Summary Basis of Decision for Macugen

Review decision

The Summary Basis of Decision explains why the product was approved for sale in Canada. The document includes regulatory, safety, effectiveness and quality (in terms of chemistry and manufacturing) considerations.


Product type:

Drug
Macugen

Pegaptanib sodium, 0.3 mg pegaptanib sodium/90 µL, Solution, Injection

Pfizer Canada Inc.

Submission control no: 094022

Date issued: 2006-03-01

Health Products and Food Branch

Our mission is to help the people of Canada maintain and improve their health.
Health Canada

HPFB's Mandate is to take an integrated approach to the management of the risks and benefits to health related to health products and food by:

  • Minimizing health risk factors to Canadians while maximizing the safety provided by the regulatory system for health products and food; and,
  • Promoting conditions that enable Canadians to make healthy choices and providing information so that they can make informed decisions about their health.

Health Products and Food Branch

Également disponible en français sous le titre : Sommaire des motifs de décision (SMD), MACUGEN*, Pegaptanib sodium par injection, 0,3 mg/90 μL, Pfizer Canada Inc., No de contrôle de la présentation 094022

Foreword

Health Canada's Summary Basis of Decision (SBD) documents outline the scientific and regulatory considerations that factor into Health Canada regulatory decisions related to drugs and medical devices. SBDs are written in technical language for stakeholders interested in product-specific Health Canada decisions, and are a direct reflection of observations detailed within the evaluation reports. As such, SBDs are intended to complement and not duplicate information provided within the Product Monograph.

Readers are encouraged to consult the 'Reader's Guide to the Summary Basis of Decision - Drugs' to assist with interpretation of terms and acronyms referred to herein. In addition, a brief overview of the drug submission review process is provided in the Fact Sheet entitled 'How Drugs are Reviewed in Canada'. This Fact Sheet describes the factors considered by Health Canada during the review and authorization process of a drug submission. Readers should also consult the 'Summary Basis of Decision Initiative - Frequently Asked Questions' document.

The SBD reflects the information available to Health Canada regulators at the time a decision has been rendered. Subsequent submissions reviewed for additional uses will not be captured under Phase I of the SBD implementation strategy. For up-to-date information on a particular product, readers should refer to the most recent Product Monograph for a product. Health Canada provides information related to post-market warnings or advisories as a result of adverse events (AE).

For further information on a particular product, readers may also access websites of other regulatory jurisdictions. The information received in support of a Canadian drug submission may not be identical to that received by other jurisdictions.

Other Policies and Guidance

Readers should consult the Health Canada website for other drug policies and guidance documents. In particular, readers may wish to refer to the 'Management of Drug Submissions Guidance'.

1 Product and submission information

Brand name:

Macugen

Manufacturer/sponsor:

Pfizer Canada Inc.

Medicinal ingredient:

Pegaptanib sodium

International non-proprietary Name:

Pegaptanib sodium

Strength:

0.3 mg pegaptanib sodium/90 µL (equivalent to the oligonucleotide moiety)

Dosage form:

Solution

Route of administration:

Injection

Drug identification number(DIN):

  • 02267225

Therapeutic Classification:

Vascular Endothelial Growth Factor antagonist

Non-medicinal ingredients:

Sodium chloride, monobasic sodium phosphate monohydrate, dibasic sodium phosphate heptahydrate, hydrochloric acid, and sodium hydroxide in water for injection

Submission type and control no:

New Drug Submission, Control No. 094022

Date of Submission:

2004-09-17

Date of authorization:

2005-05-02

* Eyetech Pharmaceuticals, Inc., Pfizer Canada Inc., licensee

2 Notice of decision

On May 2, 2005 , Health Canada issued a Notice of Compliance to Pfizer Canada Inc. for the drug product Macugen. Macugen contains the medicinal ingredient pegaptanib sodium which is a selective Vascular Endothelial Growth Factor (VEGF) antagonist that inhibits the binding of isoform VEGF165 to its VEGF receptors. VEGF induces angiogenesis, vascular permeability and inflammation, all of which are thought to contribute to the progression of the neovascular form of Age-related Macular Degeneration (AMD). Macugen is indicated for the treatment of Subfoveal Choroidal Neovascularization (SCNV) secondary to AMD.

AMD is one of the leading causes of visual impairment among older Canadians and is becoming a major public health concern due to the expected increase in incidence as a direct result of the increasing age of the population. This submission was granted Priority Review due to the unmet medical need for new therapies for exudative AMD. Available therapies are applicable only to subgroups of patients with defined lesion characteristics and there are currently no treatment options in Canada for the 70 to 80% of patients with AMD who have minimal classic or occult lesions with no classic morphology.

The market authorization of Macugen was based on adequate data from quality control studies, non-clinical, and clinical studies. Efficacy and safety were studied in two multicentre, controlled, double-masked, and identically designed randomized studies in patients with SCNV associated with AMD. A total of 1208 patients were enrolled and 1190 were treated (892 Macugen, 298 sham). Patients treated with Macugen 0.3 mg exhibited statistically significant results in both trials for the primary efficacy endpoint. The data submitted also demonstrate that Macugen can be administered safely when used under the conditions stated in the Product Monograph.

Macugen (pegaptanib sodium injection) is supplied in a single-use 1 mL glass syringe containing 0.3 mg in a 90 µL deliverable volume. Each syringe is fitted with an attached 27-gauge needle and is contained in an outer package. The accompanying plunger rod and flange are in a separate package. Macugen is administered once every six weeks by intravitreous injection into the eligible eye.

Macugen is contraindicated in patients with active or suspected ocular or periocular infection or with a known hypersensitivity to any component of this preparation. Detailed conditions for the use of Macugen are described in the Product Monograph.

Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Macugen is favourable in the treatment of subfoveal choroidal neovascularization secondary to age-related macular degeneration.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (medicinal ingredient)

Manufacturing Process and Process Controls

Pegaptanib sodium is chemically synthesized, purified, and supplied for drug product manufacturing as a white to off-white amorphous solid. The synthetic process applied for this drug substance is considered to be rational and conventional to afford the target substance. The specifications and the acceptance criteria for the starting materials and the chemical reagents/solvents are considered to be justified.

