Summary Basis of Decision for Isentress ™ *

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
IsentressTM*

Raltegravir, 400 mg, Tablet, Oral

Merck Frosst Canada Ltd.

Submission control no: 113408

Date issued: 2008-08-07

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 managing the health-related risks and benefits of 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), PrISENTRESSMD, raltégravir, 400 mg, comprimés, Merck Frosst Canada Ltd., N° de contrôle de la présentation 113408

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:

IsentressTM*

Manufacturer/sponsor:

Merck Frosst Canada Ltd.

Medicinal ingredient:

Raltegravir

International non-proprietary Name:

Raltegravir

Strength:

400 mg

Dosage form:

Tablet

Route of administration:

Oral

Drug identification number(DIN):

  • 02301881

Therapeutic Classification:

HIV integrase strand transfer inhibitor

Non-medicinal ingredients:

Tablet: microcrystalline cellulose, lactose monohydrate, calcium phosphate dibasic anhydrous, hypromellose 2208, poloxamer 407 (contains 0.01% butylated hydroxytoluene as antioxidant), sodium stearyl fumarate, and magnesium stearate.

Film coating: polyvinyl alcohol, titanium dioxide, polyethylene glycol 3350, talc, red iron oxide, and black iron oxide.

Submission type and control no:

New Drug Submission,
Control No. 113408

Date of Submission:

2007-04-18

Date of authorization:

2007-11-27

*ISENTRESS™ is a Trademark of Merck & Co., Inc. Used under license.

2 Notice of decision

On November 27, 2007, Health Canada issued a Notice of Compliance under the Notice of Compliance with Conditions (NOC/c) Policy to Merck Frosst Canada Ltd. for the drug product Isentress™. The product was authorized under the NOC/c Policy on the basis of the promising nature of the clinical evidence, and the need for confirmatory studies to verify the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions.

Isentress™ contains the medicinal ingredient raltegravir which is a human immunodeficiency virus (HIV) integrase strand transfer inhibitor.

Isentress™, in combination with other antiretroviral agents, is indicated for the treatment of HIV-1 infection in treatment-experienced adult patients who have evidence of viral replication and HIV-1 strains resistant to multiple antiretroviral agents. Raltegravir, the medicinal ingredient of Isentress™, inhibits the catalytic activity of HIV integrase, an HIV-encoded enzyme that is required for viral replication. Inhibition of integrase prevents the integration of the HIV genome into the host cell genome during the early phase of infection, thereby preventing the production of new infectious viral particles and propagation of the viral infection.

The market authorization was based on submitted data from quality (chemistry and manufacturing) studies, as well as data from non-clinical and clinical studies. The efficacy and safety of Isentress™ were evaluated based on the results of two pivotal Phase III studies and two supportive Phase II studies. The trials involved a total of 987 subjects, including 789 treatment-experienced subjects, the indicated population. Results were submitted for all of the Phase III subjects at 16 weeks, however only approximately 60% of the 24-week data was included for review; it was on this basis that the product was authorized under the NOC/c policy. Results demonstrated superior efficacy of raltegravir compared to placebo at 16 weeks, and based on the available data, virologic response and immune reconstitution appeared to be sustained through 24 weeks. Raltegravir appeared to be generally well-tolerated, with a safety profile comparable to that seen in the placebo subjects. The sponsor has committed to a risk-management plan that proposes active post-market surveillance measures to monitor malignancies and other important safety issues.

Isentress™ (400 mg, raltegravir) is presented as oral tablets. For the treatment of adult patients with HIV-1 infection, the dosage of Isentress™ is 400 mg administered orally, twice daily, with or without food. Isentress™ is to be given in a combination regimen with other antiretroviral agents. Dosing guidelines are available in the Product Monograph.Isentress™ is contraindicated for patients who are hypersensitive to any component of this medicine. Isentress™ should be administered under the conditions stated in the Product Monograph taking into consideration the potential risks associated with the administration of this drug product. Detailed conditions for the use of Isentress™ are described in the Product Monograph.

The drug submission for Isentress™ was reviewed under the Priority Review Policy. Accordingly, the data submitted in the Isentress™ Priority Review application suggested a significant increase in effectiveness with an improved benefit/risk profile compared to existing therapies for resistant HIV-1 infection, a condition that is not adequately managed by a drug marketed in Canada.

Based on the Health Canada review of data on quality, safety, and effectiveness, Health Canada considers that the benefit/risk profile of Isentress™, in combination with other antiretroviral agents, is favourable for the treatment of HIV-1 infection in treatment-experienced adult patients who have evidence of viral replication and HIV-1 strains resistant to multiple antiretroviral agents.

3 Scientific and Regulatory Basis for Decision

Isentress™ was granted a Priority Review, since, based on data submitted by the sponsor in support of the Priority Review application, it was judged that Isentress™ offered a significant increase in efficacy over existing treatments in the treatment of a life-threatening illness. Subsequent review of the sponsor's full submission led to the decision to offer the sponsor approval under the NOC/c policy, in recognition of the promising but incomplete evidence of clinical effectiveness in the submission.

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Raltegravir (as raltegravir potassium), the medicinal ingredient of Isentress™, is an HIV integrase strand transfer inhibitor. Raltegravir inhibits the activity of the HIV integrase strand transfer enzyme, which normally catalyzes the integration of HIV DNA into host cell DNA, a necessary step in HIV replication. HIV genomes that fail to integrate cannot direct the production of new infectious viral particles, thereby preventing propagation of the viral infection.

Manufacturing Process and Process Controls

Raltegravir is manufactured via a multi-step synthesis. Each step of the manufacturing process is considered to be controlled within acceptable limits:

  • The sponsor has provided information on the quality and controls for all materials used in the manufacture of the drug substance.
  • The drug substance specifications are found to be satisfactory. Impurity limits meet ICH requirements.
  • The processing steps have been evaluated and the appropriate ranges for process parameters have been established.

In-process controls performed during manufacture were reviewed and are considered acceptable. The specifications for the raw materials used in manufacturing the drug substance are also considered satisfactory.

