Summary Basis of Decision for Vantas ®

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
Vantas®

Histrelin acetate, 50 mg, Subdermal implant, Subcutaneous

Paladin Labs Inc.

Submission control no: 092567

Date issued: 2006-10-27

Health Products and Food Branch

Our mission is to help the people of Canada maintain and improve their health.
Health Canada

HPFB's Mandate is to take an integrated approach to managing the health-related risks and benefits of health products and food by:

  • minimizing health risk factors to Canadians while maximizing the safety provided by the regulatory system for health products and food; and,
  • promoting conditions that enable Canadians to make healthy choices and providing information so that they can make informed decisions about their health.

Health Products and Food Branch

Également disponible en français sous le titre : Sommaire des motifs de décision (SMD), VASOVISTMD, gadofosveset trisodique, 244 mg/mL , solution, Berlex Canada Inc. No de contrôle de la présentation 096420

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:

Vantas®

Manufacturer/sponsor:

Paladin Labs Inc.

Medicinal ingredient:

Histrelin acetate

International non-proprietary Name:

Histrelin acetate

Strength:

50 mg

Dosage form:

Subdermal implant

Route of administration:

Subcutaneous

Drug identification number(DIN):

  • 02278383

Therapeutic Classification:

Luteinizing Hormone-Releasing Hormone Analogue

Non-medicinal ingredients:

Sodium: chloride solution, stearic acid

Submission type and control no:

New Drug Submission, Control No. 092567

Date of Submission:

2004-07-02

Date of authorization:

2006-03-10

Vantas is a registered trademark of Paladin Labs Inc.

2 Notice of decision

On March 10, 2006 , Health Canada issued a Notice of Compliance to Paladin Labs Inc. for the drug product Vantas®.

Vantas® is a histrelin acetate subdermal implant which contains the medicinal ingredient histrelin, an analogue of gonadotropin releasing hormone (GnRH).

Vantas® is indicated for the palliative treatment of hormone-dependent advanced prostate cancer (Stage M1 [TNM] or Stage D2 [AUA]). Following an initial stimulatory phase, chronic continuous exposure to histrelin desensitizes the responsiveness of the pituitary gland which, in turn, causes a reduction in testicular steroidogenesis and the reduction of circulating androgens such as testosterone. Androgen deprivation causing chemical castration (testosterone concentrations less than or equal to 50 ng/dL) has been shown to be efficacious in the palliative treatment of advanced prostate cancer.

The market authorization was based on submitted data from quality (chemistry and manufacturing) studies, as well as data from preclinical and clinical studies. Clinical efficacy was demonstrated in an open-label multi-centre Phase III study with 138 prostate cancer patients treated with Vantas®. Serum testosterone was suppressed to below the castrate level ( ≤50 ng/dL) in all 134 evaluable patients (100%) on Day 28. Of all 120 patients who completed 52 weeks of treatment, a total of 115 patients had a serum testosterone measurement at Week 52. Of these, all had serum testosterone ≤50 ng/dL. In patients without a Week 52 value, castrate levels were achieved by Day 28, were maintained up to Week 52, and remained below the castrate threshold after Week 52. Vantas® should be administered under the conditions stated in the Product Monograph, taking into consideration all potential risks associated with the administration of this drug product.

Vantas® (histrelin acetate subdermal implant, 50 mg) is designed to provide continuous release of histrelin acetate at a nominal rate of 50-60 µ/g per day over 12 months. Vantas® must be removed after 12 months of therapy. At the time the implant is removed, another implant may be inserted to continue therapy. Instructions for the implant insertion and removal are provided in the Product Monograph.

Vantas® is contraindicated in patients who are hypersensitive to GnRH, GnRH agonist analogues, or to any ingredient in the formulation or component of the subdermal implant. The product is also contraindicated in pediatric patients, in women who are or may become pregnant while receiving the drug, and in nursing mothers. Detailed conditions for the use of Vantas® are described in the Product Monograph.

Based on the Health Canada review of data on quality, safety and effectiveness, Health Canada considers that the benefit/risk profile of Vantas® is favourable for the palliative treatment of hormone-dependent advanced prostate cancer (Stage M1 [TNM] or Stage D2 [AUA]).

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (medicinal ingredient)

Manufacturing Process and Process Controls

The drug substance, histrelin acetate, is synthetically derived. Materials used in the manufacture of histrelin acetate are considered suitable and meet the standards appropriate for their intended use. The manufacturing process is considered to be adequately controlled within justified limits.

Characterization

Histrelin acetate is considered to be well characterized. Elemental analysis, nuclear magnetic resonance spectroscopy, mass spectrum analysis, and other identification tests confirmed the composition and structure of the drug substance.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. The proposed limits were considered satisfactorily qualified (e.g., within recommended ICH limits, toxicological studies). Control of impurities in the drug substance is therefore considered to be acceptable.

Control of Drug Substance

Copies of the analytical methods and, where appropriate, validation reports were considered satisfactory for all analytical procedures used for release and stability testing of histrelin acetate.

The specifications are considered acceptable for the drug substance. Batch analysis results were reviewed and were within the proposed acceptance criteria.

The proposed packaging components are considered acceptable.

Stability

Based upon the long-term and accelerated stability study data submitted, the proposed re-test period, storage, and shipping conditions for the drug substance are supported and considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Vantas® is a subdermal implant that looks like a small, thin flexible tube containing a white to off-white solution. The Vantas® product consists of a 50 mg histrelin acetate drug core inside a non-biodegradable, cylindrically shaped hydrogel reservoir. The drug core also contains the inactive ingredient stearic acid. The hydrogel reservoir is a hydrophilic polymer cartridge. The polymers swell reversibly in aqueous environments and the water content increases with increasing proportions of HEMA. The hydrated implant measures 32.5-36.5 mm in length and 3.05-3.25 mm in diameter.

The insertion tool is comprised of a multi-piece plastic tool and a bevelled stainless steel cannula. The cannula is controlled by a button which is extended to hold the implant. The implant is inserted in the open end of the cannula. The implant is inserted subcutaneously in the inner aspect of the upper arm. To insert the implant, the thumb pulls the implanter button back which removes the implanter from the insertion site, leaving the implant under the skin. Histrelin diffuses via the hydrated microporous structure and the diffusion is modulated by osmotic pressure.

The implant is packaged in a 3.5 mL Type I clear glass vial containing 2.0 mL of 1.8% NaCl solution and closed with a Teflon-coated stopper and sealed with an aluminum cap seal. Caps are silver and tear-off. Operational qualifications for the packaging and the shipping validation protocol were provided and are considered acceptable. Labelling provided included the draft inner and outer labels of the implant, the package insert, Instructions for Use, and Patient Information; all met the regulatory requirements. The sponsor did not provide the insertion tool Operator's Manual, Instructions for Use, or Patient Information, however this information is not considered necessary as it is a routine device and would fall under the category of Class 1 devices.

The Vantas® subdermal implant has met the physical and mechanical design goals. Diffusion is the only mechanism involved in the drug delivery. The controlled release mechanism is considered to be acceptable.

All excipients found in the drug product are acceptable for use in drugs by the Canadian Food and Drug Regulations. The compatibility of histrelin actetate with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

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

Manufacturing Process and Process Controls

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

Data was provided in compliance with the standards listed below:

ANSI / AAMI /
ISO 11134-1993

Sterilization of Health Care Products - Requirements for Validation and Routine Control - Industrial Moist Heat Sterilization

ANSI / AAMI /
ISO 1137-1994
Sterilization of Health Care Products: Requirements for Validation and Routine Control - Radiation Sterilization
ISO 10993-4 Biological Evaluation of Medical Devices - Part 4: Selection of Tests for Interactions with Blood (Biocompatibility)
ISO 10993-5

Biological Evaluation of Medical Devices - Part 5: Tests for in vitro Cytotoxicity (Biocompatibility)

ISO 10993-10 Biological Evaluation of Medical Devices - Part 10: Tests for Irritation and Sensitization (Biocompatibility)
ISO 10993-11 Biological Evaluation of Medical Devices - Part 11: Tests for Systemic Toxicity (Biocompatibility)
EN 554-1994 Sterilization of Medical Devices - Validation and Routine Control of Sterilization by Moist Heat

The cannula is sterilized using gamma irradiation according to ANSI/AAMI/ISO 11137.

All equipment, operating parameters, in-process tests and detailed instructions are adequately defined in the documentation and considered acceptable.

Control of Drug Product

Histrelin acetate is tested to verify that its appearance, identity, potency, content uniformity, mass balance, elution rate, and levels of degradation products and related impurities are within acceptance criteria. The proposed limits for degradation products were considered satisfactorily qualified (e.g., within recommended ICH limits, toxicological studies).

Copies of the analytical methods and, where appropriate, validation reports were considered satisfactory for all analytical procedures used for release and stability testing of Vantas®. The sponsor has provided details and test results for the 52-week elution test. The data provided is considered to be supportive of the claimed daily rate of drug elution.

Data from batch analyses were reviewed and considered to be acceptable according to the specifications.

Stability

Based upon the accelerated and long-term stability study data submitted, the proposed shelf life is considered acceptable when Vantas® is protected from light and stored at 2-8 °C. The implant is susceptible to damage if frozen. Labelling and the Product Monograph have been revised to include the statement "Do not freeze".

The shelf life for the device component of this product is two years. Study results show that the cartridges and implants can withstand relatively extreme physical stress.

3.1.3 Facilities and Equipment

The design, operations and controls of the facilities and equipment that are involved in the production are considered suitable for the activities and products manufactured. All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations.

3.1.4 Adventitious Agents Safety Evaluation

N/A

3.1.5 Summary and Conclusion

This New Drug Submission is considered to meet the requirements of Division C.08.002 of the Food and Drug Regulations insofar as the Quality (Chemistry and Manufacturing) information is concerned. The Chemistry and Manufacturing information submitted for Vantas® has demonstrated that the drug substance and drug product can be consistently manufactured to meet the specifications agreed upon. 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 Physical and Compatibility Tests

The following physical tests were conducted on the cannula portion of the device: appearance, length of cannula sleeve, length of cannula extended, length mark to tip, and functionality. The following physical tests were conducted on the implant: length and diameter, residual materials, tensile properties, tonicity of implant packaging, effectiveness of leaching, compatibility of the drug product with the implant system, stability, photostability, and physical changes to the implant. All test results demonstrated that the cannula and implant have met the physical and mechanical design goals and are safe and acceptable for clinical use.

Biocompatibility testing was conducted in accordance with ISO Standard 10993-1 on the stainless steel used in the cannula. Tests included : hemolysis test, USP systemic injection, intracutaneous/intradermal reactivity test, pyrogen test, cytotoxicity, partial thromboplastin time test, murine local nymph node assay, and residual testing.

All results met acceptance criteria and successfully passed. The stainless steel used in the cannula is considered biocompatible and acceptable for the intended use.

Biocompatibility testing on the implant included the following tests: toxicology of the implant (drug/device combination), genotoxicity (in vitro and in vivo), toxicology of Hydrogel (hydroxyethyl methacrylate) polymer, and toxicity of Hydrogel implant cartridge. The implant passed all tests and is considered to be non-toxic and a negligible irritant. There was no evidence of genotoxicity under the conditions tested. The histrelin acetate subdermal implant is considered biocompatible and acceptable for the intended use.

3.2.2 Pharmacodynamics

Results from the in vitro and in vivo animal studies adequately characterized histrelin as a structural analogue of gonadotropin-releasing hormone (GnRH). Histrelin was similar to other GnRH agonists, chemically and in pharmacological response.

As with other GnRH agonists, histrelin is a modified peptide closely related to GnRH with all amino acids in the natural L-form, but with deletion and substitution of amino acids conferring higher affinity for the GnRH receptor than GnRH itself. The effects of histrelin on male reproductive function (testosterone levels, testes and prostate weight and morphology, fertility) and/or prostate tumor volume were evaluated in dogs, rats, and monkeys. The results were similar to other GnRH agonists and demonstrated the following pharmacodynamic responses:

  • Essentially inactive when administered orally.
  • Showed more potency than GnRH in vivo and in vitro.
  • Caused an initial stimulatory luteinizing hormone (LH) response with a subsequent transient increase in testosterone concentration followed by an inhibition of LH release and suppression of testosterone biosynthesis.
  • Produced pronounced effects on the prostate (greater decreases in prostate weights and prostate tumour volumes) when administered by continuous infusion as compared to daily injections.
  • Maintained activity over time. Castrate levels of testosterone were sustained over the duration of the implant (up to one year in the dog).
  • Showed no irreversible effects on the male reproductive system after dosing ceased. The reproductive functions, as well as, the morphology of the testes and accessory sex organs were restored after a recuperation time.

3.2.3 Pharmacokinetics

The characterization of the pharmakinetics of histrelin in animals was very limited and there are no data on protein binding, metabolism or excretion. Studies with dogs showed increased exposure in terms of plasma/serum concentrations with increasing number of implants and higher histrelin dose. Thus, in spite of the limitations in the characterization of the kinetics in animals, the data and the similarities of histrelin chemically and in terms of pharmacological response to the well-characterized GnRH agon ist class are sufficient and additional kinetic studies in animals are not deemed necessary.

3.2.4 Toxicology

The toxicity program for histrelin included studies for chronic toxicity, carcinogenicity, and reproductive and developmental toxicity. Single-dose studies were not submitted. The genotoxicity of histrelin itself was not assessed, however, studies did show that e xtracts from histrelin subdermal implants (containing histrelin) were not mutagenic and did not induce micronuclei. The studies are well described and were properly conducted. The studies characterized the effects of long-term administration of histrelin at doses that caused ablation of the reproductive axis. The mode of administration (subcutaneous by injection) does not entirely mimic the clinical usage but with sustained expression of effects on the reproductive axis with no identification of toxicity of concern unrelated to the pharmacology, the safety in animal models is considered adequately assessed.

Chronic Toxicity

Chronic toxicity and reversibility of histrelin-mediated ablation of the reproductive axis were assessed in monkeys and rats. All effects in monkeys can be attributed to the pharmacological effect of histrelin, and effects on the reproductive tract were reversible within 6 months of cessation of dosing. In rats there were, in addition to pharmacologically mediated effects, increased fat deposition in the bone marrow and increased hepatic vacuolation at 180 µg/kg, which was reversible in all but one animal. There was no indication that histrelin in solution was irritating when administered subcutaneously in any species evaluated.

Carcinogenicity

Carcinogenicity studies were conducted in rats and mice. In rats, dose-related decreases in mammary tumours in females and non-dose related increases in Leydig cell tumours in males were probably related to the histrelin-induced changes in hormonal-responsive tissues. The increases in pancreatic islet cell tumours in female rats were attributed to obesity as histrelin caused weight gain in females. In mice, there were none or possibly minimal changes in hormonal-responsive tissues in histrelin-treated animals and it is questionable whether the mice were exposed even to therapeutic concentrations of histrelin. In spite of the dose selection being too low and lack of exposure data, there is no need for repeat carcinogenicity studies since there are no indications that histrelin implants are genotoxic and the indication is for palliative treatment.

Reproductive and Developmental Toxicity

Histrelin, as with other GnRH agonists, reduced fertility and/or caused sterility in both males and females in the laboratory species. The effects on morphology of the reproductive tracts and/or reproductive function were reversible in rats, dogs and monkeys.

Treatment of rats and rabbits during organogenesis was associated with increased foetal mortality and the effects on the F1 generation in rats could not be assessed due to high maternal and foetal toxicity. Rodents and rabbits may be more susceptible than humans to the foetal-toxic effects of histrelin during pregnancy. Rodents and rabbits, contrary to humans, are dependent on progesterone from a functional corpus luteum and GnRH and its agonist cause regression of the corpus luteum.

Histrelin was not teratogenic in rats whereas major fetal abnormalities were observed in rabbits.

3.2.5 Summary and Conclusion

Histrelin exerts effects that are consistent with the known pharmacological mechanism of action as other GnRH analogues. The submitted data adequately characterize the effects of histrelin, showing no unexpected toxicity; with the majority of findings related to the extended pharmacology, reversible upon cessation of dosing. With the data provided, and the known safety profile for this class of compounds and the palliative indication sought, the preclinical package for histrelin is acceptable.

3.3 Clinical basis for decision

3.3.1 Clinical Program

A total of four clinical studies were conducted to demonstrate the safety and efficacy of the histrelin acetate subdermal implant for the treatment of patients with advanced prostate cancer. Study #07-03-100 was a Phase I, open study that collected single-dose pharmacokinetic data in 6 healthy male volunteers. Study BAR-002-0591A-USA was a Phase II, open, parallel-group, international study that collected continuous-dosing pharmacokinetic and dose-ranging data, as well as therapeutic efficacy and safety outcomes, in 42 males with prostate cancer. Study #301 was a Phase II/III, open-label, international, pivotal efficacy and safety study that administered a single histrelin acetate subdermal 50 mg implant every 12 months into 138 males with advanced prostate cancer, and Study #302 was a non-pivotal, Phase II/III open-label study that administered a histrelin acetate subdermal 50 mg implant every 12 months into 33 patients with advanced prostate cancer.

A total of 185 patients with metastatic prostate carcinoma or patients having failed or proven refractory to definitive therapy were administered Vantas® (50 mg histrelin acetate subdermal implant) once every 12 months. These patients were the population base for demonstrating the efficacy and safety of the histrelin acetate subdermal implant.

3.3.2 Pharmacodynamics

An indirect pharmacodynamic relationship between serum testosterone and serum histrelin concentrations was observed in 119 prostate cancer patients. This relationship was consistent with the mechanism of action . Following an initial stimulatory phase, chronic subcutaneous administration of histrelin acetate desensitizes the responsiveness of the pituitary, which in turn causes reductions in ovarian and testicular steroidogenesis, including the production of testosterone.

3.3.3 Pharmacokinetics

The pharmacokinetics of histrelin acetate has been appropriately investigated in healthy volunteers as well as in the target patient population. The pharmacokinetics were characterized in three studies: #07-03-100 (n = 6); #BAR-002-0591A-USA (n = 42), and #301 (n = 138).

From the single-dose healthy volunteer study 07-03-100, following a single 500 µg subcutaneous (SC) bolus of histrelin acetate, peak serum concentrations occurred at 0.75 to 2 hrs post-dosing, with a peak of 13.5±3.00 ng/mL (mean+SD). With prostate cancer patients enrolled in studies BAR-002-0591A and #301, peak serum histrelin concentrations were achieved more slowly with the histrelin subdermal implant (median 12 hrs post-implant insertion) while peak concentrations averaged 1.10±0.375 ng/mL. Continuous SC release was evident throughout in contrast to the rapid decline in blood levels which followed the single bolus SC injection of histrelin. The average rate of SC drug release observed in Study #301, based on residual drug content of the implant device, was >56.7  µg/day over the 52-week dosing period which is consistent to the in vitro release rate of 56-57 µg/day. This results in a bioavailability of 92% for the histrelin SC implant in males with advanced prostate carcinoma with normal renal and hepatic function.

The apparent volume of distribution after the single 500 µg SC bolus injection in healthy males was 58.4±7.86 L. The fraction of unbound drug in plasma measured in vitro was 29.5%±8.9%. The in vitro drug metabolism data for histrelin were obtained using human hepatocytes. A single metabolite was found resulting from C-terminal dealkylation. Peptide fragments resulting from hydrolysis were also likely metabolites.

Excretion data from healthy volunteers given a single SC injection of 500 µg showed an apparent histrelin clearance of 179 +37.8 mL/min with a terminal half-life of 3.92±1.01 hrs. These figures compare to corresponding figures of 174±56.6 mL/min and 20 weeks, respectively for prostate cancer patients implanted with the hydrogel system loaded with 50 mg of histrelin.

The pharmacokinetics of histrelin were evaluated in prostate cancer patients with renal or hepatic failure. Although Cmax, Tmax, Vd, and half-life were prolonged when impairment was present either at baseline or post-baseline, no dose adjustments were considered necessary as is the case with other currently employed Luteinizing Hormone-Releasing Hormone (LHRH) analogue depot products used in the same clinical setting. The wide margin of safety for histrelin appears to support this recommendation.

Study #301 enrolled patients that were Caucasian, Black, and Hispanic. A subset exploratory analysis of race indicated that race did not have an impact on the kinetics of histrelin in the target population.

No specific pharmacokinetic drug-drug interaction studies were conducted for the histrelin hydrogel implant. However, given the minimal involvement of hepatic microsomal enzymes in the metabolism of GnRH agonists, induction or inhibition of drug metabolism of other drugs is unlikely. Also, as the extent of binding is not extensive (average fraction unbound was 29.5%), protein binding interactions are unexpected. In addition, no pharmacokinetic drug-drug interactions have been reported for any other GnRH agonists.

3.3.4 Clinical Efficacy

Efficacy results were submitted from pivotal Study 301 and two non-pivotal studies, Study 302 and Study BAR-002-0591A-USA. A total of 174 patients were treated with the proposed dose for the proposed indication. In all three studies, the primary and secondary efficacy endpoints were the same, the collection of data was consistent with regard to time and frequency, and the results were summarized with the same statistical methods.

The study results indicate that the 50 mg histrelin acetate hydrogel subdermal implant device produced a rapid (within 4 weeks), consistent and sustained reduction in serum testosterone levels below castration levels (<50 ng/dL) for more than 12 months in >90% of treated males, median age 75 years with advanced prostate carcinoma. The primary endpoint of chemical castration is a well-established and accepted surrogate efficacy endpoint for hormone-dependent prostate carcinoma and its reliability is further confirmed by a mirrored final reduction in patient serum LH, PSA and PAP levels, all indicating an inhibition of the pituitary-gonadal axis characteristic of the LHRH agonist analogues. When hormone-dependent prostate tumour cells are deprived of testosterone, tumour activity is minimized or halted, and disease progression is stabilized or ceased. Disease progression (treatment failure) while receiving hormone-blockade therapy is reflected in rising testosterone and PSA levels and clinical signs and symptoms of the spreading tumour.

The submitted clinical trial efficacy data indicate that the 50 mg histrelin acetate hydrogel 12-month implant device is an effective therapy for depriving prostate cancer cells of testosterone. The observed sustained and month-to-month consistent reduction in serum testosterone to below castration levels over the 12-month lifespan of the implant in the studied target patient population confirms the sponsor-reported in vitro implant drug release data. The lack of acute-on-chronic effects from repeated implant exposure in patients who continued treatment with a second, third or fourth implant further supports the efficacy of both the active drug substance and the implant device.

Indirect subjective evidence for the efficacy of this product was found in the improved perceived clinical benefit patients reported while on therapy (i.e. performance status, pain levels, analgesic use) as well as from the fact that >80% of the enrolled and treated patients completed the 60-week study for all three trials.

The target population represents a complex, varied patient group with characteristic age-related medical co-morbidities, differing levels of age and disease-related performance status, prior prostate cancer treatment histories and varying lifestyles. The demographic and disease-related characteristics of the male prostate cancer patients enrolled in the three histrelin clinical trials are representative of the intended target population cared for in the outpatient clinic setting and therefore the reported clinical trial efficacy results are considered reliable for when the drug is administered to the larger patient population in Canada.

Data submitted in support of the therapeutic efficacy of the histrelin implant device are considered sufficiently robust and reliable based on total patient numbers studied, the characteristics of the patient population enrolled and treated, the results of the primary efficacy endpoint and corroboration by the secondary efficacy endpoint outcomes, as well as the known pharmacologic actions of this drug class. The sponsor has demonstrated, to the same standard as the other approved products for this indication in Canada, the efficacy of the 50 mg histrelin acetate 12-month implant device in the proposed target population.

3.3.5 Clinical Safety

A total of 213 patients with advanced prostate cancer were treated with the histrelin acetate 12-month implant, of which 185 were treated specifically with the 50 mg histrelin implant (Studies 301 and 302, n = 171; Study BAR-002-0591A-USA, n = 14). The two efficacy trials, Studies 301 and 302, shared an identical study design including the same schedule and nature of patient evaluation throughout the 60-week study period and therefore a combined safety analysis is appropriate. The small, Phase II dose-range finding study (Study BAR-002-0591A-USA) included higher doses of histrelin than proposed for marketing and only contributed an additional 14 patients dosed at the 50 mg level. The most important safety conclusion from that study was that although no dose-dependent therapeutic effect was observed between the 50 mg, 100 mg and 200 mg histrelin implants, the incidence of adverse reactions was higher (but not differing in nature) for the 100 mg and 200 mg histrelin implants when compared to the 50 mg dose.

For the combined efficacy trials Study 301 and Study 302 (n = 171), the study populations were reflective of the proposed target population: males, median age 74 years (range 48-92 yrs), 85% >65 yrs; ~73% Caucasian, 20% Black; with advanced or metastatic hormone-dependent prostate carcinoma. The study criteria excluded patients with prior androgen-ablative or systemic corticosteroid therapy within one year of study entry, and patients that were orchidectomized. No on-study androgen, anti-androgen or biologic response modifier concomitant therapies were permitted. The study did permit palliative procedures such as transurethral resection of the prostate and irradiation for painful bone metastases.

Adverse Reaction Profile

In the combined safety population almost 100% of the patients reported an adverse event (AE), and ~77% a treatment-related AE of which ~27% of events were rated as serious. No patient in either study discontinued the histrelin treatment due to an AE, however 2% of patients withdrew due to implant site reactions or expulsions. For the 21 patients in Study 301 who received a second implant, 95% reported an AE, 71% treatment-related and 24% experienced a serious event. The consistency in these data underscores the identical study design and on-study patient monitoring and evaluation. A limited comparison is available for the 16 patients in Study 301 that were treated with Lupron 3-month depot and the 25 patients in Study 302 treated with Zoladex 3-month depot. Approximately 94% of the Lupron and 100% of the Zoladex patients reported an AE and 31% and 28%, respectively, experienced a serious AE. Although limited by small patient numbers, the similarity in event rates is likely a reflection of the drug class rather than individual drug formulation or drug delivery system.

In the histrelin-implant population, ~65% of the patients experienced possible or probably treatment-related hot flushes (2% severe, 25% moderate and 38% mild); 13.5% insertion site or local reactions; 10% fatigue; ~5% each for mild renal impairment, gynecomastia and testicular atrophy; and ~3% each for constipation, weight increase, headache, insomnia, decreased libido, and erectile dysfunction. These are all predictable adverse events observed for this class of drug in this patient population. The nature of these AEs as well as the incidence rates are similar to data published in the approved Product Monographs for the depot formulations Lupron, Zoladex and Eligard. The incidence of hot flushes with Suprefact (buserelin acetate) was reported at ~28%.

Death resulting from an AE occurred in 12/171 (7%) of patients in the combined study set. None were judged as treatment-related by the investigator and upon evaluation of each patient's narrative history, the nature of the event leading to death, the timing in relationship to drug administration and the patient's co-morbid conditions as well as prostate cancer response to treatment, no event can be clearly related directly to the effects of histrelin alone. The causes of death in this patient population with a median age of 75 years were either cardiovascular [myocardial infarction (AMI), cardiac heart failure (CHF), cerebrovascular accident (CVA)], pulmonary, or progression or complications of a second malignancy (i.e. brain, colo-rectal). Although this drug class is associated with a slight increase in hypercholesterolemia, a direct causality to enhanced AMI or coronary artery disease (CAD) cannot be determined without studying a patient group of similar age and stage of prostate cancer without any prior CV history. Serious adverse events reported at an incidence of ≥2% included AMI (8/171 or 4.6%) and CVA (5/171 or 2.9%). The relationship of these events to a direct adverse study drug effect is difficult to determine due to the median age of this population (i.e. CV co-morbidity is common) as well as a lack of any clear pharmacological basis for the event. No investigator judged the event to be drug-related based on the timing of the event to drug administration. Also, no serious allergic or anaphylactic reaction was recorded for any study patient.

Regarding adverse events related to the actual implant insertion or implant site, for the 13.5% of patients experiencing a local or insertion-site reaction, all reactions were mild in intensity. The majority of these AEs were associated with initial insertion, or removal and insertion of a new implant, and were resolved within 2 weeks. Reactions persisted in 4 patients (2.8%) and an additional 2.8% of patients developed an application-site reaction after the first two weeks following insertion. Local reactions were characterized by pain/soreness/tenderness (3.6%), erythema (2.8%) and swelling (0.7%). No increased intensity of reaction occurred in patients with a second implant-associated reaction. In the first 12 months after initial implant insertion, 8 (4.6%) patients experienced extrusion of the implant from the insertion site. These reactions were more likely to occur when a manual technique or loading device #2 was employed and not the currently proposed insertion technique and loading device.

Changes in clinical laboratory parameters were reviewed by an independent medical reviewer and the findings take into consideration the advanced age of the patient population and the multiple medical comorbid conditions. Clinically relevant changes were values that were greater than 30% of the upper limit of normal on two consecutive occasions. Based on these criteria, the following laboratory parameter changes occurred at a frequency of <2%: ALT, BS, LDH, CrCL, testosterone, PAP, Ca, cholesterol. No histrelin-specific effects can be identified from these data and similar changes are observed for the currently marketed LHRH agonist analogues when used in this patient population.

A total of 8 patients (4.6%) were judged as experiencing treatment-related renal impairment (CrCL <60 mL/min), 5 with single isolated occurrences and 1 had 2 occasions of such an abnormality. Only 2 (1%) patients were considered as suffering clinically relevant renal impairment when CrCL values fell below 60 mL/min on 3 and 4 occasions, respectively.

The safety issue regarding immediate post-implant testosterone flare and the accompanying risk of worsening bladder outlet obstruction and bone pain, fracture or spinal cord compression remains a potential risk for this class of drugs in this patient population. Testosterone levels fell to castration levels by week 4 in >90% of all patients. Examination of testosterone levels pre-treatment and in the following 4 weeks up until castration levels were achieved, did not show evidence of a significant rise or "flare" of testosterone levels. In Study 301, for all 138 treated patients, the mean group baseline testosterone level was 376 ng/dL, increased to 531 ng/dL by approximately day 3-4 and then decreased to 86 ng/dL at week 2, and to 9.7 ng/dL at week 4. Random review of individual patient data listings did not reveal any clustering of data to confirm a high incidence of testosterone flare upon insertion of the histrelin implant. Clinically this can also be confirmed by the reported low (<0.6%) incidence of clinically significant bladder outlet obstruction and indication for transurethal resection of prostateprocedures during the study. Other clinical conditions that could indicate a significant flare reaction would be worsening bone pain, spinal cord compression or increased fracture risk. None of these events occurred with an incidence of >1%. Although no time tracking of these events was incorporated into the study plan, the rapid achievement of chemical castration associated with a minimal increase in serum testosterone levels in the 4 weeks post-implantation and the lack of a significant incidence of adverse events associated with flare reactions, it appears that histrelin implant patients are not at any increased risk for this phenomenon over the currently marketed LHRH depot formulation analogues.

Special Populations

For patients with renal impairment (CrCl <60 mL/min) at study entry (n = 55), no dose adjustment was practiced. In these patients, 27 were classified as experiencing clinically relevant renal impairment during the study. In 1.2% of the patients, investigators reported an aggravation of the baseline renal impairment which occurred with full dosing in this subset of patients. Given the advanced age and co-morbid medical conditions in this patient population, administration of histrelin acetate in the presence of renal impairment requires monitoring but a downward dose adjustment does not appear warranted based on the pharmacokinetic studies and the small subset of patients in whom the initial renal impairment deteriorated. It appears that downward dose adjustment is unlikely to be required in patients with baseline renal impairment; however, renal function monitoring would be required.

Although 7 patients with mild baseline liver impairment entered the study and received the proposed dose of histrelin acetate, no patient deteriorated in baseline hepatic functioning; neither those with initial impairment nor those with normal liver function. Multiple patients did experience elevations in various liver function parameters, however, no pattern emerged in any one patient, indicating a concern regarding hepatotoxicity. The exact effect of histrelin acetate on liver functioning and the impact of liver impairment on histrelin pharmacokinetics have not yet been clearly elucidated. The pharmacokinetic studies that did evaluate these potential effects did not involve a sufficiently large enough patient population to make any clear conclusions. Also, metastatic prostate carcinoma can in itself result in elevations in LDH and if liver metastases are present other liver function parameters can be elevated. It is considered at this time that caution should be exercised in administering histrelin acetate to patients with liver impairment and patients should be monitored for possible drug-induced liver toxicity.

The potential risk of enhanced osteoporosis while receiving any LHRH agonist on a chronic basis has been documented in the literature. The Product Monographs of the approved LHRH agonists used in this patient population in Canada carry a standard precaution. In the combined clinical trial data, the risk of osteoporosis was not specifically determined with bone marrow density studies. However, in this patient population the risk of pathological fracture secondary to bone metastases is present. The incidence of fracture in all patients treated with the histrelin implant was 1.4% (3/213): one ankle, one rib and one hip fracture with 78% of all patients achieving drug exposure of ≥12 months. It is recommended that the Product Monograph for histrelin acetate also carry the standard drug class precaution regarding the potential risk of osteoporosis.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Histrelin acetate is an LHRH agonist, a potent inhibitor of gonadotropin secretion when administered continously. Vantas® is a subdermal implant system consisting of a biocompatible, non-biodegradable hydrogel cartridge containing 50 mg of active histrelin acetate with a consistent drug release rate of 50-60 µg/day over a lifespan of 12 months. The proposed indication for Vantas® is for the palliative treatment of patients with hormone-dependent advanced prostate carcinoma (Stage M1[TNM] or Stage D2 [AUA]). Unlike the currently marketed LHRH agonists approved in Canada for the palliative treatment of advanced prostate carcinoma, the histrelin acetate subdermal implant requires surgical insertion and then removal of the empty drug delivery system every 12 months. This procedure is carried out by a physician trained in the technique using a trocar loading device and performed with or without a local anesthetic in the outpatient clinic setting. Patients do not require additional visits to monitor the actual implant once inserted. Routine patient evaluations are those considered necessary as part of the standard of care for this patient population.

The sponsor has submitted sufficient clinical efficacy and safety data to support approval of the proposed clinical use of this drug product. The proposed drug dose and formulation as well as the implant device system were studied in a large target patient population with advanced prostate carcinoma. Histrelin acetate behaves pharmacologically as an LHRH agonist by causing inhibition of the pituitary-gonadal axis. The resultant hormonal effect is rapid, sustained and continuous chemical castration as reflected by serum testosterone levels of <50 ng/dL. Hormone-dependent prostate tumour cells are thereby deprived of testosterone and tumour growth stabilized or halted. This prevents further clinical disease progression and provides palliation of symptoms related to organ disease involvement with an acceptable safety profile. The efficacy data submitted in support of this new LHRH agonist is comparable in quality to that submitted for the approval of similar products in this drug class (i.e. study design, target sample size studied, primary and secondary efficacy endpoints).

Treatment with the histrelin acetate 12-month implant results in the same degree of efficacy as the marketed LHRH agonists as reflected by the almost complete chemical castration response rate within 4 weeks of insertion. Unlike the other products that require repeat depot injections at 1, 3 or 4-month intervals, the histrelin acetate 12-month implant provides continued efficacy over the lifespan of the implant. Furthermore, a significantly large proportion of target patients maintained both chemical and clinical response with a second and third 12-month implant. Although patient convenience was not evaluated in any of the submitted clinical studies, the lifespan of this device allows for a significant reduction in visits to a clinic for the purpose of drug administration.

The safety profile of the histrelin acetate implant is characteristic of this drug class. Comparative rates of adverse reactions and drug discontinuation rates are shared by this new product and the two leuprolide acetate, one goserelin acetate and one buserelin acetate depot products currently employed in this patient population. No histrelin acetate unique drug-related safety issues were identified in the studies submitted.

No unique risks have been identified from the clinical data submitted. Standard drug class warnings and precautions are also required in the labelling of this new product. In addition, because this product requires a minor surgical procedure for the insertion and removal of the implant device, physician training is required in the event that the procedures are not performed by an urologist. In Canada, this patient population is routinely cared for by a urologist or an oncologist specializing in the treatment of prostate cancer patients. However, a family physician specifically trained in the technique for implant insertion and removal could perform such a procedure where specialist care is unavailable although this is not anticipated to be the standard of care.

3.4.2 Recommendation

The review of this submission was a joint review between the Medical Devices Bureau (responsible for the safety and efficacy of the actual physical implant material as well as the trocar used for loading and inserting the drug-filled implant) and the Bureau of Metabolism, Oncology, and Reproductive Sciences (responsible for the safety and efficacy of the drug substance/product).

The device complies with the requirements of the Medical Devices Regulations, Part 1 and so the Medical Devices Bureau has no objections to the use of this medical device component in the Vantas® drug/device combination product.

Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Vantas® is favourable for the palliative treatment of hormone-dependent advanced prostate cancer (Stage M1 [TNM] or Stage D2 [AUA] ). 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: Vantas®

Submission MilestoneDate
Pre-submission meeting2003-05-08
Request for priority status
Filed2004-03-11
Rejection issued by Bureau of Metabolism, Oncology, and Reproductive Sciences2004-03-30
Submission filed2004-07-02
Screening 1
Screening Acceptance Letter issued2004-08-20
Review 1
Medical Devices Bureau Evaluation complete2005-02-28
Quality Evaluation complete2005-06-09
Clinical Evaluation complete2005-05-13
Labelling Review complete
NON issued by Director General (quality issues)2005-06-15
Response filed2005-08-26
Screening 2
Screening Acceptance Letter issued2005-10-11
Review 2
Quality Evaluation complete2006-03-09
Labelling Review complete2006-03-09
NOC issued by Director General2006-03-10