Characterisation

The routine release-testing regimen contains tests that provide assurance that pegaptanib sodium is consistently produced exhibiting the characteristic structure. The identity test verifies that the drug substance exhibits characteristic binding to VEGF165 and the nucleoside profiling provides assurance of the correct structure. The results of these tests show that pegaptanib sodium possesses the predicted structure and that batch consistency is achieved.

Results from impurity characterization studies indicate that the methods used are sufficient to detect and measure both process-related impurities and degradation products. The impurities and degradation products that were reported and characterised were found to be within the ICH established limits and/or otherwise qualified.

Control of Drug Substance

Validation reports were satisfactorily submitted for all analytical procedures used for in-process and release testing of the drug substance, and to justify the specification of the drug substance.

The analytical procedures applied for the quality control of the drug substance are considered to be appropriate. Data from batch analyses were reviewed and considered to be acceptable according to the specification of the drug substance.

Stability

Based upon the real-time and accelerated stability data submitted, the proposed shelf-life, storage and shipping conditions for the drug substance were supported and considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Macugen (pegaptanib sodium injection) is a sterile, clear, preservative-free solution containing pegaptanib sodium for intravitreous injection. Macugen is supplied in a single-dose, pre-filled syringe to deliver a dose of 0.3 mg pegaptanib sodium (equivalent to the oligonucleotide moiety). The non-medicinal ingredients are sodium chloride, monobasic sodium phosphate monohydrate, dibasic sodium phosphate heptahydrate, hydrochloric acid, and sodium hydroxide in water for injection. Each syringe contains a nominal delivered volume of 90 µL with an overfill to ensure delivery of the required dose.

The drug product is presented in a 1 mL Type I glass barrel syringe sealed with a coated bromobutyl rubber plunger stopper. The syringe has a fixed 27-gauge needle with a rubber needle shield with a rigid plastic outer shield. A plastic syringe plunger and flange adapter are also supplied for administration purposes.

Pharmaceutical Development

Changes to the manufacturing process and formulation made throughout the development were considered acceptable upon review.

Manufacturing Process and Process Controls

All equipment, operating parameters, in-process tests and detailed instructions are adequately defined in the documentation. The manufacturing process follows standard compounding and aseptic process techniques utilizing conventional pharmaceutical equipment and facilities. The manufacturing process for commercial production is well controlled to yield a product that meets all relevant specifications.

Control of Drug Product

Macugen is tested to verify its appearance, identity, binding specificity, peak molecular weight, volume, polydispersity, pH, osmolality, viscosity, sterility, and presence of degradation products, visible particles, particulate matter, and bacterial endotoxins.

Degradation products arising from manufacturing and /or storage were reported and characterised. These products were found to be within ICH established limits and/or were otherwise qualified.

Validation reports were satisfactorily submitted for all analytical procedures used for in-process and release testing of the drug product, and to justify the specification of the drug product.

Data from final batch analyses were reviewed and considered to be acceptable according to the specification of the drug product.

Stability

Based upon the real-time and accelerated stability study data submitted, the proposed 18 month shelf-life for the drug product at 2-8oC protected from light is considered acceptable.

3.1.3 Facilities and Equipment

The design, operations, and controls of the facilities and the equipment that are involved in the production are considered to be suitable for the activities and products manufactured. All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations.

3.1.4 Adventitious Agents Safety Evaluation

All of the components used in the production of pegaptanib and pegaptanib sodium solution for injection comply with the EMEA Note for Guidance on Minimizing the Risk of Transmitting Animal Spongiform Encephalopathy Agents Via Human and Veterinary Medicinal Products, EMEA/410/01 (European Medicines Agency). The manufacturers of the starting materials and excipients have provided certification letters attesting to these claims.

3.1.5 Summary and Conclusion

The Chemistry and Manufacturing information submitted for Macugen has demonstrated that the drug substance and drug product can be consistently manufactured to meet the specifications agreed upon. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes.

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

Seven in vitro and four animal in vivo studies investigated the pharmacodynamic properties and safety profile of pegaptanib sodium. Primary pharmacology studies were performed in order to characterize the mode of action and/or effects of pegaptanib in relation to its desired therapeutic target, the VEGF165 isoform. The sequence similarity of the VEGF gene is highly conserved across species; VEGF proteins in the mouse, rat, rabbit, and dog display remarkable similarities to human VEGF. These findings allow the extrapolation of animal data to humans. For the in vitro system, h uman umbilical vein endothelial cells (HUVEC) were used as the relevant cells in the binding of labelled [125I]VEGF165, thereby complying with ICH S6 guidelines. The safety pharmacology studies were carried out in dogs, rats, and monkeys.

The in vitro studies demonstrated that pegaptanib had high specificity and affinity for the VEGF165 isoform. The binding to VEGF188 was explained as an artifact or due to the fact that the protein was partially purified; this was not further investigated. However, the amount of VEGF188 in the eye is low or undetectable. Pegaptanib did not bind to other VEGF (B-, C-, or PIGF). Pegaptanib inhibited HUVEC proliferation, calcium mobilization and tissue factor formation. Also, pegaptanib did not bind to protein (primarily albumin); thus hemorrhage present in AMD should not interfere with the effects of pegaptanib. The significance of the binding between the pegaptanib and the VEGF165 isoform is that the VEGF isoform is unable to bind to the cell receptors initiating the cascade of events resulting in increased permeability and angiogenesis of the vascular endothelial cells.

In mice with hyperoxia induced retinal neovascularization, one study was negative and the second study was positive rendering the study unreliable. However, an additional study in mice showed that pegaptanib inhibited hyperoxia induced retinal neovascularization with low intraocular tissue IC50 values. In the corneal pocket model of angiogenesis in SD rats, intravenous pegaptanib inhibited corneal angiogenesis in a dose dependent manner. In the dermal permeability study with rats, VEGF and pegaptanib were mixed in vitro previous to the intradermal injection thus only confirming previous in vitro binding studies.

The safety pharmacology studies demonstrated no meaningful compound-related effects on cardiovascular parameters (blood pressure, heart rate and electrocardiograms), respiratory parameters (respiratory rate and tidal volume), or central nervous system parameters (physiologic and behavioral indices). In addition, there were no effects on renal parameters (urinalysis and clinical chemistry) in repeat-dose toxicology studies with pegaptanib sodium given intravitreously (IVT) for 3 months (primate) and 9 months (dog). These findings were obtained from animals with a plasma concentration of pegaptanib similar to or greater than that observed in humans receiving the 3.0 mg/eye IVT dosing regimen. Thus, there is no evidence of safety pharmacology issues that would preclude the IVT administration of pegaptanib sodium in humans.