Characterization

Raltegravir is a white to off-white powder. Detailed characterization studies were performed to provide assurance that raltegravir consistently exhibits the desired characteristic structure.

The structure of raltegravir is considered to be adequately elucidated and the representative spectra have been provided. Physical and chemical properties have been described and are found to be satisfactory.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be within ICH established limits and/or were qualified from batch analysis and/or toxicological studies and therefore, are considered to be acceptable.

Control of Drug Substance

The drug substance specifications and analytical methods used for quality control of raltegravir are considered acceptable.

Copies of the analytical methods and, where appropriate, validation reports are considered satisfactory for all analytical procedures used for release and stability testing of raltegravir.

Batch analysis results were reviewed and all results comply with the specifications and demonstrate consistent quality of the batches produced.

The drug substance packaging is considered acceptable.

Stability

Based on the long-term and accelerated stability data submitted, the proposed retest period, shelf-life, and storage conditions for the drug substance are supported and considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Isentress™ is supplied as tablets, in 75 cc high-density polyethylene (HDPE) bottles with foil-induction sealed child-resistant caps and 1 g of desiccant. Isentress™ is available as 60 tablets per bottle.

Each tablet of Isentress™ contains 434.4 mg of raltegravir potassium (equivalent to 400 mg of free raltegravir) and the following excipients: microcrystalline cellulose, lactose monohydrate, calcium phosphate dibasic anhydrous, hypromellose 2208, poloxamer 407 (contains 0.01% butylated hydroxytoluene as antioxidant), sodium stearyl fumarate, and magnesium stearate. The film coating also contains polyvinyl alcohol, titanium dioxide, polyethylene glycol 3350, talc, red iron oxide, and black iron oxide.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations. The compatibility of raltegravir with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

Pharmaceutical development data, including development of the container closure system, are considered acceptable. Data provided in this section include composition of Isentress™, rationale for choice of formulation, manufacturing process including packaging, information on batches used in in vitro studies for characterization and discussion on the effect of formulation change on the safety and/or efficacy of Isentress™. Studies which justified the type and proposed concentration of excipients to be used in the drug product were also reviewed and are considered to be acceptable.

Manufacturing Process and Process Controls

The drug product is formulated, dry granulated, blended, lubricated, compressed, film-coated, and labelled using conventional pharmaceutical equipment and facilities.

All manufacturing equipment, in-process manufacturing steps and detailed operating parameters were adequately described in the submitted documentation and are found to be acceptable. The manufacturing process is considered to be adequately controlled within justified limits.

Control of Drug Product

Isentress™ is tested to verify that the identity, appearance, content uniformity, assay, levels of degradation products, dissolution, and microbiological impurities are within acceptance criteria. The test specifications and analytical methods are considered acceptable; the shelf-life and the release limits, for individual and total degradation products, are within acceptable limits.

The test specifications are considered acceptable to control the drug product, and the impurity limits were set according to ICH recommendations.

Validation reports submitted for all analytical procedures used for in-process and release testing of the drug product are considered satisfactory, and justify the specifications of the drug product.

Although impurities and degradation products arising from manufacturing and/or storage were reported and characterized, these were found to be within ICH established limits and/or were qualified from batch analysis and therefore, are considered to be acceptable.

Stability

Based on the long-term and accelerated stability data submitted, the proposed 30-month shelf-life at 15-30ºC for Isentress™ is considered acceptable. Isentress™ is photostable.

The compatibility of the drug product with the container closure system was demonstrated through compendial testing and stability studies. The container closure system met all validation test acceptance criteria.

3.1.3 Facilities and Equipment

The design, operations and controls of the facilities and equipment that are involved in the production are considered 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. All sites are compliant with Good Manufacturing Practices (GMP).

3.1.4 Adventitious Agents Safety Evaluation

Lactose monohydrate is the only excipient of animal origin in Isentress™ tablets. The lactose is derived from milk certified to originate from healthy animals and is collected in the same manner as milk fit for human consumption. The lactose is not prepared with the use of other ruminant materials, with the possible exception of calf rennet.

A letter of attestation confirming that the material is not from a BSE/TSE-affected country/area has been provided for this product indicating that it is considered to be safe for human use.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Isentress™ has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. 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

Primary Pharmacodynamics

Raltegravir was found in vitro, to be a selective inhibitor of strand transfer activity of purified recombinant HIV-1 integrase, with no HIV-1 reverse transcriptase inhibitory activity. It did not appear to have any significant inhibition of human DNA polymerases α, β, and γ, at concentrations up to 50 μM, and no in vitro activity in enzymes or receptors up to 10 µM.

Raltegravir showed potency similar to indinavir when tested against HIV-2. Furthermore, it retained activity against M-tropic (CCR5 receptors) and T-tropic strains (CXCR4 receptors), PI- and NNRTI-resistant isolates, and SI and NSI HIV-1 strains.

The sponsor believes that raltegravir will not inhibit the major double-strand DNA break-repair pathway in human cells as it was shown to inhibit strand transfer activity in vitro as cells infected with HIV-1 accumulated 2-LTR circle products.

Resistance

Among integrase mutations selected in cell culture by other structurally diverse experimental integrase inhibitors, the single mutation that caused the greatest reduction of raltegravir activity was N155S, with an approximate 19-fold resistance. The N155S mutation was shown to confer limited cross-resistance and reduced infectivity, although it appears that the reduced infectivity was specific for integrase inhibitors. Other mutations that demonstrated some level of change in the inhibitory concentration (IC50) of raltegravir were F121Y/T/125K (~9-fold); T66I/L74M/V151 (~8-fold); F121Y, S153Y/N155S, and T125K/S153Y/N155S (~5-fold); T66I, T125K, V151I, S153Y, T66I/M154, and T66I/S153Y, T66I/V151I (~1-fold).

