throbber
A Phase IIIb Study to Evaluate the Safety
`of Ranibizumab in Subjects with
`Neovascular Age-related Macular
`Degeneration
`
`David S. Boyer, MD,1 Jeffrey S. Heier, MD,2 David M. Brown, MD,3 Steven F. Francom, PhD,4
`Tsontcho Ianchulev, MD,4 Roman G. Rubio, MD4
`
`Objective: To evaluate the safety and efficacy of intravitreal ranibizumab in a large population of subjects
`with neovascular age-related macular degeneration (AMD).
`Design: Twelve-month randomized (cohort 1) or open-label (cohort 2) multicenter clinical trial.
`Participants: A total of 4300 subjects with angiographically determined subfoveal choroidal neovascular-
`ization (CNV) secondary to AMD.
`Methods: Cohort 1 subjects were randomized 1:1 to receive 0.3 mg (n ⫽ 1169) or 0.5 mg (n ⫽ 1209)
`intravitreal ranibizumab for 3 monthly loading doses. Dose groups were stratified by AMD treatment history
`(treatment-naïve vs. previously treated). Cohort 1 subjects were retreated on the basis of optical coherence
`tomography (OCT) or visual acuity (VA) criteria. Cohort 2 subjects (n ⫽ 1922) received an initial intravitreal dose
`of 0.5 mg ranibizumab and were retreated at physician discretion. Safety was evaluated at all visits.
`Main Outcome Measures: Safety outcomes included the incidence of ocular and nonocular adverse
`events (AEs) and serious adverse events (SAEs). Efficacy outcomes included changes in best-corrected VA
`over time.
`Results: Some 81.7% of cohort 1 subjects and 49.9% of cohort 2 subjects completed the 12-month study.
`The average total number of ranibizumab injections was 4.9 for cohort 1 and 3.6 for cohort 2. The incidence of
`vascular and nonvascular deaths during the 12-month study was 0.9% and 0.7% in the cohort 1 0.3 mg group,
`0.8% and 1.5% in the cohort 1 0.5 mg group, and 0.7% and 0.9% in cohort 2, respectively. The incidence of
`death due to unknown cause was 0.1% in both cohort 1 dose groups and cohort 2. The number of vascular
`deaths and deaths due to unknown cause did not differ across cohorts or dose groups. Stroke rates were 0.7%,
`1.2%, and 0.6% in the 0.3 mg and 0.5 mg groups and cohort 2, respectively. At month 12, cohort 1 treatment-
`naïve subjects had gained an average of 0.5 (0.3 mg) and 2.3 (0.5 mg) VA letters and previously treated subjects
`had gained 1.7 (0.3 mg) and 2.3 (0.5 mg) VA letters.
`Conclusions:
`Intravitreal ranibizumab was safe and well tolerated in a large population of subjects with
`neovascular AMD. Ranibizumab had a beneficial effect on VA. Future investigations will seek to establish optimal
`dosing regimens for persons with neovascular AMD.
`Financial Disclosure(s): Proprietary or commercial disclosure may be found after the references. Ophthalmology
`2009;116:1731–1739 © 2009 by the American Academy of Ophthalmology.
`
`Neovascular age-related macular degeneration (AMD) is
`characterized by new vessel growth and leakage in the
`choroidal vascular network beneath the macula, with
`extension and leakage into the subretinal space. Although
`the pathologic events that precede choroidal neovascu-
`larization (CNV) are not clearly understood, disrupting
`the activity of vascular endothelial growth factor A
`(VEGF-A), a diffusible cytokine that promotes angiogen-
`esis and vascular permeability, effectively treats CNV
`secondary to AMD.
`Ranibizumab (LUCENTIS, Genentech, Inc., South San
`Francisco, CA) is a recombinant, humanized monoclonal
`antibody antigen-binding fragment (Fab) that neutralizes all
`active forms of VEGF-A. In 2 pivotal phase III trials—
`
`Minimally Classic/Occult Trial of the Anti-VEGF Antibody
`Ranibizumab in the Treatment of Neovascular Age-Related
`Macular Degeneration (MARINA)1 and Anti-Vascular
`Endothelial Growth Factor (VEGF) Antibody for the
`Treatment of Predominantly Classic Choroidal Neovas-
`cularization (CNV) in Age-related Macular Degeneration
`(ANCHOR)2—monthly intravitreal injections of 0.3 mg or
`0.5 mg ranibizumab not only prevented vision loss but also
`improved visual acuity (VA) in patients with minimally
`classic or occult without classic and predominantly classic
`CNV, respectively. In those studies, ranibizumab treatment
`was associated with a low rate of serious adverse events
`(SAEs), including those attributable to systemic VEGF
`inhibition.
`
`© 2009 by the American Academy of Ophthalmology
`Published by Elsevier Inc.
