A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030...

8
A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration Glenn J. Jaffe, MD, 1 Dean Eliott, MD, 2 John A. Wells, MD, 3 Jonathan L. Prenner, MD, 4 Andras Papp, MD, 5 Samir Patel, MD 6 Purpose: To assess the safety and tolerability of E10030 (Fovista; Ophthotech, New York, NY), a platelet- derived growth factor (PDGF) antagonist, when administered in combination with an anti-vascular endothelial growth factor (VEGF) agent, ranibizumab (Lucentis; Genentech, South San Francisco, CA) 0.5 mg, by intravitreal injection in participants with neovascular age-related macular degeneration (NVAMD). Design: Prospective phase 1 clinical trial. Participants: A total of 23 participants diagnosed with NVAMD and aged 50 years or older were enrolled. Methods: Part 1 included 15 participants. Three participants received a single intravitreal E10030 (0.03 mg) injection and were subsequently given intravitreal ranibizumab (0.5 mg) injections at weeks 2, 6, and 10. Twelve participants (3 per group) received E10030 (0.03, 0.3, 1.5, or 3.0 mg) in combination with ranibizumab (0.5 mg) at day 0, month 1, and month 2 in an ascending manner. In Part 2 (8 participants), E10030 (0.3, 1.5, or 3.0 mg) in combination with ranibizumab (0.5 mg) was injected at day 0, month 1, and month 2. Main Outcome Measures: Safety at week 12 was the primary outcome and included assessment of vital signs, laboratory tests, and serial eye examinations. Other safety metrics included assessment through week 24 of Early Treatment of Diabetic Retinopathy Study (ETDRS) visual acuity (VA) and biomarker changes evaluated by optical coherence tomography (OCT) and uorescein angiography (FA). Results: All doses of intravitreal E10030 administered in combination with ranibizumab were well tolerated. No dose-limiting toxicities or relevant safety events were noted at any dose level during the study. Investigators did not report adverse events related to E10030 or ranibizumab. Mean VA change was a gain of 14 letters, and 59% of participants gained 15 letters from baseline at week 12. On FA at week 12, there was an 85.5% mean reduction from baseline in choroidal neovascularization (CNV) size. On OCT at the week 12 visit, there was a mean decrease in center point thickness and central subeld thickness of 38.9% and 33.7%, respectively. Conclusions: Intravitreal E10030 administered at doses up to 3 mg in combination with ranibizumab was well tolerated without evidence of systemic or ocular toxicity in participants with NVAMD. The changes in both mean VA and imaging biomarkers suggest a favorable short-term safety prole for the combination therapy of E10030 and ranibizumab. Ophthalmology 2016;123:78-85 ª 2016 by the American Academy of Ophthalmology. Supplemental material is available at www.aaojournal.org. Neovascular age-related macular degeneration (NVAMD) is the leading cause of visual loss in individuals aged more than 55 years in the western world. 1 The current standard of care for NVAMD is the intravitreal delivery of agents targeting vascular endothelial growth factor (VEGF), one of the proteins involved in the molecular orchestration of the pathologic neovascular cascade. 2 Anti-VEGF agents have greatly improved visual out- comes in patients with NVAMD. 3e5 However, a signicant unmet need exists with anti-VEGF monotherapy regardless of the dose or administration regimen. Most eyes treated with anti-VEGF monotherapy do not gain signicant visual acuity (VA) (15 Early Treatment of Diabetic Retinopathy Study [ETDRS] letters), up to 25% of eyes lose additional VA after therapy initiation, and approximately 50% of eyes do not achieve a nal VA level (20/40) that is better than or equal to that required to drive in most American states. 3 Furthermore, postregistration analyses of claims data show that on average, participants lose additional VA 3 to 4 years after initiation of therapy in a real-worldsetting. 6,7 The efcacy of anti-VEGF monotherapy in NVAMD is primarily mediated by a marked reduction in hyper- permeability associated with pathologic neovascular com- plexes. 8,9 However, VEGF antagonism does not seem to 78 Ó 2016 by the American Academy of Ophthalmology Published by Elsevier Inc. http://dx.doi.org/10.1016/j.ophtha.2015.09.004 ISSN 0161-6420/15

Transcript of A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030...

Page 1: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

A Phase 1 Study of Intravitreous E10030in Combination with Ranibizumab inNeovascular Age-Related MacularDegeneration

Glenn J. Jaffe, MD,1 Dean Eliott, MD,2 John A. Wells, MD,3 Jonathan L. Prenner, MD,4 Andras Papp, MD,5

Samir Patel, MD6

Purpose: To assess the safety and tolerability of E10030 (Fovista; Ophthotech, New York, NY), a platelet-derived growth factor (PDGF) antagonist, when administered in combination with an anti-vascular endothelialgrowth factor (VEGF) agent, ranibizumab (Lucentis; Genentech, South San Francisco, CA) 0.5 mg, by intravitrealinjection in participants with neovascular age-related macular degeneration (NVAMD).

