Summary Basis of Decision for Zeftera

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
Zeftera

Ceftobiprole medocaril, 500 mg/vial, Powder for solution, Intravenous

Janssen-Ortho Inc.

Submission control no: 112752

Date issued: 2009-08-14

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:

Zeftera

Manufacturer/sponsor:

Janssen-Ortho Inc.

Medicinal ingredient:

Ceftobiprole medocaril

International non-proprietary Name:

Ceftobiprole medocaril

Strength:

500 mg/vial

Dosage form:

Powder for solution

Route of administration:

Intravenous

Drug identification number(DIN):

  • 02313103

Therapeutic Classification:

Antibacterial agent

Non-medicinal ingredients:

Citric acid monohydrate, sodium hydroxide

Submission type and control no:

New Drug Submission, Control Number: 112752

Date of Submission:

2007-07-16

Date of authorization:

2008-06-26

* All trademark rights used under license.

2 Notice of decision

On June 26, 2008, Health Canada issued a Notice of Compliance to Janssen-Ortho Inc. for the drug product Zeftera.

Zeftera contains ceftobiprole medocaril. Ceftobiprole medocaril is the prodrug of the active antibacterial agent ceftobiprole. Ceftobiprole medocaril is rapidly converted in vivo to ceftobiprole.

Zeftera is indicated for the treatment of the following infections when caused by susceptible strains of the designated microorganisms in patients 18 years of age and older:

  • Complicated skin and skin structure infections (cSSSI), including non-limb threatening diabetic foot infections without concomitant osteomyelitis caused by: Enterobacter cloacae, Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Staphylococcus aureus (including methicillin-resistant isolates), and Streptococcus pyogenes.

Ceftobiprole has a bactericidal mode of action that involves tight binding to many common essential penicillin-binding proteins (PBPs) in both gram-positive and gram-negative bacteria. Ceftobiprole has distinctive bactericidal activity against methicillin-resistant staphylococci primarily due to its novel strong binding to the staphylococcal PBP2a, the PBP that is chiefly responsible for β-lactam resistance in methicillin-resistant staphylococci including methicillin-resistant S. aureus (MRSA).

The market authorization was based on submitted data from quality (chemistry and manufacturing) studies, as well as data from non-clinical and clinical studies. Clinical efficacy was evaluated from two Phase III studies and one open-label Phase II study, while clinical safety was based on twelve Phase I studies, one Phase II study, and two Phase III studies. Zeftera was shown to be non-inferior to vancomycin or vancomycin plus ceftazidime for the indications stated above, and was found to be safe and well tolerated administered as 500 mg using a 120-minute intravenous infusion three times daily or over a period of an hour every 12 hours daily, depending upon the condition.

Zeftera (500 mg/vial, ceftobiprole medocaril) is presented as powder for solution. The recommended dose is 500 mg administered every 8 hours as a 120-minute intravenous infusion for a duration of 7-14 days for cSSSI caused by the following organisms: Staphylococcus aureus (including methicillin-resistant isolates), Streptococcus pyogenes, Escherichia coli, Proteus mirabilis, Enterobacter cloacae, and Klebsiella pneumoniae. In cases of gram-positive bacterial infection only, 500 mg of Zeftera should be administered every 12 hours as a 60-minute intravenous infusion. The every 12-hour dosing regimen has not been studied in patients with diabetic foot infections and therefore is not recommended in these patients. Dosing guidelines are available in the Product Monograph.

Zeftera is contraindicated for patients with known hypersensitivity to ceftobiprole, any of the excipients, other cephalosporins, or in patients who have demonstrated anaphylaxis to β-lactam antibiotics. Zeftera 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 Zeftera 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 Zeftera is favourable for the indications stated above.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Zeftera contains ceftobiprole medocaril, which is the prodrug of the active antibacterial agent ceftobiprole. Ceftobiprole medocaril is rapidly converted in vivo to ceftobiprole. Ceftobiprole has a bactericidal mode of action that involves tight binding to many common essential penicillin-binding proteins (PBPs) in both Gram-positive and Gram-negative bacteria, thereby preventing completion of cell wall biosynthesis. Ceftobiprole has distinctive bactericidal activity against methicillin-resistant staphylococciprimarily due to its novel strong binding to the Staphylococcal PBP2a, the PBP that is chiefly responsible for ß-lactam resistance in methicillin-resistant staphylococci including methicillin-resistant Staphylococcus aureus (MRSA).

Manufacturing Process and Process Controls

Ceftobiprole medocaril 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 International Conference on Harmonisation (ICH) requirements.
  • The processing steps have been evaluated and the appropriate ranges for process parameters have been established.
Characterization

The structure of ceftobiprole medocaril has been 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 therefore, are considered to be acceptable.

Control of Drug Substance

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

Validation reports are considered satisfactory for all analytical procedures used for in-process testing of the drug substance.

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

Stability study results based on accelerated, long-term, and stress testing show that ceftobiprole medocaril is a stable compound when packaged as proposed over the proposed storage period.

3.1.2 Drug Product

Description and Composition

Zeftera (ceftobiprole medocaril) was developed as a prodrug, due to solubility limitations of the active moiety (ceftobiprole). Zeftera is supplied as a sterile, lyophilized powder for solution at a concentration of 500 mg ceftobiprole per vial. The drug product is presented in a clear, 20 mL Type I blow back glass vial with a 20 mm bromobutyl grey elastomer lyophilisation blow back stopper, sealed with an aluminum seal and a plastic flip-off cap.

Each vial of product contains 500 mg ceftobiprole (666.6 mg ceftobiprole medocaril) and the following excipients: citric acid monohydrate and sodium hydroxide.

The drug product is reconstituted with 10 mL of Water for Injection or 5% Dextrose (glucose) injection to a concentration of 50 mg/mL ceftobiprole prior to further dilution with intravenous infusion.

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 ceftobiprole medocaril 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 Zeftera, 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 Zeftera. 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, sterile filtered, aseptically filled into vials, lyophilized, 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.

The specifications for all of the ingredients are approved in accordance with United States Pharmacopeia/National Formulary (USP/NF) or European Pharmacopeia (Ph. Eur.) standards.

Control of Drug Product

Zeftera is tested to verify that its identity, appearance, content uniformity, assay, purity, levels of bacterial endotoxins, and sterility 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.

Validation results of the analytical method used for the determination of ceftobiprole medocaril and the drug-related impurities are considered acceptable.

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

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed 24-month shelf-life at 2-8°Celsius for Zeftera is considered acceptable. Zeftera should be protected from light.

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 of Zeftera are considered suitable for the activities and products manufactured.

All sites are compliant with Good Manufacturing Practices (GMP).

3.1.4 Adventitious Agents Safety Evaluation

Not applicable. The excipients used in the drug product formulation are not from animal or human origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Zeftera 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

The in vivo bacterial efficacy of ceftobiprole on gram-positive and gram-negative bacteria has been demonstrated in various experimental animal models. The pharmacokinetic/pharmacodynamic studies indicate that the time above the minimum inhibitory concentration (MIC) is the most important pharmacokinetic parameter that correlates with in vivo efficacy. The value for this parameter was two-fold higher for gram-negative bacteria as compared to gram-positive bacteria. Therefore the dosing schedule for gram-positive and gram-negative bacteria is expected to differ in clinical studies. Animal model studies also indicate that the activity of ceftobiprole is markedly influenced by the presence of neutrophils. The efficacy of ceftobiprole in animal model studies is either comparable or better than linezolid and vancomycin in treatment of both methicillin-susceptible and resistant strains of Staphylococcus aureus. Staphylococcus aureus which is the predominant pathogen in diabetic foot is one of the requested indications in this drug submission. The in vivo animal model study presented supports the proposed indication.

