Summary Basis of Decision for Cubicin ®

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
Cubicin®

Daptomycin, 500 mg/vial, Powder for solution, Intravenous

Cubist Pharmaceuticals, Inc.

Submission control no: 102320 / 104227

Date issued: 2008-07-31

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), PrCUBICINMD, Daptomycine, 500mg/flacon, poudre pour solution, Cubist Pharmaceuticals, Inc., N° de contrôle de la présentation 102320 / 104227

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:

Cubicin®

Manufacturer/sponsor:

Cubist Pharmaceuticals, Inc.

Medicinal ingredient:

Daptomycin

International non-proprietary Name:

Daptomycin

Strength:

500 mg/vial

Dosage form:

Powder for solution

Route of administration:

Intravenous

Drug identification number(DIN):

  • 02299909

Therapeutic Classification:

Antibacterial agent

Non-medicinal ingredients:

Sodium hydroxide

Submission type and control no:

New Drug Submission, Control No. 102320 / 104227

Date of Submission:

2005-11-14

Date of authorization:

2007-09-24

® CUBICIN used under license by Cubist Pharmaceuticals, Inc.

2 Notice of decision

On September 24, 2007, Health Canada issued a Notice of Compliance to Cubist Pharmaceuticals, Inc. for the drug product Cubicin®.

Cubicin® contains the medicinal ingredient daptomycin which is an antibacterial agent.

Cubicin® is indicated for the following infections in adults:

  • Complicated skin and skin structure infections (cSSSI) caused by susceptible strains of the following Gram-positive microorganisms: Staphylococcus aureus (including methicillin-resistant strains), Streptococcus pyogenes, and Streptococcus agalactiae.

  • Staphylococcus aureus bloodstream infections (bacteremia), including those with right-sided Staphylococcus aureus infective endocarditis (native valve) caused by methicillin-susceptible and methicillin-resistant strains.

Cubicin® is not indicated for the treatment of pneumonia. The safety and efficacy of Cubicin® in patients under the age of 18 have not been established.

Patients with cSSSI or bacteremia/endocarditis infections are initially hospitalized because of disease severity. Increasing bacterial resistance to conventional drugs is an emerging health issue with these infections. Daptomycin binds to Gram-positive bacterial membranes in a calcium-dependant manner and causes a rapid depolarization of membrane potential. This loss of membrane potential causes inhibition of protein, DNA, and RNA synthesis, which results in bacterial cell death.

The market authorization was based on quality, non-clinical, and clinical information submitted. Efficacy and safety of Cubicin® in patients with cSSSI was examined in two pivotal trials. A total of 1092 adults were treated with either Cubicin® (534) or a comparator drug (558) for 1-2 weeks; efficacy was assessed at 7-12 days post-treatment. Safety was assessed throughout the study period. Cubicin® was found to be as safe and effective against the listed causative organisms as conventional antibiotic treatments in a representative sample of infections and adults. The drug was well tolerated; most side effects were mild to moderate and reversible. One pivotal trial was used to assess the safety and efficacy of Cubicin® compared to conventional therapy in patients with S. aureus bacteremia with known or suspected S. aureus infective endocarditis. A total of 235 adult patients received study medications; 120 received Cubicin® while 115 received a comparator drug for 10-42 days. Cubicin® was found to be non-inferior to the comparator at Test-of-Cure, assessed at 6 weeks post-treatment. There was no significant difference in time to clearance of bacteremia between the groups. The drug was well tolerated; most side effects were mild to moderate and reversible.

Cubicin® (500 mg/vial, daptomycin) is presented as a powder for solution. For patients with cSSSI, 4 mg/kg of Cubicin® should be administered over a 30-minute period by intravenous (IV) infusion in 0.9% sodium chloride injection, once every 24 hours for 7-14 days. For patients with bacteremia, 6 mg/kg Cubicin® should be administered over a 30-minute period by IV infusion in 0.9% sodium chloride injection, once every 24 hours. Duration of treatment should be based on the treating physician's working diagnosis. There are limited safety data for the use of Cubicin® for more than 28 days. Dosing guidelines are available in the Product Monograph.

Cubicin® is contraindicated for patients with known hypersensitivity to daptomycin. Cubicin® 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 Cubicin®, precautions, and recommended patient monitoring are described in the Product Monograph.

Based on the Health Canada review of data on quality, safety, and effectiveness, Health Canada considers that the benefit/risk profile of Cubicin® is favourable for the indications stated above.

3 Scientific and Regulatory Basis for Decision

The initial submission for Cubicin® was submitted on November14,2005 (Submission No.102320), which was followed by a second submission (Submission No.104227) on February22,2006 for a second treatment indication. The two submissions were cross-referenced during the review process. Both submissions for Cubicin® received a Notice of Non-Compliance(NON) on February7,2007 on the basis that the sponsor did not provide sufficient data to support the manufacturing process for the drug substance and to qualify the proposed impurity limits in both the drug substance and the drug product. A response to the NON was filed by the sponsor, which was consulted on with the Clinical division. Based on the information received for the clinical consultation, the proposed impurity limits were deemed acceptable, and the issues identified in the NON were satisfactorily resolved. A single Product Monograph was developed for these submissions and information on both indications is included in this SBD.

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Daptomycin, the medicinal ingredient of Cubicin®, is a cyclic lipopeptide antibacterial agent. Daptomycin binds to Gram-positive bacterial membranes in a calcium-dependent manner and causes a rapid depolarization of membrane potential. This loss of membrane potential causes inhibition of protein, DNA, and RNA synthesis, which results in bacterial cell death.

  • Cubicin® is indicated for the following infections in adults:

  • Complicated skin and skin structure infections (cSSSI) caused by susceptible strains of the following Gram-positive microorganisms: Staphylococcusaureus (including methicillin-resistant strains), Streptococcuspyogenes, and Streptococcusagalactiae.

  • Staphylococcusaureus bloodstream infections (bacteremia), including those with right-sided Staphylococcusaureus infective endocarditis (native valve) caused by methicillin-susceptible and methicillin-resistant strains.

Cubicin® is not indicated for the treatment of pneumonia. Daptomycin activity invitro is inhibited in the presence of pulmonary surfactants.

Manufacturing Process and Process Controls

Daptomycin 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.

The manufacturing process is considered to be adequately controlled within justified limits.

Characterization

Detailed characterization studies were performed to provide assurance that daptomycin consistently exhibits the desired characteristic structure.

The structure of daptomycin 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 toxicological studies and therefore, are considered to be acceptable.

Control of Drug Substance

The drug substance specifications for daptomycin 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 daptomycin.

The levels of product- and process-related impurities were adequately monitored throughout the manufacturing process. Results from process validation reports and in-process controls indicated that the impurities of the drug substance were adequately under control. The levels of impurities reported for the drug substance were found to be within the established limits.

