week 52. More than half of brolucizumab 6 mg patients were maintained on q12w treatment interval up to week 52 immediately after the loading phase [72].
Despite the promised efficacy and increased duration between injections associated with brolucizumab treatment due to higher concentration of drug delivered, recent safety data from the HAWK and HARRIER studies as well as other case reports and case series have shown an increased risk of IOI and occlusive retinal vasculitis (RV) in these patients [73–76]. As reported at the SRIS on 17 July 2021, in KESTREL, one subject in the brolucizumab 6-mg arm had both RV and retinal vascular occlusion (RO); both events resolved without treatment and BCVA at week 52 had increased by 14 letters compared with baseline [72]. In KITE, both RO events were reported as retinal artery occlusion and were not associated with IOI or RV. Thus, despite its strong efficacy, given the potential risks and benefits, and the availability of other therapeutic options, more information is being collected and analyzed to enable clinician-scientists to determine the role of brolucizumab in the management of nv-AMD and DR [72].
3.2. Faricimab
Faricimab (Roche/Genentech, Basel, Switzerland) is a humanized antibody with a bispecific antibody design that targets both VEGF-A and angiopoietin 2 (Ang-2). Angiopoietins regulate vasculogenesis by binding to tyrosine kinase with immunoglobulin and epidermal growth factor homology domain (Tie-2) receptor. Upregulation of Ang-2 has been suggested to play a role in the pathogenesis of macular edema and other retinal vascular diseases [77,78]. By targeting both VEGF-A and Ang-2 pathways, faricimab is designed to improve efficacy and durability in patients with DME. Table 1 summarizes the structural features and pharmacodynamics of faricimab.
A phase 2 randomized, active comparator-controlled, double-masked clinical trial, BOULEVARD, compared the efficacy and safety of faricimab 1.5 mg and 6 mg with RBZ 0.3 mg. Statistically significant improvement in BCVA was achieved in the faricimab 6 mg group (mean gain of +13.9 letters) compared to RBZ 0.3 group (mean gain of +10.3 letters) in DME-naïve patients at week 24. Faricimab also demonstrated dose-dependent improvement in CST and DRSS score with a longer time to retreatment compared with RBZ [79].
Subsequently, YOSEMITE and RHINE, two phase 3 clinical trials, compared faricimab 6 mg every 8 weeks and faricimab 6 mg at a personalized treatment interval (every 4 weeks up to every 16 weeks based on disease activity) with AFB 2 mg every 8 weeks in patients with DME. Both faricimab 6 mg every 8 weeks (ΔBCVA of 10.7 letters and 11.8 letters in YOSEMITE and RHINE, respectively) and faricimab 6 mg at the personalized treatment interval (ΔBCVA of 11.6 letters and 10.8 in YOSEMITE and RHINE, respectively) were not inferior to AFB 2 mg every 8 weeks (ΔBCVA of 10.9 and 10.3 letters in YOSEMITE and RHINE, respectively) at 1 year. Data from the personalized treatment interval group showed improved VA and anatomical outcomes with extended dosing intervals, with more than 50% of patients receiving faricimab every 16 weeks at 52 weeks and more than 70% receiving faricimab every 12 weeks or longer. With regard to safety outcomes, intraocular inflammation was found to be higher in the faricimab 6 mg every 8 weeks group (YOSEMITE n = 5 [1 · 6%], RHINE n = 3 [0 · 9%]) and faricimab 6 mg at the personalized treatment interval group (YOSEMITE n = 7 [2 · 2%], RHINE n = 2 [0 · 6%]) as compared to the AFB 2 mg every 8 weeks group (YOSEMITE n = 3 [1 · 0%], RHINE n = 1 [0 · 3%]). Data from these clinical trials have led to FDA approval of faricimab for the management of DME [78].
3.3. KSI-301
KSI-301 (Kodiak Sciences, Palo Alto, CA) is a novel anti-VEGF antibody biopolymer conjugate (ABC platform), with a 950 kDa molecular weight, that is designed to maintain increased bioavailability and durability in ocular tissues for the treatment of nv-AMD and retinal vascular diseases. Table 1 summarizes the structural features and pharmacodynamics of KSI-301. A phase 1a ascending dose escalation study (1.25 mg, 2.5 mg, or 5 mg) in 9 patients with DME demonstrated a better tolerability without any adverse event in all doses at 12 weeks [80]. The ongoing NCT03790852 randomized, open label, phase 1b study is comparing 2.5 mg and 5 mg KSI 301 in patients with nv-AMD, DME, and retinal vein occlusion. The results at week 20 showed +7.4 letters mean BCVA improvement and −129 μm mean CRT change in the DME group, with 72% of the patients have been extended longer than 3 months after the loading dose without retreatment [81]. A phase 2/3 clinical trial (NCT04049266), DAZZLE, has been conducted to investigate the efficacy and safety of 5 mg KSI 301 at 12–20 weeks intervals and compare with 2 mg AFB every 8 weeks after the loading dose in patients with nv-AMD [82]. Most recently, in February 2022, Kodiak announced that the DAZZLE Study did not meet its primary endpoint of non-inferiority in BCVA, even though the majority of KSI-301-treated patients achieved durable visual gains. The failure to meet the primary endpoint is thought to be in large part due to the impact of undertreatment in some patients. Additional analyses of the DAZZLE Study are being conducted.
4. Conclusion
Although the complex pathophysiology of DR is not entirely understood, VEGF has an important role in disease progression and subsequent development of vision-threatening complications. Phase 2 and 3 clinical trials investigating the efficacy of intravitreal administration of anti-VEGFs demonstrated significant improvement in mean BCVA and reduction in mean CRT compared to traditional laser treatment, especially in patients with worse BCVA with DME. In DME, the combination of laser and anti-VEGF treatments did not show superiority compared to anti-VEGF therapy alone. Moreover, anti-VEGF therapy resulted in a remarkable reduction in DRSS score and NV in patients with NPDR and PDR. These favorable outcomes and the regression of vascular-related pathologies in DR from intravitreal anti-VEGF treatment support the crucial role of VEGF mediators in the pathogenesis of DR.