Safety Pharmacology

In vitro and in vivo studies were conducted to characterize the cardiovascular, central nervous system (CNS), and respiratory safety of ceftobiprole medocaril and ceftobiprole. The in vivo cardiovascular and CNS studies were not conducted according to the ICH guidelines as the studies were conducted early in the development of the non-clinical program, prior to finalization of the guidance. However, due to the absence of relevant findings in non-clinical and clinical studies, Janssen-Ortho did not repeat the studies.

The potential for QT/QTc delayed ventricular repolarization was examined in vitro by membrane currents in HEK293 cells stably transfected with the human ether-à-go-go-related gene (HERG). Treatment with 5 µM concentration of ceftobiprole (highest concentration attainable under the conditions of the assay) produced no statistically significant effects.

The in vivo effects of ceftobiprole on mean arterial blood pressure (MAP) and heart rate (HR) were related to high maximum plasma concentration (Cmax) values achieved in conscious rats [number (n) =4] and marmosets (n=3) with a single bolus administration of 100 mg/kg. A slight persistent increase in MAP was noted in rats and marmosets with a delayed onset following administration of a single intravenous (IV) bolus injection of ceftobiprole medocaril at 100 mg/kg. No effect was seen with single slow bolus IV administrations of 50 mg/kg in rats and 35 mg/kg dose in marmosets.

Dogs (n=1/sex) receiving a 4-hour infusion of 50 mg/kg or 100 mg/kg of ceftobiprole medocaril demonstrated transient effects on blood pressure, as well as ventricular premature complex in the male dog at 100 mg/kg. Ventricular premature complex was considered unlikely to be test drug-related as it can occur spontaneously in beagle dogs. Moreover, a toxicokinetic study in beagle dogs revealed a dose-related exposure to both ceftobiprole and ceftobiprole medocaril at the end of the 4-hour infusion period in dogs with an average Cmax of ceftobiprole of 45.0 ug/mL and 89.5 µg/mL at doses of 50 and 100 mg/kg, respectively, and 30.1 µg/mL and 46.0 µg/mL of ceftobiprole medocaril respectively. This indicates a relatively slow cleavage of the prodrug in this species. 

Ceftobiprole produced clonic and tonic convulsions in mice after IV and intra-cerebroventricular (ICV) administrations. Signs of nephrotoxicity (related to drug precipitation in distal and collecting tubules) and incidences of mortality were noted at doses ≥250 mg/kg. Delayed convulsions were observed in mice at IV doses ≥250 mg/kg were attributed to high levels of drug exposure caused by marked nephrotoxicity, which was considered to have led to reduced renal clearance of ceftobiprole, and thus, sustained elevated plasma and brain levels. Manifestation of kidney toxicity was clearly time- and dose-related, and was overall rated as minimal to moderate at 250 mg/kg IV and moderate to marked at 500 mg/kg IV.

The median effective dose (ED50) of ceftobiprole for centrally-mediated convulsions was determined to be 2.55 µg in a CNS safety study in mice. Mice were administered single ICV injections of 0.3-30 μg ceftobiprole.

IV administration over a 4-hour infusion period at doses equivalent to 125, 250, and 500 mg/kg ceftobiprole, did not affect airway resistance (RL) or dynamic lung compliance (Cdyn) when compared to vehicle-treated Sprague-Dawley rats.

3.2.2 Pharmacokinetics

Absorption

In animals and in humans, it was demonstrated both in vivo and in vitro that ceftobiprole medocaril was converted within minutes quantitatively to ceftobiprole, the active drug. In all species after IV dosing with ceftobiprole medocaril, the Cmax of ceftobiprole occurred at the first sampling point, which was five minutes post-dose. Absolute bioavailability appeared to be approximately 100%.

After administration of a single IV dose of the active drug ceftobiprole to mice, rats, rabbits, and cynomolgus monkeys, plasma concentration-time profiles showed a bi-exponentialdecline with a slight initial distribution phase.

Distribution

The volume of distribution at steady state (Vdss) was approximately 0.3-0.4 L/kg and was comparable to the extracellular space, which is typical for cephalosporins with low plasma protein binding. The in vitro binding of ceftobiprole to human plasma protein was low and concentration-independent. The in vitro plasma protein binding of ceftobiprole was 21.1% in rats, 20.5% in rabbits, 44.9% in cynomolgus monkeys, 30.2% in marmosets, and 18.9% in dogs and remained fairly constant over the concentration range tested. After IV administration of 20 mg/kg radiolabelled 14C-ceftobiprole medocaril in albino mice and pigmented rats, ceftobiprole-related radioactivity was widely distributed, with maximum concentrations at the first sampling time (0.25 hours post-dose) in all tissues. The highest radioactivity levels were found in the coagulating gland, kidney, liver, skin, and lung. As reported in mice, tissue/plasma concentration ratios were below unity in virtually all tissues except for the kidney, in which a ratio of 1.3 was observed. Penetration into the brain was minimal. Drug-related material was not retained in melanin-containing organs in pigmented rats.

Metabolism

Metabolism of ceftobiprole medocaril in vitro with heptocytes of different species revealed a very rapid hydrolytic cleavage of the carbamate group of ceftobiprole medocaril. The main metabolic reaction of ceftobiprole was the hydrolysis of the ß-lactam ring of the drug to RO65-2070; a small percentage of the drug appeared to be transformed into unknown metabolites. Starting with ceftobiprole medocaril, the metabolism of ceftobiprole within 24 hours appeared to be species-dependent: 69% in mice, 64% in marmosets, 51% in humans, 48% in rats, and 43% in dogs.

Metabolic profiles obtained in vitro in rats, dogs, marmosets, and mice were more or less similar and comparable to man. Metabolic profiles from rat liver, kidney, and brain were comparable and revealed no formation of organ-specific metabolites.

The metabolic profiles in vivo in the plasma, urine, and bile of rats and the urine of marmosets, obtained after administration of ceftobiprole and its ceftobiprole medocaril indicated that the major drug-related material excreted was unchanged ceftobiprole, together with the ring-opened product RO65-2070 and 2-3 minor metabolites of unknown structure (<5% of the dose each). Metabolic profiles of rat bile were similar to the urine.

Brain homogenates obtained from in vivo studies in mice and rats having received IV doses of ceftobiprole or ceftobiprole medocaril contained predominantly unchanged drug and very few other components in small amounts. In mice receiving ICV administration of ceftobiprole medocaril, more than 40% of ceftobiprole medocaril was cleaved to the active drug within a few minutes, but no additional peaks were observed. Since the brains were obtained when the mice showed tonic seizures, the drug itself, rather than a potential metabolite, appeared to be responsible for the observed effect on the CNS.

Excretion

The elimination half-life (t1/2) ranged from 0.29 hours in the mouse to 1.7 hours in the cynomolgus monkey. Ceftobiprole was mainly eliminated by renal excretion of unchanged drug. In rats and dogs, about 70% of the administered dose was recovered in the urine. In rats, renal excretion of ceftobiprole was shown to proceed via glomerular filtration since renal clearance of the unbound fraction was similar to the glomerular filtration rate (GFR) in this species. This finding was confirmed by the lack of effect on the clearance of ceftobiprole when probenecid was co-administered. Plasma clearance of ceftobiprole in other species was consistent with renal excretion via glomerular filtration.

