Summary Basis of Decision for Primovist ®
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:
Primovist®
Gadoxetate disodium, 181.43 mg/mL (0.25 mmol/mL), Solution, Intravenous
Bayer Inc.
Submission control no: 127609
Date issued: 2010-05-28
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:
Manufacturer/sponsor:
Medicinal ingredient:
International non-proprietary Name:
Strength:
Dosage form:
Route of administration:
Drug identification number(DIN):
- 02340666
Therapeutic Classification:
Non-medicinal ingredients:
Submission type and control no:
Control Number: 127609
Date of Submission:
Date of authorization:
2 Notice of decision
On January 14, 2010, Health Canada issued a Notice of Compliance to Bayer Inc. for the drug product Primovist.
Primovist contains the medicinal ingredient gadoxetate disodium which is a contrast enhancement agent for magnetic resonance imaging (MRI).
Primovist is indicated for intravenous (IV) use in T1-weighted MRI of the liver to detect and characterize lesions in adults with known or suspected focal liver disease.
Primovist is taken up selectively by liver cells (hepatocytes) where it accumulates and promotes enhanced visualization of liver tissue. In liver lesions with no or minimal hepatocyte function (cysts, metastases, and the majority of hepatocellular carcinomas), the accumulation of Primovist is much lower than in the surrounding tissue. This results in an increased contrast image of normal liver tissue versus liver lesions lacking normal hepatocyte function.
The market authorization was based on quality, non-clinical, and clinical information submitted. The clinical efficacy of Primovist was evaluated in four pivotal, open-label, multi-centre, Phase III studies, which included two lesion detection studies (96129 and 97610) and two lesion characterization studies (012387 and 014763). A total of 797 patients with suspected or known focal liver lesions enrolled in these studies, with 621 patients receiving Primovist. There were no control groups as each patient acted as their own control.
The primary efficacy variable for the detection studies was the sensitivity of liver lesion detection which took into account the possibility of multiple lesions within patients, as well as matching and tracking lesions within patients compared to a Standard of Reference (SOR; a combination of pathology from resected liver specimens, an intra-operative ultrasound and, if appropriate, a 3-month follow-up imaging examination). For the characterization studies, the primary efficacy variable was the proportion of correctly characterized liver lesions (correct liver lesion type) with Primovist-enhanced MRI (combined pre- and post-contrast images) compared to pre-contrast MRI. The SOR included various prospectively defined procedures, for example, histopathology of malignant lesions and specific imaging procedures for benign lesions.
After enrolment, patients underwent both the pre-defined SOR procedure and the liver MRI, which included the unenhanced MRI followed by the 0.025 mmol/kg Primovist-enhanced MRI with dynamic phase and hepatocyte phase (20 minutes after injection) imaging. For each study, three blinded radiologists evaluated the MRI images comparing pre-contrast images to combined pre- and post-contrast images obtained after Primovist use.
In all four studies, Primovist (combined pre- and post-contrast image set) led to a significant improvement in diagnostic efficacy compared to unenhanced MRI. Results from both detection studies demonstrated that the proportion of correctly detected lesions compared to the SOR were significantly higher for all three blinded radiologists (p<0.05) after the use of Primovist as compared to the proportion of correctly detected images without Primovist contrast. Results from Study 012387 demonstrated a statistically significant difference (SSD) for radiologists 1 and 2 (p<0.05), but not for reader 3 with the use of Primovist in the ability to correctly characterize lesions. Similar results were generated for Study 014763, where two of the three radiologists exhibited an SSD in lesion characterization with the use of Primovist.
Primovist [181.43 mg/mL (0.25 mmol/mL), gadoxetate disodium] is presented as a solution. It is administered by IV injection for MRI imaging, specifically for detection and characterization of lesions in the liver. The recommended dose of Primovist is 0.1 mL/kg body weight (equivalent to 0.025 mmol/kg body weight). After the injection, the patient should be kept under observation for at least 30 minutes as delayed reactions may occur, as seen with other gadolinium-based contrast agents. Dosing guidelines are available in the Product Monograph.
Primovist is contraindicated for patients who are hypersensitive to this drug or to any ingredient in the formulation or component of the container. Primovist 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 Primovist are described in the Product Monograph.