`
`ISSN 0161-6420/09/$–see front matter
`doi:10.1016/j.ophtha.2009.05.024
`
`1731
`
`Regeneron Exhibit 2032
`Page 01 of 09
`
`

`

`Ophthalmology Volume 116, Number 9, September 2009
`
`The Safety Assessment of Intravitreous Lucentis for
`AMD (SAILOR) study was a phase IIIb follow-up study
`to the MARINA and ANCHOR studies to evaluate the
`long-term safety and efficacy of ranibizumab in a large
`population of subjects with all subtypes (minimally clas-
`sic, occult without classic, and predominantly classic) of
`neovascular AMD. SAILOR included more than 5 times
`as many ranibizumab-treated subjects as the MARINA
`and ANCHOR studies combined. Thus, it is the largest
`multicenter randomized study to date to evaluate safety
`and efficacy outcomes of anti-VEGF treatment in wet
`AMD, and it
`is the only phase III study to examine
`individualized, criteria-based retreatment.
`
`Materials and Methods
`
`SAILOR was a 12-month, multicenter, phase IIIb study intended
`to further characterize the safety and efficacy profiles of intravitreal
`ranibizumab. Protocols were approved by the institutional review
`board at each study site, and the study was conducted according to the
`International Conference on Harmonisation E6 Guideline for
`Good Clinical Practice and any national requirements. All sub-
`jects provided informed consent before participation in the
`study. The SAILOR study is registered at www.clinicaltrials.
`gov (NCT00251459; accessed February 5, 2009).
`Two study cohorts were enrolled. Cohort 1 subjects were
`randomized 1:1 to receive 0.3 mg or 0.5 mg intravitreal ranibi-
`zumab. Cohort 2 subjects received open-label 0.5 mg intravitreal
`ranibizumab. Eligible subjects were ⱖ50 years of age with 20/40
`to 20/400 (Snellen equivalent) best-corrected VA in the study eye.
`Cohort 1 VA was assessed with the Early Treatment Diabetic
`Retinopathy Study (ETDRS) chart. In the interest of conserving
`time and resources, VA for cohort 2 (under a less rigorous treat-
`ment and assessment schedule) was assessed using Snellen charts.
`All subjects had angiographically determined subfoveal CNV
`(minimally classic, occult without classic, predominantly classic)
`secondary to AMD (as determined by the investigating physician),
`with evidence of recent disease progression defined by any of the
`following: loss of ⱖ5 ETDRS letters (or ⱖ1 Snellen line) within
`6 months before study initiation (i.e., day 0); 10% increase in the
`CNV lesion area determined by comparing a fluorescein angio-
`gram performed within 1 month before day 0 with an angiogram
`performed within 6 months before day 0; subretinal hemorrhage
`associated with CNV within 1 month before day 0; or classic CNV
`comprising ⬎50% of the CNV lesion area.
`Key exclusion criteria included verteporfin photodynamic ther-
`apy, pegaptanib sodium, or other AMD therapy within 30 days
`before day 0; previous submacular surgery or other surgical inter-
`vention for AMD in the study eye; participation in an investiga-
`tional drug (except vitamins and minerals) study within 30 days
`before day 0; previous participation in a ranibizumab clinical trial;
`intravitreal administration of bevacizumab within 30 days before
`day 0; or current use of systemic anti-VEGF agents. Also excluded
`were subjects with fibrosis or atrophy involving the foveal center
`of the treated eye in the absence of a new lesion; CNV in either eye
`due to other causes, such as ocular histoplasmosis, trauma, or
`pathologic myopia; a tear in the retinal pigment epithelium of the
`study eye involving the macula; or any current intraocular condi-
`tion in the study eye (e.g., cataract or diabetic retinopathy) that, in
`the investigating physician’s opinion, would require medical or
`surgical intervention during the 12-month study period or, if al-
`lowed to progress untreated, would likely contribute to the loss of
`at least 2 Snellen equivalent lines of VA over the 12-month study
`
`1732
`
`Figure 1. Study treatment and assessments. Cohort 1 subjects received 3
`loading doses of ranibizumab and were retreated on the basis of VA (⬎5
`letter decrease in VA from highest score at prior visits) or VA and/or OCT
`(⬎100 ␮m increase in CFT from the lowest measurement at prior visits)
`criteria. Cohort 2 subjects received 1 dose of ranibizumab on day 1 and
`were retreated at physician discretion. CFT ⫽ central foveal thickness;
`OCT ⫽ optical coherence tomography; VA ⫽ visual acuity.
`
`period. Subjects with a history of cardiovascular disease were not
`excluded if their disease was controlled.