Design: Prospective phase 1 clinical trial.Participants: A total of 23 participants diagnosed with NVAMD and aged 50 years or older were enrolled.Methods: Part 1 included 15 participants. Three participants received a single intravitreal E10030 (0.03 mg)

injection and were subsequently given intravitreal ranibizumab (0.5 mg) injections at weeks 2, 6, and 10. Twelveparticipants (3 per group) received E10030 (0.03, 0.3, 1.5, or 3.0 mg) in combination with ranibizumab (0.5 mg) atday 0, month 1, and month 2 in an ascending manner. In Part 2 (8 participants), E10030 (0.3, 1.5, or 3.0 mg) incombination with ranibizumab (0.5 mg) was injected at day 0, month 1, and month 2.

Main Outcome Measures: Safety at week 12 was the primary outcome and included assessment of vitalsigns, laboratory tests, and serial eye examinations. Other safety metrics included assessment through week 24of Early Treatment of Diabetic Retinopathy Study (ETDRS) visual acuity (VA) and biomarker changes evaluated byoptical coherence tomography (OCT) and fluorescein angiography (FA).

Results: All doses of intravitreal E10030 administered in combination with ranibizumab were well tolerated.No dose-limiting toxicities or relevant safety events were noted at any dose level during the study. Investigatorsdid not report adverse events related to E10030 or ranibizumab. Mean VA change was a gain of 14 letters, and59% of participants gained �15 letters from baseline at week 12. On FA at week 12, there was an 85.5% meanreduction from baseline in choroidal neovascularization (CNV) size. On OCT at the week 12 visit, there was amean decrease in center point thickness and central subfield thickness of 38.9% and 33.7%, respectively.

Conclusions: Intravitreal E10030 administered at doses up to 3 mg in combination with ranibizumab was welltolerated without evidence of systemic or ocular toxicity in participants with NVAMD. The changes in both meanVA and imaging biomarkers suggest a favorable short-term safety profile for the combination therapy of E10030and ranibizumab. Ophthalmology 2016;123:78-85 ª 2016 by the American Academy of Ophthalmology.

Supplemental material is available at www.aaojournal.org.

Neovascular age-related macular degeneration (NVAMD) isthe leading cause of visual loss in individuals aged morethan 55 years in the western world.1 The current standard ofcare for NVAMD is the intravitreal delivery of agentstargeting vascular endothelial growth factor (VEGF), oneof the proteins involved in the molecular orchestration ofthe pathologic neovascular cascade.2

Anti-VEGF agents have greatly improved visual out-comes in patients with NVAMD.3e5 However, a significantunmet need exists with anti-VEGF monotherapy regardlessof the dose or administration regimen. Most eyes treatedwith anti-VEGF monotherapy do not gain significant visual

78 � 2016 by the American Academy of OphthalmologyPublished by Elsevier Inc.

acuity (VA) (�15 Early Treatment of Diabetic RetinopathyStudy [ETDRS] letters), up to 25% of eyes lose additionalVA after therapy initiation, and approximately 50% of eyesdo not achieve a final VA level (�20/40) that is better thanor equal to that required to drive in most American states.3

Furthermore, postregistration analyses of claims data showthat on average, participants lose additional VA 3 to 4years after initiation of therapy in a “real-world” setting.6,7

The efficacy of anti-VEGF monotherapy in NVAMD isprimarily mediated by a marked reduction in hyper-permeability associated with pathologic neovascular com-plexes.8,9 However, VEGF antagonism does not seem to

http://dx.doi.org/10.1016/j.ophtha.2015.09.004ISSN 0161-6420/15

Page 2: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Jaffe et al � E10030 and Ranibizumab for NVAMD

significantly alter or induce regression of those neovascularcomplexes.10

Accordingly, pharmacologic strategies that both modifychoroidal neovascular membranes (choroidal neo-vascularization [CNV]) by inducing neovascular regressionand reduce permeability may result in improved visualoutcomes in eyes with NVAMD. Furthermore, pericyteshave been shown to be a major source of myofibroblasts,which are predominantly involved in deposition of patho-logic matrix leading to tissue fibrosis. Pericytes areincreasingly implicated in the deposition of pathologicmatrix in a number of diseases of organ fibrosis, includingthose of the liver, lung, and kidney. In addition, advancedNVAMD often includes the formation of subretinalfibrosis.3 Therefore, pericyte loss from the choroidalneovasculature may play a role in reduction of fibrousevolution of the neovascular complex associated with wetage-related macular degeneration.11,12

Multiple mechanisms have been described to explain theresistance of pathologic neovascularization to anti-VEGFmonotherapy. Pericyte coverage of endothelial cells ontheir external surface is one such mode of resistance.13