3.2.2 Pharmacokinetics

Pegaptanib plasma and vitreous pharmacokinetics were characterized after bilateral IVT injection to Dutch-Belted and New Zealand White rabbits, Beagle dogs and Rhesus monkeys. Pegaptanib sodium in phosphate buffered saline was injected into the vitreous at volumes considered to be maximal for each species. The range of the dose was between 0.2-3.0 mg/eye.

Pegaptanib was slowly absorbed from the eye into the systemic circulation with measurable plasma concentrations seen within 3 to 6 hours after IVT injection. Peak plasma concentrations occurred earlier in dogs (3 to 5 hours) compared to rabbits and monkeys (12 to 24 hours). In the animals, plasma concentrations were approximately 0.03% to 0.15% of those seen in the vitreous humor throughout the dosing interval. Local ocular concentrations were always substantially higher than those in the systemic circulation after IVT administration.

After IVT administration, the apparent terminal plasma half-life of pegaptanib was approximately 4 days, 2 days, and 4 days for rabbits, dogs and monkeys, respectively. Vitreous and plasma concentrations declined slowly. The terminal phase rate constant, in both vitreous humor and plasma, reflected the slow absorption of pegaptanib into the systemic circulation after an IVT injection.

Approximately 70% to 100% of the IVT administered dose of pegaptanib was cleared from the eye into the systemic circulation as intact drug in rabbits, dogs and monkeys based on AUC (Area Under the Curve) values determined after IV (intravenous) and IVT administration. After IVT doses, pegaptanib plasma pharmacokinetics were linear and

consistent over time. Pegaptanib did not accumulate in the plasma with repeat bilateral IVT dosing every 2 weeks in dogs or monkeys. Slight accumulation was reported in the Dutch-Belted rabbits that had IVT administration every two weeks.

Tissue distribution, following a single dose, was evaluated in male rabbits. Pegaptanib-associated radioactivity diffused in both directions; toward the anterior and posterior parts of the eye from the administration site. The fact that radioactivity was still present in the eye 1008 hours post-dose suggest that potential accumulation of pegaptanib in the eye is possible.

Pegaptanib was not extensively distributed to the other tissues. After both IVT and IV administrations of radiolabelled pegaptanib, the highest extraocular concentrations of radioactivity were obtained in the kidney followed by the spleen, bone marrow (vertebra), lymph node (mesenteric) and liver. The lowest concentrations were found in the brain, spinal cord, skeletal muscle (dorsal) and bone (vertebra). Pegaptanib crossed the placenta but only a small percentage reached the amniotic fluid. In pregnant mice, approximately 0.05% of an IV dose of pegaptanib sodium was found in the amniotic fluid.

The studies did not fully characterize the metabolic fate of pegaptanib. The presence of 2'-fluoro-deoxyuridine in rabbit plasma and urine supports the role of nucleases in the metabolism of pegaptanib. Urinary data suggest that pegaptanib is mainly excreted in the form of unchanged drug and metabolites. Studies in rabbits indicate that urinary excretion is the predominant route of elimination.

Drug interaction studies with pegaptanib sodium were not conducted. Based on information from other oligonucleotide compounds, the potential for cytochrome P450 mediated drug interactions appears to be very low. Drug interaction via plasma protein binding displacement was not investigated but is not expected based on the low systemic concentrations following IVT administration and the lack of effect of plasma on the IC50 value of pegaptanib in vitro.

3.2.3 Toxicology

Animal toxicology studies evaluated single and repeat-dose toxicity, genotoxicity, reproductive and developmental toxicity, DNA incorporation, and immunogenicity. The route of administration was the IVT route which is the intended route in human. The IV route was also investigated for potential toxic effects since pegaptanib does get absorbed into the systemic circulation.

Carcinogenicity studies were not performed. This is acceptable, because of the advanced age of the target population and the negative genotoxicity findings. In addition, the absence of neoplastic or preneoplastic effects in the eye or other organs in the 3-, 6-, and 9- month studies (in monkeys, rabbits and dogs, respectively) is strong support for not conducting carcinogenicity studies.

Toxicity tests did not investigate the impurity profile. At least three bands of impurities in the pegaptanib formulation were detected; and were found to be consistent throughout the non-clinical toxicology program and clinical development. Since the impurities were present during the animal studies, it can be concluded that the impurities were qualified based on exposures in the pivotal repeat dose and embryo-fetal toxicity studies. Another impurity of concern was an endotoxin in one batch of pegaptanib at a level which produced severe eye inflammation observed in the earlier primate subacute toxicology studies; when a lower endotoxin level batch of pegaptanib was administered intravitreously to the same primates, this observation was not repeated. The risk of adverse events due to excessive levels of endotoxin was said to be mitigated by establishing an endotoxin specification for the drug product.

Single and Repeat Dose Toxicity

The toxicology program sufficiently demonstrated that the IVT injection of pegaptanib is not expected to cause serious and exaggerated adverse effects in the eye except for those caused by the injection procedure. The doses used in the toxicology program (2 mg/eye, both eyes for rabbits; and 3 mg/eye, both eyes for dogs) were similar to the maximal dose intended for the clinical study (0.3 -3 mg /eye) but the frequency was greater. Higher doses could not be administered because of the high viscosity of the drug preparation.

Overall, the toxicity studies using the IVT route demonstrated that the adverse events detected were related to the injection procedures. Neoplastic and pre-neoplastic lesions in the eye were not found. These results were an important finding because of the high levels of pegaptanib detected in the vitreous after injection.

Toxicity studies using the IV route showed that the plasma pegaptanib concentration was nearly 1000-fold less than that found in the vitreous, indicating slow absorption from the eye to the general circulation. This and the low dose proposed for humans (0.3mg/eye, one eye only) suggest that the blood level of pegaptanib in humans will be low.

The NOAEL (the highest level at which no adverse effect is observed) in female rats was 10 mg/kg and in male rats, 1 mg/kg. Plasma concentrations at the NOAEL in males (1mg/kg/day) were > 200-fold higher than clinical exposures after a 3 mg/eye IVT dose. The proposed clinical dose of 0.3 mg/eye is 10-fold lower providing a greater safety margin.