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

The drug product, Cubicin® is supplied as a sterile, pale yellow to light brown lyophilised cake for intravenous administration following reconstitution with 0.9% sodium chloride for injection. Cubicin® is presented as vials of 500 mg of daptomycin. Vials are 10mL Type I flint glass vials, with 20mm bromobutyl rubber closures and 20mm aluminum seals with flip-off coloured plastic caps. Sodium hydroxide may also be found in Cubicin® in amounts of 25µL/g of daptomycin as a result of pH adjustment.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations. The excipients included in the formulation comprise a vehicle (water for injection), a pH adjuster (sodium hydroxide), and an inert gas employed during lyophilization (nitrogen). The compatibility of daptomycin 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 Cubicin®, rationale for choice of formulation, manufacturing process including packaging, information on batches used in invitro studies for characterization and the microbiological quality of Cubicin®. 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 bulk drug substance is a fermentation product manufactured using conventional pharmaceutical equipment and facilities. The validated process is capable of consistently generating product that meets release specifications.

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

Cubicin® is tested to verify that the identity, appearance, content uniformity, levels of degradation products, microbiological impurities, potency, and pH 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.

Data from final batch analyses were reviewed and are considered to be acceptable according to 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 36-month shelf-life at 2-8°C for Cubicin® is considered acceptable.

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 facility 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

Not applicable. Cubicin® does not contain material of animal or human origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Cubicin® 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

Pharmacodynamic (PD) studies were conducted in mouse models of soft tissue (thigh) infection using Staphylococcusaureus. These PD studies indicated that the pharmacokinetic(PK) drug exposure parameter (AUC) and the maximum plasma concentration parameter(Cmax) predicted efficacy. The AUC/MIC ratio and Cmax/MIC ratio correlated better with efficacy than the time above MIC(T>MIC), consistent with the concentration dependent bactericidal activity of daptomycin. Efficacy was obtained at clinically relevant 24-hour AUC exposures.

Monte Carlo analysis was performed to calculate probabilities of daptomycin achieving AUC/MIC criteria. Simulation for each pathogen group with the two variables applied against the total AUC/MIC pharmacodynamic standards were conducted in mouse models of thigh infection. The representative AUC/MIC criteria for efficacy used in the calculations were 180 and 160 for Staphylococcus aureus and Streptococcus species, respectively. Monte Carlo modeling indicated a high probability of efficacy with the exposure generated by the 4 mg/kg clinical dose against Staphylococcus aureus (99.9%), and Streptococcus species (99.9%).

Efficacy of daptomycin was studied in animal models of infection including the proposed clinical indication of skin and skin-structure infections and endocarditis, as well as in septicaemia, and pulmonary infections in mice, rats, rabbits, guinea pigs, and hamsters. Efficacy with 4 mg/kg q24h dose was achieved with staphylococci and streptococci in skin and soft tissue infections. Efficacy was also demonstrated with 6 mg/kg q24h in blood stream infections due to Staphylococcus aureus, with and without infective endocarditits. The clinical dosage of 4 mg/kg q24h which produced a mean Cmax of 57.8 ug.hr/mL, achieved the PD values predictive of efficacy against Staphylococcus aureus and Streptococcus species. Lack of efficacy of daptomycin has been demonstrated in broncho-alveolar pneumonia because of the interaction of daptomycin with pulmonary surfactant (a primary component of the epithelial lining fluid). Efficacy of daptomycin was similar to vancomycin for the treatment of MRSA (Methicillin-Resistant Staphylococcus aureus) osteomyelitis.

3.2.2 Pharmacokinetics

The pharmacokinetics (PK) of daptomycin were investigated in several animal species to characterize the absorption, distribution, metabolism, and excretion (ADME) profile.

Absorption

Upon bolus IV injection, daptomycin exhibits first order kinetics independent of species. Peak plasma concentrations and AUC were dose proportional. Repeated once-daily injection of daptomycin for 14 days to 6 months had no impact on pharmacokinetic parameters in dogs or monkeys.

The plasma concentrations achieved following oral administration of daptomycin in rats was low and in the range of 0.05 - 0.2 µg/mL. The overall results of studies in rats indicated that greater than 90% of orally administered daptomycin passes through the gastrointestinal (GI) tract without any absorption.Non-intravenous (IV) parenteral administration of daptomycin by subcutaneous (SC) or intraperitoneal (IP) routes at therapeutic dose levels indicated high bioavailability. In mice, the systemic bioavailability after SC administration of 1-25 mg/kg was 94-100%, but decreased to 76% when the dose level was increased to 100 mg/kg.

Distribution

The disposition of IV administered daptomycin (10 to 150 mg/kg) was investigated in mice, rats, monkeys, and dogs. In general, the plasma-drug concentration profile was very similar in different species. There was a linear dose-response up to the peak plasma concentration of daptomycin. The plasma attained the highest drug concentration as compared to any other tissue, and also had the shortest half-life.The plasma profile of daptomycin following IV administration was consistent with the two-compartmental model with a rapid distribution phase and slower elimination phase. The apparent volume of distribution of daptomycin was low. The kidney and lung attained the highest concentrations of daptomycin as compared to other tissues. The half-life of the drug in the lungs was similar to the plasma, while the kidney had a longer tissue half-life. In most of the tissues the half-life of the drug ranged between 2-4 hours.

Tissue distribution studies in rats with radiolabelled daptomycin (10 mg/kg) showed initial high concentrations of radiocarbon in the kidney and urine identifying the kidney as the primary route of elimination. Radiocarbon remained distinctly distributed within the renal cortex of the kidney indicating retention after a single dose. Lower levels of radiocarbon were also detected in the GI contents, which indicated potential elimination of daptomycin by the hepatic route.The exposure (AUC) values for most tissues were higher after multiple-dose administration as compared to single-dose administration. This indicated some degree of accumulation of the radiocarbon. However, the exposure levels of the kidney to daptomycin and/or its metabolite were 10- to 67-fold higher as compared to any other tissue following multiple-dose administration.

Metabolism

Daptomycin exhibited limited metabolism by spontaneous or enzymatic degradation in plasma, or by hepatic metabolic pathways. Across all species tested, a small amount was excreted in the urine as inactive metabolite (<20% of the dose) and the majority of the drug was excreted as intact active drug. In mice, the radioactivity profile of urine showed two peaks when administered with labelled daptomycin. One peak was daptomycin and the second peak was not identified. No metabolites have been identified in the plasma or urine of the rat, dog, or monkey.

Excretion

Daptomycin was excreted primarily in the urine in all species examined. Approximately 70-80% of the administered radioactivity was recovered in the urine by 48 hours post-dose in the mouse, rat, dog, and monkey. Studies conducted in rats with radiolabelled daptomycin indicated that approximately 15% of the drug was excreted in feces. The urine samples collected during the first 24 hours post-dose had one major peak which corresponded to the parent drug and was biologically active. It was not investigated whether any metabolites were present in fecal samples.