`Cohort 1 subjects were randomized 1:1 to receive 0.3 mg or 0.5
`mg intravitreal ranibizumab. To prevent bias in reporting AEs,
`subjects were masked to treatment dose. (Because SAILOR was
`not designed with efficacy as an objective, physicians and study
`monitors were not masked.) Randomization was stratified accord-
`ing to treatment history. “Previously treated” subjects had previ-
`ously received treatment AMD. “Treatment-naïve” subjects were
`newly diagnosed with neovascular AMD. Cohort 1 subjects
`received 3 monthly loading doses of intravitreal ranibizumab (day
`0, month 1, and month 2) with scheduled follow-up visits at
`months 3, 6, 9, and 12 (Fig 1). If, at any time, the investigating
`physician believed that the between-visit interval was too long for
`a patient to go without being assessed, an unscheduled visit could
`occur. After the 3 loading doses, retreatment was based on (1) VA
`(a ⬎5 ETDRS letter decrease in VA compared with the highest
`VA score at any prior scheduled visit) or (2) VA (same as above)
`and/or optical coherence tomography (OCT) (a ⬎100-␮m increase
`in central foveal thickness [CFT] compared with the lowest mea-
`surement at any previous scheduled study visit, with intraretinal or
`subretinal fluid present). Thus, OCT assessment was required only
`for retreatment option 2, in which case OCT data were consistently
`obtained at all study visits. Retreatment was to occur no more
`frequently than every 30 days. Before randomization, the investi-
`gating physician selected the retreatment criterion for each subject
`that was to be used throughout the study.
`Cohort 1 subjects were evaluated with a full ocular exami-
`nation and best-corrected VA (ETDRS chart at a distance of
`4 m) and safety assessments on day 0 and at all scheduled
`(months 1, 2, 3, 6, 9, and 12) visits. Visual acuity assessments
`were required at unscheduled visits if a subject was being
`evaluated for retreatment. Safety assessments were required at
`all unscheduled visits.
`Cohort 2 included both previously treated and treatment-
`naïve subjects. Subjects received 0.5 mg of ranibizumab, with
`an initial injection on day 0 and retreatment at the investigating
`physician’s discretion, no more frequently than every 30 days.
`Cohort 2 subjects were evaluated for Snellen VA at day 0 and
`months 6 and 12. At unscheduled visits, VA was assessed at the
`investigating physician’s discretion. Serious adverse events and
`adverse events (AEs) were assessed at scheduled and unsched-
`uled visits, with formal safety assessments scheduled for
`months 6 and 12.
`
`Regeneron Exhibit 2032
`Page 02 of 09
`
`

`

`Boyer et al
`
`䡠 Ranibizumab Safety in Wet AMD
`
`Adverse events included any unfavorable or unintended sign,
`symptom, or disease temporally associated with use of study drug
`or other protocol-imposed intervention. An AE was classified as an
`SAE if it caused or led to death, required or prolonged subject
`hospitalization, resulted in persistent or significant disability or
`incapacitation, or was considered to be a significant medical event
`by the investigating physician.
`One eye per subject (i.e., the study eye) was treated. After
`thoroughly cleansing the lid, lashes, periorbital area, and conjunc-
`tiva with povidone iodine, local anesthesia and antimicrobials
`(ofloxacin ophthalmic solution, trimethoprim-polymyxin B oph-
`thalmic solution, moxifloxacin ophthalmic solution, or gatifloxacin
`ophthalmic solution) were administered to the study eye. A 30-
`gauge, 0.5-inch needle attached to a low-volume syringe contain-
`ing 50 ␮L of ranibizumab solution was inserted through the
`conjunctiva and sclera, 3.5 to 4.0 mm posterior to the limbus,
`avoiding the horizontal meridian and aiming toward the center of
`the globe. The injection volume was delivered slowly. The needle
`was slowly removed, ensuring that all drug solution was in the eye.
`Immediately after the injection, antimicrobial drops were admin-
`istered, and the subject was instructed to self-administer antimi-
`crobial drops 4 times daily for 3 days. The study eye was assessed
`with a finger count test and intraocular pressure within 15 and 70
`minutes, respectively, of the ranibizumab injection.
`The primary safety end point for cohort 1 was incidence of
`ocular and nonocular SAEs evaluated through month 12. A sec-
`ondary safety end point was incidence of ocular and nonocular
`AEs evaluated through month 12. Efficacy end points for cohort 1
`included change from baseline VA, proportion of subjects who
`gained ⱖ15 VA letters from baseline, and change from baseline
`CFT across the study period.
`The primary safety end points for cohort 2 were the inci-
`dence of ocular and nonocular SAEs and AEs evaluated through
`month 12. Efficacy outcomes for cohort 2 included median
`change in Snellen VA from baseline and the proportion of
`subjects with Snellen 20/200 or worse at baseline compared
`with months 6 and 12.