In cell culture, raltegravir showed the resistance mutation Q148K which increased resistance by approximately 46-fold. When Q148K was combined with E138A, resistance increased to approximately 90-fold, and when combined with G140A, it increased to approximately 508-fold. The E138A and G140A mutations alone conferred an approximate 2-fold and zero resistance, respectively. Q148K played a major role in raltegravir resistance as it was the first mutation to occur in cell culture and all viruses with this mutation showed reduced infectivity.

In an interim analysis of 16-week data from the Phase II treatment-experienced study, Protocol 005, fifteen subjects with virologic failure from the clinical trials had full mutation profiles created. Most viruses from patients failing a raltegravir regimen tended to have a mutation at positions 148 or 155. Mutations Q148H and G140S were observed in nine subjects; Q148R was observed in three subjects; both Q148R and G140S were observed in one subject; N155H was observed in two subjects, and E92Q was observed in one subject. Secondary mutations frequently observed with the above mutations were G140S or E138A/K.

Mutations Q148H and Q148R conferred approximately 24-fold and 27-fold resistance, respectively. G140S added to the resistance if it occurred with either of the above mutations; G140S/Q148R showed approximately 405-fold increased resistance, and G140S/Q148H showed approximately 521-fold increased resistance. G140S was only observed with mutations at position 148 and so appears to be only a secondary resistance mutation, though it does appear to increase virological fitness and infectivity. The E92Q mutation, when combined with the N155H mutation, confers a 64-fold resistance and improves the infectivity of the N155H mutated virus from approximately 82% to 59%.

Three clinical isolates from virologic failure subjects were tested to ensure that integrase mutations confer resistance in the context of endogenous integrase genes and not only when introduced into a wild-type background. At virologic failure, Subject 1 had the double mutation G140S/Q148H with 211-fold reduced susceptibility to raltegravir. Subject 2 had an N155H mutation, which together with D232N, did not conform to common polymorphisms. Subject 3 was unusual in that it contained no mutations at N155 or Q148, but instead had six other changes. Substitution positions included 74, 121, and 151, and all had low-level raltegravir resistance.

The sponsor noted several important points with respect to resistance to raltegravir. It appeared that in both cell culture and clinical use, substitution of Q148 with any of the amino acids H, K, or R, conferred significant resistance and reduced infectivity of the virus. Q148 mutations appeared to be frequently linked to G140S mutations and this substantially increased resistance and improved virus infectivity. N155H alone conferred significant resistance and reduced virus infectivity but did not appear to be linked to other mutations, although its resistance was augmented and viral infectivity was increased in the presence of the E92Q mutation. It is important to watch for secondary mutations in the presence of N155H and Q148H/K/R mutations as the secondary mutations in general appeared to increase infectivity.

None of the previously identified HIV-1 integrase mutations G140S, V165I, F185K, V249I, C280S, and C280Y, were identified as conferring resistance to raltegravir. None of the mutations identified as conferring resistance to raltegravir are significant in cross-resistance to other antiretrovirals.

Safety Pharmacology

The appropriate battery of non-clinical safety pharmacology tests were conducted as per the ICH guidelines. With respect to cardiovascular effects, no meaningful effects on PR<QRS, QT, or heart-rate corrected QTc intervals were noted at any doses of raltegravir providing exposures similar or higher than those in humans. Furthermore, no significant drug-related increases in heart rate, mean arterial blood pressure, or blood flow were noted at doses of 1, 3, or 5 mg/kg in dogs.

After oral administration of 5 mg/kg raltegravir in dogs, no effect on urine output, urinary sodium, potassium excretion, or plasma electrolyte concentrations were noted. Glomerular filtration rate or filtration fraction did not appear to be affected and no emesis or changes in demeanour were noted in the 3-hour post-dose interval.

A 5 mg/kg intravenous (IV) dose of raltegravir in dogs showed no notable effect on respiratory mechanics, ventilation, blood pressure, heart rate, hemostasis, or platelet function.

Intestinal transit in the mouse was significantly increased after administration of 30 mg/kg of raltegravir compared to the vehicle alone (although the sponsor notes the vehicle group had below average transit). In a second study, gastrointestinal (GI) mobility was not significantly altered at a dose of 10 mg/kg. No adverse effects (behavioural, diarrhea) were noted.

A statistically significant but small increase in body temperature was noted in raltegravir-dosed mice during a 120-minute assessment phase. No effect on central nervous system behaviour including spontaneous activity, neural reflexes, or thermoregulation was noted after a 100 mg/kg dose.

Pharmacodynamic drug interactions

Antiviral activity was evaluated by measuring p24 antigen production in a multiple-cycle replication assay using the laboratory HIV-1 isolate H9IIIB and the human T-cell line MT4. Raltegravir did not show any antagonism with any antiretrovirals at any

concentration tested (from below-effective levels to levels at >99% inhibition). Depending on the concentrations used, interactions were all additive or synergistic. Synergism occurred at concentrations approaching IC95 and higher.

3.2.2 Pharmacokinetics

Absorption

Absorption was relatively rapid in both rats and dogs. Exposure in both species was good, with dose-proportional exposures achieved over the 40 to 120 mg/kg dose range in rats and the 5 to 45 mg/kg dose range in dogs. Absolute bioavailability in both dogs and rats was good at approximately 62% for rats and 70% for dogs. In rats it was found that the milled free phenol form of raltegravir was poorly bioavailable while the potassium salt form of the drug had reasonable and dose-proportional exposure when administered in 0.5% methylcellulose twice a day. A PEG400/water formulation yielded good exposures over the 40 to 120 mg/kg dose range without needing to be dosed twice a day, so it was selected for the sub-chronic safety studies in rats. In dogs, the potassium salt single daily dose in 0.5% methylcellulose showed good dose-proportional exposure over the dose range of 5 to 45 mg/kg. Exposure in dogs was higher than in rats.

Distribution

Single oral or IV doses of radiolabelled raltegravir were administered to rats and dogs. In rats, after a 3 mg/kg IV dose, plasma clearance was 45.5 mL/min/kg, steady-state volume of distribution was 2.2 L/kg, and half-life was 0.2 hours. After oral dosing at 6 mg/kg, Cmax was 1.7 µg at 0.4 hours post-dose. Clearance after oral dosing was 7.4 mL/min/kg in dogs.