Special Populations

The effect of renal impairment on the PK of daptomycin was investigated rats. The study demonstrated that renal impairment decreases systemic clearance of daptomycin and confirms the importance of kidney function in the excretion of daptomycin. Non-clinical studies indicated that modification of the dose regimen may be warranted in patients with severe renal impairment. There were no PK studies conducted with animal models of hepatic impairment. However, as hepatic metabolism of daptomycin is limited, hepatic impairment may have minimal effects on the PK of daptomycin.

Drug-Drug Interactions

The potential drug interactions of daptomycin and simvastatin on skeletal muscle were investigated in rats. The results of the investigation indicate that there was no observed drug interaction to have effect on skeletal muscle upon co-administration of daptomycin and simvastatin to rats at clinically relevant doses. The study has been described in detail in the Product Monograph.

The interaction of tobramycin and daptomycin on skeletal muscle was investigated in rats. The study indicated that mild skeletal muscle degeneration and/or regeneration was observed with 20 mg/kg daptomycin when administered alone. During concurrent administration with tobramycin, mild skeletal muscle changes were observed at dose levels ‑5 mg/kg daptomycin. This indicates a weak potentiating effect of tobramycin on muscle damage caused by daptomycin. This interaction is captured in the drug-drug interaction section of the Product Monograph.

The interaction between daptomycin and gentamicin with regard to nephrotoxicity was investigated in dogs. Administration of daptomycin alone did not induce nephrotoxicity. There was a slight increase in relative kidney weight. Gentamicin treatment alone was associated with expected evidence of dose-related nephrotoxicity, including changes in serum chemistry, urine chemistry, and kidney weights, as well as histopathological lesions. When high dose daptomycin (30 mg/kg) was administered with a high dose of gentamicin (30 mg/kg), blood urea nitrogen and creatinine increased two fold over that observed with gentamicin alone and the severity of the microscopic renal lesions observed was graded as minimal to moderate. It is not known if these results were due to competitive interaction of two renally excreted drugs, leading to increased systemic exposure to gentamicin. At a clinically relevant dose, the combination of daptomycin and gentamicin caused only a minor increase in the severity of the lesions with no functional consequence. This interaction is captured in the detailed pharmacology section of the Product Monograph.

3.2.3 Safety Pharmacology

Daptomycin was evaluated for its pharmacological effects on cardiovascular, respiratory, renal, gastrointestinal (GI), central nervous system (CNS), and immune systems in rodents and dogs. The effect of daptomycin on the CNS was evident at high dose levels. In mice, there was a consistent decrease in motor activity at dose levels of 100 mg/kg and greater. At 200 mg/kg, the drug reduced the hypothermic effect of apomorphine and also decreased acetic acid-induced writhing. At ≥200 mg/kg, there were tremors and convulsions. Daptomycin also prolonged hexobarbital sleeping time at a dose level of 25 mg/kg which reached statistical significance at 200 mg/kg. An inhibition of hexobarbital metabolism may explain the prolonged sleeping time. The CNS effects of daptomycin in rats were similar to those in mice. Doses of 15 and 50 mg/kg had no effect on thiopental-induced sleep time or loss of motor coordination. However, these effects were seen when the dose levels of daptomycin were increased to 150 mg/kg, which also potentiated thiopental-Na anaesthesia by 4-8 fold. In dogs, administration of 5 or 50 mg/kg of daptomycin had no marked effects on the EEG sleep-awakecycle.

The penetration of daptomycin into the CNS in animals with an intact blood brain barrier was limited. Animal model studies demonstrated that there was an increase in the penetration of daptomycin into the cerebrospinal fluid through inflamed meninges. Although the CNS effects of daptomycin were observed at much higher doses than the proposed clinical dose, the possibility of CNS effects when meninges are inflamed has not been excluded. The CNS effects of daptomycin are presented in detail in the pharmacology section of the Product Monograph.

Daptomycin exhibited neuromuscular degeneration in studies in rats and dogs, while there was no effect of daptomycin on neuromuscular transmission in rat phrenic nerve/diaphragm preparations in vitro at concentrations ranging from 10-3 to 10-9M. When the daptomycin concentration was increased to 10-2M (16 mg/mL), neuromuscular blocking activity was observed, which may have a role in the toxicity observed in laboratory animals administered high doses. Some antibiotics such as aminoglycosides have been shown to exhibit neuromuscular blockage at high concentrations and the effect can be reversed by the addition of calcium. In the study presented, the neuromuscular blocking effect of daptomycin could not be reversed by the addition of calcium. Neuromuscular blocking activity of daptomycin observed at 10-2M occurs at approximately 200 times the plasma concentration reported for human volunteers when administered 6 mg/kg dose levels. Therefore, this effect should not be a concern at therapeutic dose levels.

The pharmacological effects of daptomycin on the respiratory and cardiovascular systems were investigated in vivo and in vitro. Daptomycin administration in dogs (50 mg/kg) did not affect respiratory function (respiratory rate, minute volume, PCO2, PO2, HCO3- and arterial blood pH). There were no significant effects on cardiovascular parameters at 50 mg/kg dose levels in dogs. There was a slight increase in mean arterial pressure observed at this dose that should not be of clinical concern as the therapeutic dose is 4-6 mg/kg. In an in vitro study, daptomycin had no effect on the response of isolated atrial preparations from guinea pigs to either electrical or chemical stimulation at concentrations up to 10-4M (160 µg/mL).

The effects of daptomycin on the rapidly polarizing cardiac potassium current was investigated in vitro using cloned hERG channels cDNA expressed in human embryonic kidney cells. At concentrations of unbound drug up to 100 times the clinical plasma concentrations (i.e., 300 μM or approx. 500 μg/mL), daptomycin did not cause any inhibition of the hERG currents. The studies were conducted with an appropriate positive control that caused a 60% blockage of hERG currents. The in vitro studies indicated that daptomycin at clinical concentrations should not cause prolongation of the QT interval. Daptomycin administration in dogs (50 mg/kg) was not associated with any apparent changes in PR interval, QRS duration, or QTc interval.

Daptomycin at dose levels up to 10 mg/kg had no effect on urine volume, osmolality, or electrolyte excretion in female rats. At this dose level, daptomycin did not appear to have any functional effect on macrophages, T-lymphocytes, or B-lymphocytes in primary antibody response to a particulate antigen in mice.The dose levels studied were 1.7 to 2.5-fold the therapeutic dose.

Daptomycin was evaluated as an agonist and pharmacologic antagonist in a variety of smooth muscles from guinea pigs, rats, and rabbits. Daptomycin was generally devoid of pharmacologic activity in these preparations except for inhibition of response to oxytocin in the estrogen-primed uterus and attenuation of response to field stimulation of rat vas deferens. However, as these effects are seen at a concentration approximately 30-fold higher than the therapeutic plasma concentration, it has little clinical significance.