Pericytes are derived from the same progenitor cells asvascular smooth muscle. They are contractile cells thatintimately cover the underlying endothelial cells via ashared common basement membrane.14 This anatomicrelationship permits pericytes to locally provideendothelial cells with VEGF and other growth andsurvival factors via paracrine and juxtacrine signals.15

Therefore, it has been proposed that pericytes may protectthe endothelial cells in the face of anti-VEGF agents andplay an important role in anti-VEGF resistance.16 Preclinicalophthalmic and oncologic models of angiogenesis confirmthis pericyte-mediated anti-VEGF resistance.17,18 Otherproposed mechanisms of anti-VEGF resistance may includepersistent inflammation and upregulation of mediators otherthan VEGF that are involved in the molecular cascade thatdrives angiogenesis.19

Platelet-derived growth factor (PDGF)-BB is a homo-dimer consisting of a dimeric molecule of disulfide-bondedB-polypeptide chains. Platelet-derived growth factor-BBbinds to a dimerized protein tyrosine kinase receptor onpericytes. This ligand-receptor complex is critical for peri-cyte survival, recruitment, and maturation.20 Inhibition ofPDGF-BB results in pericyte loss in genetic deletionstudies and in vivo models of pathologic angiogenesis.21

Because pericytes protect endothelial cells from VEGFinhibition, pericyte loss within a neovascular complex mayrender the underlying endothelial cells susceptible to theeffects of VEGF blockade.18,22 E10030 is a 32-mer-pegy-lated aptamer that binds and inhibits PDGF-BB. In pre-clinical models, E10030 potently strips and induces pericyteloss.23

This report describes the phase 1 trial of an intravitreallydelivered anti-PDGF aptamer, E10030 (Fovista; Oph-thotech, New York, NY) combined with an anti-VEGFprotein. This phase 1 trial was initiated in treatment-naïveparticipants with NVAMD to assess the safety of intravitrealadministration of E10030 in a dose-escalation scheme whenadministered in combination with ranibizumab (Lucentis,

Genentech, South San Francisco, CA). The study is basedon the premise that inducing neovascular pericyte loss willenhance the effects of anti-VEGF agents on unprotectedendothelial cells and therefore will induce neovascular tissueregression and modify the underlying disease.

Methods

Study Design

This prospective study is registered at ClinTrials.gov, IdentifierNCT00569140; was conducted at 11 study sites in compliance withthe Declaration of Helsinki, US Code 21 of Federal Regulations,and the Harmonized Tripartite Guidelines for Good ClinicalPractice (1996); and was reviewed and approved by the appropriateEthics Committees or institutional review boards at each studycenter. Informed consent was obtained from all study participants.Twenty-three participants who had a diagnosis of NVAMD andwere aged �50 years were enrolled between July 2009 and April of2010. When aptamers such as E10030 are manufactured for ther-apeutic use, increasing drug concentrations cause increased vis-cosity. Doses of E10030 >3.0 mg were deemed to be too viscousfor intravitreal administration. Thus, 3.0 mg was the highestE10030 dose tested. Part 1 of the study used an ascending dosedesign that included 15 participants. Three participants received asingle intravitreal administration of E10030 (0.03 mg), and thenranibizumab (0.5 mg) was administered subsequently at weeks 2,6, and 10. This was a dose-escalating safety study, and once thesafety of 1 dose (in combination with ranibizumab) was demon-strated, the next higher dose was given. Accordingly, 12 partici-pants, 3 in each dose group, received E10030 (0.03, 0.3, 1.5, or 3.0mg) in an ascending manner in combination with ranibizumab (0.5mg) at day 0, month 1, and month 2. Part 2 of the study used aparallel dose design and included 8 participants who were givenE10030 (0.3, 1.5, or 3.0 mg) in combination with ranibizumab (0.5mg) at day 0, month 1, and month 2. The primary end point was at12 weeks, and follow-up continued through week 24. There was nocontrol group, and the study was not double-masked in this phase 1protocol. The protocol design is shown in Figure 1.

Study Population

To be included in the trial, participants had to be aged �50 years,had to have been newly diagnosed with subfoveal CNV secondaryto NVAMD, and had to have some classic CNV componentdocumented on fluorescein angiography (FA). The total area of thelesion (including blood, neovascularization, and scar/atrophy) wasrequired to be �5 disc areas, of which at least 50% was required tobe active CNV. The other main inclusion criteria were as follows:(1) ETDRS best-corrected VA in the study eye between 20/63 and20/200 inclusive; (2) presence of clear ocular media and adequatepupillary dilation to permit sufficient resolution of the stereoscopicfundus photographic images; and (3) intraocular pressure (IOP)�21 mmHg. A more detailed list of inclusion and exclusion criteriais listed in the Appendix (available at www.aaojournal.org).

One participant was enrolled erroneously because he had adiagnosis of diabetes, a protocol exclusion. The patient received asingle dose of E10030 and ranibizumab at day 0 but no furthertreatment; this patient withdrew from the study and was excludedfrom the per-protocol dataset (n ¼ 22).