The apparent clearance of ceftobiprole was 5.05 and 2.59 mL/min/kg in the marmoset and cynomolgus monkey, respectively. In the rat however, clearance was 13.0 mL/min/kg, which exceeded the GFR, and which was also higher than the value obtained after administration of ceftobiprole itself. This finding indicates a somewhat incomplete conversion of ceftobiprole medocaril to ceftobiprole, which would decrease the bioavailability of ceftobiprole.

IV administration of 14C-labelled ceftobiprole medocaril in albino mice demonstrated that elimination of radioactivity from the body was very rapid; by two hours post-dose, tissue concentrations declined on average by a factor of ten. At 24 hours post-dose, only the kidney cortex and liver still contained quantifiable levels of radioactivity. In pigmented rats, at seven hours after administration of 14C-labelled ceftobiprole medocaril, the tissue concentrations declined on average by a factor of ten. At 48 hours post-dose, most of the tissues showed radioactivity levels below or close to the quantitation limit. In rats, excretion of radioactivity in the feces was 17%. Levels of radioactivity in cage wash, gastrointestinal tract, and residual carcass were below 3% of the administered dose by 96 hours post-dose.

High exposures to ceftobiprole were achieved in plasma and urine. No significant deviations from dose-proportionality were observed at the dose range of ceftobiprole medocaril used in toxicity studies, and no apparent accumulation of ceftobiprole was noted after repeated dosing, even at the highest dose tested of ceftobiprole medocaril. In general, the pharmacokinetics (PK) of ceftobiprole were independent of time. No apparent gender-related differences with respect to plasma or urinary exposures were observed.

Drug Interactions

Under the test conditions, ceftobiprole did not seem to be a P-glycoprotein (P-gp) substrate or P-gp inhibitor and the bi-directional permeability of ceftobiprole in MDR-MDCK cell monolayers was very low.

The inhibitory potential of ceftobiprole was investigated in vitro on several cytochrome P450 isozyme-catalyzed reactions, using human liver microsomes, one substrate concentration, and several concentrations of ceftobiprole. Ceftobiprole showed slight to no inhibitory potential of CYP1A2, 2C9, 2C19, 2D6, and 3A4 in the high dose range (50-100 µM).

The effect of probenecid on the PK of ceftobiprole and ceftobiprole medocaril was studied in rats to evaluate the excretion process in the kidney. Concomitant administration of probenecid with ceftobiprole or ceftobiprole medocaril showed no significant effects on the plasma clearances and area under the curve (AUC) parameters of ceftobiprole or its prodrug ceftobiprole medocaril, suggesting a lack of relevant PK interaction. Since probenecid is known to inhibit active renal secretion processes, it appears unlikely that ceftobiprole is significantly excreted by active transport processes in the kidney, which is consistent with the fact that total plasma clearances in animals and humans after administration of ceftobiprole or ceftobiprole medocaril were similar to their respective glomerular filtration rates.

Therefore, the potential of ceftobiprole to affect the P450-dependent metabolic clearance of potential co-administered drugs is not anticipated.

3.2.3 Toxicology

Acute Toxicity

No single-dose studies with ceftobiprole were conducted. The initial bolus dose selection was based on kinetic studies in rats with single-dose administration and subsequent computer simulations of expected exposures with Cmax values in the range of 700 µg/mL for a dose of 200 mg/kg. This dose was well tolerated in an exploratory single-dose study in mice.

Studies in rats demonstrated nephrotoxicity with ≥150 mg/kg thrice daily (TID) bolus administration of ceftobiprole medocaril, while nephrotoxicity was seen at >250 mg/kg twice daily (BID) doses of ceftobiprole when administered slowly for four hours. The nephrotoxicity observed in both cases was of similar effect.

Repeat-Dose Toxicity

The repeat-dose IV toxicity of ceftobiprole medocaril was evaluated in rats, dogs, and primates for up to 13 weeks duration.

Ceftobiprole medocaril was well tolerated up to 360 mg/kg infusion (four hours, twice a day) study in male rats. At doses ≥250 mg/kg, pigment deposits and amorphous precipitates were present in the distal renal tubules and collecting tubules. At the end of the administration period, hyaline droplets in the proximal renal tubules of almost all animals administered ceftobiprole medocaril may have reflected facilitated re-uptake of excreted test article, and were considered to be toxicologically insignificant since no necrotic or degenerative changes were observed in the tubules. The treatment-related findings exhibited a trend towards complete reversibility following the 4-week recovery period. Mean plasma concentrations were dose-related.

In a four-week infusion (four hours, once daily) study, minimally increased levels of urinary N-acetyl-beta-glucosiminidase (NAG) seen in high-dose (360 mg/kg/day) male rats at Weeks 2 and 4 were comparable to levels seen in control animals at the end of the recovery period. Dose-dependent minimal to slight cytoplasmic inclusions seen in renal proximal tubules at doses ≥250 mg/kg/day were not associated with any functional or other morphological changes in the kidneys. The renal toxicity observed in rats and marmosets showed reversibility after cessation of dosing. Safety margins for renal toxicity ranged from approximately 2-7 for plasma exposures, and from approximately 4-34 for urine exposures, based on clinical exposure data for the 500 mg dose administered as a 2-hour infusion. Enlargement of the cecum, seen in rats at 360 mg/kg, was related to the pharmacological activity of the drug.

In contrast to its benign profile in the proximal tubule, ceftobiprole medocaril shows dose-related adverse effects in the distal tubule and collecting duct. These effects were more pronounced for a bolus dose, compared to the same dose administered as a four-hour infusion, indicating a relationship to Cmax. The effects were associated with the presence of precipitate in the distal tubules and collecting ducts, and were considered to be due to precipitation of the active drug within the distal tubules. The active drug was present at very high levels in the urine due to this being the predominant route of elimination. During IV infusion, there is rapid conversion of ceftobiprole medocaril to ceftobiprole in the plasma, which is then filtered and concentrated in the distal tubules resulting in urine concentrations that are higher than concentrations measured in plasma, and causing the occurrence of drug precipitates in the distal tubules and collecting ducts. At a dose level of 175 mg/kg/h a precipitate was observed in the urine during a two-week marmoset study in which dosing was performed as a four-hour infusion every 12 hours.

The precipitate was confirmed as ceftobiprole by bioanalysis. This is consistent with the microscopic observations and supports the conclusion that the precipitate observed in the distal tubules is comprised of ceftobiprole.

The renal toxicity observed in rats and marmosets showed reversibility after cessation of dosing.

Dose-dependent convulsions were observed for ceftobiprole following direct administration (ICV) into the brain in mice. In contrast to the high potency for convulsions when administered directly into the brain, convulsions were rare after systemic administration of ceftobiprole medocaril and ceftobiprole, and were associated with overdosing. Doses ≥ 250 mg/kg showed dose-related incidences of convulsions associated with signs of renal toxicity.

Incidences of convulsions were also observed in rats and marmosets. The convulsions occurred at high doses that were associated with sustained elevated plasma levels (attributed in some cases to renal toxicity and reduced clearance). In a three-day marmoset pilot study, an animal showed convulsions approximately eight hours after the second IV bolus dose of 200 mg/kg ceftobiprole medocaril. The plasma concentration for this animal was 77.4 µg/mL, which is approximately two-times higher than the concentration observed two hours after the end of a four-hour infusion dose of 200 mg/kg ceftobiprole medocaril. The prolonged elevation of plasma levels after the IV bolus dose at 200 mg/kg is attributed to reduced clearance due to renal toxicity.