3.2.4 Toxicology

To support the toxicology section of this submission, the sponsor submitted over 50 new toxicology studies. In these toxicology studies, several noteworthy effects were reported, particularly in the acute and repeat-dose toxicity studies. Skeletal muscle and peripheral nerves were identified as the primary target organs of toxicity for daptomycin.

Acute Toxicity

Acute toxicity testing identified the neuromuscular system (nervous system and/or skeletal muscle) as the target organ of daptomycin toxicity in all four species tested (i.e., mouse, rat, dog, and monkey). After single-dose IV administration (at doses ranging from ≥25 to ≥700 mg/kg) of daptomycin, clinical signs suggestive of toxicity to the skeletal muscle and/or nervous systems [central nervous system (CNS) and peripheral nerves] were evident in all four species and were accompanied by decreases in body weight and/or body weight gain in rodents and dogs. Deaths from high doses (≥140 to ≥700 mg/kg) appeared to be related to CNS toxicity. Effects, including tremors, clonic convulsions, and coma, occurred at dose levels similar to those associated with mortality. The peak plasma levels of daptomycin in animals at the minimal lethal dose ranged from approximately 30 to 100 times that of the intended clinical dose (4 mg/kg every 24 hours).

Repeat-Dose Toxicity

Repeat-dose toxicity studies were all conducted by the intravenous (IV) route of administration, the intended route of clinical administration. Although a 30-minute infusion is used clinically, daptomycin was administered by bolus injection in all but one study because infusion was impractical for the number of animals tested. However, one study also investigated administration via 30-minute IV infusion. For most studies, daptomycin was administered once daily, except in select investigative studies in which it was administered on a three times daily regimen.

Based on the neuromuscular effects observed in the safety pharmacology and acute toxicity studies of daptomycin, the repeat-dose studies often included special endpoints to characterize effects on muscle and nerves.

The results of repeat-dose and investigative studies consistently indicated the primary target organ to be skeletal muscle in adult rats and dogs, with effects observed in the peripheral nerves at higher dose levels in both species. Skeletal myopathy was usually accompanied by serum creatine phosphokinase (CPK) elevations in adult dogs, which preceded clinical effects and correlated with the severity of microscopic lesions. Moderate to severe clinical signs (abnormal posture/gait, impaired coordination, inability to stand, sternal recumbency) and functional (electrophysiology) deficits were evident. Microscopic effects were detected in peripheral nerves, dorsal ganglia, nerve roots (including left and right ventral and dorsal roots), and spinal nerves. The primary skeletal myopathy and neuropathy observed in dogs following repeat-dose administration of daptomycin appeared to be independent/unrelated effects. For instance, the two effects appeared to be related to different pharmacokinetic parameters. Whereas skeletal muscle effects were independent of the maximum plasma concentration (Cmax) and appeared primarily related to dosing frequency (time between doses) and/or AUC, Cmax appeared the key determinant for peripheral nerve effects (not related to AUC). Moreover, the skeletal muscle and nerve effects exhibited differential dose response curves and rates of recovery. The muscle effects were recovered at a faster rate than the nerve effects, which took significantly longer to recover. For instance, daptomycin-related muscle effects were reversible within 30 days following cessation of dosing. Recovery of peripheral nerve function was evident within 3 to 6 months post-dosing (consistent with the lack of effect upon the neuronal cell body), although histological changes (dorsal roots, ventral roots and spinal nerves) were evident 6 months after dosing. In all but one case, the axonal degeneration observed in these tissues was graded as very minimal and described as rare, scattered vacuoles.

In contrast to adult dogs, juvenile dogs appeared to be more sensitive to peripheral nerve lesions than skeletal myopathy, and their CPK levels were not increased. Juvenile dogs developed axonal degeneration of peripheral nerve fiber (sciatic, ulnar), spinal cord (cervical, thoracic, lumbar) and dorsal nerve root at doses lower than those associated with skeletal muscle toxicity. In addition, the skeletal muscle effects were reversed but the peripheral nerve (sciatic) and spinal cord (cervical, thoracic, lumbar) effects were only partially reversed following a 4-week recovery phase.

Nephrotoxicity and gastrointestinal (GI) effects observed in rats appear to be species-specific; these effects were not evident in either dogs or monkeys up to the highest doses tested (75 mg/kg/day and 10 mg/kg/day in dogs and monkeys, respectively).

It should be noted that only one repeat-dose study was conducted with monkeys, where daptomycin was administered only up to 10 mg/kg for one month. No noteworthy adverse effects were observed in this study but the doses tested were too low (1.7 to 2.5-fold the recommended therapeutic doses) for any meaningful observations.

Mutagenicity/Genotoxicity

Daptomycin was not mutagenic or clastogenic in a battery of genotoxicity tests, including the Ames assay, a mammalian cell gene mutation assay, a test for chromosomal aberrations in Chinese hamster ovary cells, an in vivo micronucleus assay, an in vitro DNA repair assay, and an in vivo sister chromatid exchange assay in Chinese hamsters.

Carcinogenicity

Carcinogenicity studies with daptomycin were not conducted in view of the short recommended duration of clinical therapy and the negative genotoxicity findings.

Reproductive Toxicity

Animal studies have not demonstrated harmful effects with respect to mating, fertility, pregnancy, embryo-fetal development, parturition, or post-natal development. Nevertheless, given that no clinical studies have been performed in pregnant women, a warning and precautionary statement is included in the Product Monograph.

Other Toxicity Studies

Daptomycin caused slight and transient irritation to exposed skin and slight and transient conjunctivitis upon topical application to the eye. Even though there were no specific local tolerance studies conducted, daptomycin did not appear to cause local injection site irritation following repeated administration by IV injection. Daptomycin exhibited low immunogenicity upon repeated administration.

3.2.4 Microbiology

Daptomycin inserted directly at the membrane of both growing and stationary phase of susceptible Gram-positive bacteria, which resulted in dissipation of the membrane potential and an efflux of potassium ions, and caused inhibition of protein, DNA, and RNA synthesis and bacterial cell death with negligible cell lysis. Daptomycin was not active against Gram-negative bacteria.

3.2.5 Summary and Conclusion

The pharmacokinetics of daptomycin were investigated in several animal species to characterize the ADME profile. With IV administration, there was linear dose-response up to the peak plasma concentration of daptomycin. Daptomycin exhibited limited metabolism across all species of animals tested and was excreted primarily in the urine. In rats, daptomycin accumulated in the kidney and had a long tissue half-life. Renal impairment decreased systemic clearance of daptomycin. A potential interaction with tobramycin was observed in rats, which lowered the dose of daptomycin (‑5 mg/kg) at which mild skeletal muscle changes were noted.