The safety of BVZ, RBZ and AFB has been evaluated in large-scale clinical trials. The most common ocular AEs include subconjunctival hemorrhage, eye pain, elevated intraocular pressure, and floaters. Intravitreal injection-related ocular SAE such as cataract, endophthalmitis, and rhegmatogenous retinal detachment were uncommon.
Systemic AE such as arterial thromboembolism, myocardial infarction, stroke, and hypertension due to intravitreal administration of anti-VEGF have been found to be lower in clinical trials, though these studies were not powered to assess the safety risks of SAE and have not enrolled patients at increased risk for serious SAEs.
5. Expert opinion
Intravitreal anti-VEGF treatments are the first-line treatment options for diabetic macular edema (DME). Key clinical trials BOLT investigating bevacizumab (BVZ), RIDE and RISE employing ranibizumab (RBZ), and VIVID and VISTA evaluating aflibercept (AFB) have shown significant superiority in visual acuity (VA) and anatomical improvement compared to laser treatment in patients with DME [13,36,56,57]. Protocol T is the first clinical trial comparing the efficacy and safety of currently available anti-VEGFs (BVZ, RBZ, and AFB) in the management of DME. In Protocol T, all anti-VEGF agents were effective equally for eyes with mild VA loss (20/32–20/40). However, AFB was found to be more efficacious for eyes with moderate or worse VA loss (20/50–20/320) [59]. Unlike studies investigating the efficacy of anti-VEGFs in DME with mild to severe VA loss, Protocol V study indicated that observation without treatment only if VA worsens maybe a reasonable and cost-effective strategy for eyes with good VA (>20/25) and center involved-DME [58].
Despite successful outcomes with anti-VEGF treatment in the management of DME, some eyes show incomplete anatomic and/or visual responses to treatment. In regard to inadequate response to anti-VEGF treatment, post hoc analyses of clinical trials have shown that continued treatment with RBZ injections based on the clinical trial algorithm provides a reduction in the CST or CFT and improved VA in eyes with persistent DME through 24 months [83,84]. Similarly, post hoc analysis of the Protocol T study indicated that continued treatment in accordance with the DRCR.net retreatment algorithm, particularly with AFB and RBZ, provided resolution of DME despite initial limited response following 3 or more anti-VEGF injections. The study also demonstrated considerable improvement in VA for all anti-VEGF groups with a low risk of vision loss in eyes with persistent DME through 24 weeks [85]. To address the issue of persistent DME, a phase 2 multi-center randomized clinical trial was conducted to compare combination therapy of RBZ and dexamethasone implant with RBZ treatment alone in patients who had persistent DME following at least 3 anti-VEGF injections. Although the reduction in CST was more likely in eyes treated with dexamethasone administration in addition to continued RBZ IV injections (mean, −110 ± 86 μm) compared to RBZ injections alone (mean, −62 ± 97 μm), the mean change in VA remained similar in both groups at the 24-week follow-up. Additionally, 29% of eyes developed increased intraocular pressures in the combination group, whereas no intraocular pressure elevation occurred in the RBZ group [86].
Current clinical trials have not addressed the issue of switching to other anti-VEGF treatments in eyes who have suboptimal or worsening responses to treatment with one particular anti-VEGF alone. Tachyphylaxis or tolerance has been described as the phenomenon that could be attributed to the lack of response to over repeated one anti-VEGF agent, particularly in NV-AMD. To overcome this resistance, switching agents to another anti-VEGF agent has been suggested to provide improvement in the treatment response [87,88]. Similarly, in DME, some eyes can be unresponsive or show limited response to one particular anti-VEGF but show considerable response when treated with a different anti-VEGF agent. There are studies investigating switching from BVZ and/or RBZ to AFB, which demonstrated improved functional and anatomical outcomes in DME [88–92]. On the other hand, a retrospective study that compared the efficacy of sustained single anti-VEGF and double anti-VEGF switching intravitreal injections in persistent DME following 3 initial consecutive monthly anti-VEGF injections showed no significant visual outcomes between the two treatment strategies. The retrospective nature of the study and the small number of patients poses a limitation for the study. Further prospective randomized studies are required to investigate the effect of changing anti-VEGF agents in the management of persistent DME [93].
Clinical trials for anti-VEGF treatments in DME have shown further benefits in improving the severity of diabetic retinopathy (DR) [41–43,54,55,57,59,61,62]. Most recently, the PANORAMA study (NCT02718326) demonstrated a significant improvement in the DRSS score in patients with moderately severe to severe non-proliferative DR without DME [64]. For proliferative DR (PDR), anti-VEGFs alone or as an adjunctive with pan-retinal photocoagulation (PRP) demonstrated non-inferiority to PRP alone and provides a reasonable reduction in retinal NV, PDR-related complications as well as DME [15,16,46,47,63,94]. However, cost-effectiveness and patients’ compliance for repeated treatments with anti-VEGF are the most important factors to consider compared to PRP in patients with PDR. Therefore, the use of intravitreal anti-VEGF administration may be a better treatment option for PDR patients with concurrent DME.
Among currently available three anti-VEGFs (BVZ, RBZ, and AFB), BVZ is originally developed for the treatment of meta-static colorectal cancers [95,96]. Although BVZ has not been approved for the treatment of retinal vascular diseases by the FDA, it is used off-label by physicians throughout the world due to its economic efficiency in comparison to RBZ and AFB.
Pharmacokinetics of intravitreally injected anti-VEGF agents are important considerations in determining the optimal intraocular concentration and dosing frequency to achieve the maximal therapeutic effect of anti-VEGF. Vitrectomy has been shown to increase clearance and reduce the half-life of anti-VEGF agents in both animal models and human eye studies [97–104]. The enhanced clearance of anti-VEGF after vitrectomy may have an important effect on the dosing regimen as vitrectomized eyes may need more frequent injections and closer follow-ups. However, there is some controversy as to whether the level of anti-VEGF agent may reach an adequate level and duration in vitrectomized eyes as the vitreous concentration of VEGF also reduces following vitrectomy [97,105]. In MODEVA, a prospective, non-comparative, and multicenter clinical trial, a significant VA gain and CST reduction were achieved with 5 monthly injections followed by a PRN regimen in vitrectomized eyes with DME at one year. Mean interval injection of AFB was 5.8 weeks [106]. The outcomes of clinical trials including VITCLEAR (NCT02174211) assessing a pharmacokinetic study of RBZ, AFB, and the effect of vitrectomy in age-related macular degeneration [107] will likely provide a better understanding in the management of DME in vitrectomized eyes.