After oral dosing, levels of radioactivity in plasma, blood, and blood cells reached maximum mean concentrations at 0.5 hours post-dose, with values of 601, 341, and 157 ng equivalents/g, respectively. Maximum mean concentrations were observed in most tissues at 0.5 hours post-dose. The highest mean concentrations were found in the GI tract and organs of excretion, particularly the stomach (30900 ng equivalents/g), small intestine (8970), liver (3910), kidney (1720), and urinary bladder (437). Maximum mean concentrations in other tissues ranged from 9.71 ng equivalents/g (brain) to 221 ng equivalents/g (lungs) at 0.5 hours post-dose. At 2 hours post-dose, maximum mean concentrations were observed in the small intestine and mesenteric lymph nodes, and by 6 hours post-dose, in the cecum and large intestine. Levels in most tissues were below the limit of quantitation by 24 hours post-dose. Aside from the GI tract, the highest concentrations of radioactivity were found in the liver, stomach at 0.5 hours, and small intestine at 2 hours. A single IV dose of raltegravir was administered to P-gp competent and deficient mice and concentrations in plasma and brain were determined. Raltegravir penetrated the brain poorly.

Metabolism

Raltegravir was mainly eliminated by metabolism in both rats and dogs, with changed drug in urine and bile accounting for 10 to 30% of the administered dose. The major metabolite detected was the phenolic glucuronide derivative of the parent compound (M2) which accounted for 62% of dose in rats and 31% in dogs. Other major metabolites included the glucose conjugate of the parent compound (M1) and the acetyl hydrazine derivative (M3).

No significant metabolism was observed in the liver microsomes fortified with NADPH from all three species tested. The major metabolite in rat and dog hepatocytes was the phenolic glucuronide conjugate of the parent compound (M2), and the same conjugate also was a major metabolite in hepatocytes from humans. Small quantities of the glucose derivative of the parent (M1) and an acetyl hydrazine derivative (M3) were also detected. The metabolic pathways were qualitatively and quantitatively similar in hepatocytes from all three species. The formation of the glucuronide metabolite M2 in human liver microsomes was characterized and UGT1A1 was found to play a major role in the glucuronidation of raltegravir. Raltegravir was not a potent reversible inhibitor (IC50 >100 μM) of CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, or CYP2B6. Similarly, raltegravir did not potently inhibit (IC50 > 50 μM) β-estradiol (3-OH) and AZT glucuronidation mediated by UGT1A1 and UGT2B7, respectively. Also, raltegravir (up to 10 mM) did not induce CYP3A4 RNA expression or CYP3A4-dependent testosterone 6β-hydroxylase activity. Raltegravir was modestly bound to plasma proteins from rat, dog, and human. Raltegravir was not an inhibitor of human P-gp. The major metabolite in mice after a 20 mg/kg oral dose was the phenolic glucuronide derivative of the parent compound (M2, 96.3% of the radioactivity recovered in bile). M2 was also a major circulating metabolite along with the unchanged parent compound.

Excretion

In rats, the total recovered radioactivity after IV administration was 70.2% (19.9% in urine and 50.3% in feces) and after oral administration, 87.7% (23.7% in urine and 64.0% in feces). In dogs, the recoveries were 88.4% from IV dosing (30.8% from urine and 57.6% from feces) and 87.1% from oral dosing (13.2% in urine and 73.9% in feces).

3.2.3 Toxicology

Acute Toxicity

Acute oral toxicity studies were conducted in mice and rats to determine the approximate lethal dose (LD50), and in dogs for a toxicokinetic (TK) analysis. Raltegravir administered to female and male mice as a single oral dose of up to 2000 mg/kg caused treatment-related mortality in a third of the male mice at 2000 mg/kg. The LD50 of raltegravir was ≥2000 mg/kg in mice. Raltegravir administered to three female rats by oral gavage produced no treatment-related mortality, physical signs, or effects on body weight at 2000 mg/kg. The LD50 for raltegravir in female rats was >2000 mg/kg. Administration of single-rising doses of up to 1000 mg/kg of raltegravir to dogs produced treatment-related emesis at ≥500 mg/kg and no mortality.

Repeat-Dose Toxicity

Repeat-dose toxicity studies with raltegravir were conducted in mice, rats, and dogs.

Raltegravir, administered by oral gavage once daily (OD) to female mice at doses up to 2500 mg/kg/day and male mice at doses up to 1000 mg/kg/day for approximately 5 weeks, caused mortality at ≥500 mg/kg/day. At the minimum lethal dose (MLD) of 500 mg/kg/day, the systemic exposure to raltegravir (45.2 μM•hr, or 13.56 μM•hr unbound drug for mice) was approximately 1.48-fold greater than the exposure (AUC) (54 μM•hr, or 9.18 μM•hr unbound drug) at the 400 mg, twice daily maximum recommended dose (BID MRD). The maximum-tolerated dose (MTD) in mice was exceeded at 500 mg/kg/day. Raltegravir, administered to mice at doses up to 5000 mg/kg/day for 14 weeks, produced treatment-related mortality at doses ≥500 mg/kg/day, as well as treatment-related physical signs and body weight loss, and/or decreases in body weight gain (-30%). Histomorphologic observations were consistent with the clinical signs of gastrointestinal bloating and were suggestive of an irritant effect from the compound. The no-effect level (NOEL) for mice was 50 mg/kg/day.

Repeat-dose oral toxicity studies of 5, 14, and 27 weeks duration were conducted in rats. Raltegravir doses up to 120 mg/kg/day caused post-dosing salivation. At doses up to 600 mg/kg/day for 5 weeks, minor increases in alanine aminotransferase (ALT) and evidence of irritation to the stomach mucosa were evident. Twice-daily dosing of raltegravir at 600 mg/kg/day did not provide a significant increase in systemic exposure to rats. Raltegravir caused mortality at 600 mg/kg/day in the 27-week study that was associated with body weight loss, urine staining, decreased food consumption prior to death, as well as decreases in mean body weight gain. Degeneration of the glandular mucosa of the stomach was seen at ≥120 mg/kg/day. Additional observations in rats included stomach inflammation in the 5-week study, and post-dosing salivation, audible respiratory sounds, and inflammation in the nose and nasopharynx in the 27-week study. Therefore, the NOEL in rats following 27 weeks of treatment was 30 mg/kg/day. The no-adverse-effect-level (NOAEL) was 120 mg/kg/day, which provided an exposure margin of 1.6-fold above the expected exposure in patients with 400 mg/BID.