In a two-week marmoset study, convulsions were observed in two animals from the high dose group (360 mg/kg , administered as four-hour infusions twice daily) after the first infusion on Day 1. The plasma concentration of ceftobiprole in these two animals ranged from 317-355 µg/mL, which is approximately ten times greater than the clinical Cmax.

In a 13-week rat study, convulsions were observed in an animal from the high-dose group (750 mg/kg, administered as an eight-hour infusion once daily) on Day 34, approximately one hour before the end of infusion. The plasma concentration in this animal was 511 µg/mL at 1.5 hours after the end of infusion, which is about 15 times greater than the clinical Cmax.

Treatment-related infusion site reactions were observed during repeat dose studies in marmosets and rats, including the presence of a yellowish-white fibrinous material with thrombus formation at the catheter tip and around the catheter, and signs of vascular irritation (thickening of the vessel wall, erosion of the endothelium, localized thrombosis and chronic active inflammation with fibrosis of the vein wall). These local vascular effects at the infusion site and associated thromboembolic effects, were attributed as the cause of mortality in the 13-week marmoset study and as a cause of incidences of mortality in the 13-week rat study. The infusion site reactions were further evaluated in two-week rat bridging toxicity studies that were conducted subsequent to the 13-week studies. Local vascular irritation by the test article occurred mainly in the region of the vessel apposed to the catheter, with little effect in the region of the vessel distal to the catheter. Thus, damage by the catheter may play a permissive role in these animals, with test article-related effects occurring only in the damaged regions.

Thromboembolic effects were not observed in a 13-week dog study that included serial sectioning of the brain, kidney, and heart which indicated that the test article-related effects for thromoembolic reactions appear to be more prominent in small animals.

Genotoxicity

The genotoxic potential of ceftobiprole medocaril and/or ceftobiprole was assessed by a battery of in vitro and in vivo assays: Ames test, Mouse Lymphoma Cell Mutation Test (ML/TK), human chromosome aberration assay, mouse micronucleus test, and measurement of unscheduled DNA synthesis in rat liver. In comparison to ceftobiprole, only ceftobiprole medocaril and cleavage product "diacetyl" were found to possess genotoxic activies in the ML/TK test. This genotoxic activity was presumed to have been attributable to the formation of the cleavage product 2,3-butan-dione. In the confirmatory in vivo mouse micronucleus assay, ceftobiprole medocaril did not induce chromosomal aberrations in bone marrow of the mouse. Under the above-mentioned experimental conditions, ceftobiprole medocaril did not show genotoxic potential.

Carcinogenicty

No carcinogenicity studies were conducted to evaluate the carcinogenic potential of ceftobiprole medocaril. This was considered acceptable due to the intended short-term clinical duration of therapy and the low potential for genotoxicity.

Reproductive and Developmental Toxicity

The potential effects of ceftobiprole medocaril on fertility were examined in two separate studies in male and female rats. Dose levels of 0, 175, 250, and 360 mg/kg/day ceftobiprole equivalent were used in both these studies, administered as once daily IV infusions (four hours). There was no effect on male or female reproductive performance or on early embryonic development. Abnormally coloured urine was observed, as well as a slightly suppressed body weight gain in males. The no observable adverse effect level (NOAEL) for general toxicity was 250 mg/kg/day in males, and 360 mg/kg/day in females. In both studies, the NOAEL for reproductive performance and early embryonic development was 360 mg/kg/day. Toxicokinetic data demonstrated a dose-proportional increase in mean plasma concentration of ceftobiprole between doses of 175 mg/kg/day and 360 mg/kg/day.

Embryo-foetal development toxicity studies were conducted in rats and cynomolgus monkeys. The NOAEL for dams and embryo-fetal development in rats was 360 mg/kg/day. The NOAEL for pregnant monkeys was 60 mg/kg/day, and 120 mg/kg/day for fetuses. No teratogenic potential or embryo-fetal effects were noted up to 120 mg/kg/day.

In a pre- and post-natal developmental toxicity study, doses of ceftobiprole medocaril (0, 175, 250, and 360 mg/kg/day) were administered to pregnant female rats as a once daily IV infusion (four hours) from implantation to weaning of the first generation (F1) pups on Day 21 of lactation. The number of born and live pups (F1) was slightly reduced at 360 mg/kg/day, due to post-implantation loss associated with maternal toxicity. No effect on other reproductive parameters was observed. Functional and physical development of pups was normal in all groups; mating performance of F1 animals was not affected by treatment and no effect on reproductive parameters was observed for the F1 animals. No treatment-related effect on F2 animals was observed.

The NOAEL for ceftobiprole medocaril in dams was 175 mg/kg/day for general maternal toxicity with no effects noted in body weight or food consumption, 250 mg/kg/day for reproductive toxicity with no effects noted in live birth or viability indices in Fl, and 360 mg/kg/day for developmental toxicity in the Fl and F2 generations. Enlargement of the cecum was observed in the 175 mg/kg/day groups due to general maternal toxicity.

Ceftobiprole was excreted in milk with levels amounting to approximately one fifth of the corresponding maternal plasma levels. Nursing pups were not exposed systemically to ceftobiprole, which was consistent with low absorption via the oral route of administration.

No studies in which the offspring were dosed with ceftobiprole medocaril have been conducted to date.

Local Tolerance

Two local tolerance studies were conducted in rabbits. There was a slight decrease in adverse events (AEs) between the irritancies of freshly prepared ceftobiprole medocaril (within six hours of preparation) and that of formulation prepared 30 hours previously.

Antigenicity

The antigenicity of ceftobiprole medocaril was investigated in the guinea pig in a Maximization Test, an active systemic anaphylaxis (ASA) assay, and a four-hour passive cutaneous anaphylactic (PCA) assay. The low dose was approximately the intended clinical dose and the high dose was a 5X multiple.

Ceftobiprole medocaril was found to possess antigenic potential. The positive ASA reactions (scratching, piloerection) were present in animals receiving single IV bolus doses of ≥20 mg/kg ceftobiprole medocaril five times weekly for three weeks or 50 mg/kg ceftobiprole medocaril emulsified with Freund's complete adjuvant (FCA) or Freund's incomplete adjuvant (FIA) once weekly for three weeks.

Nephrotoxicity

No nephrotoxicity was observed in male rabbits that received a single IV administration of 160 mg/kg of ceftobiprole medocaril. There was red discolouration of renal papilla.

Phototoxicity

Ceftobiprole proved to be non-phototoxic in vitro after ultraviolet (UV) irradiation and ceftobiprole medocaril did not induce a phototoxic skin reaction in vivo under the experimental conditions used.

Potential for Hemolysis and Plasma Turbidity

No significant hemolysis and plasma turbidity or precipitation was seen in dog blood under in vitro conditions in a 13.3% test solution of RO65-5788 corresponding to 1.25% of ceftobiprole. At higher concentrations, hemolysis up to 7.3% and moderate to marked plasma turbidity and precipitation were observed and were attributed to the low solubility of ceftobiprole in plasma under physiological pH conditions.

The intravenous administration of ceftobiprole medocaril formulated in reconstitution solution, whilst causing no significant hemolysis, may cause increasingly significant adverse plasma interactions at infusion solution concentrations of 12.5 to 50 mg/mL, whichever species is used. The severity of adverse plasma interactions may be exacerbated with increasing infusion volume, repeated dosing, and/or increasing infusion duration.