Results showed some decreases in serum protein and albumin levels in male rats at high doses (10 mg/kg IV). Progressive nephropathy (trace to mild) was also detected in rats after IV administration of doses up to 10 mg/kg for 13 weeks. This effect was observed in all groups including the control group. Male rats are generally susceptible to chronic progressive nephropathy; the relevance of these renal changes to humans is considered to be low.

Genotoxicity

Pegaptanib tested negative in all of the various assays: reverse mutation, forward mutation, micronucleus, or Syrian hamster embryo (SHE) cell transformation assays.

Reproductive and Developmental Toxicity

Pegaptanib sodium was not teratogenic in the range-finding studies with rabbits and mice nor in the definitive study with mice. Developmental toxicity, characterized by a mild decrease in fetal body weight and a reduction in ossification of phalanges that were within historical control ranges, was observed in the 40 mg/kg/day dose group. The NOAEL for developmental toxicity in this study was 6.5 mg/kg (IV). No maternal toxicity was observed. A very small percent (0.05%) of the maternal dose was found in the amniotic fluid. No effects on ECG parameters or cardiac histopathology were noted, suggesting normal fetal cardiac angiogenesis. The concern that pegaptanib (an anti-angiogenic agent) could affect fetal development is highly improbable in humans as the dosing of pegaptanib is intravitreous and is administered in low amounts.

DNA Incorporation

Test results showed that pegaptanib was metabolized in vivo to its nucleoside components. Studies to determine whether the incorporation of these components and individual nucleosides into DNA of rats and woodchucks were inconclusive. Of concern, were the component nucleosides detected in the liver, kidney and spleen. This issue was extensively discussed; it was agreed that the Product Monograph should state that the DNA studies were inconclusive, due to the lack of control experiments that would rule out possible contamination with unincorporated residual component nucleosides in the tissue samples.

Additional toxicology studies were conducted in rats and woodchucks with IV administration of the individual pegaptanib nucleotide components, 2'-FU and 2'-FC to investigate their potential toxicity. Considering the doses administered, the duration of exposure, and the severity and reversibility of the changes, both 2'-FU and 2'FC were considered relatively non-toxic. Overall, pegaptanib sodium and its component modified nucleotides do not exhibit mitochondrial toxicity.

Immunogenicity

Pegaptanib was non-antigenic in animals. Pegaptanib failed to elicit antibodies when administered in mice, rats or rabbits. Therefore, an immune response to pegaptanib is considered unlikely in a clinical situation.

3.2.4 Summary and Conclusion

The non-clinical pharmacology and toxicology program for pegaptanib sodium was extensive. The program included: studies to elucidate affinity and specificity of the pegylated aptamer for VEGF165; studies to verify the expected pathophysiologic effect of VEGF and its blockage by pegaptanib in animal models of angiogenesis and vascular permeability; and studies to elucidate the safety pharmacology and the potential immunogenicity of pegaptanib. Results showed that pegaptanib sodium is relatively safe, even when used at high doses over periods of 6 and 9 months in animals. There are some questions about the potential long-term consequences of the drug potentially becoming incorporated into the cellular DNA; however there was no evidence of clastogenicity by the compound. Overall the pharmacology and toxicology studies support the use of pegaptanib in humans with AMD.

3.3 Clinical basis for decision

3.3.1 Human Pharmacology

Studies in Age-related Macular Degeneration (AMD)

Protocol
Type of study
Design Dose Patients Treated Study Assessments
NX109- 01

Safety and Pharmacokinetics
Phase 1, multicentre, open-label, escalating dose, dose finding Single intravitreous injection of either 0.25, 0.5, 1, 2 or 3 mg pegaptanib sodium/eye 15 patients ≥50 years of age (y.a) with exudative AMD DLT, AEs, vital signs, BCVA, IOP, laboratory parameters, immune response, PK parameters, local ocular events.
EOP1000

Safety and Pharmacokinetics
Phase 1/2, multicentre, open-label, multiple dose in patients without PDT Total of 3 consecutive intravitreous injections of 3 mg pegaptanib sodium/eye, 28 days apart 10 patients ≥50 y.a with SCNV secondary to exudative AMD BCVA, AEs, IOP, laboratory parameters, vital signs, DLT, PK parameters, immune response, local ocular events
EOP1001

Safety and Pharmacokinetics
Phase 1/2, multicentre, open-label, multiple dose in patients following PDT Total of 3 IVT injection of 3 mg pegaptanib sodium/eye, 28 days apart. 11patients ≥50 y.a with predominant classic SCNV secondary to exudative AMD BCVA, AEs, IOP, laboratory parameters, vital signs, DLT, PK parameters, immune response, local ocular events plus requirement for PDT administration
EOP1003

Efficacy, safety
Phase 2/3 multicentre, randomized, sham-injection controlled, double mask, dose finding IVT injection of either 0.3, 1 or 3 mg pegaptanib sodium/eye or sham every 6 weeks for 54 weeks 612 patients ≥50 y.a. with Active SCNV secondary to exudative AMD BCVA, Fluorescein angiography and fundus photography, AEs, IOP, lab parameters, vital signs, PDT administration, local ocular events.
EOP1004

Efficacy, safety
Same as above Same as above 578 patients ≥50 y.a with Active SCNV secondary to exudative AMD BCVA, Fluorescein angiography and fundus photography, AEs, IOP, lab parameters, vital signs, PDT administration, local ocular events, PK, QOL.
EOP1006

Safety
Phase 2 multicentre, randomized, multiple dose, cohort IVT injections of 1 or 3 mg pegaptanib sodium/eye every 6 weeks for 54 weeks. 37 open-label (3mg) and 110 masked patients ≥50 y.a. with SCNV secondary to exudative AMD AE, local ocular events, IOP, lab parameters, vital signs PK parameters, immune response

DLT = Dose limiting toxicity; AE = Adverse event; BCVA = Best corrected visual acuity; IOP = Intraocular pressure;
PK = Pharmacokinetics; PDT = Photodynamic therapy with verteporfin; S CNV = Subfoveal choroidal neovascularization;
QOL = Quality of life

The clinical program consisted of six studies in Age-related Macular Degeneration (AMD). Five studies (NX 109-01, EOP1000, EOP1001, EOP1003 and EOP1004) were presented in support of the claim for efficacy of pegaptanib sodium in the treatment of patients with exudative AMD. All studies evaluated the safety of pegaptanib sodium. Two studies (EOP1003 and EOP1004) were classified as pivotal and are ongoing with Year 2 extensions; this priority submission only contains data from the first year. Study EOP1006 was designed to further assess the safety of pegaptanib sodium.

