Summary Basis of Decision for Stelara

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
Stelara

Ustekinumab, 45 mg/0.5 mL, 90 mg/1.0 mL, Solution, Subcutaneous

Janssen-Ortho Inc.

Submission control no: 114272

Date issued: 2009-05-27

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:

Stelara

Manufacturer/sponsor:

Janssen-Ortho Inc.

Medicinal ingredient:

Ustekinumab

International non-proprietary Name:

Ustekinumab

Strength:

45 mg/0.5 mL, 90 mg/1.0 mL

Dosage form:

Solution

Route of administration:

Subcutaneous

Drug identification number(DIN):

  • 02320673 - 45 mg/0.5 mL
  • 02320681 - 90 mg/1.0 mL

Therapeutic Classification:

Selective Immunomodulating Agent

Non-medicinal ingredients:

sucrose, L-histidine, L-histidine monohydrochloride monohydrate, polysorbate 80, and water for injection

Submission type and control no:

New Drug Submission, Control Number 114272

Date of Submission:

2007-12-28

Date of authorization:

2008-12-12

* All trademark rights used under license

2 Notice of decision

On December 12, 2008, Health Canada issued a Notice of Compliance to Janssen-Ortho Inc. for the drug product, Stelara.

Stelara, a first-in-class agent, contains the medicinal ingredient ustekinumab which is a fully human IgG1κ monoclonal antibody that binds with high affinity and specificity to the human cytokines interleukin IL-12 and IL-23, and inhibits their bioactivity by preventing these cytokines from binding to their receptor protein expressed on the surface of immune cells.

Stelara is indicated in adult patients for the treatment of chronic moderate to severe plaque psoriasis who are candidates for phototherapy or systemic therapy. Abnormal regulation of IL-12 and IL-23 has been associated with immune-mediated diseases, such as psoriasis. Stelara prevents IL-12 and IL-23 contributions to immune cell activation, such as intracellular signalling and cytokine secretion, and is believed to interrupt signalling and cytokine cascades that are central to psoriasis pathology.

The market authorization was based on quality, non-clinical, and clinical information submitted. The safety and efficacy of Stelara were assessed in two multicentre, randomized, double-blind, placebo-controlled studies in 1996 patients 18 years of age and older, with chronic plaque psoriasis, who were candidates for phototherapy or systemic therapy. Overall, Stelara demonstrated efficacy and an acceptable risk over benefit ratio in the two pivotal studies. The efficacy of Stelara was significantly superior to placebo for the key psoriasis clinical outcomes. The data submitted demonstrate that Stelara can be administered safely when used under the conditions stated in the Product Monograph.

Stelara (45 mg/0.5 mL or 90 mg/1.0 mL, ustekinumab) is presented as a sterile solution for subcutaneous injection. The recommended dose of Stelara is 45 mg administered at Weeks 0 and 4, then every 12 weeks thereafter. Alternatively, 90 mg may be used in patients with a body weight greater than 100 kg. Stelara should be used only by physicians who have sufficient knowledge of plaque psoriasis and who have fully familiarized themselves with the efficacy/safety profile of the drug. Dosing guidelines are available in the Product Monograph.

Stelara is contraindicated for patients with known hypersensitivity to Stelara or to any of its components, and also for patients with severe infections such as sepsis, tuberculosis, and opportunistic infections. Stelara 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 Stelara 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 Stelara is favourable for adult patients for the treatment of chronic moderate to severe plaque psoriasis who are candidates for phototherapy or systemic therapy.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Abnormal regulation of the human cytokines interleukin-12 (IL-12) and IL-23 is associated with immune-mediated diseases, such as psoriasis. Ustekinumab, the medicinal ingredient of Stelara, is a fully human IgG1κ monoclonal antibody that binds to the p40 protein subunit of human IL-12 and IL-23. Ustekinumab inhibits the bioactivity of human IL-12 and IL-23 by preventing these cytokines from binding to their IL-12Rβ1 receptor protein expressed on the surface of immune cells.

Manufacturing Process and Process Controls

Ustekinumab, is manufactured by continuous perfusion cell culture, and purified through a combination of chromatography steps and viral inactivation/removal steps.

In-process controls performed during the 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 ustekinumab consistently exhibits the desired characteristic structure and biological activity. The impurities that were reported and characterized were found to be within established limits.

Control of Drug Substance

The drug substance manufacturing process has been validated through the manufacture of three validation batches and representative small-scale studies. Process validation data demonstrate that the manufacturing process operates in a consistent and controlled manner, yielding product that consistently meets specifications.

Stability

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

3.1.2 Drug Product

Description and Composition

Stelara is presented as a sterile, clear to slightly opalescent, colourless to light yellow solution in a single-use (Type I) glass vial with a butyl rubber stopper. Each 1.0 mL of Stelara contains 90 mg of ustekinumab. Stelara contains the following inactive ingredients: sucrose, L-histidine, L-histidine monohydrochloride monohydrate, polysorbate 80, and water for injection.

There are two strengths of Stelara: 45 mg of ustekinumab in 0.5 mL of solution, or 90 mg of ustekinumab in 1.0 mL of solution. Stelara is available in packs of one single-use vial.

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

Pharmaceutical Development

The formulation of Stelara was optimized during development for increased stability and suitability for subcutaneous administration. Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

Stelara is manufactured from ustekinumab formulated bulk. The process essentially consists of thawing the formulated bulk, pooling, mixing, sterile filtration, and aseptic filling into 2 mL glass vials. All product-contact equipment is product-dedicated or single use. The method of manufacturing is considered acceptable and the process is considered adequately controlled within justified limits.

Control of Drug Product

Stelara is produced from a well controlled and consistent process. Test methods that are in use to assess identity, purity, and potency are reliable; and all lot release test results provided in this submission were within specifications.

The drug product process validation program evaluated the following aspects of the process: the performance of major equipment, hold times, aseptic filling, cleaning/sanitization and sterilization procedures. Data provided in the submission demonstrate that the manufacturing process operates in a consistent and controlled manner, yielding product that consistently meets specifications.

Stability

Based on the stability data submitted, the proposed 12-month shelf-life at 2-8°C for Stelara is considered acceptable.

The compatibility of the drug product with the container-closure system and container-closure system integrity was demonstrated through stability studies.

3.1.3 Facilities and Equipment

An On-Site Evaluation (OSE) was conducted at the drug substance manufacturing facility. The facility and equipment are considered acceptable for the production of ustekinumab. The facility involved in the manufacture of the drug product did not warrant an OSE since the facility was recently evaluated. The facilities and equipment are considered acceptable for the production of the drug substance and drug product.

3.1.4 Adventitious Agents Safety Evaluation

Pre-harvest culture fluid from each lot is tested to ensure freedom from adventitious agents (bioburden, mycoplasma, and viruses). Several steps of the purification process were demonstrated to remove and/or inactivate viruses, resulting in an adequate viral safety margin.

No raw materials used in the drug substance manufacture are of animal origin. All raw materials are subject to appropriate controls associated with their receipt, storage, and handling. The excipients used in the drug product formulation are not of animal or human origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Stelara 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 manufacturing processes.

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

Primary pharmacodynamic studies characterized the binding interactions of ustekinumab, as well as its mechanism of action, functional effects of neutralization, species cross-reactivity, and in vivo activity thereby supporting a psoriasis indication.

  • Ustekinumab was shown to bind to Domain 1 of the p40 protein subunit of human IL-12 and IL-23 with high affinity, specificity, and the expected 2:1 ligand to antibody ratio.
  • Ustekinumab demonstrated equivalent neutralization of IL-12 and IL-23 binding to IL-12Rβ1, which was sufficient to block binding to functional dual
    IL-12Rβ1/IL-12Rβ2 and IL-12Rβ1/IL-23R cell surface receptor complexes for
    IL-12 and IL-23, respectively.
  • Ustekinumab effectively neutralized all IL-12- and IL-23-mediated cellular responses tested using in vitro bioassays.
  • Ustekinumab demonstrated comparable species reactivity to humans and non-human primates (including cynomolgus monkeys), but only partial or no activity to mouse, rat, dog, or rabbit IL-12 or IL-23. The key amino acid residues required for ustekinumab binding are identical between human and cynomolgus monkeys.

Secondary pharmacodynamic studies analyzed the in vivo activity of ustekinumabin animal models of immune-mediated diseases other than psoriasis. These studies demonstrated successful in vivo IL-12 neutralization via ustekinumab.

In the safety pharmacology studies with cynomolgus monkeys, no adverse effects of ustekinumabwere observed following single and repeat doses of up to 45 mg/kg via intravenous (IV) or subcutaneous (SC) routes of administration. Furthermore, no adverse effects of ustekinumab were noted in cardiovascular evaluations of 5-month old juvenile monkeys exposed to ustekinumab indirectly following treatment of their respective dams.

Due to the high binding specificity of ustekinumab for its target, IL-12/23p40, ustekinumab is unlikely to have pharmacodynamic interactions with co-administered drugs. Thus, non-clinical pharmacodynamic drug interaction studies were not conducted with ustekinumab.

3.2.2 Pharmacokinetics

The pharmacokinetics of ustekinumab were investigated in cynomolgus monkeys, following single and multiple SC administrations of ustekinumab at doses ranging from 0.9 to 45 mg/kg.

Absorption

Following single and multiple SC administration in monkeys, ustekinumab was absorbed into the systemic circulation with the maximum plasma concentration (Cmax) occurring 1 to 7 days post-administration. The mean Cmax and drug exposure (AUC) values increased in an approximately dose-proportional manner. Following twice-weekly SC dosing, an estimated 5- to 10-fold accumulation in AUC and Cmax was noted. Steady-state was reached by Week 13.

Distribution

The volume of distribution of ustekinumab in monkeys was approximately 85 to 100 mL/kg suggesting that the distribution of ustekinumab was confined mainly to the intravascular space.

In pregnant monkeys, ustekinumab distributed to the foetus during gestation, having an apparent foetal/maternal distribution ratio of approximately 0.4. The exposure ratio appeared to be independent of the route of administration. Ustekinumab was also detected in the breast milk of lactating monkeys.

Metabolism and Excretion

No studies were conducted to evaluate ustekinumab metabolism or excretion. In monkeys, ustekinumab was eliminated from the circulation with mean terminal half-life values ranging from 8 to 22 days.

Immune Responses

Immune responses to ustekinumab treatment were seen in some single-dose studies but not in the multiple-dose studies. However, high serum concentrations of ustekinumab observed in the multiple-dose studies may have interfered with the detection of antibodies to ustekinumab.

3.2.3 Toxicology

Toxicology studies with ustekinumab were conducted in cynomolgus monkeys. In vitro data demonstrated that ustekinumab neutralizes the activity of IL-12 and IL-23 cytokines from humans and cynomolgus monkeys with a similar concentration response in vitro. Ustekinumab did not neutralize IL-12 and IL-23 cytokines from other common species used for toxicology studies, including rodents and dogs; therefore, no toxicology studies were conducted with these species.

Single-Dose Toxicity

Single-dose toxicity of ustekinumab was not evaluated. No evidence of acute toxicity was observed in the single-dose pharmacokinetic studies.

Repeat-Dose Toxicity

Ustekinumab was generally well-tolerated in the repeat-dose toxicology studies in monkeys following IV doses of up to 45 mg/kg/week for up to 1 month, and following twice-weekly SC doses of up to 45 mg/kg for 6 months.

The low and high doses in the toxicity studies were between 9 and 45-fold higher than the highest intended clinical dose in psoriasis patients (approximately 1 mg/kg based on a 90 kg psoriasis patient). Sub-chronic and chronic testing in cynomolgus monkeys was carried out for 1, 3 and 6 months at dose levels of 9 mg/kg (1-month study), 22.5 mg/kg (3- and 6-month studies) and 45 mg/kg (all studies). Safety pharmacology endpoints were incorporated into the design of the toxicity studies and included the evaluation of potential effects on the cardiovascular system (electrocardiograms, blood pressure, heart rate, and capillary refill time) and the central nervous system (clinical cage side observations and rectal body temperature). The dose levels were well-tolerated and the findings were consistent after 1, 3, or 6 months on study. There were no treatment-related adverse effects noted after 1, 3, or 6 months for any of the measured parameters at any dose level tested.

Genotoxicity and Carcinogenicity

Genotoxicity and carcinogenicity tests were not conducted with ustekinumab.

The range and type of genotoxicity studies routinely conducted for pharmaceuticals are not applicable to biotechnology-derived pharmaceuticals.

The risk of tumour development in humans could not be evaluated directly in the standard rodent carcinogenicity studies because ustekinumab is pharmacologically inactive in rodents. Of note, there were no tumours or pre-neoplastic changes observed in the histopathology evaluations in any of the ustekinumab toxicity studies in monkeys. The carcinogenic risk associated with ustekinumab treatment can only be evaluated in humans by monitoring malignancy in patients administered ustekinumab. This has been stated as part of the Risk Management Plan for Stelara.

Reproductive and Developmental Toxicity

No evidence of drug-related maternal toxicity, teratogenicity, or developmental delays were observed in the developmental and reproductive toxicity studies at doses up to 45 mg/kg ustekinumab in monkeys.

Immunotoxicity

Immunotoxicity testing, which was incorporated into the toxicity studies, revealed that there were no adverse effects of ustekinumab on functional immune responses, circulating lymphocyte subpopulations, or the distribution of T- and B-lymphocytes in lymphoid tissue.

3.2.4 Conclusion

The non-clinical pharmacology and toxicology studies support the use of Stelara for the proposed indication.

The non-clinical pharmacology studies demonstrated that ustekinumab acts as an interleukin 12/23 inhibitor. Ustekinumab was generally well-tolerated in the toxicity studies at doses up to 45 mg/kg, which is approximately 45-fold higher than the equivalent dose intended to be administered to psoriasis patients. In conclusion, the non-clinical toxicology database was considered to be adequate to assess the safety profile of ustekinumab and to support its use for psoriasis in humans.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Clinical results of a Phase II study were used to determine the appropriate doses and dose regimens for drug evaluation in the Phase III studies.  A dose-response relationship in efficacy was demonstrated in the reduction of the Psoriasis Area and Severity Index (PASI), specifically the PASI 75 response (a reduction in score of at least 75% from baseline in PASI). Thirty-one of the 70 patients (44.3%) who were responding inadequately at Week 16 achieved a PASI 75 response with one additional dose at Week 16. This suggests that a maximum response may not be achieved until after 16 weeks of treatment. In the range of doses studied (45 mg to 360 mg), a dose-response relationship in safety was not apparent and the adverse events (AEs) did not increase with the increasing doses.

3.3.2 Pharmacokinetics

Absorption

Ustekinumab was absorbed slowly into the systemic circulation with a median time to reach the maximum serum concentration (tmax) occurring between 7.0 days (90 mg) and 13.5 days (45 mg) after a single SC administration in patients with psoriasis.

The Cmax and AUC of ustekinumab appeared to increase proportionally with the 45 mg and 90 mg dose levels. Steady-state was achieved by Week 28 and there was no evidence of accumulation in ustekinumab concentrations over time when given every 12 weeks subcutaneously. The absolute bioavailability was approximately 57.2% in patients with psoriasis.

Serum ustekinumab concentrations appeared to be impacted by patient body weight. The median trough serum concentrations of ustekinumab in patients with higher body weight (>100 kg) in the 90 mg group were comparable to those in patients with lower body weight (≤100 kg) in the 45 mg group. When given the same dose, patients of higher body weight (>100 kg) had lower median serum ustekinumab concentrations compared with patients of lower body weight (≤100 kg).

Distribution

In patients with psoriasis, the median apparent volume of distribution during the terminal phase was 76 to 161 mL/kg following a single SC administration.

Metabolism

The exact metabolic pathway for ustekinumab is unknown. Like all proteins, ustekinumab is degraded in the lysozymes residing in the cells of all tissues.

Excretion

Ustekinumab was slowly eliminated from the circulation with a median half-life (t1/2) of approximately 19.8 days (45 mg dose) to 21.2 days (90 mg dose), after a single SC administration in patients with psoriasis.

Immune Response

Antibodies to ustekinumab developed at a low incidence rate (approximately 5%) in patients that were treated with ustekinumab. The low incidence of antibody positive patients precludes definitive conclusions on the impact of antibody status on clinical response.

3.3.3 Clinical Efficacy

The safety and efficacy of Stelara (ustekinumab) were assessed in two multicentre, randomized, double-blind, placebo-controlled pivotal studies (PHOENIX 1 and PHOENIX 2) in patients 18 years of age and older with chronic (>6 months) plaque psoriasis who had a minimum body surface area involvement of 10%, and PASI score ≥12 and who were candidates for phototherapy or systemic therapy. Patients with other forms of psoriasis (guttate, erythrodermic, or pustular psoriasis) were excluded from the studies. No concomitant anti-psoriatic therapies were allowed during the study with the exception of low-potency topical corticosteroids on the face and groin after Week 12. A total of 1996 patients were enrolled in the two studies. Stelara and the placebo were administered by SC injection. In PHOENIX 1, the duration of treatment was 52 weeks or longer. In PHOENIX 2, the duration of treatment was 28 weeks. Patients randomized to receive 45 mg or 90 mg of Stelara, received Stelara at Weeks 0, 4, and 16. Patients randomized to placebo, received 45 mg or 90 mg Stelara at Weeks 12 and 16. In PHOENIX 1, subsequent dosing regimens were determined by each patient’s response status according to the study design.

For both studies, the primary endpoint was the proportion of patients who achieved a reduction in PASI score of at least 75% (PASI 75) from baseline at Week 12. The major secondary endpoint was the Physician’s Global Assessment (PGA) that indicated the physician’s overall assessment of psoriasis focusing on plaque thickness/induration, erythema, and scaling. The Dermatology Life Quality Index (DLQI) was also assessed in both studies. Other efficacy endpoints included the Nail Psoriasis Severity Index (NAPSI) and the Itch Visual Analog Scale (VAS) in PHOENIX 1; and the Hospital Anxiety and Depression Scale (HADS), and Work Limitations Questionnaire (WLQ) in PHOENIX 2.

The proportion of patients who achieved the primary efficacy endpoint (PASI 75 response at Week 12) was significantly greater in each of the Stelara groups (45 mg and 90 mg) compared to the placebo group in both pivotal studies (PHOENIX 1 and PHOENIX 2). In PHOENIX 1, the PASI 75 response at Week 12 was 67% and 66% for the patients that received 45 mg and 90 mg of Stelara, respectively, compared to 3% for those that received placebo. In PHOENIX 2, the same efficacy endpoint was 67% and 76% for patients that received 45 mg and 90 mg of Stelara, respectively, compared to 4% for those that received placebo.

In both studies, treatment with Stelara, administered as 45 mg or 90 mg SC injections, led to rapid, substantial, significant, clinically meaningful improvements in psoriasis and was generally well-tolerated. The improvements in psoriasis were generally consistent with each Stelara group. A dose response in efficacy was observed with generally higher response rates observed in the 90 mg group versus the 45 mg group. Treatment with Stelara resulted in significantly improved Quality Of Life (QOL), as measured by the PGA and DLQI in both studies. The other endpoints (NAPSI, VAS, HADS, and WLQ) also supported the primary endpoint. In PHOENIX 1, the dose response began to emerge at Week 16, reached a maximum around Week 24, and was generally maintained through Week 40. In PHOENIX 2, the dose response reached its maximum around Week 16, and was generally maintained through Week 28. In both studies, clinical efficacy was affected by patient weight. In patients ≤100 kg, efficacy was similar with 45 mg or 90 mg dosing, whereas in patients >100 kg, higher levels of efficacy were observed with 90 mg compared to 45 mg dosing. Clinical response was generally associated with serum ustekinumab concentrations. Patients with a higher clinical response (as measured by PASI response) had higher median serum concentration of ustekinumab than those with a lower clinical response.

In addition, a multicentre, randomized, active-controlled study (ACCEPT) compared the safety and efficacy of ustekinumab and etanercept in patients 18 years of age and older with chronic (>6 months) plaque psoriasis who had a minimum body surface area involvement of 10%, PASI score ≥12, PGA score ≥3, who were candidates for phototherapy or systemic therapy, and who had an inadequate response to, intolerance to, or contraindication to cyclosporine, methotrexate, or Psoralen and Ultraviolet A (PUVA) therapy. A total of 903 patients were enrolled in the study. The percentages of patients that achieved a PASI 75 response at Week 12 were 67% and 74% for the 45 mg and 90 mg Stelara groups, respectively; compared to 57% for the etanercept group.

Overall, patients with moderate to severe psoriasis who were treated with Stelara 45 mg or 90 mg showed significant improvement in psoriasis.

3.3.4 Clinical Safety

The clinical safety of Stelara was primarily assessed from the two pivotal studies PHOENIX 1 and PHOENIX 2, described in 3.3.3 Clinical Efficacy. Additional safety data from a Phase III clinical study, ACCEPT, (also described in 3.3.3 Clinical Efficacy) provided supportive data to the original drug submission.

In both pivotal studies, Stelara was generally well-tolerated. The AE rates, AE profiles, and rates of AEs in the 45 mg and 90 mg Stelara groups were generally comparable to that observed in the placebo groups. The rates of discontinuation due to the study drug were generally comparable in the 45 mg and 90 mg groups. The rate of serious adverse events (SAEs) was low. No dose response in SAE rates or profile was apparent. The proportions of patients experiencing markedly abnormal values in haematology and chemistry laboratory test results were low and generally comparable between the placebo and Stelara groups. The incidence of injection site reactions with Stelara was low and generally mild in nature. The low incidence of antibody-positive subjects precludes definitive conclusions on the impact of antibody status on the development of injection site reactions. There was no apparent association between the development of antibodies to Stelara and the development of injection site reactions.

Stelara is a selective immunomodulator. Immunomodulating agents have the potential to increase the risk of infections and malignancies.

In both pivotal studies, the infection rates and the profile of infections observed through Week 12 were similar in the placebo and Stelara groups; and the infection rates and profiles were similar in the 45 mg and 90 mg groups. The most common AEs were nasopharyngitis and other upper respiratory tract infections. Most were considered mild and did not necessitate drug discontinuation. Rates of serious infections were generally low. No cases of tuberculosis were reported. One serious potential opportunistic infection of cutaneous disseminated herpes zoster was reported in the Stelara 90 mg group.

In PHOENIX 1, no malignancies were reported through Week 12. However, through the end of the reporting period (Week 52) malignancies occurred in 9 patients. Four non-cutaneous solid tumour malignancies (one case each: breast, prostate, thyroid cancer, and a malignant kidney neoplasm) were reported in 4 patients in the 45 mg group, and basal cell skin carcinomas were reported in 5 patients (2 patients in the placebo who later went to the 45 mg group, 1 patient in the 45 mg group, and 2 patients in the 90 mg group). In PHOENIX 2, through Week 28, 8 malignancies occurred in 7 patients. Non-melanoma skin carcinomas were reported in 6 Stelara-treated subjects. One solid tumour (hepatic neoplasm) was reported in a patient in the placebo group.

The ACCEPT study provided additional data related to the incidence of malignancies. In this study, 209 patients received 45 mg of ustekinumab, 347 patients received 90 mg of ustekinumab, and 347 patients received etanercept, for the treatment of moderate to severe plaque psoriasis. Four (0.7%) subjects in the ustekinumab groups had a treatment-emergent malignancy. Three subjects had a basal and/or squamous cell skin cancer detected in areas of psoriasis that had cleared with treatment. One subject with a familial history of breast cancer was diagnosed with breast cancer. No malignancies were observed in the etanercept group.

Longer term follow-up is required to better define the safety profile of Stelara. Changes have been made to the Product Monograph to reflect the identified safety concerns, particularly the theoretical concern that products such as Stelara might foster the development of malignancies. The sponsor has agreed to provide additional safety data as outlined in section 3.3.5 Additional Issues.

3.3.5 Additional Issues

The following commitments were requested by Health Canada and were accepted by the sponsor:

  • Provide Health Canada with a detailed Canadian Risk Management Plan (RMP) for evaluation by December 24th, 2008 and work with Health Canada to revise the RMP based on their feedback, as appropriate.
  • Perform all post-marketing activities listed in the Response to Clarifax dated October 17, 2008, including but not limited to: Pharmacovigilance Activities (Adverse Event Collection and Single Case Processing, Aggregate Reports, Safety Surveillance and Signal Detection); Evaluation of Safety Data from Clinical Trials and Post-Marketing Studies; and Medical Education Plan.
  • Submit to Health Canada, in accordance with Canadian Regulations, all serious adverse events including malignancies, other autoimmune diseases, serious infections, et cetera (etc.). that occur in the ongoing clinical trials with Stelara.
  • Submit to Health Canada the Stelara Medical Educational materials for comments and feedback by December 19, 2008.
  • Provide Health Canada with Periodic Safety Update Reports (PSUR) for Stelara every six months for the first two years post approval. Include in each PSUR an analysis of all Adverse Drug Events that may be related to the immunogenicity profile of Stelara (for example [e.g.], ADEs related to Immune System Disorder, Skin and Subcutaneous Tissue Disorders, etc.); malignancies, serious infections; blood glucose abnormalities.
  • Include Canadian patients receiving Stelara in a patient registry program, either in the international Public Service Appointment Regulations (PSOLAR) registry program or in a Canadian registry program, as will be indicated in the Canadian RMP.
  • File a Supplemental New Drug Submission as soon as the final study reports of the two pivotal studies that led to the market authorization of Stelara by Health Canada become available.
  • Submit to Health Canada any reports or correspondence regarding the PSOLAR registry program as well as the ongoing clinical trials or any other sources.
  • Provide all reports pertaining to post-approval commitments as made to major Regulatory Authorities.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Stelara (ustekinumab) is a new, first-in-class, fully human IgG1κ monoclonal antibody indicated in adult patients for the treatment of chronic moderate to severe plaque psoriasis who are candidates for phototherapy or systemic therapy. Ustekinumab binds with high affinity to the human cytokines IL-12 and IL-23 and neutralizes their bioactivity by preventing these cytokines from binding to their receptor protein expressed on the surface of immune cells. In the pivotal clinical studies, the patients that were treated with Stelara showed significant improvements in the key psoriasis clinical outcomes compared with the patients that received placebo.

The following serious adverse reactions were reported: serious infections and malignancies. The most common adverse reactions (>10%) in the Stelara clinical studies were nasopharyngitis and upper respiratory tract infection. Other adverse effects and risks pertaining to infections, malignancies, hypersensitivity reactions and antibody formation are detailed in the Product Monograph. A long term follow-up of these patients is required to better define the safety profile of this new drug.

As a selective immunomodulating agent, Stelara is contraindicated in patients with a malignancy or a major infection. The known risks associated with potentially life-long immunosuppression remain, and should be monitored. The sponsor has provided a Risk Management Plan and will submit to Health Canada all SAEs (per regulation) including malignancies, other autoimmune diseases, serious infections, etc. that occur in the ongoing clinical studies with Stelara, as well as Periodic Safety Update Reports for Stelara, every six months for the first two years post market authorization.

Overall, the studies demonstrated that Stelara was well-tolerated and associated with a manageable safety profile. Based on the safety and efficacy profile, the benefits of Stelara therapy seem to outweigh the risks. Restrictions to manage risks associated with the identified safety concerns have been incorporated into the Stelara Product Monograph, and form part of the Risk Management Plan.

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 Stelara is favourable in adult patients for the treatment of chronic moderate to severe plaque psoriasis who are candidates for phototherapy or systemic therapy. 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 (NOC) pursuant to section C.08.004 of the Food and Drug Regulations.

4 Submission Milestones

Submission Milestones: Stelara

Submission MilestoneDate
Pre-submission meeting:2007-10-16
Submission filed:2007-12-28
Screening
Screening Acceptance Letter issued:2008-02-18
Review
On-Site Evaluation:2008-05-12 - 2008-05-16
Quality Evaluation complete:2008-12-12
Clinical Evaluation complete:2008-12-12
Labelling Review complete:2008-12-03
NOC issued by Director General:2008-12-12