Favorable results from large-scale clinical trials for anti-VEGF treatments in retinal vascular diseases have led to the development of new anti-VEGF molecules. The goals for future anti VEGF molecules are to bind and inhibit VEGF receptors with a higher affinity and provide longer duration of therapeutic effect and to attack other targets that play a role in the management DR and other retinal vascular diseases. These features are expected to lessen the overall number of IVIs to reduce the treatment burden for DR patients. Brolucizumab, Faricimab, and KSI 301 are three promising anti-VEGF agents currently under investigation for the treatment of retinal vascular diseases.
Brolucizumab is a humanized single-chain variable fragment inhibitor of VEGF-A with a small molecular size of 26 kDa. Its small size enables better target-tissue penetration and allows use of a higher concentration of the agent, leading to a rapid onset of action and a longer duration of therapeutic effects [68,69]. In 2019, the United Sates Food and Drug Administration (FDA) approved brolucizumab for the management of nv-AMD based on the successful results from HAWK and HARRIER phase 3 clinical trials [70,71]. However, the same studies have reported higher IOI rates for brolucizumab compared to AFB (1.4–2.2% vs 0–0.3%, respectively). In addition to mild–moderate IOI that has been reported in these HAWK and HARRIER trials, on 23 February 2020, the American Society of Retina Specialists alerted members to reported 14 cases of RV, of which 11 were identified as occlusive RV after brolucizumab treatment [76]. More recently, the 52-week outcomes of the KITE and KESTREL for DME were reported, demonstrated clear efficacy of brolucizumab for DME [72]. Safety profile at 52-week showed less evidence of IOI than in HAWK and HARRIER [71]. Therefore, a careful evaluation of any active IOI sign is recommended before the initiation of brolucizumab due to increased risk of IOI associated with occlusive RV. If IOI occurs, based on what have been reported, treatment with anti-inflammatory therapy should be initiated as promptly and aggressively as possible.
Faricimab is a bispecific antibody that inhibits both VEGF-A and angiopoietin-2 (Ang-2) pathways. The angiopoietins, Ang-1 and Ang-2, bind to tyrosine kinase with immunoglobulin and epidermal growth factor homology domain (Tie-2) receptor. While Ang-1 provides vascular stability and feedback inhibition of Ang-2 production, Ang-2 has been shown to inhibit the stabilization or maturation features of Ang-1 [108,109]. Therefore, dual blockade of VEGF-A and Ang-2 pathways may act synergistically and result in increased vascular stability and improved outcomes in DME and other retinal vascular disorders. BOULEVARD, a phase 2 clinical trial, showed a significant improvement in BCVA with 6 mg faricimab compared to RBZ 0.3 mg in patients with DME [79]. Subsequently, YOSEMITE and RHINE, two phase 3 clinical trials, established comparable results with intravitreal injection of 6 mg faricimab every 8 weeks, 6 mg of faricimab with personalized treatment interval, and 2 mg AFB every 8 weeks in the management of DME. The incidence of intraocular inflammation was found to be numerically greater in both faricimab groups as compared to AFB group in the same two phase 3 clinical trials [78].
KSI 301 is a novel anti-VEGF antibody biopolymer conjugate, a combination of monoclonal anti-VEGF antibody and a high molecular weight phosphorylcholine biopolymer, that is designed to increase durability and bioavailability in ocular tissue. Phase 1a and ongoing phase 1b studies showed promising results on efficacy, safety and durability of intravitreal KSI 301 administration in patients with DME, nv-AMD and retinal vein occlusion [80,81]. Based on the successful outcomes from phase 1 studies, a double-masked, randomized, multicenter phase 2 trial (NCT04049266), DAZZLE, has been conducted to compare the efficacy and safety of 5 mg KSI 301 at 12–20 weeks intervals and 2 mg AFB every 8 weeks after the loading dose in patients with nv-AMD [82]. However, in February 2022, it was announced that the DAZZLE Study did not meet its primary endpoint; additional analyses are being conducted.
Article highlights.
Vascular endothelial growth factor (VEGF), particularly VEGF-A, is known to be a key mediator in the pathophysiology of diabetic retinopathy.
Intravitreal anti-VEGF therapy is the first-line treatment for center-involved diabetic macular edema.
Currently available intravitreal anti-VEGF agents, bevacizumab, ranibizumab, and aflibercept, have shown significant superiority in visual acuity (VA) and anatomical improvement compared to laser treatment for center-involved diabetic macular edema in the key clinical trials.
Intravitreal anti-VEGF agents have demonstrated additional benefits in improving severity of diabetic retinopathy in patients with both non-proliferative and proliferative diabetic retinopathy.
Brolucizumab, faricimab, and KSI 301 are three novel anti-VEGF pharmacologic agents that have demonstrated promising results on efficacy and durability in patients with diabetic macular edema as well as other retinal vascular diseases.
Recent safety data from the HAWK and HARRIER studies as well as the KITE and KESTREL studies, and other case reports and case series, have shown an increased risk of intraocular inflammation and occlusive retinal vasculitis following intravitreal brolucizumab administration, more among eyes with neovascular age-related macular degeneration than eyes with diabetic macular edema.
Funding
This paper was not funded.