79

Page 3: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Figure 1. Flowchart showing the dosing regimen for subjects enrolled in the study.

Ophthalmology Volume 123, Number 1, January 2016

Drug Administration Procedure

Intravitreal injections were given according to standard-of-caretechniques used in modern retinal practice. Briefly, a sterile lidspeculum was placed, and local anesthesia was administered. Theconjunctiva and ocular adnexa were prepared with 5% povidoneiodine. A 27-gauge needle was used for all injections, which weregiven 3.5 mm from the limbus. In patients in whom both intra-vitreal ranibizumab and E10030 were given, the injections weregiven consecutively (50 ml each), and ranibizumab was adminis-tered first. In these eyes, the IOP was measured 30 minutes after thefirst injection was given. The IOP had to be �21 mmHg or within 5mmHg of the baseline IOP on the day of injection, before theE10030 injection could be administered. The IOP was monitoredafter the second injection until it was less than 30 mmHg.

Safety Assessments

Drug tolerability and safety metrics were monitored closely duringthe trial. A thorough clinical evaluation was performed at each visitand included slit-lamp examination of the anterior segment andlens, and biomicroscopic and ophthalmoscopic examination of theposterior segment; data from each of these examinations wererecorded. In addition, IOP, adverse ocular and systemic events, andlaboratory test result data were collected. Laboratory tests per-formed in the clinical trial included the following: hematology:hemoglobin, platelet count, white blood cell, and differential; renalfunction: serum creatinine and blood urea nitrogen; hepatic func-tion: serum bilirubin, alkaline phosphatase, gamma-glutamyltransferase, serum glutamic oxaloacetic transaminase/aspartatetransaminase, and serum glutamic pyruvic transaminase/alanineaminotransferase; electrolytes: sodium, potassium, chloride,

80

bicarbonate, calcium, and phosphate; and complete urinalysis.Safety biomarker data also were collected. These biomarkersincluded changes in visual function, as measured by VA, quanti-tative retinal thickness changes as determined by optical coherencetomography (OCT), and CNV lesion size assessed by FA. AStratus OCT system (Zeiss Meditec, Dublin, CA) was used toobtain OCT scans. The OCT images were obtained with the fastmacular thickness map protocol at investigator study sites. In-vestigators recorded quantitative imaging data including the centerpoint value and central subfield value onto standardized case reportforms. Those case report form data were confirmed and thenanalyzed internally. Stereo FA was obtained on field 2 of amodified 3-field imaging protocol at early (as soon as dye was firstseen in the eye), mid (1e3 minutes), and late (5 minutes) phases.Two independent retinal specialists used Adobe Photoshop CS4Extended software (Adobe Systems, Inc, San Jose, CA) to pre-cisely outline the area(s) of CNV on FAs at standardized magni-fication. The software contains measurement and analytic tools thatwere used to quantify (in pixel area) the corresponding CNV size atbaseline and week 12. The CNV size was determined during theFA mid-phase when the neovascular membrane is well defined, butbefore the development of significant leakage (Fig 2). The primaryend point was at 12 weeks, and follow-up continued throughweek 24.

Results

Baseline Study Population Characteristics

The mean participant age was 75 years (range, 56e88 years), 13were women, and 14 right eyes and 9 left eyes were enrolled. The

Page 4: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Figure 2. Example showing the method used to determine choroidal neovascularization (CNV) area on fluorescein angiography images. Pixelations seen onimages correspond to the quantified area of CNV. The numbers reflect the CNV area in m2.

Jaffe et al � E10030 and Ranibizumab for NVAMD

mean best-corrected VA was 49.2 letters (range, 24e73.5) on theEarly Treatment Diabetic Retinopathy (ETDRS) chart. The averageOCT central subfield thickness by OCT was 386.5 mm (range,249.5e599 mm).

Safety

E10030 was well tolerated. There was no dose-limiting toxicity atany dose level, and no safety concerns were identified. There wereno ocular serious adverse events. As would be expected in a trial ofintravitreally delivered medications, many participants developedocular adverse events related to the injection itself, such as ocularirritation and subconjunctival hemorrhage (Table 1). However, noocular adverse events were attributed to E10030 or ranibizumab.

Table 1. Ocular Adverse Event

Adverse Event 0.03 mg (n ¼ 6) 0.3 mg (n ¼Patients with at least 1 event 6 (100%) 5 (63%)Foreign body sensation in eyes 4 (67%) 0 (0%)Conjunctival hemorrhage 2 (33%) 3 (38%)Eye irritation 0 (0%) 2 (25%)Myodesopsia 1 (17%) 0 (0%)Punctate keratitis 2 (33%) 0 (0%)Anterior chamber cell 1 (17%) 0 (0%)Conjunctival edema 0 (0%) 1 (13%)Macular degeneration* 0 (0%) 0 (0%)

*Macular degeneration refers to the development of neovascular age-related m

The combination of E10030 and ranibizumab did not result incomplications of the anterior segment, such as corneal toxicity,lens toxicity, or ocular inflammation. Furthermore, posteriorsegment complications, including inflammation and abnormalitiesof the normal vasculature, were not observed.