Raltegravir was tested in oral toxicity studies of 5, 14, 27, and 53 week's duration in dogs. Doses up to 45 mg/kg/day were administered for 14 weeks, then subsequently doses were administered at 500 mg/kg/day for 5 weeks or 360 mg/kg/day for 53 weeks. There were no treatment-related deaths or post-mortem changes. Treatment-related changes included transient and/or intermittent emesis with or without body weight loss. Therefore, the NOEL in dogs, with the exception of emesis, following 53 weeks of treatment, was ≥360 mg/kg/day. The exposure margin was 9-fold above the expected human exposure at the 400 mg/BID MRD.

Genotoxicity

Raltegravir was not mutagenic when tested in vitro in bacteria with and without metabolic activation. It did not induce a significant increase in DNA single strand breaks in the in vitro rat hepatocyte DNA alkaline elution assay. Raltegravir treatment groups did not show statistically significant increases in the frequency of chromosome aberrations in the Chinese hamster ovary cell chromosome aberration assay with and without metabolic activation. In an in vivo mouse micronucleus assay at doses up to 1500 mg/kg, there was no evidence of an increase in micronuclei or on the proportion of polychromatophilic erythrocytes at any of the doses tested.

Carcinogenicity

The carcinogenicity studies in mice and rats are ongoing. The in-life phase of these studies is to be completed in 4Q 2007 with final study reports planned to be completed in 3Q-4Q 2008. In mice, doses up to 400 mg/kg/day (females) or 250 mg/kg/day (males) were used. In rats, doses up to 300 mg/kg/day (males) or 600 mg/kg/day (females) were selected. Preliminary results up to 76 weeks showed increasing mortality of up to 56% in female mice and 46% in female rats. Effects of drug irritation to the nose/nasopharynx including inflammation, epithelial hyperplasia, and squamous metaplasia were seen in rats. Six neoplasms of the nose/nasopharynx were detected in rats. Similar effects of drug irritation to the nose, nasopharynx, and trachea including inflammation, epithelial hyperplasia, and squamous metaplasia were seen in mice; however, no malignancies were detected. In a 27-week TK study with mice, systemic exposure at the high dose was approximately 1.8-fold (females), or equal to (males), the human AUC at the 400 mg/BID MRD. A 26-week parallel TK study in rats indicated that at 300-600 mg/kg/day (female), or 150-300 mg/kg/day (male), systemic exposure was approximately 10-fold (female), or 1.7-fold (male), of the human AUC at the 400 mg/BID MRD.

Reproductive and Developmental Toxicity

Administration of raltegravir orally OD to rats [females: for 14 days prior to and during co-habitation, and through gestation day (GD) 7; males: from prior to co-habitation for approximately 8 weeks] with doses up to 600 mg/kg/day did not demonstrate any treatment-related effects on mating performance, fertility parameters, F1 embryonic/fetal survival, sperm count, or sperm motility. No treatment-related gross changes in the thoracic or abdominal cavities, testicular weights, or gross or microscopic changes in the testes or epididymides were observed. The NOEL for effects of raltegravir on fertility in F0 female and male rats was ≥600 mg/kg/day.

Raltegravir, administered orally OD to pregnant rats at doses up to 600 mg/kg/day from GD 6 through lactation day (LD) 20, did not cause treatment-related changes. Following raltegravir administration to female rats, doses up to 600 mg/kg/day from GD 6 to 20 or from GD 6 to LD 20 showed no treatment-related findings in the F0 and F2 generation. Treatment-related increases in incidences of supernumerary ribs were observed in the F1 generation of the 600 mg/kg/day group. The NOEL for maternal toxicity was ≥600 mg/kg/day. The NOEL for developmental toxicity was 300 mg/kg/day (3.4-fold the exposure in patients receiving the 400 mg/BID MRD).

There were no indications of maternal or developmental toxicity following oral administration OD to rabbits at doses up to 1000 mg/kg/day from GD 7 to 20. The NOEL of raltegravir for maternal and developmental toxicity in rabbits was ≥1000 mg/kg/day.

Doses up to 600 mg/kg/day were orally administered to juvenile rats from post-natal day (PND) 5 to postnatal week (PNW) 8. No treatment-related changes were observed except for treatment-related histomorphologic findings consisting of vacuolation of the nonglandular mucosa at the limiting ridge and inflammation at ≥200 mg/kg/day. The NOEL for juvenile rats was 50 mg/kg/day. The findings in juvenile rats were consistent with the stomach irritation effects seen in adult rats. There were no additional toxicities noted in juvenile rats indicating that, overall, raltegravir effects were similar between juvenile and adult rats.

Local Tolerance

Raltegravir potassium was non-irritating based on 104.2% cell viability in an in vitro human epidermal skin culture system (EpiDerm™).

Based upon assessment of the induction of opacity and changes in permeability to fluorescein (irritation score) with raltegravir tested as a 20% solution, raltegravir free base was classified as a mild irritant and raltegravir potassium salt was classified as a severe irritant in the bovine corneal opacity (BCOP) in vitro assay.

Groups of mice were treated topically with raltegravir, later injected with tritiated thymidine or BrdU, sacrificed and incorporation of radioactivity in the auricular lymph node measured as an index of sensitization potential. No significant increase in cell proliferation in the lymph node was detected, and as such, raltegravir, as either free base or potassium salt, was not considered a dermal sensitizer using the local lymph node assay.