All studies were performed in accordance with Good Clinical Practice (GCP) and the Declaration of Helsinki. It was indicated that the studies were designed, conducted, analyzed and reported to a high standard in concordance with current clinical research practices. All studies clearly identified patient populations and employed prospectively-defined standards and accepted measures for the assessment of safety and efficacy.

The development program in humans was conducted entirely in patients; the reason being that the intravitreous mode of administration imposed some risks that are not justifiable in healthy volunteers who derive no benefit.

3.3.2 Pharmacodynamics

Pharmacodynamic studies were not conducted. Based on literature findings on the role of VEGF165 or VEGF164 on humans and animals, and on the performed non-clinical pharmacology studies, it was postulatedthat pegaptanib sodium will inhibit pathological Choroidal Neovascularization (CNV), without an effect on physiological aspects of VEGF in the eye or in the systemic circulation. Hence, the VEGF165-specific antagonist pegaptanib is expected to treat exudative AMD, and this should prevent relative deterioration of visual acuity. The design of the clinical trials included assessments of AMD lesion characteristics before and after treatment with pegaptanib sodium.

3.3.3 Pharmacokinetics

The pharmacokinetics (PK) of pegaptanib in humans was characterized in plasma only and was therefore not fully characterized. The plasma kinetics of pegaptanib were characterized following single and multiple IVT dosing in patients with AMD. For more information on the study designs, see Section 3.3.1Human Pharmacology. The dose of 3 mg/eye, which is 10x the proposed therapeutic dose, was used for the determination of the PK parameters; doses less than 3 mg/eye resulted in plasma levels close to or below the limit of quantification. A validated, Good Laboratory Practices (GLP), dual hybridization method was used to quantify plasma pegaptanib concentrations except for study NX-109-01 where a non-GLP but validated method was used.

Absorption

Results from studies EOP1000, 1001 and 1006 showed similar plasma PK parameters following single or multiple IVT doses of 3 mg/eye of pegaptanib. From these studies, the overall mean Cmax was 76 ng/mL (range of individual values 19-136 ng/mL, mean Tmax 1-2.5 days) and 77 ng/mL (range of individual values 27-200 ng/mL, mean Tmax 1-2.5 days) after the first dose, and the third or fourth dose, respectively. Also, the AUC inf range of individual values was 4-49 µg·h/mL (mean of 25 µg·h/mL) after the first dose and the AUCtau range of individual values was 13-41µg·h/mL (mean of 25 µg·h/mL) between the second and third dose and, between the third and fourth dose.

These results provide evidence that pegaptanib is unlikely to accumulate in the plasma following repeated dosing (IVT administration every 4 or 6 weeks for 12 or 24 weeks respectively) with a dose of 3 mg/eye. Importantly, results from study EOP1004 provide evidence that there is no plasma accumulation following the administration of the proposed therapeutic dose of 0.3 mg/eye every 6 weeks up to 42 weeks; 100% of the patients (n = 64) with trough samples had non-measurable pegaptanib plasma concentrations (LOQ of 8 ng/mL; samples taken approx. 6 weeks following IVT injection).

Absolute systemic/plasma bioavailability of pegaptanib following IVT administration could not be evaluated since there were no clinical studies with IV administration. Results from animal studies, however, suggest that 70 to 100% of the intravitreously administered dose would be absorbed in the plasma.

Distribution

The terminal half-life was estimated to be 10 ± 4 days in humans (study EOP1006). This half-life provides support for a slow transfer from the eye/vitreous to the systemic circulation as observed in animals. Relative to animal data, where mean half-life of 2-5 days was observed following IVT administration, the longer terminal half-life observed in humans may suggest slower passage from the eye to the systemic circulation, lower plasma metabolic activity and/or slower renal elimination. The fact that levels of pegaptanib were close to or below the limit of quantification 4 weeks after IVT administration (3 mg/eye) suggest low residual levels of pegaptanib in the eye/vitreous at the end of the proposed dosing interval (6 weeks) and dose (0.3 mg/eye). Nevertheless, the possibility of accumulation of pegaptanib or one of its metabolites in the eye cannot be excluded. In light of the favourable non-clinical and clinical safety profile of pegaptanib, the potential accumulation of small amounts of pegaptanib and or metabolites does not appear to be problematic.

Information from a pegaptanib tissue distribution study in rabbits, albeit the small number of animals involved (n = 1/sample time collection; 5 sample collection over 42 days), showed that pegaptanib was distributed to the highly perfused organs (liver, kidney, lymph node, spleen, bone marrow).

Metabolism

Pegapatnib is believed to be metabolized by endo- and exonucleases to shortmers and/ or nucleotides. No measurements of pegaptanib metabolites were made in the vitreous, plasma and urine of patients. Metabolism of pegaptanib by nucleases in human is supported by the presence of 2'-fluoro-deoxyuridine, a modified nucleotide of pegaptanib also found in the plasma and urine of pegaptanib-treated rabbits. Also, there was in vitro evidence suggesting that pegaptanib can be metabolized by nucleases in human plasma (study 400014), albeit the metabolic rate in human plasma appears to be much lower relative to other species (dogs and rabbits). Fomivirsen, an oligodeoxynucleotide, was shown to be metabolized by nucleases in human vitreous in vivo. The possibility that pegaptanib (a pegylated modified oligonucleotide) could be metabolized in the human vitreous/eye cannot be excluded.

Elimination

The excretion of pegaptanib and its breakdown products were not evaluated in humans. A study in rabbits, however, indicate that urinary excretion is the predominant route of elimination. The fact that renal clearance affects AUC and C max provides evidence of renal excretion of pegaptanib in humans.

Special Populations

Subjects involved for PK parameters determination (Studies EOP1000, 1001 and 1006) were elderly (mean age range 75.7-78.3; overall range 53-92), mainly female (64% of total subjects) and were afflicted by AMD. Study EOP1006 involved some subjects previously treated with photodynamic therapy (PDT) or had PDT during the study. Study EOP1001 involved subjects scheduled to have PDT treatment in the course of the study. There were a limited number of patients that were below 60 and above 85 years old (EOP1006). Overall, the data did not provide any evidence that the plasma PK parameters were significantly affected by gender, age or PDT treatment.