There was a small mean IOP increase after each injection wasgiven; the IOP returned to pre-dose levels by the following visit. Atweeks 12 and 24, mean IOP was again similar to baseline levels.There was no indication of a cumulative increase in IOP aftermultiple injections.

There were no systemic serious adverse events with 1 excep-tion: an 87-year-old woman developed atrial fibrillation. Thisparticipant in the multiple-dose arm received E10030 0.3 mg andreceived the first intravitreous injections of ranibizumab and

s (Occurring in >1 Patient)

Dose Group

8) 1.5 mg (n ¼ 5) 3.0 mg (n ¼ 4) All (n ¼ 23)

3 (60%) 4 (100%) 18 (78%)0 (0%) 2 (50%) 6 (26%)0 (0%) 0 (0%) 5 (22%)1 (20%) 1 (25%) 4 (17%)1 (20%) 1 (25%) 3 (13%)1 (20%) 0 (0%) 3 (13%)0 (0%) 1 (25%) 2 (9%)1 (20%) 0 (0%) 2 (9%)1 (20%) 1 (25%) 2 (9%)

acular degeneration in the fellow eye.

81

Page 5: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Ophthalmology Volume 123, Number 1, January 2016

E10030 on June 23, 2008. Subsequent injections were given perprotocol on July 21 and August 14, 2008, without incident. OnSeptember 17, 2008, atrial fibrillation developed and was judged tobe mild in severity and not related to study drug or injection pro-cedure. The atrial fibrillation was noted on a routine study elec-trocardiogram, and the patient was asymptomatic.

There were no significant changes in measured laboratoryvalues for the study population at the tested drug doses. As aresult of this overall safety profile and because it was notpossible to evaluate doses higher than 3.0 mg, as described inthe “Methods” section, a maximum tolerated dose was notidentified.

Figure 4. Percentage of study eyes that gained �15 letters at each timepoint. The percentages at each time point are indicated above each cor-responding bar on the graph.

Safety Biomarkers

Visual Acuity. There were no adverse VA safety signals iden-tified in the study group. There was a general trend toward a meanincrease in VA (number of ETDRS letters) at all time points,when compared with baseline, for both the overall population andall treatment groups. At week 12, there was a mean increase inVA of 14 letters (standard deviation, 11.3) from baseline. Themajority of participants gained VA at all time points during thestudy period. At week 12 (1 month after the final injection formost participants), 59% of participants gained �15 ETDRS letters(Figs 3 and 4).

Optical Coherence Tomography. Adverse safety signalswere not identified by OCT. Mean center point and central subfieldthickness decreased at all time points for all treatment groups. Theabsolute thickness changes from baseline are summarized inFigures 5 and 6. At the week 12 visit, 1 month after the lastscheduled drug administration, there was a mean decrease incenter point thickness and central subfield thickness of 38.9%and 33.7%, respectively (Fig 5, Table 2).

Fluorescein Angiography. One of the 22 participants inthe per-protocol dataset was allergic to fluorescein and had onlya baseline FA performed. Therefore, 21 participants provideddata for the FA analyses. All participants analyzed demonstratedregression in CNV area from baseline to week 12 (Figs 6 and 7).Mean reduction in CNV area was 85.5%. Five participants hadcomplete (100%) CNV regression. Fifteen of the 21 participants(71%) had >90% CNV area regression (Table 2).

Figure 3. Mean visual acuity change over time. The mean Early Treat-ment of Diabetic Retinopathy Study (ETDRS) letters seen by all partici-pants in the study group at each time point are indicated on the graph.N ¼ 22 at each time point.

82

Discussion

In the present investigation, escalating intravitreal doses ofan anti-PDGF pegylated aptamer E10030 (0.03, 0.3, 1.5,and 3.0 mg) were administered in combination with rani-bizumab, an anti-VEGF antibody fragment. This is the firstclinical trial to target PDGF and VEGF via intravitrealcombination therapy in treatment-naïve subjects withNVAMD. The experimental regimen was well tolerated. Nolocal or systemic adverse safety signals were observed afteradministration of the individual investigational drug or thecombination of active agents.

According to the study design, E10030 was first given inisolation, followed by ranibizumab at later time points. Theinitial dose of E10030 was given in isolation rather than incombination with ranibizumab to ensure that an adversereaction to Fovista monotherapy did not occur; this studywas the first human ocular exposure to the drug, and thisconservative approach was taken to ensure patient safety.Once it was relatively clear that E10030 monotherapy waswell tolerated, the clinical paradigm of same-day combina-tion was subsequently used. This latter approach reflects ourbelief that same-day or sequential combination therapy maybe the regimen used in future studies or the clinical setting.