The dermal irritation potential of raltegravir as a free base or potassium salt was determined following application of the drug to the intact skin of New Zealand White (NZW) rabbits. No evidence of dermal irritation was found in treated rabbits. Raltegravir, as either a free base or potassium salt, was not considered a dermal irritant based on the results of this study.

Other Toxicity Studies

The phototoxicity potential of raltegravir was determined after oral administration to female mice at doses up to 2000 mg/kg as a single dose then exposing mice to UVB, UVA, and visible light. Treatment-related mortality, body weight effects, or phototoxic effects were not noted. Treatment-related decreased activity was observed at 2000 mg/kg. Raltegravir was considered non-phototoxic at doses up to 2000 mg/kg.

Raltegravir was negative for in vitro hemolysis of human, dog, or rat erythrocytes prior to initiation of the intravenous (IV) toxicity studies. Raltegravir administered as a single IV injection to female rats at doses up to 1600 mg/kg caused treatment-related mortality associated with physical signs at ≥200 mg/kg. Evidence of drug irritation at the injection site was observed at >100 mg/kg. In a range-finding study, dogs received IV rising doses of 40 and 100 mg/kg/day for three consecutive days each. One dog died on Day 1 during IV administration of the 40 mg/kg dose. The death was attributed to cardiac arrhythmia secondary to the amount of administered potassium. Increases in alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) at 100 mg/kg/day were observed. Raltegravir IV administered to dogs OD at doses of 30 or 100 mg/kg/day for seven days produced primary changes at both doses including transient post-dosing emesis/retching and local changes at the injection sites, as well as body weight loss, minimal increases in serum urea nitrogen, increases in ALT (up to 1967%), ALP (up to 374%), and cholesterol, and very slight, multifocal tubular dilatation in the cortex of the kidneys at 100 mg/kg/day. At the NOEL, the safety margin was 6.5-fold that of the expected exposure of the 400 mg/BID MRD in humans.

Raltegravir was administered to rats at 30, 120, 600 mg/kg/day by oral gavage OD for approximately 5 weeks. Treatment-related salivation at the mid- and high-dose levels was observed. Raltegravir caused very slight irritation to the limiting ridge of the non-glandular stomach accompanied by a very slight increase in inflammation at 600 mg/kg/day. The NOEL for the local effect on the stomach was 120 mg/kg/day. Administration of raltegravir containing 0.03% of the acid impurity/degradate to rats for one month did not cause any unique toxicity relative to raltegravir alone, nor was the NOEL for the gastric irritation significantly altered. Results from this study support a Drug Product Specification limit for the acid impurity/degradate up to 0.3% (on mg/kg basis).

No toxicity studies were conducted to assess antigenicity, immunotoxicity, or dependence. No studies were conducted to evaluate specific metabolites.

3.2.4 Summary and Conclusion

Raltegravir was found, in vitro, to be a selective inhibitor of strand transfer activity of purified recombinant HIV-1 integrase with no HIV-1 reverse transcriptase inhibitory activity. The non-clinical pharmacology and pharmacokinetic studies were considered to be thorough and acceptable. No major areas of concern were identified.

The toxicity profile of raltegravir after repeated oral dosing was characterized in rats, mice, and dogs and demonstrated that raltegravir was well tolerated in animals. Doses used in toxicity studies provided adequate systemic exposure and margins of safety were determined for each of the toxicities identified. Raltegravir was not genotoxic in a battery of in vitro assays in bacteria and mammalian cells or in vivo in mice designed to detect mutagenicity, direct DNA damage, or elastogenicity. An assessment of the carcinogenic potential of raltegravir is ongoing, preliminary results up to Week 76 indicated increased mortality, evidence of drug irritation, and neoplasms. The relevance of these findings to human safety will be assessed in the clinical studies. Raltegravir was shown not to pose a significant hazard to reproduction or to the developing fetuses based on studies in rats and rabbits.

3.3 Clinical basis for decision

3.3.1 Human Pharmacology

The clinical pharmacology program characterized the initial safety and tolerability as well as pharmacokinetic (PK) characteristics of raltegravir. The program included 18 Phase I studies and included healthy male and female subjects, patients with hepatic insufficiency, and patients with renal insufficiency, and a full PK evaluation of a subset of HIV patients enrolled in a Phase II study. The PK analysis included a composite analysis across many of the Phase I studies as well as a limited population PK analysis of data from the Phase I and the Phase II/III program in HIV-infected individuals. One study was conducted to assess the effect of raltegravir on the QTc interval.

Raltegravir was generally tolerated up to 1600 mg administered as a single dose and 800 mg administered every 12 hours (q12 hr) for 10 days. There were no serious adverse events (SAEs) observed. All adverse events were generally transient and mild to moderate in intensity. There were two discontinuations due to study drug (including raltegravir and co-administered agents). No clinically important abnormalities were noted in routine blood and urine chemistry panels, complete blood count, electrocardiograms, and physical examinations including vital signs. In the thorough QTc study, there was no statistically significant effect on QTc intervals relative to placebo following a single dose of 1600 mg raltegravir. In an oral clinical absorption, distribution, metabolism, and excretion (ADME) study, approximately 32% of the radiolabelled oral dose was recovered in urine. Similarly, the animal data suggested that the bioavailability may be on the order of 60 to 70%.

The data indicated that the main mechanism of clearance of raltegravir in humans is UGT1A1-mediated glucuronidation, with a small component (~9% of the administered dose eliminated as parent compound) of elimination via renal excretion.

Raltegravir displayed dose proportional PK over the dose range of 100 to 800 mg. At the clinical dose of 400 mg, the apparent terminal elimination half-life was approximately 9 hours with a shorter α-phase half-life (~1 hour) accounting for much of the AUC. Steady-state was generally reached in approximately two days and accumulation was slight with multiple-dose administration; the estimated AUC accumulation ratio was 1.05 with twice daily dosing. Raltegravir was relatively rapidly absorbed with a Tmax of approximately 3 hours in the fasted state.