3.2.4 Microbiology

Ceftobiprole is an expanded-spectrum cephalosporin with activity against gram-positive and gram-negative aerobic bacteria. Ceftobiprole exerts its antibacterial activity by binding to essential PBPs in both gram-positive [including methicillin-resistant Staphylococcus aureus (MRSA)] and gram-negative bacteria. In gram-positive bacteria, ceftobiprole differs from other cephalosporins and ß-lactams due to its unique high affinity for PBP2a from methicillin-resistant staphylococcus. Ceftobiprole has high affinity for the essential PBP3 and PBP2 of Escherichia coli.In vivo ceftobiprole treatment in animal models of infection did not produce resistant mutants of Staphylococcus aureus, Enterobacter cloacae, and extended spectrum b-lactamase (ESBL)-negative Klebsiella pneumoniae.

Ceftobiprole also appears to be hydrolyzed by b-lactamases from anaerobic bacteria. Ceftobiprole demonstrated reduced clinical efficacy against infections with Pseudomonasaeruginosa with a minimum inhibitory concentration (MIC) of >4 ug/mL, and as such, use of Zeftera against Pseudomonas aeruginosa is not recommended.

Ceftobiprole MICs against both gram-positive and gram-negative bacteria were not affected by more than one doubling dilution by such factors as divalent calcium, 2% sodium chloride, 5% carbon dioxide, and pH ranging from 6.4-8.2. Inoculum effects were generally not seen.

The efficacy of ceftobiprole was examined in skin and soft tissue infection models septicemia infection models, and in a rabbit model of MRSA osteomyelitis.

  • Ceftobiprole achieved a >1.7 log10 colony forming units/gram (CFU/g) reduction of methicillin-sensitive Staphylococcus Aureus (MSSA) in skin tissue compared to the initial inoculum at doses of 1.6 to 100 mg/kg/day. Against MRSA, ceftobiprole showed similar or improved reduction in bacterial load at doses of 1.6 to 100 mg/kg/day against comparators. Ceftobiprole was effective in reducing the lesion volume at the infection site in animals infected with either MSSA or MRSA .
  • Ceftobiprole was effective in treating experimental septicemia. The strains included MSSA, MRSA (except MRSA 8525), Escherichia coli, Klebsiella pneumoniae, and Proteus mirabilis.
  • Ceftobiprole provided effective parenteral treatment of osteomyelitis caused by MRSA in the rabbit osteomyelitis infection model. Following ceftobiprole treatment bacterial titres in all infected left tibiae from evaluable rabbits were below the level of detection, whereas 73% of infected left tibiae from comparator treated animals had bacterial titres below the level of detection. Mean ceftobiprole titres were 3-5 times higher in the infected left tibias compared to the uninfected right tibiae.

3.2.5 Conclusion

Appropriate non-clinical toxicology studies were performed and are considered adequate. The toxicology findings were consistent with the pharmacological effects of ceftobiprole medocaril/ceftobiprole. The non-clinical pharmacology and toxicology studies support the use of Zeftera for the proposed indication.

3.3 Clinical basis for decision

3.3.1 Human Pharmacology

Two formulations of ceftobiprole medocaril were used in Phase 1, 2, and 3 clinical studies. Both formulations (designated as Phase 2 and Phase 3 formulations) were used in the studies summarized in the submission; the commercial formulation was not used in any study. Differences between the formulations related to the amount of citric acid, the pH, and the presence of mannitol. No clinically relevant differences in PK is expected between these formulations.

Bioavailability

Since ceftobiprole (as ceftobiprole medocaril) is administered intravenously, its bioavailability is assumed to be 100% and therefore no bioequivalence studies to compare the various formulations were conducted or are planned. The systematic exposure associated with these formulations was compared across several Phase 1 studies. Peak drug concentrations (Cmax) were dependent on the infusion duration, such that shorter infusion durations (0.5 and 1 hour) resulted in higher Cmax estimates than the longer (2 hour) infusion durations. The half-life of ceftobiprole was similar across all studies. Systemic exposure and clearance of ceftobiprole were generally similar in studies that utilized the formulations with and without mannitol.

In Vitro Studies

Two studies were conducted in pooled human plasma samples at 0.5-100 µg/mL ceftobiprole or freshly obtained (within 72 hours of blood collection), non-pooled human plasma samples at 3.5 and 35 µg/mL ceftobiprole. The mean percentages of binding were 17.3% and 15.4% in the two studies, respectively, resulting in a mean plasma protein binding value of approximately 16%.

3.3.2 Pharmacokinetics

Eleven Phase 1 studies in healthy volunteers and one Phase 2 open-label study in patients with cSSSI infections with Gram-positive bacteria were conducted. PK data was also made available from a limited number of patients enrolled in two Phase 3 pivotal trials. The PK of ceftobiprole was studied by a cross-over ascending dose study and single and multiple ascending dose studies.

Conversion from the prodrug form (ceftobiprole medocaril) to the active ceftobiprole occurred rapidly and was mediated by plasma Type A esterases, such that prodrug concentrations were low and are measurable only during infusion administration. Systemic exposure (AUC and Cmax) of ceftobiprole increased linearly at the dose range of 125-1000 mg ceftobiprole (infused as ceftobiprole medocaril) for 30 minutes by the IV route. The mean half-life (t1/2) estimate was approximately three hours. The steady-state volume of distribution (18.4 L) approximated the extra-cellular fluid volume in humans, which is typical of cephalosporin antibiotics and suggests that intra-cellular penetration of ceftobiprole does not occur to an appreciable extent. Estimates of the volume of distribution at steady state (Vss) were consistent across the dose ranges studied (125-1000 mg) and remained consistent following single- and multiple-dose administrations. Accumulation was negligible following repeated continuous-dose administration according to a regimen of dosing once every 12 hours.The in vitro studies showed a mean human plasma protein binding of approximately 16%.

Ceftobiprole underwent minimal metabolism to the open-ring metabolite. Total systemic clearance was approximately 5 L/h and was consistent across the dose range tested. Approximately 80-90% of the dose was recovered in urine as unchanged ceftobiprole. Urinary excretion of ceftobiprole was collectively based on urine concentrations of ceftobiprole and its open-ring metabolite. The metabolite exhibited a slightly longer half-life (5 hours) than ceftobiprole (3 hours). Approximately 5% of the administered dose was excreted in the urine as the open-ring metabolite. The PK of ceftobiprole were consistent following 30 minutes continuous dosing of 750 mg ceftobiprole via IV infusion for 12 days, indicating time-invariant PK.

Following the 250 mg infusion, the time during which the plasma concentrations of ceftobiprole exceeded the 90% minimum inhibitory concentration (MIC90) of 4 µg/mL was approximately four hours. For the 500, 750 mg, and 1000 mg doses, the mean percent time >MIC for unbound ceftobiprole concentrations above an MIC of 4 µg/mL was 58%, 81%, and 85%, respectively, assuming a dosing interval of eight hours, and 37%, 55%, and 57%, respectively, assuming a dosing interval of 12 hours. These results demonstrated that a reduction in dose, together with a prolonged infusion time, produced similar T>MIC (the time the concentration of an antibiotic remains above the MIC) values.In the treatment of MRSS, T>MIC90 for 40-50% of the dosage interval is assumed to be sufficient.One gram of a once-daily dose of ceftobiprole given as a 90-minute infusion for eight days demonstrated that unbound ceftobiprole plasma concentrations (based on 16% protein binding) were above the MIC of 4 µg/mL for a mean time of eight hours, equivalent to the dosing interval.

