Summary Basis of Decision for Nplate ™

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
NplateTM

Romiplostim, 250 µg/0.5 mL and 500 µg/1.0 mL, Powder for solution, Subcutaneous

Amgen Canada Inc.

Submission control no: 117327

Date issued: 2009-07-21

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:

NplateTM

Manufacturer/sponsor:

Amgen Canada Inc.

Medicinal ingredient:

Romiplostim

International non-proprietary Name:

Romiplostim

Strength:

250 µg/0.5 mL and 500 µg/1.0 mL

Dosage form:

Powder for solution

Route of administration:

Subcutaneous

Drug identification number(DIN):

  • 02322854
  • 02322862

Therapeutic Classification:

Thrombopoiesis-stimulating protein

Non-medicinal ingredients:

L-histidine, mannitol, sucrose, polysorbate 20, hydrochloric acid, and sterile water for injection

Submission type and control no:

New Drug Submission, Control Number: 117327

Date of Submission:

2007-11-16

Date of authorization:

2009-02-19
2 Notice of decision

On February 19, 2009, Health Canada issued a Notice of Compliance to Amgen Canada Inc. for the drug product, Nplate™.

Nplate™ contains the medicinal ingredient romiplostim which is a thrombopoiesis-stimulating protein.

Nplate™ is indicated to increase the platelet levels in adult patients with chronic immune (idiopathic) thrombocytopaenic purpura (ITP):

  • who are non-splenectomised and have had an inadequate response or are intolerant to corticosteroids and/or immunoglobulins;
  • who are splenectomised and have had an inadequate response to splenectomy.

Nplate™ has been used alone or in combination with other ITP therapies such as corticosteroids, azathioprine, or danazol.

Patients with ITP have blood that does not clot properly due to a low number of blood cells called platelets. Nplate™ increases platelet production through binding and activation of the thrombopoietin receptor, a mechanism analogous to endogenous thrombopoietin (eTPO).

The market authorization was based on quality, non-clinical, and clinical information submitted. The results of two pivotal, Phase III, randomized, double-blind, placebo controlled studies were submitted to support the efficacy and safety of Nplate™ in the treatment of adult thrombocytopaenic patients with ITP. One study included patients who were refractory to splenectomy while the other included non-splenectomised patients refractory to or intolerant of conventional therapies. In both studies, patients entered the studies with platelet counts ≤30 × 109/L. The primary efficacy endpoint of the two pivotal studies was durable platelet response: at least 6 weekly platelet responses (platelet counts ≥50 × 109/L) during the last 8 weeks of treatment, in the absence of rescue medication at any time during the 24-week treatment period. A statistically significant effect was seen for Nplate™ relative to the placebo, regardless of whether or not the patients had undergone splenectomy. A total of 61.0% of Nplate™-treated patients who had undergone splenectomy achieved a durable platelet response, as did 38.1% of Nplate™-treated patients who were refractory to splenectomy. Patients in both pivotal studies were permitted rescue medication (consisting mainly of corticosteroids, immunoglobulins, and platelet transfusions) at any time during the study. An overall reduction of 38.0% in the proportion of patients receiving rescue medication was observed for the Nplate™-treated patients relative to the placebo in the combined analysis. Rescue medication was required by 17.1% of the non-splenectomised subjects and 26.2% of the splectomised subjects who received Nplate™.

Nplate™ (250 µg/0.5 mL and 500 µg/1.0 mL, romiplostim) is supplied as a powder for solution. It is administered as a weekly subcutaneous injection with dose adjustments based upon platelet count response. The recommended initial dose for Nplate™ is 1 µg/kg based on actual bodyweight. Platelet counts should be assessed weekly until a stable platelet count (≥50 × 109/L for at least 4 weeks without dose adjustment) has been achieved. Thereafter, platelet counts should be taken monthly. A maximum weekly dose of 10 µg/kg should not be exceeded. Dosing guidelines are available in the Product Monograph.

Nplate™ is contraindicated for patients who are hypersensitive to this drug or to any ingredient in the formulation or any component of the container, or with a known history of sensitivity or allergy to any E. coli-derived product. Nplate™ 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 Nplate™ are described in the Product Monograph.

Priority Review status was granted for the evaluation of Nplate™ as it appeared to provide promising evidence of clinical efficacy for a serious, life-threatening, and severely debilitating disease that is not adequately managed by a drug marketed in Canada.

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

3 Scientific and Regulatory Basis for Decision

A Notice of Deficiency (NOD) was issued on June 10th, 2008 for this New Drug Submission based on the identification of deficiencies revealed in the quality (chemistry and manufacturing) evaluation. Furthermore, it was requested that the sponsor adequately address various non-clinical and clinical issues as well as their labelling implications in the NOD response.

The NOD response was received on September 12, 2008. The response was found to be acceptable and a screening acceptance letter was sent on September 19, 2008. The submission entered the review stream with a review target of March 16, 2009.

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Romiplostim, the medicinal ingredient of Nplate™ is a recombinant non-glycosylated Fc (fragment, crystallisable) fusion protein (termed peptibody) produced in E. coli. It is comprised of a human immunoglobulin G1 (IgG1) Fc domain, with each single chain subunit covalently linked at the C-terminus to a peptide chain containing two thrombopoietin (TPO) receptor binding domains. Romiplostim stimulates platelet production by a mechanism similar to that of endogenous TPO (eTPO). The proposed indication for romiplostim is the treatment of thrombocytopaenia in adult patients with chronic immune (idiopathic) thrombocytopaenic purpura (ITP).

Manufacturing Process and Process Controls

The drug substance is generated by recombinant technology. Romiplostim is expressed in E. coli as inclusion bodies using fed-batch fermentation. The downstream process includes a solubilization step, refolding, and purification through a series of ultrafiltration/diafiltration and chromatography steps. Process validation data demonstrate that the manufacturing process operates in a consistent manner, yielding product of acceptable quality.

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

Characterization

Detailed characterization studies were performed to provide assurance that romiplostim consistently exhibits the desired characteristic structure and biological activity.

Appropriate tests are adequately controlling the levels of product- and process-related impurities.

Control of Drug Substance

The drug substance manufacturing process has been scaled-up and optimized during development. The process changes introduced at each generation of the process were adequately described and comparatively assessed. Lot release, characterization, and stability data have also been used to support the comparability assessments.

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

The specifications are considered acceptable for the drug substance. Data from the batch analyses were reviewed and are within the proposed acceptance criteria.

The drug substance packaging is considered acceptable.