Declaration of interests
QD Nguyen serves on the scientific advisory boards for Bausch and Lomb, Bayer, Genentech/Roche, Regeneron, Novartis and Santen. QD Nguyen has also chaired the steering committee for the RISE and RIDE studies and was on the steering committee for the VISTA Study, as well as other studies sponsored by Genentech and Regeneron. DV Do serves on the scientific advisory boards for Allergan, Kodiak, Genentech and Regeneron. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Abbreviations
- AE
adverse events
- AFB
aflibercept
- Ang-2
Angiopoietin-2
- ATE
arterial thromboembolic events
- BCVA
best-corrected visual acuity
- BVZ
bevacizumab
- CHO
Chinese hamster ovary
- CMT
central macular thickness
- CRT
Central retinal thickness
- DME
diabetic macular edema
- DR
diabetic retinopathy
- DRCR
diabetic retinopathy clinical research
- DRSS
diabetic retinopathy severity scale
- ETDRS
early treatment diabetic retinopathy study
- FA
fluorescein angiography
- Fab
fragment antigen-binding
- Fc
fragment crystallizable
- FDA
Food and Drug Administration
- HF
heart failure
- IOI
intraocular inflammation
- IVIs
intravitreal injections
- kDa
kilodalton
- NPDR
non-proliferative diabetic retinopathy
- NV
neovascularization
- nv-AMD
neovascular age-related macular degeneration
- PACORES
Pan-American Collaborative Retina Study Group
- PDR
proliferative diabetic retinopathy
- PRP
panretinal photocoagulation
- PRN
pro re nata
- RBZ
ranibizumab
- RO
retinal vascular occlusion
- RV
retinal vasculitis
- SAE
systemic adverse events
- T&E
treat-and-extend
- VA
visual acuity
- VEGF
vascular endothelial growth factor
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 1.Fong DS, Aiello L, Gardner TW, et al. Retinopathy in diabetes. Diabetes Care. 2004;27Suppl 1:S84–7. [DOI] [PubMed] [Google Scholar]
- 2.Antonetti DA, Klein R, Gardner TW. Diabetic retinopathy. N Engl J Med. 2012 Mar 29;366(13):1227–1239. [DOI] [PubMed] [Google Scholar]
- 3.Cheung N, Mitchell P, Wong TY. Diabetic retinopathy. Lancet. 2010 Jul 10;376(9735):124–136. [DOI] [PubMed] [Google Scholar]
- 4.Moss SE, Klein R, Klein BE. The 14-year incidence of visual loss in a diabetic population. Ophthalmology. 1998;105(6):998–1003. [DOI] [PubMed] [Google Scholar]
- 5.Stitt AW, Curtis TM, Chen M, et al. The progress in understanding and treatment of diabetic retinopathy. Prog Retin Eye Res. 2016;51:156–186. [DOI] [PubMed] [Google Scholar]
- 6.Calderon GD, Juarez OH, Hernandez GE, et al. Oxidative stress and diabetic retinopathy: development and treatment. Eye. 2017;318:1122–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Stewart MW. Anti-vascular endothelial growth factor drug treatment of diabetic macular edema: the evolution continues. Curr Diabetes Rev. 2012 Jul 1;8(4):237–246. [DOI] [PubMed] [Google Scholar]
- 8.Boyer DS, Hopkins JJ, Sorof J, et al. Anti-vascular endothelial growth factor therapy for diabetic macular edema. Ther Adv Endocrinol Metab. 2013;46:151–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.US Food and Drug Administration. Oncologic Drugs Advisory Committee Meeting Briefing Document ABP 215, a proposed bio-similar to Avastin 2017. [cited 2020 January 20]. Available from: https://www.fda.gov/media/106549/download
- 10.Lode HE, Gjolberg TT, Foss S, et al. A new method for pharmaceutical compounding and storage of anti-VEGF biologics for intravitreal use in silicone oil-free prefilled plastic syringes. Sci Rep. 2019 Dec 2;9(1):18021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Retinopathy Clinical Research N D, Scott IU, and Edwards AR, et al. A phase II randomized clinical trial of intravitreal bevacizumab for diabetic macular edema. Ophthalmology. 2007;114 (10):1860–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This was the first randomized clinical trial that gave data on the short-term effect of intravitreal bevacizumab for diabetic macular edema (DME).
- 12.Arevalo JF, Sanchez JG, Fromow-Guerra J, et al. Comparison of two doses of primary intravitreal bevacizumab (Avastin) for diffuse diabetic macular edema: results from the Pan-American Collaborative Retina Study Group (PACORES) at 12-month follow-up. Graefe’s archive for clinical and experimental ophthalmology. 2009. Jun;247(6):735–743. [DOI] [PubMed] [Google Scholar]
- 13.Rajendram R, Fraser-Bell S, Kaines A, et al. A 2-year prospective randomized controlled trial of intravitreal bevacizumab or laser therapy (BOLT) in the management of diabetic macular edema: 24-month data: report 3. Arch Ophtalmol. 2012;130(8):972–979. [DOI] [PubMed] [Google Scholar]
- 14.Photocoagulation treatment of proliferative diabetic retinopathy. Clinical application of Diabetic Retinopathy Study (DRS) findings, DRS report number 8 the diabetic retinopathy study research group. Ophthalmology. 1981. Jul;88(7):583–600. [PubMed] [Google Scholar]
- 15.Tonello M, Costa RA, Almeida FP, et al. Panretinal photocoagulation versus PRP plus intravitreal bevacizumab for high-risk proliferative diabetic retinopathy (IBeHi study). Acta Ophthalmol. 2008;864:385–389. [DOI] [PubMed] [Google Scholar]
- 16.Ernst BJ, Garcia-Aguirre G, Oliver SC, et al. Intravitreal bevacizumab versus panretinal photocoagulation for treatment-naive proliferative and severe nonproliferative diabetic retinopathy. Acta Ophthalmol. 2012;90(7):e573–4. [DOI] [PubMed] [Google Scholar]
- 17.Arevalo JF, Lasave AF, Wu L, et al. INTRAVITREAL BEVACIZUMAB FOR PROLIFERATIVE DIABETIC RETINOPATHY: results From the Pan-American Collaborative Retina Study Group (PACORES) at 24 months of follow-up. Retina. 2017;37 (2):334–343. [DOI] [PubMed] [Google Scholar]; • This was the first randomized clinical trial that gave data on the short-term effect of intravitreal bevacizumab for diabetic macular edema (DME).