Figure 5. Mean centerpoint and central subfield thickness, as determinedby optical coherence tomography, over time. The corresponding thicknessvalues are shown on the graph at each time point.

Page 6: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Figure 6. Reduction in choroidal neovascularization (CNV) area at week12 when compared with baseline. Data shown indicate the percentage ofresidual CNV area, when compared with baseline value, as measured byfluorescein angiography for each study participant.

Jaffe et al � E10030 and Ranibizumab for NVAMD

Adverse events specific to the intravitreal delivery ofaptamers are not known, to the best of our knowledge. Asexpected, no such adverse events were evident in this trial.In particular, local ocular adverse events, including intra-ocular inflammation, media opacification, retinal orchoroidal vasculopathy, or other evidence of retinal tissuetoxicity, were not seen. Transient changes known to beassociated with the intravitreal injection procedure itselfwere expected and observed (e.g., ocular irritation andsubconjunctival hemorrhage). Furthermore, no ocularE10030/ranibizumab drugedrug interaction side effectswere identified, and systemic adverse events due to theexperimental regimen were not seen. Increased drug vis-cosity at higher concentrations limited additional doseescalation. Therefore, 3.0 mg was the highest dose tested,and the maximum tolerated dose was not identified.

Theoretically, PDGF inhibition could result in strippingof pericytes associated with the normal host vasculature.Therefore, a resulting phenotype analogous to that seen indiabetic retinopathy is plausible.24 However, on meticulousfundus photograph and angiographic image evaluation at the3- and 6-month follow-up time points, no morphologic al-terations, such as microaneurysms suggestive of an incipientdiabetic retinopathy-like picture, were observed. Lack ofsuch vascular alterations is consistent with the preclinicalstudies of ocular and tumor angiogenesis in which normalhost pericytes are resistant to PDGF antagonism.18 The

Table 2. Safety Biomar

Dose Group (mg) N Baseline VA Mean VA

0.03 5 53.6 þ10.3 8 44.3 þ11.5 5 52.3 þ3.0 4 49.1 þ

CNV ¼ choroidal neovascularization; CST ¼ central subfield thickness; VA ¼

mechanisms related to this form of normal hostvasculature resistance are unknown. Pericytes share acommon basement membrane with the underlyingendothelial cells and have strong intercellular junctionalcomplexes.13 Therefore, an underlying mechanicalresistance to pericyte “stripping” may explain thesefindings. Alternatively, proliferating pericytes may expressPDGF receptors, which are loosely associated with theneovascular endothelial cells, whereas quiescent pericyteswith strong junctional processes to the underlyingendothelial cells on the host vasculature may not expressPDGF receptors.13 Such differential receptor expressionmay result in stripping of neovascular pericytes whilesparing the pericytes on normal host vessels. However, alonger duration of follow-up with chronic therapy isrequired to adequately assess the potential pericyte loss innormal host vasculature consequent to PDGF inhibition.

Pericytes function to stabilize vessels and help to inducea state of neovascular quiescence. Inhibition of pericytefunction theoretically could result in unwanted leakage byaltering the neovasculature barrier properties or inducingendothelial cell activation. This phenomenon was not seenin this study by imaging studies (OCT and FA). However,the potent antipermeability property of the co-administeredanti-VEGF agent may have modified or masked anyenhanced permeability alteration. Furthermore, this studywas of relatively short duration, and longer-term confirma-tory studies with E10030 are required to confirm thefavorable safety profile seen thus far.

Functional visual outcome data were obtained as part ofthe safety assessment (i.e., adverse effects on VA). Thisphase 1 study used a dose-escalation scheme resulting in asmall and imbalanced sample in each dose group (Table 1).The small sample size resulted in a lack of statistical powerto permit subgroup analysis, particularly in the presence of amarked variability in baseline factors that affect visualoutcomes and absence of a uniform regimen in the lowerdose groups in this heterogeneous disease. Therefore, thevisual outcome data and biomarkers were analyzed in apooled fashion. No safety signals related to the loss ofmean VA were identified in these pooled data.

Imaging biomarkers assessed by FA and OCT also didnot show any drug-related adverse effects. Center point andcentral subfield thickness on OCT decreased by 38.9% and33.7%, respectively, at week 12. Combining ranibizumabwith E10030 did not reduce this beneficial effect of anti-VEGF therapy.

Qualitative and quantitative analysis of FA data demon-strated regression in CNV area from baseline to week 12

kers by Dose Group

Change Mean CST Change % CNV Regression

3.4 �140.7 88.69.8 �183.3 84.19.0 �87.9 94.49.5 �100.9 75.3

visual acuity.

83

Page 7: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Figure 7. Case showing choroidal neovascularization (CNV) regression 12 weeks after combination antieplatelet-derived growth factor/antievascularendothelial growth factor therapy, associated with a 4.5 Early Treatment of Diabetic Retinopathy Study (ETDRS) letter gain.