Safety pharmacology studies in animals indicated effects on the central nervous system at high doses. Appropriate statements on these potential effects have been included in the Product Monograph. Daptomycin is not expected to have an effect on the QTc interval.

Noteworthy effects were also reported in the toxicology studies at high doses, notably in the acute and repeat-dose toxicity studies. Appropriate statements are included in the Product Monograph to convey the risks. Should increased clinical treatment durations be proposed in the future, a re-evaluation of the need for carcinogenicity studies and of the appropriate duration of repeat-dose toxicity studies is recommended.

3.3 Clinical basis for decision

3.3.1 Pharmacokinetics

Absorption

Daptomycin pharmacokinetics were generally linear and time-independent at doses of 4 to 12 mg/kg q24h. Steady-state trough concentrations were achieved by the third daily dose. The mean (standard deviation) steady-state trough concentrations attained following administration of 4, 6, 8, 10 and 12 mg/kg q24h were 5.9 (1.6), 6.7 (1.6), 10.3 (5.5), 12.9 (2.9) and 13.7 (5.2) μg/mL, respectively. The mean AUC and Cmin (minimum plasma concentration) of daptomycinduring once-daily dosing with 6, 8, 10 and 12 mg/kg were dose proportional; however, the mean Cmax (maximum plasma concentration) was slightly less than dose proportional. Total clearance was unchanged across 4 to 12 mg/kg q24h.

Distribution

Daptomycinis reversibly bound to human plasma proteins, primarily to serum albumin, in a concentration-independent manner. The overall mean binding at doses from 4 to 12 mg/kg ranged from 90 to 93%. The apparent volume of distribution (Vd) of daptomycinat steady-state in healthy adult subjects was low, approximately 0.1 L/kg at doses of 4 to 12 mg/kg, consistent with distribution primarily within the extracellular space. Studies conducted in healthy subjects with cantharides-induced blisters demonstrated that daptomycin penetrated into inflammatory exudates. Daptomycin penetrated the inflammatory exudates at a moderate rate with time to maximum concentration (Tmax) occurring approximately three hours later than in plasma. The penetration of daptomycin into the inflammatory exudates was good and the maximum concentration (Cmax) achieved in inflammatory fluid was 27.6 µg/mL. The concentration of daptomycin achieved at the inflammatory foci in the skin was well above the MIC90 for the majority of potential Gram-positive pathogens. The population PK study indicated that the volume of distribution increased in subjects with acute bacterial infection relative to uninfected subjects. This is consistent with the pathophysiology of acute bacterial infection which is characterized by an inflammatory response associated with increased vascular permeability and collection of extracellular fluid at the site of infection.

Clinical pharmacology studies indicated that 90-93% of daptomycin binds to plasma proteins. In vitro studies indicated that daptomycin binds to serum albumin in a concentration-independent fashion. The protein binding is relatively weak and readily reversible. Serum protein binding in subjects with varying degrees of renal impairment was comparable to healthy subjects; however, protein binding was decreased in subjects with end-stage renal disease (ESRD) that were on dialysis.

Metabolism

In healthy subjects, no products of daptomycin metabolism or degradation were detected in plasma following administration of radiolabelled drug. After 24 hours, 65% of the radioactivity was recovered in the urine. One-hundred and forty-four (144) hours following administration, 78% of labelled drug was recovered in urine and an additional 5% and 3% of radioactivity was recovered in the feces and the expired air, respectively. In a second non-radiolabelled study, there were four minor metabolites observed in urine samples at low concentrations (each <5% of the daptomycin peak) but none in the plasma samples. Two out of the four metabolites observed in the urine appeared to be Phase-1 oxidative metabolites. It was not reported whether these metabolites were active.

Studies conducted in vitro indicated that daptomycin was not metabolized by human liver microsomes under the conditions tested. Further in vitro studies examined the effect of daptomycin on induction and activity of hepatic cytochrome-P450 isoenzymes. The doses used in these studies were appropriate to the therapeutic concentration. There was no biologically significant inhibition or induction of the enzymes. A possibleenzyme induction of CYP2C9 was observed with 400 µg/ml daptomycin, a concentration that exceeded the expected clinical concentration by 4 to 8x. The effect was not consistently observed in donor tissue.The study presented demonstrated that daptomycin should not adversely influence CYP3A4-mediated metabolism in vivo and that CYP-mediated drug-drug interactions are unlikely.

Excretion

Systemic clearance of daptomycin occurred primarily by renal excretion. In a clinical study using radiolabelled daptomycin, 75-80% of the administered dose was excreted in the urine. The fecal route accounted for an additional 4-6% of the administered dose. Administration of probenecid had no effect on the PK of daptomycin. This suggests that renal tubular secretion, blocked by probenecid, is not a component in explaining daptomycin elimination. Glomerular filtration was the major elimination route for daptomycin.

Special Populations

Three clinical studies examined the PK of daptomycin in subjects with renal impairment. All studies clearly indicated that plasma clearance of daptomycin significantly correlated with creatinine clearance. PK parameters related to renal elimination of daptomycin were significantly influenced in patients with creatinine clearance of <30 mL/min. The drug exposure (AUC) in this group was about 2.5-fold higher as compared to healthy volunteers. In subjects with severe renal impairment, a dosing regime of 4 mg/kg daptomycin once every 48 hours for cSSSI and 6 mg/kg daptomycin every 48 for bacteremia or infective endocarditis has been approved. These doses are derived from PK modelling studies but have not been tested for efficacy or safety. This has been clearly stated in the Product Monograph. Further, it is also stated under Dosage and Administration that "Cubicin should only be used in patients whose creatinine clearance is <30 mL/min when it is considered that the expected clinical benefit outweighs the potential risk and for whom there are no further therapeutic options". In subjects with creatinine clearance >30 mL/min but <50 mL/min, the mean AUC increased by 1.65-fold and the 24-hour serum levels were about 2-fold higher than the control group. In subjects with renal failure, there was a trend towards an increased free fraction of daptomycin but the difference was not statistically significant.

No dose adjustment is required for subjects whose estimated creatinine clearance is >30 mL/min, but for patients whose estimated creatinine clearance is <30 mL/min, a change in the dosage regimen is necessary. As both trough levels and AUC of daptomycin increase in subjects with a creatinine clearance of <50 mL/min, the potential for elevation of serum CPK levels and possible muscle toxicity increases. It is indicated in the Warnings and Precautions section of the Product Monograph that CPK levels should be closely monitored in all patients with some degree of renal insufficiency.