- 18.ClinicalTrials.gov. A 3-month study to assess the safety of ONS-5010 in subjects with visual impairment due to retinal disorders. [cited 2021. November 16]. [updated 2020 Apr 28; cited 2021 Nov 16]. Available from: https://clinicaltrials.gov/ct2/show/NCT04516278
- 19.Fasih U, Shaikh N, Rahman A, et al. A one-year follow-up study of ocular and systemic complications of intravitreal injection of bevacizumab (Avastin). J Pak Med Assoc. 2013; 636: 707–710. [PubMed] [Google Scholar]
- 20.Goldberg RA, Flynn HW Jr., Isom RF, et al. An outbreak of streptococcus endophthalmitis after intravitreal injection of bevacizumab. Am J Ophthalmol. 2012;1532:204–208 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tranos P, Gemenetzi M, Papandroudis A, et al. Progression of diabetic tractional retinal detachment following single injection of intravitreal Avastin. Eye. 2008;226:862. [DOI] [PubMed] [Google Scholar]
- 22.Afarid M, Sadegi Sarvestani A, Rahat F, et al. Intravitreal Injection of Bevacizumab: review of our previous Experience. Iran J Pharm Res. 2018;17(3):1093–1098. [PMC free article] [PubMed] [Google Scholar]
- 23.Sangroongruangsri S, Chaikledkaew U, Kumluang S, et al. Real-world safety of intravitreal bevacizumab and ranibizumab treatments for retinal diseases in Thailand: a Prospective Observational Study. Clin Drug Investig. 2018;389:853–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ferrara N, Damico L, Shams N, et al. Development of ranibizumab, an anti-vascular endothelial growth factor antigen binding fragment, as therapy for neovascular age-related macular degeneration. Retina. 2006;268:859–870. [DOI] [PubMed] [Google Scholar]
- 25.Nguyen QD, Shah SM, Heier JS, et al. Primary End Point (Six Months) Results of the Ranibizumab for Edema of the mAcula in diabetes (READ-2) study. Ophthalmology. 2009;116(11):2175–81 e1. [DOI] [PubMed] [Google Scholar]; • This was the pivotal prospective, randomized, interventional, multicenter clinical trial to show superiority of ranibizumab for DME through comparison between ranibizumab, photocoagulation, and both combination.
- 26.Nguyen QD, Shah SM, Khwaja AA, et al. Two-year outcomes of the ranibizumab for edema of the mAcula in diabetes (READ-2) study. Ophthalmology. 2010;11711:2146–2151. [DOI] [PubMed] [Google Scholar]
- 27.Do DV, Nguyen QD, Khwaja AA, et al. Ranibizumab for edema of the macula in diabetes study: 3-year outcomes and the need for prolonged frequent treatment. JAMA Ophthalmol. 2013;1312:139–145. [DOI] [PubMed] [Google Scholar]
- 28.Sepah YJ, Sadiq MA, Boyer D, et al. Twenty-four-month outcomes of the ranibizumab for edema of the macula in diabetes - protocol 3 with high dose (READ-3) Study. Ophthalmology. 2016;12312:2581–2587. [DOI] [PubMed] [Google Scholar]
- 29.Massin P, Bandello F, Garweg JG, et al. Safety and efficacy of ranibizumab in diabetic macular edema (RESOLVE Study): a 12-month, randomized, controlled, double-masked, multicenter phase II study. Diabetes Care. 2010;3311:2399–2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Retinopathy Clinical Research N D, Elman MJ, Aiello LP, et al. Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology. 2010;1176:1064–1077 e35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Elman MJ, Bressler NM, Qin H, et al. Expanded 2-year follow-up of ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology. 2011;1184:609–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Retinopathy Clinical Research N D, Elman MJ, Qin H, et al. Intravitreal ranibizumab for diabetic macular edema with prompt versus deferred laser treatment: three-year randomized trial results. Ophthalmology. 2012;11911:2312–2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Elman MJ, Ayala A, Bressler NM, et al. Intravitreal Ranibizumab for diabetic macular edema with prompt versus deferred laser treatment: 5-year randomized trial results. Ophthalmology. 2015;1222:375–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.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;1184:615–625. [DOI] [PubMed] [Google Scholar]
- 35.Ishibashi T, Li X, Koh A, et al. The REVEAL study: ranibizumab monotherapy or combined with laser versus laser monotherapy in asian patients with diabetic macular edema. Ophthalmology. 2015;1227:1402–1415. [DOI] [PubMed] [Google Scholar]
- 36.Nguyen QD, Brown DM, Marcus DM, et al. Ranibizumab for diabetic macular edema: results from 2 phase III randomized trials: RISE and RIDE. Ophthalmology. 2012;119(4):789–801. [DOI] [PubMed] [Google Scholar]; • These phase III, multicenter, double-masked, sham injection-controlled, randomized studies reported visual and anatomical improvement by ranibizumab in patients with DME.