Stability

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

3.1.2 Drug Product

Description and Composition

Nplate™ (romiplostim) is supplied as a sterile, preservative-free, lyophilized white powder in single-dose vials for reconstitution. Each Nplate™ vial contains L-histidine, mannitol, sucrose, polysorbate 20, and dilute hydrochloric acid for pH adjustment. Nplate™ (500 µg) is reconstituted with 1.2 mL of sterile Water for Injection (WFI), United States Pharmacopeia (USP) and Nplate™ (250 µg) is reconstituted with 0.72 mL of Sterile WFI, USP. Reconstitution yields a clear, colourless, iso-osmotic solution of Nplate™ for subcutaneous (SC) injection. The container closure system consists of a Type I glass vial, a rubber stopper, and an aluminium seal with a plastic flip-off cap.

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 romiplostim with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

The proposed excipients used in the formulation of Nplate™ drug product are common for injectable preparations.

Pharmaceutical Development

Manufacturing changes implemented throughout development involved modifications in presentation, process scale, and changes in the site of manufacture. The various changes were adequately described and comparatively assessed. Assessment of drug product comparability included lot release testing, biochemical, biological, biophysical, and structural comparisons as well as stability testing.

Changes to the manufacturing process made throughout the pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

The drug product manufacturing process consists of thawing the drug substance, followed by formulation, aseptic filtration, filling of the vials, lyophilization and capping using conventional pharmaceutical equipment and facilities. Process validation data demonstrate that the manufacturing process operates in a consistent manner, yielding product of acceptable quality.

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 Pharmacopoeia (Ph. Eur.) standards.

Control of Drug Product

Nplate™ is tested to verify that its appearance, identity, purity, sterility, pH, moisture content, protein content, osmolality, and level of bacterial endotoxins are all within acceptance criteria.

Concerns regarding the system suitability/assay acceptance criteria of one of the Nplate™ release/stability assays resulted in the issuance of a NOD. In the sponsor's response to the NOD, all of the quality concerns that led to the NOD were satisfactorily resolved.

The test specifications are considered acceptable to control the drug product, and the impurity limits were set according to International Conference on Harmonisation (ICH) recommendations.

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed 36-month shelf-life at 2-8°C for Nplate™ is considered acceptable.

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

3.1.3 Facilities and Equipment

An on-site evaluation (OSE) of the facilities involved in the manufacture and testing of romiplostim drug substance has been successfully conducted by the Biologics and Genetic Therapies Directorate, Health Canada. The OSE and the review of the responses to the Exit Notice observations were found to be satisfactory.

An OSE of the facilities involved in the manufacture and testing of Nplate™ drug product was not warranted since the facility was recently evaluated for another product produced at this site. Given the history of this site, the OSE was waived for this submission.

The design, operations and controls of the facility and equipment that are involved in the production of Nplate™ are considered suitable for the activities and products manufactured.

3.1.4 Adventitious Agents Safety Evaluation

An assessment was performed on all materials used in the commercial manufacturing process to determine the risk associated with any material of animal origin. In addition, animal-derived materials not directly used in the process, but which may come into contact with the product during manufacture or in packaging, have also been subjected to risk assessment. This assessment concluded that the associated transmissible spongiform encephalopathy (TSE) risk from this product is negligible, since the raw materials used in this product are from tissues with no detected infectivity per the European Medicines Agency (EMEA) Committee for Medicinal Products for Human use (CHMP) guideline, "Note for Guidance on Minimizing the Risk of Transmitting Animal Spongiform Encephalopathy Agents via Medicinal Products".

There are no human-derived materials utilized in the production of romiplostim.

3.1.5 Conclusion

All issues identified during the review have been adequately addressed and resolved.

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

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

Pharmacodynamic (PD) studies were conducted with romiplostim utilizing doses that permitted the evaluation of a full range of dose-response activity. As there are currently no available animal models which are entirely analogous to ITP in humans, the clinical dose could not be selected directly from the animal pharmacology data.

Romiplostim has been evaluated in vitro and/or in vivo in several animal models including mice, rats, rabbits, and non-human primates (rhesus monkeys, cynomolgus monkeys, and baboons).

Romiplostim is a thrombopoiesis stimulating peptibody that increases platelet production through the activation of the TPO receptor (c-Mpl). Romiplostim mimics the activity of eTPO by binding to c-Mpl and transducing a similar cascade of intracellular events, thereby triggering the same biological outcome.

In primary PD studies, the binding ability of romiplostim was evaluated using Biacore, a technology used for studying biomolecular interactions. Results demonstrated that the dissociation constant (KD) of TPO for human-c-Mpl was 33 nM, whereas the KD of romiplostim for human-c-Mpl fell in the range of 14 to 51 nM.

In vitro studies demonstrated that romiplostim effectively stimulates megakaryocyte colony-forming cell proliferation from cynomolgus monkey, baboon, and human haematopoietic progenitor cells. Secondly, romiplostim binds to rat, cynomolgus monkey, and human platelets. Binding to human and monkey platelets was adequate and fully displaceable. For monkeys and humans, respectively, there was an 11-fold and 17-fold lower binding affinity when compared TPO. Furthermore, a study evaluating human platelet function in vitro found that at concentrations of 10 or 100 ng/mL romiplostim increased the adenosine diphosphate-generated aggregation response of platelets in a similar manner to the positive control megakaryocyte growth and development factor (MGDF), a c-Mpl ligand. The data suggest that the effects of romiplostim to stimulate platelet production are mediated via c-Mpl.

Romiplostim was screened in vitro for receptor binding affinity to sixty-three different receptors, enzymes, or ion channel targets. Results indicated that it is unlikely that romiplostim would cause any substantive pharmacologic activity in vivo other than the desired pharmacodynamics.

When tested in vivo, the administration of romiplostim caused increases in platelet counts in mice, rats, and non-human primates. These increases were dose-dependent, but dosing-route independent. The response in non-human primates was less pronounced than that seen in rodents, requiring higher doses to elicit a similar response.

Treatment with romiplostim in a mouse model of idiopathic ITP was effective in pre-splenectomy animals, following splenectomy (in animals that responded to splenectomy), in non-responders, and in responding animals who later relapsed, hence supporting its development for the use of romiplostim to treat patients with ITP.

3.2.2 Pharmacokinetics

Enzyme-linked immunosorbent assay (ELISA) assays were developed and validated for measuring romiplostim in the serum of rats, rabbits, rhesus monkeys, and cynomolgus monkeys. All relevant assays were shown to be sensitive, specific, reproducible and accurate enough for the assessment of pharmacokinetic (PK) and PD studies, and to support the toxicology studies of romiplostim in the relevant species.