In rabbits, pegaptanib and 2'-fluoro-deoxyuridine (a nucleotide and metabolite of pegaptanib) were eliminated mainly via the kidney/urine. Results of study EOP1006 showed that renal function may affect the PK parameters of pegaptanib. These changes in the PK of pegaptanib were considered of no clinical consequence as the variation in plasma levels caused by the renal insufficiency were within levels attained with a dose of 3 mg/eye, a dose 10x higher than the proposed therapeutic dose and shown to be safe in animal and human studies. Patients with creatinine clearance less than 20 mL/min have not been studied.

Drug Interaction Studies

Pharmacokinetic drug interaction studies were not conducted. There is no information suggesting that o ligonucleotides are metabolized by the cytochrome P450 (CYP450) system. Following IVT administration, only low concentrations of pegaptanib transit through the liver making the possibility of a classic drug-drug interaction through liver metabolizing enzymes unlikely. As well, even though several antisense oligonucleotides have been specifically designed to inhibit CYP450 enzymes, non-specific control oligonucleotides have been shown to have no effect.

The potential plasma protein interactions/binding with pegaptanib were not investigated. A drug interaction through plasma protein displacement is unlikely given the low systemic exposure following IVT administration. As well, plasma protein binding is not believed to be extensive as results show a lack of effect of 50% plasma on the IC 50 value of pegaptanib in vitro.

3.3.4 Clinical Efficacy

Six studies were submitted to support the use of Macugen for the treatment of exudative AMD. Their design descriptions can be found in Section 3.3.1Human Pharmacology. The two pivotal studies (EOP1003 and EOP1004) were designed to last 2 years. This current submission contains data from only the first year. Both studies had identical designs except that EOP1004 included assessments of the drug plasma concentration measurements and the National Eye Institute Visual Functioning Questionnaire (VFQ 25).

The two pivotal clinical studies were well conducted. One study was conducted internationally including Canada and USA. Of the main efficacy variables used, the last observation carried forward to impute data for missing values, is a potential concern in a condition with declining vision. The concern was greatly mitigated by the low proportion of patients who withdrew from the studies. There was an ethical concern to prohibit treating patients with the currently approved therapy, photodynamic therapy (PDT). Administration of PDT was at the investigators' discretion, making analysis of the concomitant PDT and pegaptanib data flawed (not reported). In addition, the selection of the control group posed difficulties because of the invasive administration procedure, intravitreous injection. Therefore, a sham was selected as control. The problem with sham treatment is that neither the effect of the injection, the drug or the solvent can be segregated in the results for efficacy/adverse events. In addition, the injection alone may result in some type of body reaction that will not be controlled in sham.

In general, the study population reflected the demographic of AMD. The medical and surgical histories reflected the age of the AMD population; the most common vascular conditions were hypertension, cardiac disorders, and muscular-skeletal conditions. Baseline applanation tonometry revealed that patients had normal or medically controlled intraocular pressure of 15.2 (SD 2.9) mm Hg. There was a similar distribution in ocular conditions among the groups. The number of patients that received prior PDT treatment was similarly distributed but the proportion in study 1004 (sham 11%, 0.3 mg 13%, 1mg 14% and 3 mg 14%) was larger than in study 1003 (sham 3%, 0.3 mg 4%, 1 mg 6% and 3 mg 4%). The majority of patients (80%) had a visual acuity 20/30 to 20/100. All lesion sizes were ≤ 12 disc area. The study groups had a similar proportion of patients with the three AMD subtypes. Among the lesions the "classic only" was present in about 25% of the population. Similar proportions of patients had "minimally classic" and "occult only" AMD. The studies were not designed to analyze the efficacy data in each of the AMD subtypes.

In both pivotal studies, the primary efficacy endpoint was the proportion of Responders, defined as patients losing < 15 letters of best-corrected visual acuity (VA) in the study eye from baseline up to 54 weeks, using the Early Treatment Diabetic Retinopathy Study chart at 2 meters. This measurement is considered to be the most meaningful and clinically relevant measure of visual outcome in patients with exudative AMD. Secondary efficacy endpoints were i) the proportion of patients gaining at least 15 letters of vision from baseline to 54 weeks; ii) the proportion of patients gaining at least 0 letters of vision from baseline to 54 weeks and iii) the change in mean VA from baseline to 6, 12 and 54 weeks.

Both pivotal studies demonstrated that Macugen was efficacious in patients suffering from SCNV secondary to AMD (SCNV-AMD) as assessed from results of the primary and secondary variables. The proportion of patients who lost < 15 letters at week 54 in the Macugen 0.3 mg group was statistically significantly higher as compared with the sham group in both studies (Study EOP1003: Macugen 0.3 mg 73% vs. Sham 59%, p-value = 0.0105; Study EOP1004: Macugen 0.3 mg 67% vs. Sham 52%, p-value = 0.0031). Results showed that the proportion of patients losing less than 15 letters at week 54 were similar irrespective of baseline characteristics, including lesion sub-type, prior PDT usage, visual acuity, lesion size, gender, iris pigment and age as assessed by similarity in results (eye bold), not by statistical analyses. The calculated Therapeutic Gain (treated minus sham) in the 0.3 mg dose group was about 15%. It can be concluded that Macugen 0.3 mg treatment over a one-year period reduced the risk of vision loss in patients with SCNV-AMD. However, the benefit over sham was only about 15%.

Secondary variables demonstrated that patients in both Macugen 0.3 mg and sham continued to experience vision loss, but Macugen slows down the disease progression assessed by the number of letters that patients were able to read during the one year study period. Furthermore, the proportion of patients having severe vision loss during the study period, defined as a loss of ≥ 30 letters of VA over 54 weeks, was reduced by more than a half in the Macugen-treated patients (10% of patients experienced severe vision loss) compared with sham (22% of patients).

3.3.5 Clinical Safety

The clinical program consisted of six studies in Age-related Macular Degeneration (AMD) and all evaluated the safety of pegaptanib sodium administration. Two studies (EOP1003 and EOP1004) were classified as pivotal and were designed to last two years. The data for this priority submission was taken from the first year. Study EOP1006 was designed to further assess the safety. The study design of these trials can be found in Section 3.3.1 Human Pharmacology.