- 37.Brown DM, Nguyen QD, Marcus DM, et al. Long-term outcomes of ranibizumab therapy for diabetic macular edema: the 36-month results from two phase III trials: RISE and RIDE. Ophthalmology. 2013;120(10):2013–2022. [DOI] [PubMed] [Google Scholar]
- 38.Schmidt-Erfurth U, Lang GE, Holz FG, et al. Three-year outcomes of individualized ranibizumab treatment in patients with diabetic macular edema: the RESTORE extension study. Ophthalmology. 2014;1215:1045–1053. [DOI] [PubMed] [Google Scholar]
- 39.Pearce I, Banerjee S, Burton BJ, et al. Ranibizumab 0.5 mg for diabetic macular edema with bimonthly monitoring after a phase of initial treatment: 18-month, multicenter, phase IIIB RELIGHT Study. Ophthalmology. 2015;1229:1811–1819. [DOI] [PubMed] [Google Scholar]
- 40.Payne JF, Wykoff CC, Clark WL, et al. Randomized trial of treat and extend ranibizumab with and without navigated laser for diabetic macular edema: TREX-DME 1 Year Outcomes. Ophthalmology. 2017;1241:74–81. [DOI] [PubMed] [Google Scholar]
- 41.Ip MS, Domalpally A, Hopkins JJ, et al. Long-term effects of ranibizumab on diabetic retinopathy severity and progression. Arch Ophtalmol. 2012;130(9):1145–1152. [DOI] [PubMed] [Google Scholar]
- 42.Ip MS, Domalpally A, Sun JK, et al. Long-term effects of therapy with ranibizumab on diabetic retinopathy severity and baseline risk factors for worsening retinopathy. Ophthalmology. 2015;1222:367–374. [DOI] [PubMed] [Google Scholar]
- 43.Sadiq MA, Hassan M, Soliman MK, et al. Effects of two different doses of ranibizumab on diabetic retinopathy severity. Ophthalmol Retina. 2017;1(6):566–567. [DOI] [PubMed] [Google Scholar]
- 44.Hassan M, Sadiq MA, Halim MS, et al. Short-Term effects of ranibizumab on diabetic retinopathy severity and progression. Ophthalmol Retina. 2018;27:749–751. [DOI] [PubMed] [Google Scholar]
- 45.Bressler SB, Odia I, Glassman AR, et al. CHANGES IN DIABETIC RETINOPATHY SEVERITY WHEN TREATING DIABETIC MACULAR EDEMA WITH RANIBIZUMAB: DRCR.net Protocol I 5-Year Report. Retina. 2018;3810:1896–1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Research N, Gross JG, Glassman AR; Writing Committee for the Diabetic Retinopathy Clinical, et al. Panretinal Photocoagulation vs Intravitreous Ranibizumab for Proliferative Diabetic Retinopathy: a Randomized Clinical Trial. Jama. 2015. Nov 24;314 (20):2137–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gross JG, Glassman AR, Liu D, et al. Five-year outcomes of panretinal photocoagulation vs intravitreous ranibizumab for proliferative diabetic retinopathy: a randomized clinical trial. JAMA Ophthalmol. 2018 Oct 1;136(10):1138–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Li X, Dai H, Li X, et al. Efficacy and safety of ranibizumab 0.5 mg in Chinese patients with visual impairment due to diabetic macular edema: results from the 12-month REFINE study. Graefe’s archive for clinical and experimental ophthalmology. 2019. Mar;257 (3):529–541. [DOI] [PubMed] [Google Scholar]
- 49.Yanagida Y, Ueta T. Systemic safety of ranibizumab for diabetic macular edema: meta-analysis of randomized trials. Retina. 2014;34 (4):629–635. [DOI] [PubMed] [Google Scholar]
- 50.Balaratnasingam C, Dhrami-Gavazi E, McCann JT, et al. Aflibercept: a review of its use in the treatment of choroidal neovascularization due to age-related macular degeneration. Clin Ophthalmol. 2015;9:2355–2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Stewart MW, Rosenfeld PJ. Predicted biological activity of intravitreal VEGF Trap. Br J Ophthalmol. 2008;92(5):667–668. [DOI] [PubMed] [Google Scholar]
- 52.Sarwar S, Bakbak B, Sadiq MA, et al. Fusion Proteins: aflibercept (VEGF Trap-Eye). Dev Ophthalmol. 2016;55:282–294. [DOI] [PubMed] [Google Scholar]
- 53.Do DV, Nguyen QD, Shah SM, et al. An exploratory study of the safety, tolerability and bioactivity of a single intravitreal injection of vascular endothelial growth factor Trap-Eye in patients with diabetic macular oedema. Br J Ophthalmol. 2009;932:144–149. [DOI] [PubMed] [Google Scholar]
- 54.Do DV, Schmidt-Erfurth U, Gonzalez VH, et al. The DA VINCI Study: phase 2 primary results of VEGF Trap-Eye in patients with diabetic macular edema. Ophthalmology. 2011;1189:1819–1826. [DOI] [PubMed] [Google Scholar]
- 55.Do DV, Nguyen QD, Boyer D, et al. One-year outcomes of the da Vinci Study of VEGF Trap-Eye in eyes with diabetic macular edema. Ophthalmology. 2012;1198:1658–1665. [DOI] [PubMed] [Google Scholar]
- 56.Brown DM, Schmidt-Erfurth U, Do DV, et al. Intravitreal aflibercept for diabetic macular edema: 100-week results from the Vista and VIVID Studies. Ophthalmology. 2015;122 (10):2044–2052. [DOI] [PubMed] [Google Scholar]; •• In both VISTA and VIVID, randomized, phase 3 trials, the 52-week visual and anatomic superiority of intravitreal aflibercept injection for DME over laser control was sustained through week 100.
- 57.Heier JS, Korobelnik JF, Brown DM, et al. Intravitreal Aflibercept for Diabetic Macular Edema: 148-Week Results from the Vista and VIVID Studies. Ophthalmology. 2016;12311:2376–2385. [DOI] [PubMed] [Google Scholar]
- 58.Baker CW, Glassman AR, Beaulieu WT, et al. Effect of initial management with aflibercept vs laser photocoagulation vs observation on vision loss among patients with diabetic macular edema involving the center of the macula and good visual acuity: a randomized clinical trial. Jama. 2019 May 21;321(19):1880–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wells JA, Glassman AR, Ayala AR, et al. Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema: two-year results from a comparative effectiveness randomized clinical trial. Ophthalmology. 2016;123(6):1351–1359. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• The prospective randomized multi-center trial conducted by DRCR.net was the only trial to directly compare all three anti-VEGF drugs.
- 60.Glassman AR, Wells JA 3rd, Josic K, et al. Five-Year Outcomes after Initial Aflibercept, Bevacizumab, or Ranibizumab Treatment for Diabetic Macular Edema (Protocol T Extension Study). Ophthalmology. 2020;1279:1201–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Mitchell P, McAllister I, Larsen M, et al. Evaluating the impact of intravitreal aflibercept on diabetic retinopathy progression in the VIVID-DME and Vista-DME Studies. Ophthalmol Retina. 2018;210:988–996. [DOI] [PubMed] [Google Scholar]
- 62.Bressler SB, Liu D, Glassman AR, et al. Change in diabetic retinopathy through 2 years: secondary analysis of a randomized clinical trial comparing Aflibercept, Bevacizumab, and Ranibizumab. JAMA Ophthalmol. 2017 Jun 1;135(6):558–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sivaprasad S, Prevost AT, Vasconcelos JC, et al. Clinical efficacy of intravitreal aflibercept versus panretinal photocoagulation for best corrected visual acuity in patients with proliferative diabetic retinopathy at 52 weeks (CLARITY): a multicentre, single-blinded, randomised, controlled, phase 2b, non-inferiority trial. Lancet. 2017 Jun 3;389(10085):2193–2203. [DOI] [PubMed] [Google Scholar]
- 64.Brown DM, Wykoff CC, Boyer D, et al. Evaluation of Intravitreal Aflibercept for the Treatment of Severe Nonproliferative Diabetic Retinopathy: results From the PANORAMA randomized clinical trial. JAMA Ophthalmol. 2021 Sep 1;139(9):946–955. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• In the PANORAMA clinical trial, treatment of severe NPDR with intravitreal aflibercept injections improved the severity of retinopathy and reduced the risk of progression to CI-DME and vision-threatening complications vs sham treatment with observation.