Based on the Health Canada review of data on quality, safety, and efficacy, Health Canada considers that the benefit/risk profile of Primovist is favourable for the intravenous use in T1-weighted MRI of the liver to detect and characterize lesions in adults with known or suspected focal liver disease.
3 Scientific and Regulatory Basis for Decision
3.1 Quality Basis for Decision
3.1.1 Drug Substance (Medicinal Ingredient)
General Information
Gadoxetate disodium, the medicinal ingredient of Primovist, is a contrast enhancement agent for MRI. It is taken up selectively by liver cells (hepatocytes) where it accumulates and promotes enhanced visualization of liver tissue.
Manufacturing Process and Process Controls
The drug substance, gadoxetate disodium, is formed by reaction of gadolinium oxide with ethoxybenzyl diethylenetriamine pentaacetic acid (EOB-DPTA) and sodium hydroxide. To ensure that no ionic gadolinium (Gd) occurs in the finished product, the product is formulated to contain an excess of complexing agent, as calcium trisodium salt (caloxetate trisodium).
The specifications for the raw materials used in manufacturing the drug substance are considered satisfactory. In-process controls are performed throughout the in-situ preparation of the drug substance during the manufacture of the drug product and are considered adequate.
Characterization
Detailed characterization studies were performed to elucidate the structure of gadoxetate disodium. Gadoxetate disodium is not isolated during the manufacture of Primovist.
Potential impurities in gadolinium oxide are other metals, especially other rare earth metals. The potential impurities aluminum, arsenic, and lead are controlled in the release test specification for gadolinium oxide. Related substances of EOB-DTPA are controlled in the specifications for the complexing agent, which also include limits on ionic substances which could compete with gadolinium during the complexing reaction, and a test for enantiomeric purity.
Control of Drug Substance
Gadoxetate disodium is not isolated during the manufacture of Primovist. Quality of the drug substance is ensured by adequate specifications for the starting materials (gadolinium oxide and EOB-DTPA), and by controls during the manufacturing process for Primovist.
Stability
No degradation of gadolinium oxide is to be expected when stored in closed containers at temperatures <100°C. Therefore, it was decided that formal stability studies on gadolinium oxide would not be conducted.
EOB-DTPA has been shown to be stable for 60 months under International Conference on Harmonisation (ICH) long-term and intermediate storage conditions.
The stability of gadoxetate disodium is established in the drug product.
3.1.2 Drug Product
Description and Composition
Primovist is supplied as a sterile, non-pyrogenic, clear, colourless to pale yellow, aqueous solution containing 181.43 mg/mL (0.25 mmol/mL) of gadoxetate disodium, caloxetate trisodium, trometamol, and water for injection. Sodium hydroxide and/or hydrochloric acid are added to adjust the pH. No preservative is added.
Primovist is supplied in 6-mL single-use vials containing 5 mL of solution, and in 10-mL single-use vials containing 7.5 mL and 10 mL of solution. The vials are made of colourless glass Type I [conforms to European Pharmacopoeia (Ph.Eur.)/United States Pharmacopeia (USP)/Japanese Pharmacopeia (JP)] and are fitted with black stoppers and bordered caps with protective disks. The materials are Type I chlorinated butyl rubber (Ph.Eur.) for the stoppers, lacquered aluminum for the bordered caps, and polypropylene for the cover disks.
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 gadoxetate disodium with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.
Pharmaceutical Development
Changes to the manufacturing process and the formulation made throughout the pharmaceutical development are considered acceptable upon review. Parameters relevant to the performance of the drug product were not affected by the changes described.
Manufacturing Process and Process Controls
The manufacturing process for Primovist allows formation of the drug substance
Gd-EOB-DTPA in the course of the preparation of the drug product by complexing
EOB-DTPA with gadolinium oxide. Due to the involved heating/cooling steps as well as the required reaction time for formation of Gd-EOB-DTPA the total manufacturing procedure for Primovist takes three days. The final bulk solution is filtered and filled into vials, which are then capped and sealed, and subjected to terminal steam sterilization.