Romiplostim was administered as a single intravenous (IV) administration to normal mice [30, 100 and 300 µg/kg bodyweight (bw)], neonatal Fc receptor (Fc-Rn)-deficient mice (100 and 1000 µg/kg bw) and thrombocytopaenic mice (3 and 30 µg/kg bw). In normal mice, the PKs of romiplostim were non-linear. Clearance and the volume of distribution at steady-state decreased with increasing dose with serum half-life values ranging from 6.1 hours (low dose) to 15.0 hours (high dose). Platelet response was dose-dependent and maximum platelet counts correlated with overall exposure to romiplostim. In Fc-Rn deficient mice, romiplostim was eliminated much faster than in normal mice, indicating that the platelet response was more pronounced in normal mice. This data suggest that the Fc-Rn receptor acts as a salvage receptor for romiplostim by maintaining romiplostim serum levels. In thrombocytopaenic mice, the effect was dose-dependent.  For example, at 3 μg/kg bw, thrombocytopaenic mice had a higher exposure than normal mice, while at 30 µg/kg bw, similar exposures were observed in normal and thrombocytopaenic mice suggesting that platelets play a role in the elimination of romiplostim, through a saturable process of target-mediated elimination.

Romiplostim was also administered as a single IV administration to normal rats, splenectomised rats, or bilaterally nephrectomised rats, at dose levels of 30, 100, 300 and/or 1000 µg/kg bw. The PK and PD profiles of romiplostim in control, sham-operated and splenectomised rats were similar, indicating that the spleen is not involved in the removal of romiplostim. In nephrectomised rats, exposure to romiplostim was significantly higher when compared to normal or sham-operated animals. In addition, the estimated apparent half-lives were significantly longer in nephrectomised animals than in sham-operated animals, suggesting that the kidney plays a significant role in the clearance of romiplostim during the first 12 hours.

Single dose IV and SC PK studies were conducted in rats, cynomolgus monkeys, and rhesus monkeys over a dose range of 30 to 300 µg/kg bw in rats and 500 to 5000 µg/kg bw in monkeys. After a single SC dose, bioavailability of romiplostim was approximately 21 to 28% in rats across the 30 to 300 µg/kg bw dose range and 19% in cynomolgus monkeys at 5000 µg/kg bw. For rhesus monkeys, bioavailability decreased from 74% at 500 µg/kg bw to 45% at 5000 µg/kg bw. Maximum concentration was achieved at a median time of peak concentration (tmax) of 8 to16 hours and 4 to 8 hours post-dose in rats and monkeys, respectively. The apparent terminal elimination t½ was 19 to 21 hours in rats. In monkeys, the terminal elimination t½ was 296 hours and 110 to 195 hours, for cynomolgus and rhesus monkeys, respectively, representing a slower absorption process into the systemic circulation. The single IV dose PKs were dose proportional for all three species. The estimated systemic clearance was approximately 6.3 to 8.4, 17.3 to 20.1 and 7.4 to 7.8 mL/hr/kg bw in rats, cynomolgus monkeys, and rhesus monkeys, respectively. The terminal elimination t½ was approximately 17 to 19 hours in rats, 68 to 96 hours in cynomolgus monkeys, and 102 to 143 hours in rhesus monkeys.

Tissue distribution studies revealed that romiplostim distributed into all tissues. The highest concentrations of radioactivity were observed in the thyroid, blood components, kidneys, bone marrow, and liver at 0.5 hours post-dose. The highest percent values were seen in the blood, liver, muscle, skin and bone. Concentrations of radioactivity declined slowly and were detectable in all tissues at all collection time points (up to 168 hours). Radioactivity was detected in the brain at low levels, suggesting that the drug-derived radioactivity crossed the blood/brain barrier. Most of the radioactivity associated with serum, spleen, kidneys, heart, ovaries and liver was trichloacetic acid (TCA)-precipitable at all time points. Serum contained over 91% TCA-precipitable radioactivity throughout the study, and analyzed tissues exceeded 83% at all time points. It was determined that urine was the primary route of elimination with 87.7% of the administered dose being excreted in the urine and 6.56% excreted in faeces. However, only 10.7 to 11.3% of the radioactivity in urine and 45.9 to 49.2% of the radioactivity in faeces was TCA-precipitable.

Multiple dose SC PKs were evaluated in rats, cynomolgus monkeys, and rhesus monkeys. The test material was administered over a dose range of 10 to 100 µg/kg bw in rats, 500 to 5000 µg/kg bw in cynomolgus monkeys, and 100 to 5000 µg/kg bw in rhesus monkeys.

For rats, there was no appreciable sex difference in concentration profiles, and the sex-combined data showed no apparent accumulation over a 4-week dosing period. Exposure increased about 8- to 11-fold over the range of 30-100 µg/kg bw. Most of the serum samples in the 10 µg/kg bw group fell below the assay quantification limit. The incidence of anti-romiplostim binding and neutralizing antibodies was substantial, however, anti-TPO antibodies were not observed. When pregnant rats were dosed with romiplostim from gestational days 7 to 19 over the dose range of 10 to 100 µg/kg bw, there was no accumulation of romiplostim. The test material readily transferred across the placenta to the foetus, possibly mediated by the Fc-Rn-receptors. The average foetal serum concentration was approximately one-half of the average maternal serum concentration. A greater than dose proportional increase in area under the concentration-time curve (AUC) and the observed maximum serum concentration (Cmax) was observed in the maternal and foetal serum. The romiplostim concentrations in the amniotic fluid were more variable relative to serum. The exposure in amniotic fluid only represented a fraction of the maternal serum exposure, that is to say (i.e.), 44% at 10 µg/kg bw, 17% at 30 µg/kg bw and 11% at 100 µg/kg bw. Exposure in pregnant rats appeared to be lower than in non-pregnant rats.

For monkeys, there were no appreciable sex differences in exposure. Exposure over a one-month dosing period increased less than dose proportionally for rhesus monkeys and increased approximately dose proportionally in cynomolgus monkeys. Exposure in rhesus monkeys was approximately twice as high as that observed in cynomolgus monkeys. The estimated accumulation ratios (ARs) were similar in rhesus monkeys and cynomolgus monkeys which ranged between 1.40 and 2.11 for the dose range of 300-5000 µg/kg bw. At 100 µg/kg bw, the exposure decreased following one month of dosing (AR = 0.437) which was most likely related to the elevated platelet counts. This hypothesis is supported by the finding that platelets play a role in the elimination of romiplostim and this role decreased with increasing dose. Neutralizing antibodies to romiplostim were not observed although binding antibodies occurred in both cynomolgus and rhesus monkeys. The presence of binding antibodies had no influence on the kinetics in cynomolgus monkeys, whereas the influence of binding antibodies could not be assessed in rhesus monkeys as all rhesus monkeys developed antibodies only after the treatment phase was completed.