The safety population in the two pivotal studies, EOP1003 and EOP1004 consisted of 612 and 578 patients, respectively, who received at least one treatment. Less than 14% of the patients withdrew from the studies during the first year and approximately 90% of patients received 8 to 9 treatments (pooled data from the two studies). Patients were mostly white, a median age of 75 years of age and female, reflecting the general population with AMD. The different groups (Macugen-treated groups and sham-treated groups) were similarly distributed with respect to gender, age, race, performance status, height and weight. The groups were also well-balanced in terms of ophthalmic, medical and surgical history. The most prevalent baseline disorders were hypertension Not Otherwise Specified (NOS, 48-54%), arthritis NOS (19-21%), hypercholesterolemia (18-23%) and drug hypersensitivity (16-23%). There were no imbalances in baseline medication across treatment groups: nearly 85% received medications affecting the sensory organs (mostly anticholinergics), 64-72% received cardiovascular medications (mostly HMGCoA inhibitors), and 52-59% received medications affecting the alimentary tract and metabolism (vitamins and calcium). No patients had baseline electrocardiograms (ECG) with clinically significant findings.

The two studies showed no clinically meaningful imbalances across the groups with respect to the ophthalmic history. Baseline lesions subtypes were classified based on fluorescein angiographic appearance with 38-40% having 0% classic AMD (occult), 34-38% having from 1-49% classic AMD (minimally classic), and 24-27% having ≥ 50% classic AMD (predominantly classic). For the overall baseline ocular history 12-14% of patients across treatment groups had received prior PDT with verteporfin (≥ 8 weeks from first treatment) and 5-6% had received prior systemic treatment for AMD. Subretinal hemorrhages were present at baseline in 71-73% (69% in EOP1003; 75% in EOP1004 of the safety population that included all treatment groups) of all patients.

The rate of patient discontinuation was low. The most frequent reason for discontinuation was "patient request" (5%), adverse events (1%), and death (2%). Death was not unexpected in this age group; none were related to treatment.

Adverse events were reported in nearly all patients (95-100% of patients in study EOP1003 and 99-100% of patients in study EOP1004). The proportion of patients having at least 1 serious adverse event (SAE) was < 20% and only 1-2% of the SAE led to discontinuation.

It should be kept in mind that Macugen was planned to be administered intravitreously (IVT) at 0.3 mg, 1 mg, 3 mg dose per eye in a total volume of 90uL every 6 weeks during Year 1. The planned number of IVT injections was nine. As mentioned previously, 8 to 9 injections were administered to about 90% of the patients. The control group (sham) did not receive IVT injections but were subjected to all the previous procedures and were simulated to receive the IVT injection. They did not know whether they received the injection or not. The adverse events, elicited differentially between the sham group and Macugen groups, could be assessed as being related to the IVT injection or to the drug (pegaptanib sodium). It is difficult to separate injection from drug- related events and this is therefore a deficiency of the study, but accepted because of ethical concerns.

Ocular Adverse Events

The majority of all the adverse events (AEs) were eye disorders (82-88% in study EOP1003 and 92-97% in study EOP1004). Systemic AEs were infrequent, as expected with a locally administered treatment.

Approximately 90% of the patients reported ocular AEs in the study eye; the majority of the events were mild to moderate in severity. With few exceptions, ocular AEs were elicited similarly among all three active treatments (0.3 mg, 1 mg, 3 mg dose per eye) which suggest a relationship to the injection procedure rather than the drug itself.1 The ocular AEs that appeared to be Macugen dose-dependent were corneal edema, cataract, conjunctival edema, and eye lid edema, but, the incidences in the 0.3 mg group were similar to or lower than in the sham group.

The safety data described below summarize the experience of 295 patients exposed for up to one year (total number of injections = 2478, mean number of injections/patient = 8.4) at the recommended dose of 0.3 mg.

  Incidence (%)
MedDRA Preferred Term 0.3 mg Pegaptanib Sodium N=295 Sham N=298
Eye pain 34 29
Punctate keratitis 33 27
Vitreous floaters 31 8
Visual acuity reduced 28 28
Cataract 22 23
Vitreous opacities 19 10
Anterior chamber inflammation 16 6
Intraocular pressure increased 14 3
Visual disturbance NOS 14 13
Eye discharge 11 8
Corneal oedema 9 7
Vision blurred 9 5
Lacrimation increased 8 10
Macular degeneration 8 12
Conjunctival haemorrhage 8 6
Abnormal sensation in eye 8 10
Eye irritation 7 7
Photophobia 7 8
Blepharitis 7 6
Eye pruritus 7 8
Eye redness 7 7
Photopsia 7 3
Vitreous disorder NOS 6 2
Corneal epithelium disorder 4 6
Dry eye NOS 6 5
Ocular discomfort 6 4
Retinal haemorrhage 5 10
Conjunctivitis 5 3
Vitreous detachment 4 5
Conjunctival oedema 4 4
Corneal epithelium defect 3 5
Eyelid oedema 2 4
Conjunctival hyperaemia 2 3
Vitreous haemorrhage 2 0
Endophthalmitis 2 0
Retinal exudates 2 2
Eye haemorrhage NOS 2 0
Periorbital Haematoma 2 2
Keratitis 1 3
Ocular hypertension 1 2
Corneal dystrophy 1 1
Eyelid ptosis 1 2
Eyelids pruritus 1 0
Mydriasis 1 0
Eye swelling 1 0
Meibomianitis 1 0
Conjunctivitis allergic 1 0
Retinal degeneration 1 0
Pupillary reflex impaired 1 2
Retinal artery embolism 1 1
Eye inflammation NOS 1 0
Corneal abrasion 1 0

1 Animal studies, which had IVT injection control groups, demonstrated that repeat intraventricular injection resulted with ocular adverse effects (ocular discharge, conjunctival irritation, vitreal floaters, cataracts, retinal detachment) across treatment groups including controls suggesting that these ocular events were injection-related.

The following ocular AEs were reported as serious in any Macugen group: endophthalmitis (12 cases, 1%), retinal detachment (4 cases, < 1%), retinal hemorrhage (3 cases, < 1%), cataract (3 cases, < 1%), traumatic cataract (3 cases, < 1%), vitreous hemorrhage (2 cases, < 1%), glaucoma NOS (1 case, < 1%), uveitis NOS (1 case, < 1%), and increased intraocular pressure (1 case, < 1%).