The PK of daptomycin (with the exception of Cmax) was significantly different in geriatric subjects; exposure was 58% higher as compared with younger subjects. Renal clearance was lower in geriatric subjects and changes in daptomycin PK may be attributed to changes in renal function. Despite the fact that exposure was higher in geriatric subjects, a 4 mg/kg single-dose of daptomycin was well tolerated and there were no clinically significant changes in hematology, clinical chemistry, or urinalysis. No dosage adjustment is warranted for elderly subjects with normal renal function based on age alone.

There was no effect of mild to moderate hepatic impairment on the PK of daptomycin. The pharmacokinetics of daptomycin in patients with severe hepatic insufficiency have not been evaluated. Compared with non-obese controls, plasma clearance of daptomycin was approximately 15% lower in moderately obese subjects and 23% lower in extremely obese subjects while the AUC increased by approximately 30% and 31%, respectively. The population PK analysis indicated a modest influence of gender on the clearance of daptomycin. The clinical significance of this observation is uncertain. No dosage adjustments are required based on obesity or gender alone.

Drug Interactions

The pharmacokinetics of daptomycin and aztreonam when administered alone or in combination was investigated in healthy volunteers. A comparison of Cmax, AUC0-∞ and Fedose indicated that daptomycin and aztreonam do not show a PK drug-drug interaction with each other. The safety profile of 6 mg/kg of daptomycin administered as a single IV dose did not differ when administered in combination with 1000 mg of aztreonam.

A clinical study evaluated the effects of daptomycin at steady state on the single-dose PK of the R- and S-warfarin enantiomers and the pharmacodynamics (PD) of warfarin in healthy subjects. Comparison of Cmax and AUC indicated that daptomycin does not have significant effect on the PK of either R- or S-warfarin. In addition, the baseline-corrected INR supports the absence of an effect of daptomycin on the PD of warfarin.The study presented did not examine the PK of daptomycin at the steady state concentration of warfarin. However, the design used in this study minimized the risk of exposure of healthy volunteers to a longer anticoagulant effect, and a single dose of warfarin should be able to detect if there is any interaction with the PK of daptomycin. Comparison of Cmax and AUC of daptomycin in this study indicated that the PK of daptomycin was not significantly altered with warfarin administration. Results of safety monitoring during this study did not raise any new safety concerns related to daptomycin administration.

A clinical study evaluated the safety of daptomycin when administered once daily, for 14 consecutive days, in subjects on a stable daily dose of 40 mg of simvastatin. The results indicated that the trough concentration of daptomycin in subjects treated with simvastatin was similar to the concentration observed in other Phase I studies. Multiple-dose administration of daptomycin in subjects receiving concomitant treatment of simvastatin was not associated with a higher incidence of adverse events. Further concomitant administration of daptomycin and simvastatin had no effect on serum CPK levels. There did not appear to be any interaction between simvastatin and daptomycin.

Experience with co-administration of HMG-CoA reductase inhibitors and daptomycin in patients is limited, and also the study design for the interaction study excluded subjects on simvastatin whose CPK levels were >1.5 times the upper limit of normal (ULN). Consideration should be given to temporarily suspending use of HMG-CoA reductase inhibitors in patients receiving daptomycin. A warning has been included in the Product Monograph regarding the use of HMG-CoA reductase inhibitors in patients receiving Cubicin®.

A clinical study evaluated whether the PK of daptomycin and tobramycin are affected when they are concurrently administered. The results indicated that there was a slight decrease in renal clearance and excretion of daptomycin with concurrent administration of tobramycin. The interaction study of tobramycin and daptomycin was conducted with doses of daptomycin lower than the doses requested for clinical indications. Renal excretion of daptomycin is an important route of elimination. The reviewer is concerned that if therapeutic doses of daptomycin were used in these studies, a possible significant change in renal clearance would have been observed when administered with tobramycin. In the Product Monograph, it is clearly stated that the interaction between these two drugs at clinical dose is not known. Results from the drug interaction studies in animals demonstrated that tobramycin may have a weak potentiating effect on muscle damage caused by daptomycin which is captured in the drug-drug interaction section of the Product Monograph.

3.3.2 Additional Pharmacology Studies

Clinical pharmacology studies were conducted to assess the effect of daptomycin on cardiac repolarization and peripheral nerve conductivity. The study presented found no evidence that exposure to daptomycin at 6 mg/kg once every 24 hours for 14 days caused any clinically meaningful change in cardiac repolarization as measured by QTcB.

The exposure of healthy volunteers to daptomycin at 6 mg/kg once every 24 hours for a period of 14 consecutive days was not associated with either myopathy or neuropathy as judged by objective electrophysiological measures for nerve motor function in the hand or vibration perception threshold (VPT) in the great toe. However, the small fibre sensory function as assessed by a subjective neurological questionnaire revealed the possibility of some degree of peripheral nerve damage. During the 14-day follow-up period more subjects in the daptomycin group (8) compared to the normal saline group (5) reported symptoms of tingling, numbness and weakness. Precautionary statements to monitor for signs and symptoms of neuropathy during therapy with daptomycin have been included in the Product Monograph.

3.3.3 Clinical Efficacy

Complicated Skin and Skin Structure Infections (cSSSI) Treatment

In two similar comparative pivotal two arm cSSSI trials (Studies 9801 and 9901) conducted internationally (US, Europe, South Africa, Israel, Australia), 1092 adults were exposed in approximately equal numbers to either Cubicin® (daptomycin) or comparator drugs. In each of the two study arms, patients were pre-randomized according to the likelihood or not of exhibiting Staphylococcus aureus resistance to methicillin and/or penicillin intolerance. In the first arm, patients received daptomycin. In the second arm, patients received either semi-synthetic penicillins (i.e., oxacillin, nafcillin, cloxacillin or flucloxacillin) or alternatively, vancomycin if resistance to penicillin or penicillin intolerance was suspected. The control drugs were currently approved for the treatment of cSSSI caused by susceptible pathogens in the countries where they were used. Patients in both study arms could be switched to oral antibiotics after a minimal period of parenteral therapy or immediately to vancomycin in the control arm if resistance was discovered. Aztreonam and/or metronidazole could be administered if infections included Gram-negative and/or anaerobic bacteria.

The primary endpoints of the studies addressed clinical efficacy (improvement, cure) at 7-12 days after treatment with 4 mg/kg daptomycin IV every 24 hours for 7-14 days. Safety was assessed throughout the study period.

A successful microbiologic response for an infecting pathogen was defined as definite eradication (failure to isolate the pathogen from a culture of the primary site of infection at the test-of-cure assessment) or presumed eradication (healing of the primary site of infection with explicit indication that there was "nothing to culture"). Subjects were designated as microbiologic failures if culture data were not available for any other reasons or if one or more of the original infecting Gram-positive pathogens were explicitly isolated (documented persistence). Missing data were designated as presumed persistent failures. Overall, therapeutic success was defined as a subject who had an outcome of clinical success and, if an infecting pathogen had been isolated at baseline, also had a successful microbiologic response. A subject who was a clinical failure, was non-evaluable or was a clinical success with microbiologic failure, was designated as a therapeutic failure.