Ophthalmology Volume 123, Number 1, January 2016

with an overall mean decrease of 85.5%. The combinationof CNV regression and mean VA increase of 14 letters frombaseline, and the finding that 59% of participants gained�15 letters at week 12 may indicate potential bioactivitywith the dual antagonism of PDGF and VEGF in NVAMD.However, great caution should be exercised in the inter-pretation of functional VA data given the small sample size,absence of a control group, relatively short-term follow-up,and high variability of VA measurement in eyes withNVAMD. However, choroidal neovascular membraneregression observed in this study stands in contrast to mostprospective studies of anti-VEGF monotherapy, in whichCNV size is not significantly reduced over time.10,25 Thisconcordance with preclinical studies demonstrating similarneovascular regression may signal the modification of 1mode of anti-VEGF resistance, namely, pericyte-mediatedendothelial cell protection.

Given the potential for pericyte-mediated anti-VEGFresistance in NVAMD, this phase 1 study was designed toaddress the safety of combined VEGF and PDGF inhi-bition in NVAMD. Therefore, it was logical to combinean anti-PDGF agent such as E10030 with an anti-VEGFagent. The anti-PDGF agent has the potential toactively strip pericytes and inhibit their function,rendering the CNV more susceptible to anti-VEGF ther-apy.18 E10030 injection in doses up to 3 mg was welltolerated without evidence of systemic or oculartoxicity in participants with NVAMD. The absence ofsafety signals, the VA benefit, and the potential tomodify the disease, thereby causing CNV regression inthe acute phase and reduced fibrosis in the chronic

84

phase, support the approach of combining E10030 andranibizumab and call for a prospective study with alarger sample size and longer follow-up. Additionalinvestigation will help build evidence for the concept ofdual VEGF and PDGF antagonism to improve visualoutcomes and address unmet need in NVAMD.

References

1. Klein R, Chou CF, Klein BE, et al. Prevalence of age-relatedmacular degeneration in the US population. Arch Ophthalmol2011;129:75–80.

2. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992;267:10931–4.

3. Comparison of Age-related Macular Degeneration TreatmentsTrials Research Group; Martin DF, Maguire MG, et al.Ranibizumab and bevacizumab for treatment of neovascularage-related macular degeneration: two-year results. Ophthal-mology 2012;119:1388–98.

4. Schmidt-Erfurth U, Kaiser PK, Korobelnik JF, et al. Intra-vitreal aflibercept injection for neovascular age-related maculardegeneration: ninety-six-week results of the VIEW studies.Ophthalmology 2014;121:193–201.

5. Busbee BG, Ho AC, Brown DM, et al. Twelve-month efficacyand safety of 0.5 mg or 2.0 mg ranibizumab in patients withsubfoveal neovascular age-related macular degeneration.Ophthalmology 2013;120:1046–56.

6. Holekamp NM, Liu Y, Yeh WS, et al. Clinical utilization ofanti-VEGF agents and disease monitoring in neovascular age-related macular degeneration. Am J Ophthalmol 2014;157:825–833.e1.

Page 8: A Phase 1 Study of Intravitreous E10030 in Combination ......A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration

Jaffe et al � E10030 and Ranibizumab for NVAMD

7. Rasmussen A, Bloch SB, Fuchs J, et al. A 4-year longitudinalstudy of 555 patients treated with ranibizumab for neovascularage-related macular degeneration. Ophthalmology 2013;120:2630–6.

8. Kang HM, Kwon HJ, Yi JH, et al. Subfoveal choroidalthickness as a potential predictor of visual outcome andtreatment response after intravitreal ranibizumab injections fortypical exudative age-related macular degeneration. Am JOphthalmol 2014;157:1013–21.

9. Sizmaz S, Kucukerdonmez C, Kal A, et al. Retinal andchoroidal thickness changes after single anti-VEGF injectionin neovascular age-related macular degeneration: ranibizumabvs bevacizumab. Eur J Ophthalmol 2014;24:904–10.

10. Brown DM, Michels M, Kaiser PK, et al. Ranibizumab versusverteporfin photodynamic therapy for neovascular age-relatedmacular degeneration: two-year results of the ANCHORstudy. Ophthalmology 2009;116:57–65.e5.

11. Humphreys BD, Lin SL, Kobayashi A, et al. Fate tracing re-veals the pericyte and not epithelial origin of myofibroblasts inkidney fibrosis. Am J Pathol 2010;176:85–97.

12. Lin SL, Kisseleva T, Brenner DA, Duffield JS. Pericytes andperivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am JPathol 2008;173:1617–27.

13. Benjamin LE, Hemo I, Keshet E. A plasticity window forblood vessel remodelling is defined by pericyte coverage of thepreformed endothelial network and is regulated by PDGF-Band VEGF. Development 1998;125:1591–8.