For the purposes of assessing the impact of extrinsic and intrinsic factors (e.g., concomitant drugs, gender, age, body mass index, hepatic function, renal function, race, and HIV status) on raltegravir PK, a ≥2-fold increase in raltegravir AUC or a ≥60% decrease C12hr was considered a clinically meaningful alteration. C12hr is considered to be the most sensitive PK parameter to predict viral response. Within the concentration range studied, the virologic response was similar for patients with lower C12hr values compared to those with higher C12hr values. On the other hand, AUC or Cmax are the PK parameters most likely to be associated with toxicity. There were no safety issues identified that were associated with the observed high Cmax values within the dose range studied. Therefore, AUC was considered to be the most appropriate PK parameter from the safety perspective. During the Phase I and II studies, AUC values twice that achieved with the proposed 400 mg dose were observed and were well tolerated. This suggests that a 2-fold increase in AUC can be justified as there was no increased incidence of adverse events; however, this rationale does not seem to be clinically relevant based on the available clinical experience and the high range of within-subject variability in PK values for raltegravir. The Product Monograph highlights areas of uncertainty and duly cautions prescribers of the risks.

Gender, age, body mass index, race, HIV status, moderate hepatic insufficiency, and severe renal insufficiency had no clinically meaningful effect on raltegravir PK. No dose adjustment is required for raltegravir based on any of these demographic factors.

Administration with Food

The effect of food on the absorption of raltegravir was examined in clinical studies. The extent of absorption of the FMI formulation, as assessed by area under the concentration time curve from 0 to infinity (AUC0-∞), was similar in the fed (high-fat meal) and fasted states, although food appeared to slow the rate of absorption and extend the duration of absorption. This resulted in an approximately 34% decrease in maximum plasma concentration (Cmax), an 8.5-fold increase in trough plasma concentration at 12 hours (C12hr), and a 7.3 hour delay in time to maximum plasma concentration (Tmax). A cross-study comparison of multiple-dose PK suggested that the magnitude of the food effect was diminished following multiple-dosing and when administered with a standard, rather than high-fat, meal. Food appeared to increase PK variability somewhat over the fasted state and consequently, a similar lower range of individual C12hr values was observed in the cross-study comparison. This suggests that food does not consistently increase C12hr values. On the basis of the clinical significance criteria, food does not have a clinically meaningful effect on the PK profile of raltegravir, relative to fasted administration.

Drug Interactions

In vitro results indicated that raltegravir was not an inhibitor of the major cytochrome P450 (CYP) isozymes, including CYP3A4, major UDP-glucuronosyltransferases (UGTs), and P-glycoprotein. Additionally, raltegravir was not an inducer of CYP3A4. In clinical studies, raltegravir did not meaningfully alter the PK of midazolam, suggesting that raltegravir has a low propensity for causing drug interactions with other substrates of CYP3A4. Additionally, raltegravir did not meaningfully alter the PK of tenofovir (TFV), TMC125, and lamivudine (3TC). A number of clinical studies were conducted with agents known to be broad inducers (e.g. ritonavir, efavirenz, rifampin and tipranavir+ritonavir combination) and inhibitors (e.g. atazanavir alone or in combination with ritonavir) of drug metabolizing enzyme UGT1A1. As anticipated, raltegravir plasma levels were increased following co-administration with atazanavir alone or in combination with ritonavir. However, no change in safety profile of raltegravir was observed and the administered concentrations were well tolerated. Based on these data, raltegravir may be co-administered with atazanavir and no dose adjustment is required.

Co-administration with rifampin and tipranavir+ritonavir resulted in the reduction of raltegravir exposure, which could be due to the combination of UGT1A1 and P-gp induction. Due to the observed high variability in raltegravir PK parameters, a dose adjustment for raltegravir based on these results can not be extrapolated to other inducers such as phenytoin and phenobarbitol until further data is available.

Co-administration of raltegravir with tipranavir in combination with ritonavir also resulted in decreased plasma raltegravir exposure; however, efficacy was not altered.

Efavirenz had a modest influence on the PK profile of raltegravir. There was a modest reduction in C12hr (21%), which was probably due to slight induction of UGT1A1. The 90% confidence interval (CI) for these effects indicated that the magnitude is small and not likely to be clinically meaningful. The lack of substantive effect on AUC, Cmax, and Tmax suggested that the overall effect was minor and unlikely to be clinically meaningful. Based on this data, efavirenz may be co-administered with raltegravir without dose adjustment.

3.3.2 Clinical Efficacy

The clinical safety and efficacy of Isentress™ were assessed based on two pivotal Phase III studies (Protocols 018 and 019) and two supportive Phase II studies (Protocols 004 and 005). Protocol 004 was conducted in treatment-naïve subjects, and therefore primarily contributed safety data. The studies involved 1075 subjects, including 877 treatment-experienced subjects. The two Phase III pivotal studies enrolled 699 treatment-experienced triple-class resistant subjects, including 462 raltegravir-treated subjects. Triple-class-resistance was defined as documented resistance to at least one drug from each of the three classes of approved oral antiretrovirals (NRTIs, NNRTIs, and PIs).

In the pooled results of Protocols 018 and 019, raltegravir was superior to placebo when both were combined with individually optimized background treatment (OBT). For the primary efficacy endpoint, at Week 16 based on Non-Completer=Failure (NC=F), 77.3% of raltegravir-treated subjects had HIV RNA levels <400 copies/mL, compared to 41.8% of placebo-treated subjects. Response was sustained in the approximately 60% of subjects for whom 24-week data was available, with 75.5% of the raltegravir subjects having HIV RNA <400 copies/mL, versus 39.3% of the placebo subjects. Results were comparable for the secondary endpoint of HIV RNA <50 copies/mL, with 61.7% of raltegravir-treated subjects having a viral load of <50 copies/mL at Week16 compared to 34.6% for the placebo subjects. Again, this was sustained through 24 weeks, based on the partial results, with 62.6% of raltegravir subjects having HIV RNA <50 copies/mL, versus 33.3% of the placebo subjects.