Drug-Drug Interactions

Ceftobiprole is neither a substrate nor inhibitor of p-glycoprotein, thus transport-related interactions are not anticipated. Ceftobiprole is not extensively metabolized, does not induce cytochrome P450 isoenzymes, and minimally inhibits cytochrome P450 isoenzymes, therefore metabolic drug-drug interactions are not anticipated. The overall potential for ceftobiprole to interact with other agents and the potential for other agents to interact with ceftobiprole is considered very low. Exploratory population PK screening after IV administration of ceftobiprole in healthy volunteers and patients with cSSSI indicated that the administration of the following concomitant medications did not affect the PK of ceftobiprole: fentanyl, lidocaine, paracetamol (acetaminophen), diclofenac, acetylsalicylic acid (aspirin), heparin, diphenhydramine, propofol, hydromorphone hydrochloride, methadone, hydrocodone bitartrate, metamizole sodium, and furosemide.

Special Populations
Renal Impairment

One study was conducted to study the PK, following a single 30-minute administration of 250 mg ceftobiprole, in volunteers with normal renal function, and patients with mild, moderate, and severe renal impairment. Systemic exposure in terms of AUClast was 29% higher in patients with mild renal impairment and 2.5-fold and 3.3-fold higher in patients with moderate and severe renal impairment, respectively, compared to subjects with normal renal function. Total systemic clearance (CLS) and renal clearance (CLR) decreased such that the greatest reductions were noted in the moderately (62% for CLS and 78% for CLR) and severely (75% for CLS and 91% for CLR) renally impaired patients. Urinary recovery ranged from 32-74% in patients with mild to severe renal impairment. Estimates of Cmax and Vss were similar across the varying degrees of renal impairment.Elimination half-life increased with decreasing renal function, such that patients with severe renal impairment exhibited the longest t1/2, 11 hours.

Hepatic Impairment

No studies were conducted to examine the effect of ceftobiprole on subjects with hepatic impairment as ceftobiprole does not appear to undergo significant hepatic metabolism

and its systemic clearance is not expected to be significantly affected by hepatic impairment. This was considered to be acceptable.

QTc Prolongation

Two Phase 1 studies were conducted to assess the impact of ceftobiprole on QTc prolongation in healthy volunteers. One study with single 2-hour infusions of 500 mg and 1000 mg ceftobiprole (given as ceftobiprole medocaril) in 32 male and 28 female healthy volunteers reported no discernable relationship between drug exposure and QTc prolongation. From the second study no conclusions could be drawn in regards to QTc prolongation due to the limited number of subjects with serial electrocardiogram (ECG) data (4/20). A high frequency (10 subjects, 50%) of infusion site reactions were reported as adverse events and study was discontinued.

Age

The PK of ceftobiprole in patients under the age of 18 has not yet been established.

In the Phase 3 clinical studies the effect of age on the PK of ceftobiprole was explored as a covariate in the population PK analysis of 211 elderly subjects (≥65 years old),including 63 subjects who were ≥75 years old. Age was not identified as a statistically significant covariate in the final population PK model when creatinine clearance explained the apparently lower clearance in elderly subjects. Therefore, no dosage adjustments are recommended based on age alone; however, since elderly patients often have impaired renal function, this should be taken into consideration when determining dose in elderly patients.

Gender

The effect of gender on the PK of ceftobiprole was evaluated in two clinical pharmacology studies. Systemic exposure was higher in females (21% for Cmax, 15% for AUC ) than in males. Estimates of CLS and Vss were 12% and 29% lower, respectively, in females compared with males. Half-life estimates were similar in both sexes (3 hours).

Renal clearance and urinary excretion of ceftobiprole were similar in both males and females, such that the amount excreted unchanged in the urine ranged from a mean of 92-96%.

No dosage adjustments are recommended based on sex.

Race

No specific clinical studies were conducted to investigate the effect of race on the PK of ceftobiprole. The population PK analysis demonstrated that there was no difference in systemic clearance of ceftobiprole among the various race groups.

Pharmacokinetic/Pharmacodynamic Parameters (from human trials)

Population PK/PD data from Phase 1 trials demonstrated a fractional target attainment rates of approximately 90% or greater for ceftobiprole MICs ≤4 µg/mL against Gram-positive pathogens that require a 30% T>MIC. For Gram-negative pathogens with ceftobiprole MICs ≤4 µg/mL, the fractional target attainment for the 50% T>MIC target was 89% or greater when calculated for a ceftobiprole dose of 500 mg every 8 hours with a 2-hour infusion.

3.3.3 Clinical Efficacy

The clinical program supporting the effectiveness of ceftobiprole in the treatment of cSSSI included two Phase 3 studies (BAP00154 and BAP00414) and one open-label Phase 2 study (BAP00034). All studies were conducted in accordance with the ICH guidelines for Good Clinical Practice. The major pathogens isolated from SSSI and examined in these studies included: Staphylococcus aureus (including methicillin-resistant isolates), Pseudomonas aeruginosa, Enterococcus spp., Escherichia coli, Enterobacter spp., and Klebsiella spp.

Study BAP00154

Study BAP00154 was a Phase 3, randomized, double-blind, multicenter, efficacy and safety study designed to assess the non-inferiority of ceftobiprole in comparison to vancomycin with respect to the clinical cure rate of cSSSI caused by proven or suspected Gram-positive pathogens that required therapy with an anti-MRSA antibiotic. Approximately 790 subjects at least 18 years of age were hospitalized with a diagnosis of infection consistent with a cSSSI (defined as infection either involving deeper soft tissue or requiring significant surgical intervention), and one or more of the following: an infection within 30 days of surgery/trauma, abscess (without open wound), or cellulitis. Subjects were randomized in a 1:1 ratio to receive 500 mg of ceftobiprole or 1000 mg of vancomycin by a 60-minute IV infusion twice daily for 7-14 days, with a possible extension up to 28 days if deemed necessary by the investigator (the original protocol allowed prolongation of therapy beyond 14 days up to a maximum of 28 days if approved by the medical monitor; a subsequent amendment near the end of enrolment prohibited prolongation of therapy beyond 14 days).

BAP00154 achieved the primary and secondary endpoints of non-inferiority of ceftobiprole compared with vancomycin for clinical cure rate, microbiological eradication rate, clinical relapse rate, and microbiological relapse rate in subjects with cSSSI infections due to Gram-positive pathogens.

Clinical cure rates at the test-of-cure (TOC) visit were similar for ceftobiprole and vancomycin in the co-primary clinically evaluable analysis set (93.3% and 93.5%, with a 2-sided 95% confidence interval (CI) for the difference of -0.2% (-4.4%; 3.9%), the intent-to-treat (ITT) analysis set was (77.8% and 77.5%, respectively) with a 2-sided 95% CI for the difference of 0.3% (-5.5%; 6.1%). The Non inferiority of ceftobiprole compared with vancomycin was demonstrated for both co-primary populations. Similar results were observed with the mITTand microbiologically evaluable populations.

Non-inferiority of ceftobiprole compared with vancomycin was demonstrated for clinical relapse rates. Clinical relapse rates at the lost to follow-up (LFU) visit in the clinically evaluable analysis set were 1.6% for ceftobiprole and 0.8% for vancomycin, with a 2-sided 95% CI for the difference of 0.8% (-1.15%, 2.68%). 