`
`Statistical Analysis
`
`Safety and efficacy analyses included all subjects who received at
`least 1 injection of ranibizumab. Incidence of ocular and nonocular
`SAEs and AEs and 95% 2-sided confidence intervals for key SAEs
`were determined for both cohorts and each dose group. No formal
`hypothesis testing was conducted to compare cohorts, dose groups,
`or treatment-naïve and previously treated subjects. A sample of
`2378 cohort 1 subjects and 1922 cohort 2 subjects was considered
`sufficient to estimate rates of uncommon SAEs and AEs.
`Efficacy results for cohort 1 were stratified by dose group and
`treatment history. Estimated proportions were obtained for dichot-
`omous end points. Continuous end points were evaluated using
`descriptive statistics, including mean, median, standard deviation,
`standard error, and range.
`To further evaluate stroke rates across cohorts and dose groups,
`each subject’s medical history was reviewed, and subjects were
`classified by preexisting conditions that may have been associated
`with the incidence of stroke during the 12-month study. These
`included prior stroke, myocardial infarction (MI), hypertension,
`transient ischemic attack, coronary artery disease, arrhythmias,
`valve malfunction, congestive heart failure, angioplasty, deep vein
`thrombosis, diabetes, endocardectomy, cardiac inflammation, prior
`stent, and use of aspirin, lipid-lowering drugs, anticoagulants, or
`platelet aggregation inhibitors. A univariate Cox proportional haz-
`ard regression model was used to identify which of those were
`significant (i.e., Pⱕ0.05) risk factors for stroke in SAILOR. In
`
`addition, models that included the interaction of dose with each of
`the significant risk factors were fit separately.
`
`Missing Data
`Missing data were not imputed for safety end points. For cohort 1,
`missing values for efficacy end points were imputed using the
`last-observation-carried-forward method. For cohort 2, missing
`Snellen values were not imputed.
`
`Results
`
`From November 2005 to June 30, 2006 (when ranibizumab was
`approved for the treatment of neovascular AMD by the Food and
`Drug Administration), 2378 cohort 1 subjects were randomly
`assigned to receive 0.3 mg (n ⫽ 1169) or 0.5 mg (n ⫽ 1209)
`intravitreal ranibizumab at 105 US centers. Cohort 1 subjects had
`an average age of 79 years, and 59% were female (Table 1).
`Approximately 60% of cohort 1 subjects in each dose group had
`been previously treated for AMD. The types of previous treatment
`were similar across dose groups and included photodynamic ther-
`apy (33%), intravitreal pegaptanib sodium (30%), intravitreal tri-
`amcinolone acetonide (17%), and laser photocoagulation (10%).
`Investigating physicians elected to use the VA plus OCT retreat-
`ment criterion for approximately 81% of the subjects in each dose
`group.
`Previously treated and treatment-naïve subjects had similar
`baseline ocular characteristics, with the exception that previously
`treated subjects had a longer time since first diagnosis and lower
`baseline VA (Table 2). Approximately 18% of cohort 1 subjects in
`each dose group discontinued the study before the month 12 visit
`(Table 3). Baseline ocular characteristics of subjects who com-
`
`Table 1. Subject Baseline Characteristics
`
`Characteristic
`
`Age (yrs)
`Mean ⫾ SD
`Range
`Sex
`Female
`Race
`Caucasian
`AMD treatment
`history
`Treatment naïve
`Previously treated
`Retreatment criteria
`VA
`VA plus OCT
`Systolic BP
`Mean ⫾ SD
`Range
`Diastolic BP
`Mean ⫾ SD
`Range
`
`Cohort 1
`
`0.3 mg
`(n ⫽ 1169)
`
`0.5 mg
`(n ⫽ 1209)
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`78.7⫾7.6
`51–97
`
`78.7⫾8.6
`52–101
`
`78.7⫾8.1
`45–99
`
`59.9
`
`96.6
`
`39.5
`60.5
`
`19.3
`80.7
`
`58.1
`
`97.1
`
`40.5
`59.5
`
`18.4
`81.6
`
`137.4⫾17.3
`90–213
`
`137.8⫾18.0
`80–220
`
`76.2⫾9.7
`48–118
`
`77.0⫾9.7
`48–110
`
`61.6
`
`96.2
`
`—
`—
`
`—
`—
`
`—
`—
`
`—
`—
`
`AMD ⫽ age-related macular degeneration; BP ⫽ blood pressure; OCT ⫽
`optical coherence tomography; SD ⫽ standard deviation; VA ⫽ visual
`acuity.
`Values are percentages except where otherwise noted.