The serious events that could be preventable or tightly-controlled are endophthalmitis and traumatic cataracts due to the intravitreous injection procedure. The incidence of endophthalmitis was reduced with improved aseptic conditions, but not eliminated. Adherence to strict aseptic procedures, as well as patient follow-up during the first week after any injection should be emphasized. The signs and symptoms of endophthalmitis should be clearly annotated in the Product Monograph, as well as a description of the procedure before and after IVT injection. To avoid traumatic cataracts, the Product Monograph should instruct doctors to avoid touching the lens with the syringe needle.

The ocular AEs that may be possibly related to undesirable VEGF inhibition in the eye, based on experience with systemic agents that inhibit VEGF, are thromboembolic or bleeding events. Vitreous hemorrhage was the only event that occurred in greater proportion in the Macugen-treated group compared to sham. But the incidence of vitreous hemorrhage did not increase with higher doses of pegaptanib sodium, and did not reoccur in 15 of the 16 patients as would be expected with chronic VEGF inhibition. Fluorescein angiography revealed no retinal vascular abnormalities that are unexpected in the natural history of exudative AMD. There were no notable delays in arterial-venous transit time, abnormalities in choroidal perfusion, or arteriolar occlusions.

The possibility that ophthalmic bleeding could be exacerbated or elicited by co-administered medications such as anticoagulants or platelet aggregation inhibitors was of interest. Study results showed no evidence for an increased risk of vitreous hemorrhage in Macugen-treated patients associated with concomitant anticoagulation medications, platelet inhibitors or non-steroidal anti-inflammatory drugs.

Systemic Adverse Events

Non-ocular AEs related to the systemic inhibition of VEGF were unexpected in the pegaptanib sodium program because of the specificity of pegaptanib sodium for VEGF165 and the low or undetectable systemic exposure from intravitreous dosing . In addition, the preclinical assessment of pegaptanib sodium showed no evidence of physiological changes or toxic consequences that would be regarded as reasonably related to VEGF-antagonism, either systemically or in the eye. Preclinical exposures from the intravenous or intravitreous routes achieved exposures considerably higher than exposures achieved in humans administered with 3 mg in one eye. Toxicology and safety pharmacology studies showed no evidence of hypertension, proteinuria, thromboembolic, or bleeding phenomenon.

Systemic AEs were infrequent in the safety studies. The types and incidence of the systemic AEs were not unexpected for an elderly population and most events had similar incidences in all treatment groups. The most common events in the 0.3 mg dose group were hypertension (14/295, 4.7%), nasopharyngitis (19/295, 6.4%), headache (19/295, 6.4%), and bronchitis (16/295, 5.4%). Arthralgia and nausea, and upper respiratory tract infections had an incidence of 4% each.

Systemic thrombotic cardiovascular/pulmonary and hemorrhagic SAEs were few and similar between Macugen groups and the sham group. A review of the safety narrative for these patients indicated that all but one of these cases had strong pre-existing risk factors for thromboembolic cardiac and central nervous system (CNS) disorders; namely hypertension or pre-existing vascular occlusive disease. There is no indication that hypertension/increase blood pressure and myocardial infarction (MI) are related to anti-VEGF effect. Anti-VEGF therapy could potentially inhibit/delay recovery from MI, inhibiting the development of collaterals associated with MI/stroke, inducing or accelerating angina. No time frame for anti-VEGF therapies could be offered if patients recently had an episode of MI/stroke.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

This priority New Drug Submission (NDS) contains data in support of a novel compound, Macugen (pegaptanib sodium), for the treatment of Subfoveal Choroidal Neovascularization associated to Age-related Macular Degeneration (SCNV-AMD). Previously, therapy for the treatment of SCNV-AMD was only applicable to subgroups of exudative-AMD patients with predominantly classic lesion characteristics as determined by fluorescein angiography. There was no effective treatment for patients with other lesion types of exudative AMD such as minimally classic and occult-only lesions.

Pegaptanib was shown to bind with high specificity and sensitivity to vascular endothelium growth factor isoform 165 (VEGF165), which is believed to be involved in angiogenesis and increased permeability of the new blood vessels that develop in the macular portion of the retina. It is possible that this mechanism is one among others contributing to the increased permeability of blood vessels for all lesion subtypes; all demographic and lesion characteristics. Pegaptanib sodium reduced moderate and severe vision loss consistently across two large-scale pivotal studies in the investigated population.

From the safety perspective, pegaptanib sodium was tolerated at all doses studied and few patients withdrew from the studies due to adverse events. Ocular adverse events were common and, with the exception of infrequent events related to intravitreous injection, were mild to moderate in severity and were resolved with time.

The risk of serious ocular adverse events was low (3%). The majority of serious ocular adverse events: endophthalmitis, traumatic cataract and retinal detachment were related to the intravitreous injection procedure rather than the drug substance. Hence they are understandable, predictable and may be reduced in practice. Aseptic technique is important to keep the incidence of endophthalmitis low.

Intravitreous injection of pegaptanib sodium was associated with the risk of elevated intraocular pressure (IOP). Mild, transient increases in IOP were manageable and did not require intervention in the majority of cases. However, there were some patients (9%) who developed very high IOP. Therefore, monitoring is required during the first week after injection.

Vitreous hemorrhage was detected in 2% of the patients treated with Macugen. It is possible that vitreous hemorrhage in many patients were secondary to the underlying AMD process, but may also be related to the injection procedure. In most cases, the vitreous hemorrhage resolved without sequelae.

The high completion rate seen in the studies is indicative of the ability of the patient population to tolerate prolonged intravitreous injection-based therapies at the prescribed regimen despite the discomfort and inconvenience associated with the intravitreous injection preparation and administration procedures.

Macugen has shown a favourable risk/benefit profile. The benefits associated with the treatment outweigh the risks of the injection procedure.

3.4.2 Recommendation

Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Macugen is favourable in the treatment of Subfoveal Choroidal Neovascularization secondary to Age-related Macular Degeneration. The New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations.

4 Submission Milestones

Submission Milestones: Macugen

Submission MilestoneDate
Pre-submission meeting:2004-02-09
Request for priority status
Filed:2004-08-13
Approval issued by the Director:2004-09-14
Submission filed:2004-09-16
Screening 1
Screening Acceptance Letter issued:2004-11-10
Review 1
Quality Evaluation complete:2005-04-06
Clinical Evaluation complete:2005-04-15
Labelling Review complete:2005-04-13
NOC issued by Director General:2005-05-02