Raltegravir-treated subjects also displayed superior immune reconstitution, with a mean increase of 83.9 CD4 cells/mm3 seen at Week 16 for the raltegravir group versus 35.6 CD4 cells/mm3 for the placebo group. At Week 24, this had increased to 89.2 CD4 cells/mm3 for the raltegravir subjects versus 35.1 CD4 cells/mm3 for the placebo subjects.

Efficacy results for the Phase II trial Protocol 005, supported the Phase III studies with 71.1% of subjects in the raltegravir + OBT group having HIV RNA <400 copies/mL compared to 15.6% of subjects in the placebo + OBT group at Week 24. The lower response rate seen in the placebo group of Protocol 005 compared to Protocols 018 and 019 is likely due to the inclusion of darunavir and tipranavir (investigational drugs at the time of the studies) in the OBT group in the Phase III studies but not in Protocol 005.

3.3.3 Clinical Safety

The clinical safety of Isentress™ was assessed based on two pivotal Phase III studies (Protocols 018 and 019) and two supportive Phase II studies (Protocols 004 and 005) as described above in Section 3.3.2 Clinical Efficacy.

Raltegravir was generally well tolerated, with a side-effect profile comparable to placebo + OBT-treated subjects. Initially, concern arose regarding an excess of malignancies in raltegravir-treated subjects, however the imbalance in rates of malignancies seen in the original submission has not been sustained with monitoring of the ongoing clinical trials. Analysis is consistent with an initial paucity of tumours in the placebo-treated subjects; moreover, the majority of malignancies reported in both treated and placebo groups are those expected in a population with advanced HIV-infection (i.e. Kaposi's sarcoma, lymphoma, etc.). A slight excess of rash associated with raltegravir was also seen in the Phase III studies, however the cases were generally mild-to-moderate in severity, and did not result in drug discontinuation. No severe rash events (i.e. Stevens-Johnson Syndrome) were reported, and the pattern of rash adverse events does not suggest a hypersensitivity phenomenon. A slight excess of elevations of creatinine kinase was noted in the raltegravir-treated subjects. A single episode of rhabdomyolysis was seen in both the placebo and the raltegravir-treated groups during the trial period covered by the submission, however at least two additional cases of rhabdomyolysis and two cases of myopathy have since been reported in raltegravir-treated subjects in ongoing trials. The occurrence of CK elevations, myopathy, and rhabdomyolysis is mentioned specifically in the Adverse Reactions section of the Product Monograph. There was no evidence of an increased risk of elevations of hepatic transaminases in raltegravir-treated subjects, although, like the comparator subjects, elevations were slightly more common in hepatitis B and/or C co-infected subjects.

The sponsor has submitted a risk-management plan to satisfy the requirements of other regulators. In addition to standard pharmacovigilance measures, the sponsor proposes active surveillance in the form of a prospective post-licensure observational cohort surveillance study to monitor malignancies and other important safety issues. The Risk Management Plan has been requested as part of the Notice of Compliance with Conditions requirements for full NOC.

3.3.4 Additional Issues

Isentress™ has demonstrated promising evidence of efficacy and safety, however several conditions are required as part of the market authorization. As part of the authorization with conditions for Isentress™, the sponsor has agreed to the following conditions:

  • Submit confirmatory studies that include safety and efficacy results at 48-weeks for clinical studies P018 and P019, and the follow-up of adverse events from these studies as well as events noted in post-marketing data and any other major safety concerns identified.
  • Provide a Risk Management Plan for Isentress™.
  • Submit Periodic Safety Update Reports (PSURs) semi-annually until the conditions have been fulfilled and removed from the NOC/c by Health Canada.
  • Provide an analysis of any medication error reports occurring with Isentress™ semi-annually until the conditions have been fulfilled and removed from the NOC/c by Health Canada.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk assessment

Raltegravir (Isentress™) is the first inhibitor of the HIV integrase enzyme to be considered for licensing in any jurisdiction. Based on the evidence of safety and efficacy of raltegravir for the treatment of HIV in treatment-resistant patients with limited option, the benefits of raltegravir are felt to outweigh its possible risks. Results demonstrated superior efficacy of raltegravir compared to placebo at 16 weeks, and based on the available data, virologic response and immune reconstitution appeared to be sustained through 24 weeks. Raltegravir appeared to be generally well tolerated, with a safety profile comparable to that seen in the placebo subjects. A Risk Management Plan has been requested as part of the Notice of Compliance with Conditions requirements for full NOC, which will provide active post-market surveillance measures to monitor malignancies and other important safety issues.

3.4.2 Recommendation

Based on the Health Canada review of data on quality, safety and effectiveness, Health Canada considers that the benefit/risk profile of Isentress™, in combination with other antiretroviral agents, is favourable in the treatment of HIV-1 infection in treatment-experienced adult patients who have evidence of viral replication and HIV-1 strains resistant to multiple antiretroviral agents. This New Drug Submission (NDS) qualifies for authorization under the Notice of Compliance with Conditions Policy.

As part of the authorization with conditions for Isentress™, the sponsor has agreed to the following conditions:

  • Submit confirmatory studies that include safety and efficacy results at 48-weeks for clinical studies P018 and P019, and the follow-up of adverse events from these studies as well as events noted in post-marketing data and any other major safety concerns identified.
  • Provide a Risk Management Plan for Isentress™.
  • Submit Periodic Safety Update Reports (PSURs) semi-annually until the conditions have been fulfilled and removed from the NOC/c by Health Canada.
  • Provide an analysis of any medication error reports occurring with Isentress™ semi-annually until the conditions have been fulfilled and removed from the NOC/c by Health Canada.

4 Submission Milestones

Submission Milestones: IsentressTM*

Submission MilestoneDate
Request for priority status
Filed2007-03-15
Approval issued by Director, BGIVD2007-04-02
Submission filed2007-04-18
Screening
Screening Acceptance Letter issued2007-05-14
Review 1
Quality Evaluation complete2007-11-07
Clinical Evaluation complete2007-11-06
Labelling Review complete2007-11-13
NOC/c-QN issued2007-11-09
Response filed2007-11-14
NOC issued by Director General under the NOC/c Policy2007-11-27