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
Primovist is tested to verify that its identity, appearance, assay, pH, viscosity, particulates, sterility, filling weight, osmolality, colour, levels of degradation products, drug-related impurities, foreign particulate matter, bacterial endotoxins, and microbiological impurities are within acceptance criteria. The test specifications and analytical methods are considered acceptable; the shelf-life and the release limits, for individual and total degradation products, are within acceptable limits.
Validation results of the analytical method used for the determination of gadoxetate disodium and the drug-related impurities are considered acceptable.
Stability
Based on the long-term, and accelerated stability data submitted, the proposed 60-month shelf-life at 15 to 30°C for Primovist is considered acceptable. Primovist is chemically and physically stable; however, it should be used immediately after opening.
3.1.3 Facilities and Equipment
The design, operations, and controls of the facility and equipment that are involved in the production of Primovist are considered suitable for the activities and products manufactured.
The proposed manufacturing site complies with the requirements of Division 2 of the Food and Drug Regulations and is compliant with Good Manufacturing Practices.
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 Primovist has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes.
3.2 Non-Clinical Basis for Decision
3.2.1 Pharmacodynamics
Primary Pharmacodynamics
Gadoxetate disodium is a paramagnetic compound that develops a magnetic moment when placed in a magnetic field. The relatively large magnetic moment produced results in a local magnetic field, yielding enhanced relaxation rates (shortening of relaxation times) of water protons in the vicinity of the paramagnetic agent. This leads to an increase in signal intensity (brightening) of blood and tissue.
In MRI, visualization of normal and pathological tissue depends in part on variations in the radiofrequency signal intensity that occur with differences in proton density; differences of the spin-lattice or longitudinal relaxation time (T1); and differences in the spin-spin or transverse relaxation time (T2). When placed in a magnetic field, gadoxetate disodium decreases the T1- and T2-relaxation time in target tissue. At the recommended dose, the effect is observed with greatest sensitivity in T1-weighted magnetic resonance (MR) sequences.
In non-clinical pharmacodynamic (PD) studies, the relaxation times of protons in water molecules were used to assess the paramagnetic effect of gadoxetate, in water, plasma, bile, and liver tissue. The T1- and T2-relaxation times in water and plasma (bovine) were measured in samples containing 0, 0.25, 0.50, and 1.0 mmol/mL of gadoxetate as its disodium salt or a reference. The T1-relaxation times in liver were determined following a single IV injection of 0.25 mmol/kg gadoxetate as its dimeglumine salt in rats. Some animals were sacrificed without receiving an injection. All other animals were sacrificed at 5, 10, 20, 30, or 60 minutes after administration. The livers were excised and divided into three parts. The T1-relaxation time and the gadolinium concentration were determined in each fraction of the livers immediately after excision. The T1-relaxation times in bile were determined after single IV injection of 0.5 mmol/kg gadoxetate as its dimeglumine salt in rats. The bile was collected in fractions while the animals were under anaesthesia. The T1-relaxation time and the gadolinium concentration were determined in the bile fractions.
Both relaxation times (T1 and T2) were markedly reduced in all the studies, demonstrating the pronounced paramagnetic effect of gadoxetate. A further increase in T1 relaxivity (the slope of the linear plot of relaxation rate in sec -1 versus the concentration of contrast agent in mmol/L) was observed in liver tissue as gadoxetate disodium is selectively taken up by hepatocytes. As liver lesions have no or minimal hepatocyte function (cysts, metastases, and the majority of hepatocellular carcinomas), the concentration of Primovist is much lower than in the surrounding tissue, resulting in an increased contrast image of normal liver tissue versus liver lesions lacking normal hepatocyte function in MRI.
In a separate study, the relaxivity of gadoxetate disodium was measured in a 2.0-Tesla field. The contrast agent decreased the relaxation time of protons of water and increased the relaxivity. As a reference, the relaxivities of Magnevist® (gadopentetate dimeglumine) in water and plasma were determined using the same procedure. Gadoxetate disodium decreased both T1- and T2-relaxation times in water and plasma. The relaxivities of gadoxetate disodium were higher than those of Magnevist® in water and plasma.