- 65.Ziemssen F, Schlottman PG, Lim JI, et al. Initiation of intravitreal aflibercept injection treatment in patients with diabetic macular edema: a review of VIVID-DME and Vista-DME data. Int J Retina Vitreous. 2016;2(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Retinopathy Clinical Research N D, Wells JA, Glassman AR, et al. Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema. N Engl J Med. 2015 Mar 26;372(13):1193–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Andrade GC, Dias JR, Maia A, et al. INTRAVITREAL INJECTIONS OF ZIV-AFLIBERCEPT FOR DIABETIC MACULAR EDEMA: a Pilot Study. Retina. 2016;369:1640–1645. [DOI] [PubMed] [Google Scholar]
- 68.Holz FG, Dugel PU, Weissgerber G, et al. Single-Chain antibody fragment VEGF Inhibitor RTH258 for neovascular age-related macular degeneration: a randomized controlled study. Ophthalmology. 2016;1235:1080–1089. [DOI] [PubMed] [Google Scholar]
- 69.Nguyen QD, Das A, Do DV, et al. Brolucizumab: evolution through preclinical and clinical studies and the implications for the management of neovascular age-related macular degeneration. Ophthalmology. 2020 Jan 17;127(7):963–976. [DOI] [PubMed] [Google Scholar]
- 70.Dugel PU, Jaffe GJ, Sallstig P, et al. Brolucizumab versus aflibercept in participants with neovascular age-related macular degeneration: a randomized trial. Ophthalmology. 2017;1249:1296–1304. [DOI] [PubMed] [Google Scholar]
- 71.Dugel PU, Koh A, Ogura Y, et al. HAWK and HARRIER: phase 3, Multicenter, Randomized, Double-Masked trials of brolucizumab for neovascular age-related macular degeneration. Ophthalmology. 2020;1271:72–84. [DOI] [PubMed] [Google Scholar]
- 72.Brown DM, Wolf S, Garweg J, et al. Brolucizumab for the treatment of visual impairment due to diabetic macular edema: 52-week results from the KITE and KESTREL studies. Invest Ophthalmol Vis Sci. 2021;62(8):1045. [Google Scholar]
- 73.Jain A, Chea S, Matsumiya W, et al. Severe vision loss secondary to retinal arteriolar occlusions after multiple intravitreal brolucizumab administrations. Am J Ophthalmol Case Rep. 2020;18:100687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Haug SJ, Hien DL, Uludag G, et al. Retinal arterial occlusive vasculitis following intravitreal brolucizumab administration. Am J Ophthalmol Case Rep. 2020;18:100680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Baumal CR, Spaide RF, Vajzovic L, et al. Retinal Vasculitis and Intraocular Inflammation after Intravitreal Injection of Brolucizumab. Ophthalmology. 2020;12710:1345–1359. [DOI] [PubMed] [Google Scholar]
- 76.American Society of Retina Specialists. Beavu update for ASRS members Published February 23, 2020 and 2020. Mar 30. Available from: https://www.asrs.org/clinical/clinical-updates
- 77.Sharma A, Kumar N, Kuppermann BD, et al. Faricimab: expanding horizon beyond VEGF. Eye. 2020;345:802–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wykoff CC, Abreu F, Adamis AP, et al. Efficacy, durability, and safety of intravitreal faricimab with extended dosing up to every 16 weeks in patients with diabetic macular oedema (YOSEMITE and RHINE): two randomised, double-masked, phase 3 trials. Lancet. 2022 Jan 21;399(10326):741–755. [DOI] [PubMed] [Google Scholar]; •• YOSEMITE and RHINE, randomized, double-masked phase 3 trials with comparison between facirimab vs aflibercept demonstrated the potential for the new drug of faricimab to extend the durability of treatment for patients with DME.