Analysis of the microbiological outcome at the TOC visit showed that the pathogens in 94% of microbiologically evaluable subjects in both the ceftobiprole and vancomycin treatment arms were considered to be Eradicated or Presumed Eradicated (94.2% and 93.5%, respectively) with a 2-sided 95% CI for the difference of 7% (-3.8%; 5.2%). Similar results were observed in the mITT population (68.3% and 67.4%, respectively) with a 2-sided 95% confidence interval for the difference of 0.8% (-6.6%; 8.2%); a lower limit greater than or equal to -10% indicates that ceftobiprole is not inferior to vancomycin. Microbiological eradication rates in the microbiologically evaluable population for subjects with infection isolates positive for Staphylococcus aureus (MSSA and MRSA) were similar.

Non-inferiority of ceftobiprole compared with vancomycin was demonstrated at the LFU visit for microbiological relapse rates. The evaluable analysis set were 2.8% for ceftobiprole and 4.9% for vancomycin with a 2-sided 95% CI for the difference of -2.1% (-5.8%; 1.6%).

The results of this study showed that 1) ceftobiprole is as effective as vancomycin in the treatment of cSSSI due to gram-positive infections, and 2) ceftobiprole administered at 500 mg using a 60-minute intravenous infusion twice daily for 14 days is safe and well tolerated.

Study BAP00414

Study BAP00414 was a Phase 3, randomized, double-blind, multicentre study to assess the efficacy and safety of ceftobiprole in comparison to vancomycin plus ceftazidime in subjects with cSSSI including DFI, caused by Gram-positive and/or Gram-negative pathogens. Approximately 816 subjects hospitalized with cSSSI, at least 18 years of age, were to be enrolled to achieve 570 clinically evaluable subjects. Subjects were randomized in a 2:1 ratio to receive 500 mg ceftobiprole every eight hours plus placebo twice daily, or 1000 mg vancomycin every 12 hours plus 1000 mg ceftazidime every 8 hours for 7-14 days.

BAP00414 achieved the primary and secondary endpoints of non-inferiority compared with vancomycin plus ceftazidime for clinical cure rate, clinical relapse rate, microbiological eradication rate, and microbiological relapse rate in subjects with cSSSI infections, including diabetic foot infections, due to Gram-positive, Gram-negative, or mixed infections.

The clinical cure rates were similar between the treatment groups in the clinically evaluable at the TOC visit for the clinically evaluable analysis set (90.5% and 90.2%), the 2-sided 95% CI for the difference in the cure rates was 0.4 (-4.2% to 4.9%) for ceftobiprole and comparator) and for the ITT analysis set, (81.9% and 80.8%); the 2-sided 95% CI being 1.1 (-4.5% to 6.7%) for ceftobiprole and comparator, respectively). Noninferiority was demonstrated between the two treatments within the 10% noninferiority margin.

Clinical cure rates in the microbiologically evaluable analysis set for subjects in whom Staphylococcus aureus (MRSA and MSSA ) were identified as the pathogen causing disease were 89.7% and 93.8%, respectively, for ceftobiprole and 86.1% and 93.3%, respectively, for vancomycin plus ceftazidime with a 2-sided 95% CI being 3.5 (-9.4% to16.5%) and 0.4 (-6.0% to 6.8% respectively).

The clinical cure rates for subjects with diabetic foot infections were (86.2% for ceftobiprole and 81.8% for vancomycin plus ceftazidime groups); with a 2-sided 95% CI of 4.4 (-5.9; 14.7). The rates were lower in both treatment groups compared with subjects with other infection types.

Clinical relapse rates at the LFU visit in the clinically evaluable analysis set were 2.1% for ceftobiprole and 0.5% for vancomycin plus ceftazidime. The 2-sided 95% CI for the difference of the clinical relapse rates was 1.7 (-0.0; 3.3). The upper limit less than or equal to 10% indicates that ceftobiprole is noninferior to vancomycin/ceftazidime.

Microbiological eradication rates were similar for ceftobiprole compared with vancomycin plus ceftazidime in the microbiologically evaluable analysis set (88.0% and 88.9%, respectively). The 2-sided 95% CI was -1.0 (-6.4; 4.5), and 79.3% and 79.0%, respectively for the mITT analysis set with a 2-sided 95% CI for the difference of 0.2 (-6.3; 6.8). Noninferiority was demonstrated between the two treatment groups.

Microbiological relapse rates at the LFU visit in the microbiologically evaluable analysis set were 2.5% for ceftobiprole and 0% for vancomycin plus ceftazidime. The upper limit of the 2-sided 95% CI for the difference in microbiological relapse rates for ceftobiprole minus vancomycin plus ceftazidime was less than the non-inferiority margin of 10%.

The results of this study demonstrate that 1) ceftobiprole is as effective as vancomycin plus ceftazidime for subjects with cSSSI due to gram-positive and gram-negative pathogens, including subjects with diabetic with foot infections, and 2) ceftobiprole administered at 500 mg using a 120-minute intravenous infusion 3 times daily is safe and well-tolerated. Overall systemic exposure of ceftobiprole appears to be lower in cSSSI subjects with normal renal function compared with healthy volunteers; however, %T>MIC, which is considered to be the critical pharmacodynamic parameter associated with favourable clinical outcome, was similar in both populations. The pharmacokinetics of ceftobiprole in subjects with cSSSI will be further evaluated in a population pharmacokinetic analysis.

Study BAP00034

Study BAP00034 was a Phase 2, open-label, non-comparative, multicentre study designed to assess the clinical cure rate and microbiological eradication/presumed eradication rate of cSSSI caused by Gram-positive pathogens, including MRSA, after treatment with ceftobiprole. Subjects enrolled in this study were 18-75 years of age with a diagnosis of cSSSI, with or without bacteremia, involving surgical incision or site of trauma (including burns), abscess without open wound requiring surgical intervention, or cellulitis. Subjects received 750 mg of ceftobiprole as an IV infusion over 30 minutes or 60 minutes twice daily for 7-14 days. Subjects were evaluated before the start of therapy, during therapy on Days 1, 2, 3, 5, and 7, and if therapy was extended beyond 7 days, on Days 10 and 14 (or within 24 hours after the end of therapy visit). If treatment was prolonged, assessments were scheduled on Days 17 and 21 (or within 24 hours after the end of therapy).

In subjects with Gram-positive cSSSI infections, clinical cures were reported for all 34 (100%) clinically evaluable subjects, including four subjects with cases of MRSA . Microbiological eradication was reported for 21/23 (91%) microbiologically evaluable subjects, including three of the four cases of MRSA . In the microbiologically evaluable analysis set, the mean time to eradication was 5.5 days overall. An improvement over time of clinical signs and symptoms was observed for all parameters after the start of therapy. At TOC assessment, the majority of subjects in the ITT analysis set (79%) had no clinical signs and symptoms of the infection and the size of the primary site of infection was substantially reduced.

Other Pathogens

Enterococcus faecalis, Staphylococcus epidermidis, and Streptococcus agalactiae were not approved, because of insufficient clinical efficacy data.

Data from PK/PD modeling, as well as clinical studies from patients with infections due to Pseudomonas aeruginosa with an MIC >4 µg/mL, indicated efficacy in only 50% of patients, and therefore were not approved.

Although the number of subjects with Streptococcus pyogenes was low in diabetic foot infections epidemiological data suggested that the chances of getting Streptococcus pyogenes in cSSSI is <5%. Current clinical trials demonstrated 7% of group A streptococcal isolates were isolated from cSSSI without DFI, and <1% from cSSSI with DFI. Surveillance data on MIC90 (0.06), breakpoint for Streptococcus species (<0.5 µg/mL), and historical background information on activity of cephalosporins against Streptococcus pyogenes was used in allowing inclusion of Streptococcus pyogenes for the proposed indication of cSSSI without diabetic foot infection only.