The efficacy of gadoxetate disodium to enhance the liver was studied in female rats by means of MRI images taken using a 2.0-Tesla animal imager. Standard imaging sequences (T1-weighted spin-echo sequences) were used. Even after the lowest dose of 10 µmol gadolinium (Gd)/kg, there was a marked increase in contrast between the tumour and the liver parenchyma. The contrast increased further as the dose was increased and only after the highest dose of 60 µmol Gd/kg was there no additional increase in contrast.
A pharmacological characterization study was conducted to characterize the individual gadoxetate disodium enantiomers. There was no statistically significant difference between either enantiomer in binding to human plasma. In rats, the median lethal dose (LD50) was estimated to be between 7.5 and 10.0 mmol/kg for both enantiomers. The relaxivity of both enantiomers was not seen to differ.
Secondary Pharmacodynamics
Pharmacological parameters such as protein binding, influence on the morphology of erythrocytes, enzyme inhibition, haemodialysis and complement activation were studied in in-vitro tests.
The protein binding of gadoxetate was found to be exceedingly low and vary between 7.7 and 9.1%. A study was conducted to evaluate if the low protein binding of gadoxetate disodium would interfere with hemodialysis. Gadoxetate disodium is quickly and completely removable from plasma by dialysis through a commercial hemodialysis device. In a separate study, it was determined that there was no difference between the effects of gadoxetate disodium and Magnevist® in terms of their influence on the morphology of erythrocytes.
Safety Pharmacology
Safety pharmacological investigations studied the potential effects of gadoxetate disodium on several organ functions such as blood coagulation (rat), kidney (rat, rabbit), cardiovascular (in-vitro and in-vivo, dog), central nervous system (CNS) (mouse, rat) and respiratory (rabbit).
Gadoxetate disodium did not exhibit any major effect on renal function; however, the single-dose study which examined renal function had no control group making the results difficult to interpret. Urine flow was increased at 2 hours and returned to baseline at 6 hours (2.7 to 6.9 mL/h). Proteinuria was increased with four of five rabbits having increased urine protein content (30 mg/dL). An increase in creatinine clearance was observed at 2 hours which returned to baseline at 6 hours (4.92 mL/min to 17.14 mL/min). As well, lactate dehydrogenase (LDH) was increased in urine from 2 to 48 hours. Gadolinium is taken up by bone tissue, as seen by the low concentrations found in bone at Day 7 of the study.
Gait disturbances were noted in mice at 800 mg/kg 30 minutes post dose. These effects were not observed in the control group.
There was a dose-dependent increase in action potential duration, and a significant change in membrane potential (p = 0.015) of the action potential at 30% repolarization (ADP30) at a dose of 10 mmol/L and at 1 Hz stimulation in isolated guinea pig papillary muscle. This concentration is about 38-times higher than the clinical exposure (0.26 mmol/L at 2 minutes).
An increase in QTcF and QTcQ was seen with the use of gadoxetate disodium at 0.1 and 0.5 mmol/kg; however, the study only used four animals. Nevertheless, the effect of gadoxetate disodium on the prolongation of the QTc interval cannot be excluded by this study. On further investigation and consultation with a clinical expert, it was concluded that Primovist has a minimal effect on the QTc interval.
An effect of drug interaction on the MRI enhancement properties of gadoxetate disodium was investigated in rats. The results demonstrated that compounds belonging to the class of rifamycins block the hepatic uptake of gadoxetate disodium, thus reducing the hepatic contrast effect. In this case, the expected benefit of an injection of Primovist may be limited. Interactions with other medications are not known.
3.2.2 Pharmacokinetics
Biodistribution studies conducted in rats using radiolabelled gadoxetate disodium demonstrated that the highest levels of gadoxetate were found in the liver, kidney, and bowel (due to biliary excretion). Gadoxetate was still detectable in the femoral bone and testis 72 hours post-administration.
In rats, the maximum concentration of radioactivity in the foetus and amniotic fluid were below one one-hundredth of that in maternal plasma, and radioactivity disappeared completely by 24 h. There was evidence that gadoxetate disodium is transferred via milk. The amount transferred to a neonate via milk was below 0.5% of the dose administered to the dam.
Plasma binding in both rat and dog was observed to be ~10%. The data presented suggests that the study agent is dialyzable.
There was no incidence of biotransformation of gadoxetate disodium as examined by mass spectroscopy, with high-performance liquid chromatography (HPLC), or inductively coupled plasma atomic emission spectroscopy (ICP-AES).
Both dog and rat studies showed non-linear and dose-dependent excretion by renal and hepatic routes. The renal excretion was linear and only limited by the glomerular filtration rate (GFR). The biliary excretion route (uptake by hepatocytes, excretion into bile, then to faeces) was the rate-limiting step. In dog studies, recovery of gadoxetate disodium was approximately 97% after 7 days.
Rat studies showed that the hepatic elimination was blocked with sulfobromophthalein, suggesting that the organic ion transport mechanism is involved. The enterohepatic circulation in rats was reported to be 3.89% (± 2.12%) of the injected dose.
Pharmacokinetic (PK) data derived from the pharmacologic characterization study indicated that the enantiomers were bioequivalent with respect to the amount eliminated in faeces. Complete bioequivalence was not seen in area under the concentration time curve (AUC), renal clearance, and amounts recovered in urine; these were not statistically bioequivalent. This could be due to the higher renal excretion rate of the R-enantiomer; however, this is not likely of clinical relevance.
3.2.3 Toxicology
Toxicity studies were conducted with gadoxetate disodium supplied in 0.25 mmol/mL and 0.5 mmol/mL formulations.
Single-dose Toxicity
Single-dose toxicity studies were conducted in mice, rats, and dogs. The results indicate that following IV administration, gadoxetate disodium was tolerated without lethality at doses up to 7.5 mmol/kg (mice), 10.0 mmol/kg (rats), 5.0 mmol/kg (weaned rats), and 3.0 mmol/kg (dogs). Higher doses were not tested in dogs as the dose of 3.0 mmol/kg already exceeded the envisaged diagnostic dose in humans by approximately 120-fold on the basis of body weight. Mortality was observed at doses of 10.0 mmol/kg (mice), 12.5 mmol/kg (rats), and 7.5 mmol/kg (weaned rats).
Repeat-dose Toxicity
In 4-week, repeated IV dose toxicity studies conducted with rats and dogs, relatively minor clinical signs and effects on body weight, food consumption, and clinical pathology were observed mainly with mid-doses (0.3 mmol/kg) and high-doses (1.0 mmol/kg) in dogs; however, the most significant finding was dose-dependent reversible renal tubular vacuolation in mid- (0.6 mmol/kg) and high-dose (2.0 mmol/kg) rats and high-dose dogs. This reversible vacuolation has also been observed with other gadolinium-based contrast agents (GBCAs).
The no observable effect level (NOEL) in rats (0.2 mmol/kg) and dogs (0.1 mmol/kg) to human (0.025 mmol/kg) dose ratios were 8-fold based on body weight and 1.3-fold based on body surface area in rats and 4-fold based on body weight and 2.2-fold based on body surface area in dogs.
The margins of safety are considered acceptable as Primovist is to be used as a single-dose agent.
Mutagenicity
Gadoxetate disodium was not mutagenic or clastogenic in a battery of in vitro and in vivo tests that exceed current ICH guideline recommendations.
Reproductive and Developmental Toxicity
The reproductive and developmental studies were consistent with current ICH guidelines. Gadoxetate disodium was not teratogenic in rats, at multiples of the 0.025 mmol/kg human dose up to 200-fold based on body weight and 32-fold based on body surface area. Further testing in rabbits also demonstrated that gadoxetate disodium was not teratogenic; however, it did cause embryo-foetal toxicity at multiples of the human dose of 80-fold based on body weight and 26-fold based on body surface area, with the NOEL at 20-fold based on body weight and 6.5-fold based on body surface area.
No effects on fertility were observed in rats at doses up to 40-times the human dose based on body weight or 6.5-fold based on surface area. In a peri- and post-natal development study, maternal toxicity was observed, but no developmental effects on their progeny were seen at doses up to 48-times the human dose based on body weight or 7.8-fold based on surface area.
Local Tolerance
Evaluation of local tolerance revealed that the 0.25 mmol/mL formulation was generally well tolerated when administered to rabbits and/or rats by the IV, paravenous, and intra-arterial routes. This was in contrast to the 0.5 mmol/mL formulation which caused reactions (slight to moderate reddening of the skin with vessel injection) in the congested vein. No intolerance reactions were seen in the uncongested vein. Intramuscular (IM) injection of either formulation caused injection site reactions. Direct injection of the 0.5 mmol/mL formulation into the liver, mammary gland, and prostate gland did not cause any local reactions. Dermal application of gadoxetate disodium was well tolerated by rabbits as was 10 mg, but not 100 mg, applied to the conjuctival sac of rabbits.
The 0.25 mmol/mL formulation of gadoxetate disodium did not show antigenic potential in a mouse-rat test system and in guinea pigs or delayed hypersensitivity potential in guinea pigs.
3.2.4 Summary and Conclusion
The results of the non-clinical PD studies support the proposed indication for Primovist for the detection and characterization of lesions in the liver. Safety pharmacology studies suggest the potential for gadoxetate disodium to affect vital functions in humans is low.
The PK profile of gadoxetate disodium has been adequately characterized in the non-clinical studies. It is excreted primarily by renal and hepatic routes.
The toxicity profile of gadoxetate disodium has been characterized. The results of the toxicity studies support the intended single IV dose of a 0.25 mmol/mL formulation for diagnostic imaging purposes.
3.3 Clinical basis for decision
3.3.1 Pharmacodynamics
A study was conducted in healthy subjects to demonstrate the difference in signal intensity in the liver before and after four increasing doses of gadoxetate disodium. Doses of 0.01, 0.025 (the clinical dose), 0.05 and 0.10 mmol/kg were administered. The % MR signal enhancement relative to pre-dose baseline value was the PD measure. Safety and laboratory parameters were evaluated 4- and 24-hours post infusion.
Images were taken using a 1.5 Tesla MRI. An identical imaging protocol was used for each patient to visualize the liver and upper abdomen, with images taken up to 6 hours post-infusion. Some subjects in higher dose groups also had images taken at 24 and 48 hours. Two T1-weighted pulse sequences were used for imaging before and after contrast application: spin-echo (SE) and heavily T1-weighted gradient-echo (GRE) with 256 frequency encoding points for both sequences and defined time points.
Despite the fact that vital signs fluctuated in some subjects, these fluctuations were not considered to be clinically significant or affected by the study agent.
There were no clinically significant changes seen in haematololgy results, liver function tests or creatinine levels. Other chemistry results were deemed to be non-clinically significant.
The AUC and maximum signal intensity after 2 hours [relative signal intensity (RSI) max] were not significantly improved by the highest dose in this study (0.10 mmol/kg) but there were dose-response increases in signal with the first three doses. The study determined that the choice for further study rests between the 0.025 and 0.50 mmol dose.
3.3.2 Pharmacokinetics
After a bolus injection (0.01 - 0.10 mmol/kg), the rapid removal of gadoxetate disodium from the circulation was attributed to rapid renal elimination and uptake by the liver. The AUC(0-4 h) accounted for approximately 90% of the AUC(0-infinity).
Although the hepatic uptake and disposition of gadoxetate disodium is known to be an active carrier mediated process, the mean terminal half-life (range: 1.1 - 1.6 h), the total clearance (range: 224 - 272 mL/min), and dose-independent faecal and urinary excretion (50:50 proportion) over the dose range (0.01 - 0.10 mmol/kg body weight) indicated that at up to a 4-fold higher dose than the suggested clinical dose, the disposition processes are not saturated.
Linear pharmacokinetics were observed in doses between 25 and 100 µmol/kg). At higher doses (up to 20-times the clinical dose), PK parameters were not dose-proportional.
The study agent does not appear to be metabolized. Renal filtration and extra-hepatic clearance are the main pathways of elimination (50:50 proportion).
The extent of liver enhancement was decreased in patients with high bilirubin levels. Decreased liver enhancement was also seen in patients with end-stage renal disease (ESRD) due to high ferritin levels. PK analysis showed that patients with ESRD have marked higher values of certain parameters (AUC, terminal half-life, and total clearance) when compared to patients with moderate renal impairment.
3.3.3 Clinical Efficacy
The clinical efficacy of Primovist was assessed in four pivotal, open-label, multi-centre, Phase-III studies which included two lesion diagnostic studies (96129 and 97610) and two lesion characterization studies (012387 and 014763). A total of 797 adult patients with suspected or known focal liver lesions who were to undergo surgery enrolled in these studies. A total of 621 of these patients received a single IV dose of Primovist. No control groups were formed in any of the studies as each patient acted as their own control.
In all studies, patients underwent both a pre-defined SOR procedure and a liver MRI, which included an unenhanced MRI followed by the 0.025 mmol/kg Primovist-enhanced MRI with dynamic phase and hepatocyte phase (20 minutes after injection) imaging. For each study, three blinded radiologists evaluated MRI images comparing pre-contrast images to combined pre- and post-contrast images obtained after Primovist use.
Lesion Detection Studies
The primary efficacy variable for the detection studies was the sensitivity of liver lesion detection which took into account the possibility of multiple lesions within patients, as well as matching and tracking of lesions within patients compared to the SOR (a combination of pathology from resected liver specimens, an intra-operative ultrasounds and, if appropriate, a 3-month follow-up imaging examination).
For Study 96129, the intent to treat (ITT) population included 136 patients, while the per protocol (PP) population included 129. A total of 302 liver lesions were verified by SOR in total. When compared to those detected with the SOR, the proportion of correctly detected lesions after pre- and post-contrast images with use of Primovist was statistically significantly higher for all three blinded radiologists (p<0.05) as compared to the proportion of correctly detected lesions from images taken without Primovist contrast.
Study 97610 had an ITT population of 169 and a PP population of 131, with 316 lesions detected in 126 patients based on the SOR. Similar to Study 96129, the results of 97610 demonstrated statistical significance (p<0.05) for all three blinded radiologists for the proportion of correctly detected lesions with the use of Primovist (p<0.05).
Lesion Characterization Studies
The primary efficacy variable for the lesion characterization studies was the proportion of correctly characterized liver lesions (correct liver lesion type) with Primovist-enhanced MRI (combined pre- and post-contrast images) compared to pre-contrast MRI alone. The lesions were verified based on the SOR which included various prospectively designed procedures, for example, histopathology of malignant lesions and specific imaging procedures for benign lesions. The types of lesions characterized included metastatic lesions, hemangiomas, focal nodular hyperplasia, liver cysts, and hepatocellular carcinoma.
For Study 012387, there was a statistically significant difference for radiologists 1 and 2 (p<0.05), but not for reader 3 in their ability to correctly characterize lesions with the use of Primovist.
The results were similar for Study 014763 in that two out of three radiologists exhibited an SSD for correct lesion characterization with the use of Primovist.
3.3.4 Clinical Safety
Clinical safety was assessed in the four pivotal studies described in section 3.3.3 Clinical Efficacy.
Lesion Detection Studies
Nausea was the most frequently reported adverse (AE) observed for Study 96129, followed by dyspnoea, headache, and vasodilatation. Three serious adverse events (SAEs) occurred: peritonitis, dyspnoea, and a cardiovascular disorder. None of the SAEs were considered to be related to Primovist.
No major changes were noted in vital signs, including mean blood pressure. Fluctuations were observed in clinical laboratory parameters, many of which were expected in this patient population [for example (e.g.) altered liver function tests]. There was little to no indication that abnormal test results were related to Primovist.
No deaths occurred in either study.
Lesion Characterization Studies
The most common AEs for Study 012387 were headache and paresthesia with abdominal pain, tremor and dyspnoea. Parasthesia was classified as definitely related for one patient, while other AEs were possibly related (chills, asthenia, vomiting, headache, and pruritis). Dyspnoea and anxiety (in one patient), hemothorax, and pneumothorax were listed as SAEs; however, these were not considered to be related to Primovist. Fluctuations were observed for clinical laboratory parameters; however, there was no indication that these were related to Primovist. Some patients experienced changes in PQ, QRS, and QTc interval readings. The only clinical AE related to change in heart rhythm was in one patient who experienced an atrioventricular (AV) block.
In Study 014763, AEs that were considered possibly or probably related included taste perversion, headache, vasodilation, nausea, bundle branch block, rash, dizziness, diarrhoea, and dry mouth. One SAE was reported for an abscess; however, this was not considered to be related to Primovist.
Clinically significant changes in laboratory values were noted for Study 014763. It is not possible to conclude if these were related to Primovist. Some patients experienced increases in cardiac intervals, including QTc intervals.
Due to cardiac AEs that occurred in each of these studies, it is not possible to conclude that Primovist is without cardiac effect for this patient population.
QT Prolongation
Examination of the electrocardiograms (ECGs) during these studies did not show evidence of a systemic QTc prolongation with the use of Primovist.
In addition, the sponsor provided a synopsis of an expert opinion sought to examine the effect of Primovist on the QT interval. Upon review of this synopsis, Health Canada agreed with the conclusions of the expert that the torasdogenic potential of Primovist is low; however, non-clinical studies did demonstrate some potential for QT prolongation and individual patients in the clinical trials did have QT prolongation (without any adverse events). This information has been presented in the Product Monograph.
Laboratory Parameters
Laboratory analysis did not show any significant incidence of adverse changes. Bilirubin was increased for some patients, likely due to the fact that the agent is taken up into hepatocytes. The increased bilirubin level was reversible.
Renal Toxicity
Safety reports were reviewed regarding Nephrogenic Systemic Fibrosis (NSF), which is a serious effect seen with the use of GBCAs in patients with renal disease. NSF can result in death. This is felt to be a class effect, and all GBCAs approved by Health Canada have been labelled as such. To date, there have been no reports of NSF associated with the use of Primovist. This could be associated with the differing physiochemical properties of the Primovist molecule, which renders it more stable than some of the other GBCAs currently in use.
3.4 Benefit/Risk Assessment and Recommendation
3.4.1 Benefit/Risk Assessment
Clinical safety and efficacy studies conducted with Primovist revealed that it is efficacious and generally well tolerated. In addition, patients administered GBCAs have a reduced incidence of contrast media nephropathy, which is the third leading cause of hospital acquired acute renal failure, accounting for 12% of all cases. It has been documented that contrast media nephropathy is associated with an increased risk of morbidity and death. With the use of iodine-based contrast agents, patients with pre-existing renal disease and diabetes are at highest risk (if not given standard hydration protocol, the risk of renal failure is 12-26%). Lower-risk patients (without risk factors) have a 3.3% incidence of acquired renal failure.
Another advantage of Primovist is that like other GBCAs, its administration does not require arterial puncture, which carries its own set of complications. In addition, less exposure to ionizing radiation is required with the use of GBCAs. Compared to iodine-based contrast agents, there is a lower incidence of severe allergic and anaphylactic reactions with GBCAs, although these severe reactions can occur.
Primovist is excreted from the body via a dual renal and biliary pathway. If there is a decrease in one route of excretion, excretion by the other route is increased. Increased uptake by hepatocytes may be of benefit in more precisely defining hepatic pathology.
One risk of GBCAs is the occurrence of NSF which, although rare, can be fatal. In the past, NSF has been observed in patients with severe renal disease who have been exposed to GBCAs. Appropriate warnings have been included in the Product Monograph. To date, no occurrence of NSF has occurred with the use of Primovist. Theoretically, this could be due to differences in structure and stability as compared to other GBCAs.
Based on the evaluation of the submitted data, the benefit/risk profile for Primovist is favourable for the proposed indication.
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 Primovist is favourable for intravenous use in T1-weighted MRI of the liver to detect and characterize lesions in adults with known or suspected focal liver disease. 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: Primovist®
Submission Milestone | Date |
---|---|
Submission filed: | 2009-02-02 |
Screening | |
Screening Acceptance Letter issued: | 2009-03-20 |
Review | |
Quality Evaluation complete: | 2010-01-12 |
Clinical Evaluation complete: | 2010-01-11 |
Labelling Review complete: | 2010-01-13 |
Notice of Compliance (NOC) issued by Director General: | 2010-01-14 |