- 79.Sahni J, Patel SS, Dugel PU, 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;1268:1155–1170. [DOI] [PubMed] [Google Scholar]
- 80.NJ PSS, Qudrat A, Do DV, et al. Phase 1 first-in-human study of KSI-301: a novel anti-VEGF antibody biopolymer conjugate with extended durability. Invest Ophthalmol Vis Sci. 2019;60:3670. [Google Scholar]
- 81.Jd PSS, Miller B, Ehrlich JS, et al. Updated results of phase 1b Study of KSI-301, an Anti-VEGF antibody biopolymer conjugate with extended durability, in wAMD, DME, and RVO. Invest Ophthalmol Vis Sci. 2020;61(7):4286. [Google Scholar]
- 82.ClinicalTrials.gov. A study to evaluate the efficacy and safety of ksi-301, an anti-vegf antibody biopolymer conjugate, versus aflibercept in patients with Neovascular (Wet) age-related macular degeneration (DAZZLE). [updated 2020. Nov 19; cited 2020 Nov 30]. Available from: https://clinicaltrials.gov/ct2/show/NCT04049266
- 83.Bressler SB, Ayala AR, Bressler NM, et al. Persistent macular thickening after ranibizumab treatment for diabetic macular edema with vision impairment. JAMA Ophthalmol. 2016;1343:278–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Pieramici DJ, Wang PW, Ding B, et al. Visual and anatomic outcomes in patients with diabetic macular edema with limited initial anatomic response to ranibizumab in RIDE and RISE. Ophthalmology. 2016;1236:1345–1350. [DOI] [PubMed] [Google Scholar]
- 85.Bressler NM, Beaulieu WT, Glassman AR, et al. Persistent Macular Thickening Following Intravitreous Aflibercept, Bevacizumab, or Ranibizumab for Central-Involved Diabetic Macular Edema With Vision Impairment: a Secondary Analysis of a Randomized Clinical Trial. JAMA Ophthalmol. 2018 Mar 1;136(3):257–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Maturi RK, Glassman AR, Liu D, et al. Effect of adding dexamethasone to continued ranibizumab treatment in patients with persistent diabetic macular edema: a DRCR network phase 2 randomized clinical trial. JAMA Ophthalmol. 2018 Jan 1;136(1):29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Binder S Loss of reactivity in intravitreal anti-VEGF therapy: tachyphylaxis or tolerance? Br J Ophthalmol. 2012;96(1):1–2. [DOI] [PubMed] [Google Scholar]
- 88.Wallsh JO, Gallemore RP. Anti-VEGF-resistant retinal diseases: a review of the latest treatment options. Cells. 2021 Apr 29;10 (5):1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Klein KA, Cleary TS, Reichel E. Effect of intravitreal aflibercept on recalcitrant diabetic macular edema. Int J Retina Vitreous. 2017;3(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Salimi A, Vila N, Modabber M, et al. One-year outcomes of Aflibercept for refractory diabetic macular edema in Bevacizumab nonresponders. Indian J Ophthalmol. 2021;692:360–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Laiginhas R, Silva MI, Rosas V, et al. Aflibercept in diabetic macular edema refractory to previous bevacizumab: outcomes and predictors of success. Graefe’s archive for clinical and experimental ophthalmology. 2018. Jan;256(1):83–89. [DOI] [PubMed] [Google Scholar]
- 92.Liu Y, Cheng J, Gao Y, et al. Efficacy of switching therapy to aflibercept for patients with persistent diabetic macular edema: a systematic review and meta-analysis. Ann Transl Med. 2020;86:382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sorour OA, Liu K, Mehta N, et al. Visual and anatomic outcomes of sustained single agent anti-VEGF treatment versus double anti-VEGF switching in the treatment of persistent diabetic macular edema. Int J Retina Vitreous. 2020;6(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Arevalo JF, Liu TYA. Pan-American Collaborative Retina Study G. Intravitreal bevacizumab in diabetic retinopathy. recommendations from the pan-American collaborative retina study group (PACORES): the 2016 Knobloch Lecture. Asia Pac J Ophthalmol (Phila). 2018;7(1):36–39. [DOI] [PubMed] [Google Scholar]
- 95.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004 Jun 3;350(23):2335–2342. [DOI] [PubMed] [Google Scholar]
- 96.Loupakis F, Cremolini C, Masi G, et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014 Oct 23;371(17):1609–1618. [DOI] [PubMed] [Google Scholar]
- 97.Edington M, Connolly J, Chong NV. Pharmacokinetics of intravitreal anti-VEGF drugs in vitrectomized versus non-vitrectomized eyes. Expert Opin Drug Metab Toxicol. 2017;13(12):1217–1224. [DOI] [PubMed] [Google Scholar]
- 98.Yanyali A, Aytug B, Horozoglu F, et al. Bevacizumab (Avastin) for diabetic macular edema in previously vitrectomized eyes. Am J Ophthalmol. 2007;1441:124–126. [DOI] [PubMed] [Google Scholar]
- 99.Mehta S, Blinder KJ, Shah GK, et al. Intravitreal bevacizumab for the treatment of refractory diabetic macular edema. Ophthalmic Surg Lasers Imaging. 2010;413:323–329. [DOI] [PubMed] [Google Scholar]
- 100.Laugesen CS, Ostri C, Brynskov T, et al. Intravitreal ranibizumab for diabetic macular oedema in previously vitrectomized eyes. Acta Ophthalmol. 2017;951:28–32. [DOI] [PubMed] [Google Scholar]
- 101.Koyanagi Y, Yoshida S, Kobayashi Y, et al. Comparison of the effectiveness of intravitreal ranibizumab for diabetic macular edema in vitrectomized and nonvitrectomized eyes. Ophthalmologica. 2016;236(2):67–73. [DOI] [PubMed] [Google Scholar]
- 102.Do DV, Rhoades W, Nguyen QD. pharmacokinetic study of intravitreal aflibercept in humans with neovascular age-related macular degeneration. Retina. 2020;40(4):643–647. [DOI] [PubMed] [Google Scholar]
- 103.Krohne TU, Eter N, Holz FG, et al. Intraocular pharmacokinetics of bevacizumab after a single intravitreal injection in humans. Am J Ophthalmol. 2008;1464:508–512. [DOI] [PubMed] [Google Scholar]
- 104.Krohne TU, Liu Z, Holz FG, et al. Intraocular pharmacokinetics of ranibizumab following a single intravitreal injection in humans. Am J Ophthalmol. 2012;1544:682–686 e2. [DOI] [PubMed] [Google Scholar]
- 105.Bressler SB, Melia M, Glassman AR, et al. Ranibizumab plus prompt or deferred laser for diabetic macular edema in eyes with vitrectomy before anti-vascular endothelial growth factor therapy. Retina. 2015;3512:2516–2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Tran THC, Erginay A, Verdun S, et al. One-year outcome of aflibercept intravitreal injection in vitrectomized eyes with diabetic macular edema. Clin Ophthalmol. 2021;15:1971–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.ClinicalTrials.gov. A pharmacokinetic study: ranibizumab, aflibercept and the effect of vitrectomy (VITCLEAR). updated 2021 Apr 30; cited 2022. Feb 11].
- 108.Sharma A, Kumar N, Parachuri N, et al. Faricimab: two in the bush is proving better than one in the hand? Ocul Immunol Inflamm. 2021;9:1–3. [DOI] [PubMed] [Google Scholar]
- 109.Cai J, Kehoe O, Smith GM, et al. The angiopoietin/Tie-2 system regulates pericyte survival and recruitment in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2008;495:2163–2171. [DOI] [PubMed] [Google Scholar]