`
`1733
`
`Regeneron Exhibit 2032
`Page 03 of 09
`
`

`

`Ophthalmology Volume 116, Number 9, September 2009
`
`Table 2. Baseline Ocular Characteristics
`
`Treatment Naive
`
`Cohort 1
`
`0.3 mg
`(n ⫽ 462)
`79.9⫾7.9
`0.3⫾1.4
`
`32.0
`19.7
`45.5
`
`55.0⫾12.5
`
`20/80
`12.2
`312⫾104
`15.3⫾3.2
`
`0.5 mg
`(n ⫽ 490)
`75.8⫾8.0
`0.3⫾0.7
`
`29.4
`20.2
`48.6
`
`48.9⫾13.8
`
`20/80
`15.0
`322⫾116
`15.3⫾3.2
`
`Previously Treated
`0.3 mg
`0.5 mg
`(n ⫽ 707)
`(n ⫽ 719)
`79.9⫾7.5
`79.9⫾7.5
`1.4⫾2.0
`1.3⫾1.7
`
`30.6
`26.2
`38.6
`
`53.8⫾13.8
`
`20/100
`22.9
`315⫾113
`15.7⫾3.3
`
`31.7
`23.5
`40.6
`
`50.0⫾14.3
`
`20/100
`23.0
`310⫾113
`15.4⫾3.4
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`—
`—
`
`—
`—
`—
`
`—
`
`20/100
`39
`—
`—
`
`Age at diagnosis (yrs)
`Time since diagnosis (yrs)
`CNV type (%)
`Predominantly classic
`Minimally classic
`Occult without classic
`VA
`ETDRS letters
`Snellen
`Median
`20/200 or worse (%)
`Central foveal thickness (␮m)
`Intraocular pressure (mmHg)
`
`CNV ⫽ choroidal neovascularization; ETDRS ⫽ Early Treatment Diabetic Retinopathy Study; VA ⫽ visual acuity.
`Values are mean ⫾ standard deviation except where otherwise noted.
`
`pleted the study and those who discontinued were similar. All
`cohort 1 subjects received their assigned dose of ranibizumab on
`day 0, and approximately 96% of cohort 1 subjects received their
`assigned dose at months 1 and 2 (Fig 2). Cohort 1 subjects received
`an average of 4.6 injections during the 12-month study (the pro-
`tocol required 3 initial injections). The average number of visits
`was 8.8 (the protocol required 7 scheduled visits). During months
`that visits were not scheduled (i.e., months 4, 5, 7, 8, 10, and 11),
`approximately 40% of the subjects made unscheduled visits, and
`approximately 16% of those subjects received an injection of
`ranibizumab at the unscheduled visit (relative to the number of
`subjects remaining in the study that month) (Fig 2).
`From March 2006 to June 30, 2006, 1922 cohort 2 subjects
`were enrolled at 104 US centers and received 0.5 mg intravitreal
`ranibizumab (Table 1). Approximately 50% of cohort 2 subjects
`discontinued the study before the month 12 visit (Table 3). All
`cohort 2 subjects received the protocol-required injection on day 0
`and received an average of 3.6 injections during the 12-month
`study (the protocol required 1 injection). The average number of
`visits for cohort 2 subjects was 4.9 (the protocol required 3
`scheduled visits). During months that visits were not required
`
`Table 3. Reasons for Discontinuation
`
`Cohort 1
`0.3 mg
`0.5 mg
`(n ⫽ 1169)
`(n ⫽ 1209)
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`18.6
`
`18.0
`
`1.7
`2.6
`0.7
`6.7
`3.4
`0.2
`0.6
`2.7
`
`0.1
`
`2.3
`2.2
`0.9
`5.8
`2.8
`0.1
`0.9
`3.1
`
`0
`
`50.1
`
`1.5
`1.8
`2.0
`29.0
`9.4
`0.3
`0.9
`5.3
`
`0
`
`Discontinued early (%)
`Reason for early
`discontinuation (%)
`Death
`Adverse event
`Loss to follow-up
`Subject decision
`Physician decision
`Sponsor decision
`Subject noncompliance
`Subject’s condition mandated
`other therapeutic
`intervention
`Reason not provided
`
`1734
`
`(i.e., all but months 6 and 12), the percentage of subjects who
`remained in the study that made unscheduled visits ranged from
`65% at month 2 to 17.4% at month 11. The percentage of
`subjects receiving injections ranged from 64% at month 2 to
`16.5% at month 11.
`
`Figure 2. Visits and treatment. The percentage of cohort 1 (upper) and
`cohort 2 (lower) patients making visits and receiving ranibizumab treat-
`ment during each month of the 12-month study are shown. Cohort 1 visits
`were scheduled for day 0 and months 1, 2, 3, 6, 9, and 12. Cohort 2 visits
`were scheduled for day 0 and months 6 and 12. Data from cohort 1 0.3 and
`0.5 mg dose groups are combined. Values are based on the percentage of
`subjects remaining in the study at each time point. Treatment received at
`month 12 was in violation of the protocol.