14. Hellstrom M, Kalen M, Lindahl P, et al. Role of PDGF-B andPDGFR-beta in recruitment of vascular smooth muscle cellsand pericytes during embryonic blood vessel formation in themouse. Development 1999;126:3047–55.

15. Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyteinteractions. Circ Res 2005;97:512–23.

16. Jain RK. Normalizing tumor vasculature with anti-angiogenictherapy: a new paradigm for combination therapy. Nat Med2001;7:987–9.

17. Bergers G, Song S, Meyer-Morse N, et al. Benefits of targetingboth pericytes and endothelial cells in the tumor vasculaturewith kinase inhibitors. J Clin Invest 2003;111:1287–95.

18. Jo N, Mailhos C, Ju M, et al. Inhibition of platelet-derivedgrowth factor B signaling enhances the efficacy of anti-vascular endothelial growth factor therapy in multiplemodels of ocular neovascularization. Am J Pathol 2006;168:2036–53.

19. Balasubramanian SA, Krishna Kumar K, Baird PN. The role ofproteases and inflammatory molecules in triggering neo-vascular age-related macular degeneration: basic science toclinical relevance. Transl Res 2014;164:179–92.

20. Andrae J, Gallini R, Betsholtz C. Role of platelet-derivedgrowth factors in physiology and medicine. Genes Dev2008;22:1276–312.

21. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte lossand microaneurysm formation in PDGF-B-deficient mice.Science 1997;277:242–5.

22. Erber R, Thurnher A, Katsen AD, et al. Combined inhibitionof VEGF and PDGF signaling enforces tumor vessel regres-sion by interfering with pericyte-mediated endothelial cellsurvival mechanisms. FASEB J 2004;18:338–40.

23. Akiyama H, Kachi S, Silva RL, et al. Intraocular injection ofan aptamer that binds PDGF-B: a potential treatment for pro-liferative retinopathies. J Cell Physiol 2006;207:407–12.

24. Hall AP. Review of the pericyte during angiogenesis and itsrole in cancer and diabetic retinopathy. Toxicol Pathol2006;34:763–75.

25. Brown DM, Kaiser PK, Michels M, et al. Ranibizumab versusverteporfin for neovascular age-related macular degeneration.N Engl J Med 2006;355:1432–44.

Footnotes and Financial Disclosures

Originally received: March 22, 2015.Final revision: September 4, 2015.Accepted: September 6, 2015.Available online: October 20, 2015. Manuscript no. 2015-470.1 Department of Ophthalmology, Duke University, Durham, NorthCarolina.2 Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston,Massachusetts.3 Palmetto Retina Center, Columbia, South Carolina.4 New Jersey Retina, Rutgers Medical School, Newark, New Jersey.5 Semmelweis University, Budapest, Hungary.6 Ophthotech, New York, New York.

Financial Disclosure(s):The authors have made the following disclosures: G.J.J.: Consultant eAlcon, Novartis, Neurotech, Roche, Heidelberg.

D.E.: Consultant e Alcon, Alimera, Allergan, Arctic, Bausch & Lomb,Biogen, Genentech, Ophthotech, Regeneron, Avalanche, FoundationFighting Blindness; Research funds e Ocata, Neurotech, Juvenile DiabetesResearch Foundation; Personal fees e Thrombogenics; Acucela ScientificAdvisory Board.

J.A.W.: Grants e Ophthotech, Regeneron, Genentech, Iconic Pharmaceu-ticals, LPath, Allergan, Alcon, PanOptica; Consultant e Iconic Pharma-ceuticals, PanOptica.

J.L.P.: Consultant and shareholder e Ophthotech and PanOptica;Consultant e Regeneron.

A.P.: Consultant e Bayer, Novartis.

S.P.: Cofounded and serves on the Ophthotech Board of Directors, and hasfiled patent applications related to E10030.

Ophthotech (New York, NY) funded the study reported in this article.Ophthotech participated in the design of the study, conducting the study,data collection, data management, data analysis, and interpretation of thedata. S.P., an Ophthotech employee, reviewed the article.

Author Contributions:

Conception and design: Patel

Data collection: Wells, Prenner, Papp, Patel

Analysis and interpretation: Jaffe, Eliott, Wells, Prenner, Papp, Patel

Obtained funding: Not applicable

Overall responsibility: Jaffe, Eliott, Wells, Prenner, Papp, Patel

Abbreviations and Acronyms:CNV ¼ choroidal neovascularization; ETDRS ¼ Early Treatment ofDiabetic Retinopathy Study; FA ¼ fluorescein angiography;IOP ¼ intraocular pressure; NVAMD ¼ neovascular age-related maculardegeneration; OCT ¼ optical coherence tomography; PDGF ¼ platelet-derived growth factor; VA ¼ visual acuity; VEGF ¼ vascular endothelialgrowth factor.

Correspondence:Glenn J. Jaffe, MD, Department of Ophthalmology, Duke University EyeCenter, Box 3802, Durham, NC 27710. E-mail: [email protected].

85