Susceptibility Testing

The ceftobiprole MICs against both Gram-positive and Gram-negative bacteria were not affected by more than one doubling dilution by such factors as divalent calcium, 2% sodium chloride (NaCl), 5% carbon dioxide (CO2), and pH ranging from 6.4 to 8.2. Inoculum effects were generally not seen.

3.3.4 Clinical Safety

A total number of 1593 patients were studied in the clinical safety. The incidences of adverse events (AEs), treatment-limiting AEs, and serious adverse events (SAEs) in ceftobiprole-treated subjects were similar to the incidences that occurred in the comparators. Subjects in both Phase 3 studies (including diabetic foot patients in study BAP00414), were treated between 14-21 days, and 21-28 days. There were 18 subjects who received ceftobiprole for more than 14 days in the safety analysis set. There were no patients treated with ceftobiprole for more than 21 days. There were two patients who were clinically evaluable in BAP00414 and 10 patients who were clinically evaluable in BAP00154 who received therapy for more than 14 days. The types of AEs reported in these subjects were generally similar to those subjects who received shorter courses of therapy. The safety profile of subjects who received more than 14 days of therapy was similar to the safety profile of cSSSI subjects who received 14 days or less of therapy. Staphylococcus aureus was the most common pathogen isolated (8/18 subjects).

The most common adverse drug reactions in patients treated with ceftobiprole were: nausea (9.1%); dysgeusia (5.6%); vomiting (4.8%); diarrhea (4.8%); and headache (4.5%). The majority (92.6%) of AEs were reported as mild to moderate in severity. Ceftobiprole was discontinued due to an AE in 3.8% of subjects compared with 4.1% for all comparators.

During clinical trials, adverse drug reactions that led to ceftobiprole discontinuation included: rash (0.6%); nausea (0.5%); vomiting (0.4); hypersensitivity reactions (0.3%); and hyponatremia (0.3%). For the comparators, adverse drug reactions leading to discontinuation were: rash (0.9%); hypersensitivity reactions (0.9%); nausea (0.5%); infusion site reactions (0.5%); and diarrhea (0.5%).

In individuals receiving ß-lactam antibiotics and with a history of sensitivity to multiple allergens, anaphylaxis can be serious and occasionally fatal. Hypersensitivity reactions with ceftobiprole were not more likely than comparators to result in treatment discontinuation or be considered a serious adverse event. Patients with pre-existing central nervous system (CNS)/seizure disorders may experience seizures during treatment with ß-lactams, and caution should be observed when treating these patients. The concomitant administration of aminoglycosides and some cephalosporins has been observed to cause nephrotoxicity.

The concomitant administration of aminoglycosides and some cephalosporins has been observed to cause nephrotoxicity. In patients with moderately or severely impaired renal function [Creatinine Clearance (CrCl) 10 to <50 mL/min ], dosage adjustment is required. There is no experience in patients with end stage renal disease (CrCl <10 mL/min) or in patients on dialysis. The concomitant administration of aminoglycosides and some cephalosporins has caused nephrotoxicity. Concomitant administration of Zeftera with known nephrotoxic agents has not been studied.

Elderly patients with normal renal function do not require a dosage adjustment. Since elderly patients may have impaired renal function, the CrCl should be taken into consideration when determining the dosing regimen in the elderly.

Overall, the safety results of the clinical studies demonstrated that ceftobiprole administered at 500 mg using a 120-minute IV infusion three times daily is safe and well tolerated.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Zeftera (ceftobiprole medocaril for injection) is a new broad-spectrum cephalosporin which has in vitro activity against most of the common Gram-negative and Gram-positive pathogens. Ceftobiprole is a new unique cephalosporin with respect to its expanded activity against MRSA. In gram-positive bacteria, ceftobiprole differs from other cephalosporins and β-lactams due to its unique high affinity for PBP2a from methicillin-resistant staphylococci. Ceftobiprole also has a high affinity for the essential PBP3 and PBP2 of Escherichia coli.

This New Drug Submission supports the efficacy and safety of Zeftera for the treatment of cSSSI including Non Limb Threatening Diabetic Foot Infections without Concomitant Osteomyelitiscaused by the following organisms: Staphylococcus aureus (including methicillin resistant isolates), Streptococcus pyogenes, Escherichia coli, Proteus mirabilis, Enterobacter cloacae, Klebsiella pneumoniae.

The dosing instructions for cSSI patients differ depending on the type of infection and type of causative organism. Once the organism is identified, antimicrobial therapy should be adjusted as per the instructions given in the Zeftera Product Monograph.

Complicated skin and skin-structure infections (cSSSI) include deep wounds; including surgical incisions, bites and lacerations that have become infected, major abscesses, and infected ulcers which might complicate if accompanied by a significant underlying disease. Skin and skin-structure infections occur frequently in clinical practice. Post-operative surgical-site infections represent up to 25% of all nosocomial infections, and their treatment generally requires hospitalization and substantial medical expense.

The emergence of resistant pathogens, especially MRSA, with decreased susceptibility, has created a need for modifying these regimens to expand the spectrum of antibacterial activity. Treatment presents a clinical challenge to the physician, particularly with the increase in multi drug-resistant strains and widespread cross-resistance to antibiotic treatment.

The safety profile of Zeftera is comparable to that of vancomycin with or without ceftazidime and was safe for the treatment of subjects with cSSSI infections, including non-limb-threatening diabetic foot infections.

A label warning has been included indicating that serious and occasionally fatal hypersensitivity (anaphylactic) reactions have been reported in patients receiving ß-lactam antibiotics, and are more likely to occur in individuals with a history of sensitivity to multiple allergens. Anaphylaxis has been observed with Zeftera. Before therapy is instituted, careful inquiry should be made to determine whether the patient has had a previous hypersensitivity reaction to other cephalosporins, penicillins or other allergens.

In patients with moderately or severely impaired renal function, dosage adjustment is required. Since elderly patients may have impaired renal function, the creatinine clearance should be taken into consideration when determining the dose in elderly patients.

Seizures associated with Zeftera have occurred most commonly in patients with pre-existing CNS/seizure disorders. Therefore caution is advised when treating these patients with Zeftera.

Infusion site reactions, including pain, phlebitis, and thrombophlebitis have been observed in clinical trials.

Hyponatremia was observed in the clinical trials with Zeftera. For patients at risk of hyponatremia, consideration should be given to the choice of infusion solution for Zeftera.

Overall, the clinical studies demonstrated that Zeftera is non-inferior to the comparator, and is well-tolerated and associated with a manageable safety profile. Based on the safety and efficacy profile, the benefits of Zeftera therapy seem to outweigh the risks. Restrictions to manage risks associated with the identified safety concerns have been incorporated into the Zeftera Product Monograph.

3.4.2 Recommendation

Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Zeftera is favourable in the treatment of the following infections when caused by susceptible strains of the designated microorganisms in patients 18 years of age and older:

  • Complicated skin and skin-structure infections (cSSSI), including non-limb-threatening diabetic foot infections without concomitant osteomyelitis caused by: Enterobacter cloacae, Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Staphylococcus aureus (including methicillin-resistant isolates), and Streptococcus pyogenes.

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: Zeftera

Submission MilestoneDate
Submission filed:2007-07-16
Screening
Screening Acceptance Letter issued:2007-08-31
Review
Quality Evaluation complete:2008-06-23
Clinical Evaluation complete:2008-06-26
Labelling Review complete:2008-06-26
Notice of Compliance issued by Director General:2008-06-26