`
`Regeneron Exhibit 2032
`Page 04 of 09
`
`

`

`Boyer et al
`
`䡠 Ranibizumab Safety in Wet AMD
`
`Table 4. Key Ocular Serious Adverse Events
`
`Event, %
`
`Presumed endophthalmitis*
`Uveitis
`Retinal detachment
`Retinal tear
`Retinal hemorrhage
`Detachment of retinal
`pigment epithelium
`Vitreous hemorrhage
`Cataract
`
`Cohort 1
`0.3 mg
`0.5 mg
`(n ⫽ 1169)
`(n ⫽ 1209)
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`0.2
`0.1
`0.1
`0
`0.9
`0
`
`0.3
`0.1
`
`0.4
`0.2
`0
`0.1
`0.9
`0.2
`
`0.1
`0.1
`
`0.1
`0
`0.1
`0
`0.3
`0.1
`
`0.2
`0.1
`
`*Includes 2 cases of uveitis and 1 case of iridocyclitis that were treated
`with antibiotics.
`
`Safety
`
`Ocular safety. The rates of individual key ocular SAEs in cohort
`1 were ⬍1% and similar across dose groups (Table 4). Two
`subjects (0.2%) in the 0.3 mg group and 5 subjects (0.4%) in the
`0.5 mg group developed endophthalmitis or presumed endoph-
`thalmitis (i.e., ocular infection treated with antibiotics). One sub-
`ject in each cohort 1 dose group had a serious cataract event. The
`rates of individual key ocular SAEs in cohort 2 were ⬍1%. One
`cohort 2 subject developed endophthalmitis, and 1 subject had a
`serious cataract event (Table 4).
`The incidence of ocular inflammation AEs, including iritis,
`uveitis, vitritis, and iridocyclitis, was 1.0% in the 0.3 mg group,
`1.5% in the 0.5 mg group, and 0.5% in cohort 2. The overall
`incidence of cataract AEs was 5.4% in the 0.3 mg group, 6.0%
`in the 0.5 mg group, and 2.8% in cohort 2, and was similar when
`broken down by nuclear, subcapsular, and cortical subtypes.
`Nonocular safety. The rates of key nonocular SAEs were sim-
`ilar across cohort 1 dose groups (Fig 3; Table 5). Nonvascular
`death, stroke, and hemorrhage rates were numerically higher in the
`0.5 mg group. Eight subjects (0.7%) in the 0.3 mg group and 15
`subjects (1.2%) in the 0.5 mg group had a stroke during the
`
`Figure 3. Key nonocular SAEs. The rates of individual events are de-
`picted as point estimates with 2-sided Blyth-Still-Casella 95% confidence
`intervals. Antiplatelet Trialists’ Collaboration ATEs include vascular
`deaths and deaths due to unknown cause, nonfatal MI, and nonfatal
`stroke. APTC ⫽ Antiplatelet Trialists’ Collaboration; ATE ⫽ arterial
`thromboembolic event; SAE ⫽ serious adverse events.
`
`Table 5. Nonocular Adverse Events Potentially Related to
`Anti-Vascular Endothelial Growth Factor Therapy
`
`Classification, %
`
`Arterial thromboembolic
`events
`All
`Serious
`Hypertension
`All
`Serious
`Nonocular hemorrhage
`All
`Serious
`Proteinuria
`All
`Serious
`Other
`All
`Serious
`
`Cohort 1
`0.3 mg
`0.5 mg
`(n ⫽ 1169)
`(n ⫽ 1209)
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`3.8
`2.5
`
`9.0
`0.1
`
`2.9
`0.9
`
`0.1
`0
`
`0.7
`0.3
`
`4.1
`3.1
`
`10.3
`0.1
`
`3.1
`1.5
`
`0
`0
`
`0.4
`0.2
`
`2.4
`1.6
`
`3.0
`0
`
`1.4
`0.6
`
`0
`0
`
`0.1
`0.1
`
`VEGF ⫽ vascular endothelial growth factor.
`
`12-month study period. The incidence of MI and Antiplatelet
`Trialists’ Collaboration (APTC)3 arterial thromboembolic events
`(ATEs), which include vascular death and death of unknown
`cause, nonfatal MI, and nonfatal cardiovascular accidents, were
`similar across cohort 1 dose groups.
`Rates of key nonocular SAEs in cohort 2 were generally
`lower than those in cohort 1, which may be a result of under-
`reporting because of the large number of cohort 2 subjects who
`discontinued. The incidence of nonocular AEs potentially re-
`lated to anti-VEGF therapy was low and comparable across
`cohorts and dose groups.