Exposure of cynomolgus monkeys over a 3- or 6-month period was approximately dose proportional with no appreciable sex differences. The AR over the dosing period was approximately 1.05-1.65. The observed maximum serum concentrations occurred at 4 hours. The incidence of both binding and neutralizing antibodies to romiplostim were observed. Binding antibodies did not have an apparent influence on exposure. One monkey was found to have neutralizing antibodies on week 4 and had a reduced exposure during Week 13. No anti-TPO antibodies were observed.

3.2.3 Toxicology

All definitive toxicology studies utilized the SC route of administration as this is the intended clinical route of administration. Doses chosen were those that would provide a substantial challenge to the test animals and elicit systemic effects.

Single-Dose Toxicity

The toxicity of romiplostim was evaluated when administered as a single-dose via SC injection in Sprague-Dawley rats. The acute non-lethal dose of romiplostim was determined to be >1000 µg/kg bw (the highest dose tested), which represents a dose 33-fold greater than the highest anticipated clinical dose. All animals survived the duration of the study period. Treatment-related findings were considered non-adverse and reflected the pharmacological activity of the test material, i.e., increased platelet count, increased spleen weight, and increased splenic extramedullary haematopoiesis.

Cardiovascular

A single-dose SC cardiovascular safety study in cynomolgus monkeys at dose levels of 500, 1000 or 5000 µg/kg bw did not reveal any treatment-related effects on blood pressure, heart rate or body temperature. All electrocardiograms (ECGs) were qualitatively and quantitatively within normal limits. There were no alterations in morphology, rhythm or ECG intervals attributable to romiplostim.

Central Nervous System

A single-dose SC central nervous system (CNS) safety study in rats at dose levels of 10, 30 or 100 µg/kg bw did not reveal any treatment-related effects on the functional observational battery, body temperature, or motor activity.

Repeat-Dose Toxicity

Sub-chronic testing was conducted in rats, rhesus monkeys, and cynomolgus monkeys. Doses chosen provided continuous serum concentrations that were sufficiently higher than human concentrations, thus allowing for a meaningful evaluation of safety.

Sub-chronic testing in Sprague-Dawley rats was carried out for 1 month at dose levels of 0, 10, 30 or 100 µg/kg bw. Treatment-related mortality was evident at all dose levels tested. Although the specific cause of death could not definitively be determined, it was considered to most likely be the result of megakaryocytic thrombosis and increased blood viscosity related to the massive increases in platelet counts. Additional treatment-related findings (observed at all dose levels) were primarily related to the pharmacological activity of romiplostim and included: increased platelet count, increased spleen weights with correlating histopathological findings in the spleen (megakaryocyte hyperplasia, lymphocytic depletion), liver (megakaryocytosis, extramedullary haematopoiesis), lungs (megakaryocytosis) and bone marrow (megakaryocyte hyperplasia, myelofibrosis and hyperostosis). It was also noted that there was a generalized stimulatory effect on leukocyte production and a slight decrease in red blood cell count, haematocrit and haemoglobin. However, the values for the affected parameters still fell within the normal range. Platelet aggregation was slightly reduced for females at all dose levels, and for males at dose levels of 30 µg/kg bw and higher. In addition, there was a dose-related increase in the incidence of dermal, muscle, and subcutis chronic inflammation at the injection site. This was considered to be the result of slight trauma associated with the injection procedure and/or minor local irritation. All treatment-related findings were reversed following a 1-month recovery period. A high incidence of anti-romiplostim neutralizing antibodies was observed at doses ≥30 µg/kg bw. Consequently, it was deemed more appropriate to conduct longer-term studies in non-human primates as neutralizing antibodies rarely develop in these species. It is important to note that thrombocytopaenia was not observed in antibody-positive (Ab+) rats, indicating that antibody to romiplostim did not cross-react with eTPO.

Myelofibrosis was also observed in the 1-month rat study and was dose dependent both in terms of incidence and severity. However, the fibrosis resolved after cessation of drug administration and there were no clinical sequelae of this finding for the period observed.

Sub-chronic testing in cynomolgus and rhesus monkeys was carried out for 1 month as well as for 3 and 6 months in cynomolgus monkeys. The effective doses required in non-human primates to elicit a similar response to romiplostim as that seen in humans and rats were substantially higher. Hence, the dose levels chosen for these studies were 500, 1000 and 5000 µg/kg bw. Dose levels were generally well-tolerated and findings were consistent after 1, 3, or 6 months of study. Treatment-related findings were directly or indirectly related to the thrombopoietic activity of romiplostim. Stimulation of platelet production in the bone marrow was evident from microscopic observations of megakaryocyte hyperplasia and from the marked thrombocytosis that occurred at all dose levels. There was a dose-dependent decrease in red blood cell counts and related parameters, but the degree of anaemia was not clinically significant at any dose level. Increased spleen weight was noted at all dose levels and was of uncertain toxicological significance since there were no corresponding histopathological findings. Myelofibrosis was not observed in monkeys.

In addition, at the SC injection sites there was a dose-related increase in the incidence and severity of perivascular mononuclear cellular infiltrate. In general, all of the test article-related changes were completely or partially reversed during the 4-week (1-month study) or 8-week (3- and 6- month study) recovery period.

Genotoxicity

Romiplostim is composed entirely of naturally-occurring amino acids and contains no organic linkers or other non-protein portions. This suggests that it is unlikely that romiplostim would react directly with deoxyribonucleic acid (DNA) or other chromosomal material. Therefore, consistent with current ICH guidelines for genotoxicity testing for biotechnology-derived pharmaceuticals, these studies were not conducted.

Carcinogenicity

Studies to assess carcinogenicity of romiplostim were not conducted. However, as mentioned above, it is unlikely that romiplostim would react directly with DNA or other chromosomal material, therefore, mutagenic potential would not be anticipated.

Since romiplostim is highly immunogenic in rats, a traditional chronic oncogenicity study could not be conducted. However, it was considered unlikely that romiplostim would induce tumour development or growth since its mechanism of action is stimulation of platelet production via the Mpl receptor. In addition, there was no evidence of hyperplasia or pre-neoplastic lesions in the 6-month monkey study.

It was noted that there is a theoretical concern relative to the tumour stimulation capacity of TPO, and therefore of romiplostim, for certain subsets of acute myeloid leukemia (AML) cells.

Although it was stated that stimulatory effects were seen at TPO concentrations ranging from 20 to 200 ng/mL, which exceed the concentrations expected in the clinical studies, cell proliferation studies were not conducted by the sponsor. In the absence of an appropriate validated in vivo animal model for ITP, or an animal homologue for romiplostim, there remains concern regarding the long-term carcinogenic potential of romiplostim. For this reason, patients should be carefully monitored, and follow-up long-term safety data should be submitted when available.

Reproductive and Developmental Toxicity

A study was conducted to determine the effects of romiplostim on fertility when administered by SC injection to Sprague Dawley rats. Doses of 0, 10, 30, and 100 µg/kg bw were administered three times per week. Mortality was observed at all dose levels. All deaths were considered to be due to a combination of the pharmacological effect of romiplostim (increased platelet count) and the bleeding procedure. When the blood collection procedures were amended, no other deaths associated with blood collection occurred. Treatment with romiplostim did not have any effect on reproductive performance or fertility at any dose level tested.

Embryo-foetal developmental studies were conducted in mice and rats at dose levels of 0, 10, 30, and 100 µg/kg bw. For mice, romiplostim was embryo/foetal toxic in the absence of maternal toxicity at the highest dose of 100 µg/kg bw, based on an increased incidence of post-implantation loss. For rats, there were no adverse maternal or foetal effects noted at any dose level tested.

A pre- and post-natal developmental study was conducted in Sprague Dawley rats at dose levels of 0, 10, 30, and 100 µg/kg bw. Treatment of the initial parent generation (F0) dams with romiplostim induced neutralizing antibodies to the test material in approximately 70% to 80% of the dams in each group. This created two subgroups, Ab+ and antibody-negative (Ab-), which were evaluated separately and compared as appropriate. Treatment-related maternal mortality was noted at 30 and 100 µg/kg bw shortly after blood collection procedures, which was considered secondary to the increased blood viscosity resulting from markedly elevated platelet counts. As well, an increase in the incidence of stillborn pups was noted in the 100 µg/kg bw group for both the Ab+ and Ab- subgroups. There was also an increase in pup loss during the early lactation period. A total litter loss also occurred at 30 µg/kg bw, in the Ab+ subgroup, contributing to the incidence of both stillborn and early post-parturient decedent pups. Despite the small increase in peri-natal pup mortality, there was no notable effect on overall live litter size. Also, there was no effect on post-weaning functional development (open field assessment, motor activity, learning test), preputial separation/vaginal opening, estrous cycling or gross necropsy.

Based on the data from the reproductive and developmental studies conducted, it is recommended that special consideration be given to women who are being treated with romiplostim who may wish to become pregnant. It is recommended that romiplostim not be used during pregnancy unless the potential benefit to the mother justifies the potential risk to the foetus or embryo.

Local Tolerance

Local tolerance studies were not conducted; however, no evidence of substantive irritation was identified at the sites of injection in the repeat-dose toxicity studies. As such, it is unlikely that the formulated romiplostim would pose a meaningful risk for reaction at the site of injection.

Antigenicity

In an antigenicity study conducted in mice, it was determined that although 60 to 80% of the mice generated an antibody response to romiplostim within 2 weeks of a single exposure, endogenous platelet production was not impacted within the period evaluated. The strongest serum interactions were to thrombopoietin mimetic peptide (TMP).

Subsequent exposure to romiplostim resulted in reduced efficacy to lower doses of romiplostim. Increased doses of romiplostim were able to stimulate a platelet response even in the presence of antibodies to romiplostim.

Overall, it appears that anti-romiplostim antibodies may reduce the effectiveness of the drug, but do not lead to thrombocytopaenia, which would be the expected outcome if these antibodies cross-neutralized eTPO.

3.2.4 Summary and Conclusion

The non-clinical PD and PK data provided support for the mechanism of action of romiplostim to increase platelet production through the activation of the TPO receptor c-Mpl.

Non-clinical toxicity testing demonstrated that romiplostim was generally well-tolerated in rats, mice, cynomolgus monkeys and rhesus monkeys. Treatment-related findings were primarily related to the pharmacological activity of the test material. The non-clinical toxicology data base was considered adequate to assess the safety profile of romiplostim and to support its use in humans, provided adequate safety precautions are taken, as described above.

3.3 Clinical basis for decision

3.3.1 Human Pharmacology

Nplate™ (romiplostim) is a thrombopoiesis stimulating peptibody that increases platelet production through binding and activation of the TPO receptor, a mechanism analogous to eTPO.

The authorization of Nplate™ (romiplostim) was based in part on the results of two pivotal clinical studies as well as data from non-pivotal clinical supportive studies. The two Phase III placebo-controlled, double-blind pivotal studies provided the basis for establishing the pharmacodynamics, pharmacokinetics, safety, and efficacy of Nplate™ in adult patients with chronic ITP who had completed at least one treatment prior to study entry.

Pivotal Study Inclusion Criteria

For both studies, inclusion criteria required that patients be ≥18 years of age with a diagnosis of ITP according to the American Society of Hematology (ASH) guidelines. In addition, patients must have completed at least 1 previous treatment for ITP and have had a mean of 3 platelet counts during screening and pre-treatment periods that were ≤30×109/L, with no individual count >35×109/L. At study entry, patients could not be receiving any treatment for ITP except corticosteroids, azathioprine, or danazol administered at a constant dose and schedule. Haemoglobin of at least 9.0 g/dL was required at baseline. It was compulsory that patients ≥60 years of age had a documented history of chronic ITP with a bone marrow report to support the diagnosis. Patients with a known history of bone marrow stem cell disorder were excluded.

Study Design

The two pivotal studies were designed as follows:

Study S1 (212)

This study evaluated patients who were non-splenectomised and had an inadequate response or were intolerant to prior therapies. Patients had a median of 3 (range = 1 to 7) treatments for ITP prior to study entry. Study S1 (212) included 62 non-splenectomised patients refractory to or intolerant of conventional therapies (Nplate™: n = 41; placebo: n = 21).

Study S2 (105)

This study evaluated patients who were splenectomised (≥4 weeks prior to study entry) and continued to have thrombocytopaenia. In addition to a splectomy, patients had a median of 6 (range = 3 to 10) treatments for ITP prior to study entry. Study S2 (105) included 63 patients who were refractory to splenectomy (Nplate™: n = 42; placebo: n = 21 patients).

Pivotal studies S1 (212) and S2 (105) were similar in design. Both studies were Phase III, randomized, double blind, placebo-controlled, 24-week studies that assessed the efficacy and safety of Nplate™ in the treatment of adult thrombocytopaenic patients with ITP. Most of the non-splenectomised patients (53/62; 85%) had received 1 to 4 previous treatments. In comparison, the splenectomised patients were extensively treated, with 51/63 (81%) having received 5 to 10 previous treatments including splenectomy. Prior to study entry, approximately 95% of patients in both pivotal studies had received corticosteroids and 80% had received intravenous immunoglobulin (IVIG). In addition, 29% of non-splenectomised patients and 71% of splenectomised patients had received rituximab. Despite these treatments, platelet counts at study entry were still well below 30 × 109/L, indicating a seriously ill population in both studies; for non-splenectomised patients, the median platelet count at baseline was 19.3 × 109/L (range = 2 to 31 × 109/L), and for splenectomised patients, median platelet count at baseline was 14.0 × 109/L (range = 2 to 29 × 109/L).

Patients were randomized in a 2:1 ratio to receive a starting dose of Nplate™ (1 µg/kg) or placebo. The initial dose was followed by weekly SC injections for 24 weeks. Doses were adjusted to maintain platelet counts between 50 and 200 × 109/L. The most frequently used weekly dose for splenectomised patients was between 2 to 7 µg/kg and for non-splenectomised patients was between 1 to 3 µg/kg.

Patients in both pivotal studies were permitted rescue medication (consisting mainly of corticosteroids, immunoglobulins, and platelet transfusions) at any time during the study. Rescue therapies were permitted at the discretion of the treating physician for bleeding, wet purpura, or if the patient was at immediate risk.

3.3.2 Pharmacodynamics

The primary pharmacodynamics of Nplate™ were demonstrated in both pivotal studies. Among all ITP patients treated with Nplate™ in study S1 (212) and S2 (105) during the 24-week treatment period, the mean number of weeks with platelet response (platelet count ≥50 × 109/L without rescue therapy within 8 weeks) was 15 for non-splenectomised patients and 12 for splenectomised patients. Rescue therapy is defined as any therapy administered to raise platelet counts.

Secondary pharmacodynamics studies were not conducted for Nplate™.

3.3.3 Pharmacokinetics

The pharmacokinetics of Nplate™ involve target-mediated disposition through binding to the TPO receptors on the platelets and megakaryocytes, so that the volume of distribution and clearance are nonlinear with dose. As expected with this type of binding, the serum concentrations varied among patients and did not correlate with the dose administered.

As well, the elimination of serum Nplate™ is in part dependent on the TPO receptor on platelets, and as a result, for a given dose, patients with high platelet counts demonstrate low serum concentrations of Nplate™ and vice versa. The half-life values ranged from 1 to 34 days (median 3.5 days), and in the clinical studies no accumulation in serum concentrations was observed after weekly administration of 3 µg/kg Nplate™ for 6 weeks.

In patients with ITP who received chronic weekly treatment of Nplate™ subcutaneously, the pharmacokinetics of Nplate™ over the dose range of 3 to 15 µg/kg indicated that peak serum concentrations were observed approximately 7 to 50 hours post-dose (median ∼14 hours). Given the lag times between dosing and platelet response (7 to 12 days from population PK simulations), and the nonlinearity of the PK/PD relationship, the clinician must resort to a titration procedure based on a weekly regimen.

3.3.4 Clinical Efficacy

The authorization of Nplate™ was based in part on the efficacy results of the two pivotal clinical studies described in section 3.3.1 Human Pharmacology. In addition, other non-pivotal studies were included with the submission that provided additional information for the efficacy of Nplate™, however, efficacy data was mainly derived from the 125 patients who took part in the two pivotal studies.

Primary Efficacy Endpoint

The primary efficacy endpoint for both of the pivotal studies was the proportion of patients who achieved a durable platelet response (defined as a weekly platelet count ≥ 50×109/L six or more times during the last 8 weeks of treatment), in the absence of rescue medication at any time during the 24-week treatment period. A statistically significant effect was seen for Nplate™ relative to placebo, regardless of whether or not patients had undergone splenectomy.

The primary efficacy analysis of the two pivotal studies was based on the full analysis set, consisting of all randomized patients analyzed according to their randomized treatment group. In study S1 (212), a total of 25 of the 41 patients who were treated with Nplate™ (61.0%), who had not undergone splenectomy, achieved a durable platelet response as compared to 1 patient of 21 in the placebo group (5.0%). In comparison, results from Study S2 (105), indicated that a total of 16 of 42 patients (38.1%) treated with Nplate™ who were refractory to splenectomy achieved a durable platelet response as compared to 0 of the 21 patients in the placebo group (0%).

Secondary Efficacy Endpoints

Several secondary efficacy endpoints were also evaluated in the pivotal studies. These included the overall platelet response of each patient, defined as the mutually exclusive categories of durable platelet response plus transient platelet response. Transient platelet response was defined as at least 4 weekly platelet responses without durable platelet response. Secondly, the number of weekly platelet responses during the treatment period was considered. A weekly platelet response was defined as a platelet count of ≥50 × 109/L on the weekly scheduled dose day. The total number of weekly platelet responses was counted each week, from week 2 to 25. Next, the incidence of achieving a durable platelet response with stable-dose (i.e. a dose maintained within ± 1 µg/kg during the last 8 weeks of treatment) was assessed. This endpoint captured the ability of patients to maintain adequate platelet production control while on a steady dose of Nplate™. Finally, the proportion of subjects requiring rescue medications was quantified.

The incidences of durable and overall platelet responses, the proportion of subjects requiring rescue medication, and the incidence of subjects achieving a durable platelet response with stable-dose were compared between the Nplate™ and placebo groups by using the Cochran Mantel-Haenszel test stratified by baseline concurrent ITP therapy (yes or no) and by study when data were combined. Importantly, the use of rescue medication may have unpredictable effects on platelet count and could confound the interpretation of results. For the analysis of the incidence of durable platelet response, subjects who received rescue medications at any time during the treatment period were not considered to have had a durable platelet response. For transient platelet response, and to discount the possible platelet effect associated with rescue medications, subjects were considered to have had no platelet response for 8 weeks after any administration of rescue medications.

Overall, treatment with Nplate™ demonstrated significant improvements compared to placebo in both clinical studies for all efficacy endpoints in all patients randomized to the studies based on the intention-to-treat analysis. In particular, the mean number of weeks with platelet response (as defined above) was significantly higher in both studies for patients treated with Nplate™ versus those who received placebo:

  • Study S1 (212)
    • Nplate™: mean = 15.2 weeks, standard deviation (SD) = 7.5 weeks
    • Placebo: mean = 1.3 weeks, SD = 3.5 weeks;
  • Study S2 (105)
    • Nplate™: mean = 12.3 weeks, SD = 7.9 weeks
    • Placebo: mean = 0.2 weeks, SD = 0.5 weeks.

Furthermore, the proportion of patients who achieved a durable platelet response with stable-dose (as defined above) was significantly higher in both pivotal studies in patients who received Nplate™ as compared to the placebo:

  • Study S1 (212)
    • Nplate™: 21/41 (51.2%)
    • Placebo: 0/21 (0%);
  • Study S2 (105)
    • Nplate™: 13/42 (31.0%)
    • Placebo: 0/21 (0%).

Finally, the total incidence of rescue therapy was considerably higher for patients treated with placebo than with Nplate™ in both splenectomised and non-splenectomised patients. An overall reduction of 38.0% in the proportion of patients requiring rescue medication during the treatment period was observed for Nplate™-treated patients relative to placebo in the combined analyses: 59.5% of placebo-treated patients received rescue medication compared with 21.7% of patients treated with Nplate™.  Rescue medication was required by 17.1% of the non-splenectomised patients and 26.2% of the splenectomised patients who received Nplate™.

Long-term Extension Study

Patients who had previously completed a Nplate™ ITP study (including the pivotal studies) and whose platelet counts fell below 50 × 109/L after cessation of treatment were eligible to enroll in a long-term open-label extension study S3 (213). At the time of the authorization of Nplate™, this study was ongoing.

Patients who had taken part in the previous Phase I/II ITP studies (20000137 or 20010218) entered and were administered an initial weekly dose of 1 µg/kg. Those patients who entered from the blinded studies (S1 212 and S2 105) were unblinded. Patients from these studies who had previously received Nplate™, entered the study receiving the same weekly dose as they did in the prior study whereas those who had received placebo began at an initial weekly dose of 1 µg/kg and were adjusted accordingly. Patients continued with weekly dosing and individual dose adjustments of Nplate™ based on their platelet counts. A total of 137 patients were enrolled in the extension study; of the 137 patients enrolled, 136 patients received at least 1 dose of Nplate™. In the interim analysis, the 136 patients had been on treatment for a median of 39 weeks (range = 1 to 122 weeks).

For the interim analysis, a platelet response (doubling of platelet count and platelet count ≥50 × 109/L at any time on study in the absence of rescue medication within 8 weeks) was achieved in 82% of the subjects [95% confidence interval (95% CI): 75%, 88%]. Due to the heterogeneity of the population with regard to inclusion criteria, disease baseline characteristics, treatment history, concurrent medication, Nplate™ dose received, and length of treatment included in this study, data on the long-term efficacy and safety of Nplate™ should be interpreted with caution.

3.3.5 Clinical Safety

The authorization of Nplate™ was based in part on the safety results of the two pivotal clinical studies described in section 3.3.1 Human Pharmacology. In addition, other non-pivotal studies were included with the submission that provided additional information for the safety of Nplate™, however, safety data was mainly derived from 125 patients who took part in the two pivotal studies.

The primary safety pool in the integrated analysis of safety consisted of all subjects who received at least 1 dose of Nplate™ in the two pivotal studies (Phase III ITP Safety Set, n = 125). These two studies were combined with other studies in subjects with ITP as a secondary analysis of safety (ITP Safety Set, n = 215). As well, safety data were also presented from 78 patients who received Nplate™ in healthy subject studies, 20 patients with myelodysplastic syndromes (MDS) who received Nplate™, and 4 patients who received Nplate™ in a study considering chemotherapy induced thrombocytopaenia (AMG 531 Safety Set, n = 317).

Based on the Phase III ITP Safety Set, the most frequently reported AEs were headache (31.7% placebo, 34.5% Nplate™), fatigue (29.3% placebo, 33.3% Nplate™), and epistaxis (24.4% placebo, 32.1% Nplate™).

In the ITP Safety Set, 62 patients reported at least one serious adverse event (SAE) during an ITP clinical study: 10 (21.7%) placebo patients and 52 (25.5%) Nplate™ patients. A total of 28 treatment-related SAEs occurred in 18 patients (all were receiving Nplate™ at the time of the event), and 15 of these patients were splenectomised prior to enrollment in the study. Serious adverse events with the highest study duration-adjusted event rates were thrombocytopaenia [0 placebo, 16 (8.6 per 100 patient years) Nplate™] and platelet count decreased [8 (40.4 per 100 patient years) placebo, 4 (2.1 per 100 patient years) Nplate™].

Eight patients died during the Nplate™ ITP clinical program (ITP Safety Set); 3 (6.5%) placebo patients and 5 (2.5%) Nplate™ patients. The causes of death in patients treated with Nplate™ were: intracranial hemorrhage, pneumococcal pneumonia, cardiac arrest, hepatic and renal failure in a patient with malignant hepatic neoplasm, and acute respiratory distress syndrome.

Bleeding Events

In the Phase III ITP Safety Set combined analysis, bleeding events (BEs) occurred in 25 (61.0%) placebo patients and 48 (57.1%) Nplate™ patients. The patient incidences were 29 (63.0%) placebo and 128 (62.7%) Nplate™. In post-hoc analyses, the incidence of clinically significant BEs (grade 3 or 4) was numerically higher in patients receiving placebo. In the Phase III ITP Safety Set, the patient incidence was 5 (12.2%) placebo and 6 (7.1%) Nplate™ patients; and in the ITP Safety Set, 6 (13.0%) placebo and 19 (9.3%) Nplate™patients. Reanalysis of data comparing BE rates instead of patient incidence of BEs showed the following:

  • For patients treated with Nplate™, durable responders had both a lower total number of BEs and lower study duration adjusted BE incidence rate compared with non-durable responders. However, a comparison of the BE rates between non-durable responders in the placebo and Nplate™ groups revealed that the BE rate is higher among Nplate™-treated non-durable responders compared to placebo treated non-durable responders.
  • For BEs presented by severity status for study S2 (105), Nplate™-treated non-durable responders have higher BE rates for the four categories: mild, moderate, severe, and life-threatening. There was 1 fatal BE among the placebo-treated non-durable responders. A similar pattern was observed for study S1 (212) for the mild and moderate categories. There was 1 fatal BE in the Nplate™-treated non-durable responders.
  • For the overall Phase III ITP Safety Set, the trend observed in the individual studies was clear, with Nplate™-treated non-durable responders having higher BE rates for the 4 categories: mild, moderate, severe, and life threatening.

Some patients in the pivotal studies had platelet counts that increased and decreased to extreme levels within short periods of time. Throughout the study, these patients often required multiple rescue medications and numerous Nplate™ dose adjustments. As a result of the many severe declines in platelet counts, these patients experienced numerous BEs, including severe and serious BEs, and a life-threatening hemorrhage. Although this is only a proportion of the overall study population, these 9 patients highlight the individual variability that is found in ITP and the challenges of managing patients whose platelet counts cannot be stabilized, in contrast to patients who were able to achieve a stable-response.

Discontinuation of Nplate™ may result in thrombocytopaenia of greater severity than was present prior to Nplate™ therapy. This exacerbated thrombocytopaenia may increase the patient's risk of bleeding, particularly if Nplate™ is discontinued while the patient is being treated with anticoagulants or antiplatelet agents. In clinical studies of patients who discontinued Nplate™ treatment, 4 of 57 patients developed thrombocytopaenia of greater severity than was present prior to Nplate™ therapy.

Thrombotic/Thromboembolic Events

In the ITP safety set, a similar percentage of patients in the two treatment groups reported a thrombotic/thromboembolic event, 2 (4.3%) placebo patients and 9 (4.4%) Nplate™ patients.

Bone Marrow Abnormalities

Nplate™ administration increases the risk for the development or progression of reticulin deposition within the bone marrow. In the ITP safety set, no patient in the placebo group and 6 patients (2.9%) in the Nplate™ group had a bone marrow abnormality event. Nplate™ treatment was discontinued in 4 of the 271 patients because of bone marrow reticulin deposition. Six additional patients had reticulin observed upon bone marrow biopsy. Progression to marrow fibrosis with cytopaenias was not reported in the pivotal studies. In the extension study, 1 patient with ITP and haemolytic anaemia developed marrow fibrosis with collagen during Nplate™ therapy.

Malignancies and Progression of Malignancies

Stimulation of the TPO receptor on the surface of haematopoietic cells may increase the risk for haematologic malignancies. In controlled clinical studies among patients with chronic ITP, the incidence of haematologic malignancy was low and similar between Nplate™ and placebo. In a separate single-arm clinical study of 44 patients with MDS, 11 patients were reported as having possible disease progression, among which 4 patients had confirmation of AML during follow-up. Nplate™ is not indicated for the treatment of thrombocytopaenia due to MDS or any cause of thrombocytopaenia other than chronic ITP.

Warnings and Precautions

The following Black Box Warnings have been included in the Product Monograph for Nplate™:

  • Nplate™ should not be used in patients with myelodysplastic syndromes, outside of a clinical research study, because of the possibility of potentiating the development of myeloid leukemia in such patients.
  • Despite ongoing treatment with Nplate™, serious bleeding could occur and patients should be closely monitored during treatment. Rescue medications including platelet transfusions might be required, especially for patients with unstable platelet counts.
  • Recurrence of thrombocytopaenia, sometimes markedly below pre-treatment baseline levels, and serious life-threatening or fatal bleeding after discontinuation of Nplate™ have been reported.

Nplate™ should be prescribed and monitored only by qualified healthcare providers taking into consideration the following:

  • Nplate™ should be used only in patients with ITP whose degree of thrombocytopaenia and clinical condition increase the risk for bleeding.
  • Nplate™ should not be used in an attempt to normalize platelet counts.

The long-term risk for progression to myelofibrosis is unknown.
Discontinue Nplate™ if the platelet count does not increase to ≥50 × 109/L or to a level sufficient to avoid clinically important bleeding after 4 weeks at the highest weekly dose of 10µg/kg.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Nplate™ provides an effective approach for the treatment of ITP. As demonstrated in two multicentre, randomized, placebo-controlled, Phase III studies, Nplate™ robustly maintains platelet counts at clinically meaningful levels in both splenectomised and non-splenectomised patients, as compared to patients receiving only standard of care ITP medications, with response rates generally better than those medications. Nplate™ appears to maintain platelet levels ≥50 × 109/L in durable responders from both splenectomised and non-splenectomised populations. An open-label extension study also provided long-term safety results, re-treatment experience, and confirmed the ability to maintain platelet levels in durable responders over extended periods of time.

The administration of Nplate™ seems to reduce the need for acute rescue intervention with IVIG infusions or anti-D immunoglobulins as compared to placebo. Nplate™ has demonstrated efficacy in patients with chronic ITP who have not achieved satisfactory response to other therapies.

Safety concerns include:

  • Some patients are refractory to Nplate™ and appear to be at an increased risk of bleeding, sometimes severe; non-responders must be identified early to avoid such risks.
  • Patients may exhibit particularly low platelet levels after discontinuing Nplate™ treatment.
  • Some patients exhibited highly variable platelet counts during Nplate™ treatment.
  • The long-term risk of myelofibrosis and the progression of existing malignancies (including AML) from Nplate™ therapy is unknown.

All of the above problems will require vigilant patient care.

There is a potentially different risk-benefit profile for the two patient populations (splenectomised and non-splenectomised), with the profile being worse in splenectomised patients. This has been addressed in the labelling, the patient registry, and the Risk Management Plan.

Given the expectation to increase the platelet count in patients with ITP and its apparently benign safety record, the benefits of Nplate™ appear to outweigh its safety concerns, although the risk of myelofibrosis remains unresolved.

Priority Review status was granted for the evaluation of Nplate™ as it appeared to provide promising evidence of clinical efficacy for a serious, life-threatening, and severely debilitating disease that is not adequately managed by a drug marketed in Canada.

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 Nplate™ is favourable to increase platelet levels in adult patients with chronic immune (idiopathic) thrombocytopaenic purpura (ITP):

  • who are non-splenectomised and have had an inadequate response or are intolerant to corticosteroids and/or immunoglobulins;
  • who are splectomised and have had an inadequate splenectomy.

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: NplateTM

Submission MilestoneDate
Pre-submission meeting2007-11-15
Request for priority status
Filed2007-09-26
Approval issued by Director, Centre for Evaluation of Radiopharmaceuticals and Biotherapeutics2007-10-26
Submission filed2007-11-16
Screening
Screening Acceptance Letter issued2007-12-13
Review
On-Site Evaluation2008-01-14 - 2008-01-18
Biopharmaceutics Evaluation complete2008-06-10
Quality Evaluation complete2008-06-10
Clinical Evaluation complete2008-06-10
Biostatistics Evaluation complete2008-06-10
Notice of Deficiency (NOD) issued by Director General (quality issues)2008-06-10
Response filed2008-09-12
Screening 1 or 2
Screening Acceptance Letter issued2008-09-19
Review
Biopharmaceutics Evaluation complete2009-02-18
Quality Evaluation complete2009-02-16
Clinical Evaluation complete2009-02-18
Biostatistics Evaluation complete2009-02-18
Labelling Review complete2009-02-18
Notice of Compliance issued by Director General2009-02-19