In Study 9801, one or more Gram-positive pathogens were cultured at baseline in >81% of intent to treat patients (ITT) and in that subgroup of modified intent to treat patients (MITT), Staphylococcus aureus, Streptococcus pyogenes, Streptococcus a galactiae, and Enterococcus faecalis were recovered from approximately 70%, 16.2%, 9.3% and 13.8% of patients, respectively. Overall, 13% of the culture-positive subjects were infected with both Staphylococcus aureus and the β-hemolytic streptococci Streptococcus pyogenes or Steptococcus agalactiae. During the trial period, approximately 32% of ITT patients received protocol-approved antibiotics for Gram-negative and anaerobic organisms, and approximately 40% underwent adjunct procedures such as incisions, drainage or wound debridement.

In Study 9901, one or more Gram-positive pathogens were cultured at baseline in >83% ITT patients and, in those patients S taphylococcus aureus, Streptococcus pyogenes, Streptococcus a galactiae, and Enterococcus faecalis were recovered from approximately 68.9%, 27.1%, 6.6%, and 10.3% respectively. Overall, 20.7% of the culture-positive subjects were infected with both Staphylococcus aureus and the β-hemolytic streptococci Streptococcus pyogenes or Streptococcus agalactiae. During the trial period, approximately 19.8% of ITT patients received protocol-approved antibiotics for Gram-negative and anaerobic organisms, and approximately 19.7% underwent adjunct procedures such as incisions, drainage or wound debridement.

In Study 9801, the overall clinical success rates as determined in integrated analyses were 65.1% in the daptomycin arm and 64.8% in the comparator arm for the MITT population and 76.0% and 76.7%, respectively, for the clinically evaluable (CE) subgroup of the ITT population, demonstrating the non-inferiority of daptomycin to comparators. In Study 9901, the clinical success rates were also comparable for the MITT (84.0% vs. 83.1%) and CE (89.9% vs. 90.4%) groups.

The pooled microbiological successes (organism eradication or presumed eradication ) in the microbiologically evaluable (ME) patient population of studies 9801 and 9901 were similar for daptomycin and comparator arms (74.2% vs. 75.5%). These results were slightly lower in study 9801 (67.8% vs. 67.6%) than Study 9901 (80.1% vs. 81.8%). The pooled microbiological successes in the ME population were comparable for daptomycin and comparator arms relative to the infecting baseline pathogen: methicillin-susceptible Staphylococcus aureus (MSSA) (73.2% vs. 75.8%), methicillin-resistant Staphylococcus aureus (MRSA) (53.6% vs. 58.3%), Streptococcus pyogenes (90.5% vs. 81.8%) and Streptococcus agalactiae (77.8% vs. 65.5%). Microbiological success corroborated clinical success.

Rates of clinical relapse or recurrence at late follow-up (>20 days after end of treatment) were low and there was no evidence of emergence of resistance to daptomycin during or following treatment.

Daptomycin was shown in two pivotal trials to be effective in the treatment of adult cSSSI infections caused by Staphylococcus aureus, Streptococcus agalactiae, and Streptococcus pyogenes. The claim for vancomycin-sensitive Enterococcus faecalis was not considered to be supported by either the trial design or results.

Summaries of fourteen supportive non-pivotal early or developmental trials were also submitted by the sponsor, five of which directly related to cSSSI.

Bacteremia Treatment

One pivotal trial (non-inferiority), a Phase III multicentre, randomized, open-label, comparative study, was used to assess the safety and efficacy of daptomycin compared to conventional therapy in the treatment of subjects with Staphylococcus aureus bacteremia with known or suspected Staphylococcus aureus infective endocarditis (according to modified Duke criteria). Of the 246 patients enrolled, 235 received study medication: 120 patients received daptomycin 6 mg/kg IV once daily, and 115 received comparator (vancomycin 1 gIV every 12 hours or semi-synthetic penicillin 2 g IV every 4 hours, with an initial dose of gentamycin 1 mg/kg IV every 8 hours, for four days). Primary efficacy endpoint success was based on the Independent External Adjudication Committee Outcome at Test-of-Cure (TOC) in the Intent to Treat (ITT) population pooled across diagnostic strata (left-sided infective endocarditis [LIE], complicated right-sided infective endocarditis [RIE], uncomplicated RIE, and complicated bacteremia), and was a composite endpoint based on clinical as well as microbiological success. The main secondary efficacy endpoint was time-to-clearance of bacteremia in the ITT and Per Protocol (PP) populations.

The overall success rates at TOC in the ITT population were 44.2% (53/120) in patients treated with daptomycin,and 41.7% (48/115) in patients treated with comparator. The success rates in the PP population were 54.4% (43/79) in patients treated with daptomycin, and 53.3% (32/60) in patients treated with comparator. Success rates in the overall ITT population showed that daptomycin efficacy was nearly identical irrespective of methicillin-susceptibility of the baseline Staphylococcus aureus infections.

Although the study cohort was representative of patients with Staphylococcus aureus bacteremia and known or suspected IE, multiple exclusion criteria reduced the generalizability of the efficacy results. There were notable study exclusions including, but not limited to, patients with prosthetic valves, pneumonia, known osteomyelitis, meningitis, polymicrobia bloodstream infection, intravascular foreign material not planned for removal within four days of dosing (except vascular stents in place for >6 months or permanent pacemakers), and severe renal failure.

The clinical benefit of daptomycin in clinically and microbiologially evaluable patients with a final diagnosis of IE (53) was supported by limited data for RIE [success in 8/19 (42.1%) vs. 7/16 (43.8%) of patients in the daptomycin vs. comparator arms, respectively]. However, efficacy/clinical benefit of daptomycin was not demonstrated in patients with LIE [1/9 (11.1%) vs. 2/9 (22.2%)].

Efficacy data for diagnostic subgroups were limited and not statistically powered due to a heterogeneous population, discrepancy between entry and final diagnosis of IE, and small numbers. The recommended dose and schedule were justified in patients with normal renal function or mild renal impairment. Poorer outcome was observed in daptomycin patients with moderate renal impairment (creatinine clearance of 30-50 mL/min). Clinical efficacy in patients with severe renal failure was not studied. Efficacy issues have been managed through statements in the Product Monograph.

Two further non-pivotal studies were conducted that addressed bacteremia. The first was a Cubist-sponsored open study comparing daptomycin 4 mg/kg/day, 6 mg/kg/day, and 3 mg/kg twice a day, with a comparator (vancomycin 1 g twice a day or the semi-synthetic penicillins nafcillin or oxacillin 4-13 g/day in divided doses) administered for 7-14 days to Gram-positive bacteremic patients. Robust conclusions were hard to draw but efficacy was comparable for the mid daptomycin dose and comparators (68.8% vs. 73.3% clinical success). Other doses appeared less effective, possibly due to uncontrolled mitigating factors.

The second study was an older Lilly study in which daptomycin 3 mg/kg twice a day following a 6 mg/kg loading dose was compared to conventional treatment for Gram-positive endocarditis/bacteremia. There were only 40 and 14 evaluable patients, and the overall clinical efficacy was 80% vs. 78% for daptomycin vs. comparator, respectively. Daptomycin was not as effective in endocarditis (68.8% vs. 80.0%).

3.3.4 Clinical Safety

Complicated Skin and Skin Structure Infections (cSSSI) Treatment

The integrated analysis of adverse events (AEs) for the two cSSSI pivotal trials showed similar distributions for both daptomycin and comparator groups. The overall AE rate in all patients treated was 51.3% vs. 52.5% for daptomycin vs comparator, with the most frequently occurring AEs being related to the gastrointestinal tract. The majority were mild to moderate and considered unrelated to treatment. Creatine phosphokinase (CPK) increases of 2.1% vs. 1.4% were considered drug-related. The issue of muscle toxicity is considered rare however, and on the whole has not been demonstrated in trials to be particularly problematic. Hypersensitivity and neuropathy were also observed in rare cases, and should be monitored. Precautionary statements are included in the Product Monograph. There did not appear to be any other clinically important trends indicative of daptomycin toxicity. No dose adjustments are considered necessary based on age, race, gender, or degree of hepatic impairment.

In the non-pivotal studies, daptomycin appeared to be well tolerated at the doses proposed for use in cSSSI. Drug-related neurotoxicity and peripheral musculotoxicity appeared to be very rare and reversible even at higher doses. There did not appear to be any drug-induced cardiac toxicity. Experience in patients with underlying severe renal disease, central or peripheral neurological impairments or musculopathies is limited.

Bacteremia Treatment

Safety data from the pivotal study was based on experience in a safety population (n=236) of whom 120 received at least one dose of daptomycin (6 mg/kg every 24 hours). The safety profile was based on varying durations of drug exposure (range 10-42 days, mean 14 days) and a 12-week post-treatment follow-up period. Fourteen patients received >28 days of daptomycin treatment.

The most frequent treatment emergent adverse events (TEAE) observed in the pivotal Phase III clinical study were System Organ Class (SOC): Infections; and Gastrointestinal disorders (including diarrhea, vomiting, constipation, and nausea). Most TEAE were considered unrelated to study drug and were mild to moderate in severity. The most common drug-realted TEAE with daptomycin use were: CPK increase (5%), loose stools (3.3%), blood phosphorous increase (2.5%), dyspepsia (2.5%), and rash (2.5%). Possible/probably drug-related discontinuations were due to elevated CPK [3 (2.5%)], rash [3 (2.5%)], and one report each (<1%) of vomiting, renal failure, thrombocytopenia, and cardiac arrest. Possible/probably drug-related serious adverse events (SAE) included elevated CPK (resolved upon dechallenge), atrial fibrillation/flutter, renal impairment (underlying renal insufficiency), and cardiac arrest leading to death (multiple confounders). Increased SAEs reported in patients with moderate renal impairment who received 6 mg/kg every 24 hours suggest cautious use in this cohort. The safety of daptomycin in patients with severe renal impairment has not been established. Cases of drug-related paresthesia in the daptomycin group were reported uncommonly and resolved despite continued treatment.

Risk management of these potential events includes monitoring clinical response, CPK and renal function, symptoms of myopathy or neuropathy, and discontinuation of drug/temporarily suspending HMG CoA reductase inhibitor or other drugs associated with myopathy. Safety issues have been managed through statements in the Product Monograph.

There are limited long-term safety data available (i.e. >28 days); no unique safety issues were identified in patients with longer courses of therapy.

In the non-pivotal trials, tolerability appeared to be similar for all daptomycin doses and comparators. One patient on 4 mg/kg/day had a slight daptomycin-related CPK elevation (500-750 U/L) during the third week of treatment which was associated with muscle weakness and fatigue. The patient was discontinued from the study and CPK levels promptly recovered.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk assessment

Therapeutic options for Staphylococcus aureus, particularly methicillin-resistant Staphylococcus aureus (MRSA) are limited, and the data in this application demonstrate that daptomycin is active against Staphylococcus aureus independent of their susceptibility or resistance to methicillin. Daptomycin provides a good addition to the antibiotic armamentarium in clinically treating cSSSI, including staphylococcal forms now resistant to conventional therapies. Neuro-muscular toxicity has a low but acceptable risk, though continued information gathering is warranted and ongoing. For treatment of patients with Staphylococcus aureus bacteremia, including those with right-sided Staphylococcus aureus endocarditis (native valve), daptomycin shows a favourable benefit/risk ratio. Potential risks are manageable through monitoring and an intervention of discontinuation. Use of daptomycin in Staphylococcus aureus bacteremia that is associated with other commonly associated conditions has not been specifically studied, nor has use in the pediatric population.

Overall, the benefit of using daptomycin as per protocol and as per the wording in the Product Monograph outweighs the risk incurred.

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 Cubicin® is favourable in the treatment of complicated skin and skin structure infections (cSSSI) caused by susceptible strains of the following Gram-positive microorganisms: Staphylococcus aureus (including methicillin-resistant strains), Streptococcus pyogenes, and Streptococcus agalactiae; and Staphylococus aureus bloodstream infections (bacteremia), including those with right-sided Staphylococus aureus infective endocarditis. 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: Cubicin®

Submission MilestoneDate
Submission No. 102320
Submission filed2005-11-14
Screening 1
Screening Deficiency Notice issued2005-12-29
Response filed2006-02-07
Screening Acceptance Letter issued2006-03-23
Review 1
Quality Evaluation complete2007-02-01
Clinical Evaluation complete2007-01-17
Labelling Review complete2006-11-09
NON issued by Director General due to quality issues2007-02-07
Response filed2007-04-20
Screening 2
Screening Acceptance Letter issued2007-04-27
Review 2
Quality Evaluation complete2007-04-27
Clinical Evaluation complete2007-09-20
Labelling Review complete2007-09-19
NOC issued by Director General2007-09-24
Submission No. 104227
Submission filed2006-02-22
Screening 1
Screening Acceptance Letter issued2006-04-13
Review 1
Quality Evaluation complete2007-02-01
Clinical Evaluation complete2007-01-17
Labelling Review complete2006-11-08
NON issued by Director General due to quality issues2007-02-07
Response filed2007-04-20
Screening 2
Screening Acceptance Letter issued2007-04-27
Review 2
Quality Evaluation complete2007-09-18
Clinical Evaluation complete2007-09-20
Labelling Review complete2007-09-19
NOC issued by Director General2007-09-24