`Prior stroke, history of arrhythmias, and history of congestive
`heart failure were significant risk factors for stroke (Fig 4). Al-
`though the numbers were small, there was a nonstatistically sig-
`nificant trend toward higher incidence of stroke in the cohort 1 0.5
`mg group subjects with a history of stroke. Seven of the 73
`subjects (9.6%) with a history of stroke in the 0.5 mg group
`experienced a stroke during the study compared with 2 of the 73
`subjects (2.7%) with a history of stroke in the 0.3 mg group. None
`of the cohort 2 subjects with a history of stroke experienced a
`stroke during the study (Fig 4).
`Twenty subjects (1.7%) in the cohort 1 0.3 mg group, 29
`subjects (2.4%) in the cohort 1 0.5 mg group, and 33 subjects
`(1.7%) in cohort 2 died during the 12-month study (Table 6). The
`number of vascular deaths and deaths due to unknown cause did
`not differ across cohorts or dose groups.
`
`Efficacy
`Cohort 1 efficacy results were stratified by dose and previous
`treatment for AMD. For all groups, study eye VA increased with
`3 loading doses of ranibizumab (day 0, month 1, month 3) (Fig 5).
`At month 3, treatment-naïve subjects in the 0.3 mg group had
`gained an average of 5.8 VA letters and those in the 0.5 mg group
`had gained an average of 7.0 VA letters. From months 3 to 12, with
`protocol-defined retreatment, VA tended to decrease. At month 12,
`treatment-naïve subjects in the 0.3 mg group had gained an aver-
`age of 0.5 VA letters and those in the 0.5 mg group had gained an
`average of 2.3 letters. A similar pattern was observed for previ-
`
`1735
`
`Regeneron Exhibit 2032
`Page 05 of 09
`
`

`

`Ophthalmology Volume 116, Number 9, September 2009
`
`ously treated subjects. At month 3, previously treated subjects in
`the 0.3 mg group had gained an average of 4.6 VA letters and those
`in the 0.5 mg group had gained an average of 5.8 VA letters. At
`month 12, previously treated subjects in the 0.3 mg group had
`gained an average of 1.7 VA letters and those in the 0.5 mg group
`had gained an average of 2.3 letters.
`In all cohort 1 groups, the proportion of subjects who gained
`ⱖ15 letters from baseline VA increased with 3 loading doses of
`ranibizumab (Fig 6). At month 3, 19.4% of treatment-naïve sub-
`jects in the 0.3 mg group and 20.1% in 0.5 mg group had gained
`ⱖ15 letters. The proportion of those who gained ⱖ15 letters
`tended to be maintained for the duration of the 12-month study,
`with 14.6% of 0.3 mg group subjects and 19.3% of 0.5 mg subjects
`gaining ⱖ15 VA letters at month 12. A similar pattern was
`observed for previously treated subjects. At month 3, 16.0% of
`previously treated subjects in the 0.3 mg group and 18.6% in the
`0.5 mg group had gained ⱖ15 letters; and at month 12, 15.8% of
`0.3 mg group subjects and 16.5% of 0.5 mg group subjects had
`gained ⱖ15 VA letters.
`Study eye CFT of cohort 1 subjects for whom OCT data
`were available decreased with 3 loading doses of ranibizumab,
`increased from months 3 to 6, and remained stable from months
`6 to 12 (Fig 7). For treatment-naïve subjects, CFT had de-
`creased an average of 107.0 ␮m in the 0.3 mg group and 122.0
`␮m in the 0.5 mg group at month 3. At month 12, the average
`decrease from baseline CFT was 72.0 ␮m in the 0.3 mg group
`and 92.0 ␮m in the 0.5 mg group. For previously treated
`subjects, CFT had decreased an average of 98.0 ␮m in the 0.3
`mg group and 108.0 ␮m in the 0.5 mg group at month 3. At
`month 12, the average decrease from baseline CFT was 71.0
`␮m in the 0.3 mg group and 76.0 ␮m in the 0.5 mg group.
`Because of the large number of cohort 2 subjects who discon-
`tinued, the last-observation-carried-forward method was not used
`to impute missing efficacy values, and observed results are re-
`ported. This should be kept in mind when interpreting the results.
`
`Table 6. Cause of Death
`
`Cohort 1
`0.3 mg
`0.5 mg
`(n ⫽ 1169)
`(n ⫽ 1209)
`
`Cohort 2
`0.5 mg
`(n ⫽ 1922)
`
`All deaths, %
`Deaths due to unknown
`cause, %
`Vascular deaths, %
`Cardiovasculara
`Strokeb
`Nonvascular deaths, %
`Respiratory: pneumonia,
`respiratory failure
`pulmonary failure
`pulmonary edema
`Accident, injury, intracranial
`bleed secondary to fall
`Renal failure
`Cancer
`Infection (septic shock,
`sepsis, urosepsis), liver
`failure due to hepatitis
`Postoperative bowel
`obstruction
`Vasculitis
`
`1.7
`0.1
`
`0.9
`0.8
`0.2
`0.7
`0.3
`
`0
`
`0
`0
`0.3
`
`0.1
`
`0
`
`2.4
`0.1
`
`0.8
`0.5
`0.3
`1.5
`0.6
`
`0.2
`
`0.1
`0.4
`0.2
`
`0
`
`0
`
`1.7
`0.1
`
`0.7
`0.7
`0.1
`0.9
`0.5
`
`0.1
`
`0
`0.3
`0
`
`0
`
`0.1
`
`aIncludes ischemic cardiomyopathy, coronary heart disease, cardiac arrest,
`MI, saddle pulmonary embolism, and heart failure.
`bIncludes stroke, acute ischemic stroke, intracerebral hemorrhage, cere-
`brovascular disease, and brain hemorrhage secondary to fall. Three 0.5 mg
`subjects with preexisting cancer had previously received cancer treatment.
`
`Snellen VA in cohort 2 subjects improved from a median of
`20/100 at baseline to 20/80 at months 6 and 12. The proportion of
`subjects with a Snellen equivalent of 20/200 or worse decreased
`from approximately 39% at baseline to 31% at month 6 and 32%
`at month 12.
`
`Discussion
`
`SAILOR is the largest study to date to evaluate safety
`(primary objective) and efficacy (secondary objective) of
`
`Figure 4. Stroke rate by risk factor. Point estimates and 2-sided Blyth-
`Still-Casella 95% confidence intervals for stroke rate when the risk factor
`was present or absent are shown. We evaluated the impact of 21 factors on
`the incidence of stroke. The 5 risk factors that had the greatest effect on
`stroke rates are presented.
`
`Figure 5. Change from baseline VA (cohort 1). For all groups, VA
`increased with 3 loading doses of ranibizumab (day 0, month 1, month 3).
`From months 3 to 12, with protocol-defined retreatment, VA tended to
`decrease. Error bars are ⫾1 standard error. ETDRS ⫽ Early Treatment
`Diabetic Retinopathy Study; VA ⫽ visual acuity.
`
`1736
`
`Regeneron Exhibit 2032
`Page 06 of 09
`
`

`

`Boyer et al
`
`䡠 Ranibizumab Safety in Wet AMD
`
`In SAILOR there was not a difference between doses in
`APTC ATEs overall, which is consistent with our current
`understanding of ranibizumab pharmacology. As a Fab,
`ranibizumab has low systemic bioavailability (⬃1/90,0000
`of intravitreal concentration) and a half-life of only several
`hours (Kubler P, Xu L, Jumbe N, et al. Population pharma-
`cokinetics of ranibizumab in patients with age-related mac-
`ular degeneration. Presented at: American Society of Retina
`Specialists Annual Meeting, December 1–5, 2007; Indian
`Wells, California).
`Certain subgroups of subjects (e.g., those with prior
`cardiovascular accidents) may experience higher rates of
`systemic SAEs. We observed that the incidence of stroke
`was greater for cohort 1 subjects who had a history of
`stroke, congestive heart failure, or arrhythmias. However,
`the low incidence of stroke in SAILOR made it difficult to
`draw meaningful conclusions about the relationship be-
`tween risk factors and stroke. Although the results of clin-
`ical trials cannot be directly compared with epidemiology
`studies in AMD, epidemiology stroke rates can provide a
`reference that aids in understanding stroke rates in SAILOR.
`The annual stroke rate for new-onset neovascular AMD in a
`large sample of Medicare subjects was 3.8%, and the annual
`ischemic stroke rate was 56.4% for those subjects who had
`experienced an ischemic stroke in the year before study
`entry.7 Both of these rates are higher than those observed in
`SAILOR.
`Ranibizumab treatment was associated with a net gain
`in VA in the cohort 1 0.3 mg and 0.5 mg dose groups.
`However, consistent with the results of MARINA and
`ANCHOR, 0.5 mg doses of ranibizumab tended to have a
`slightly greater VA benefit than 0.3 mg doses in subjects
`with neovascular AMD. Ranibizumab also tended to be
`
`Figure 7. Change from baseline CFT (cohort 1). In cohort 1 subjects with
`OCT data, CFT decreased with 3 loading doses of ranibizumab. Central
`foveal thickness then increased from months 3 to 6 and remained stable
`from months 6 to 12. Error bars are ⫾1 standard error. CFT ⫽ central
`foveal thickness; ETDRS ⫽ Early Treatment Diabetic Retinopathy Study;
`OCT ⫽ optical coherence tomography.
`
`1737
`
`Figure 6. Subjects gaining ⱖ15 letters from baseline VA (cohort 1). The
`proportion of cohort 1 subjects who gained ⱖ15 letters increased with 3
`loading doses of ranibizumab and was th

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket