Summary Basis of Decision for Velcade
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:
Velcade
Bortezomib, 3.5 mg/vial, Lyophilized Powder for Injection, Intravenous
Janssen-Ortho Inc.
Submission control no: 90084
Date issued: 2005-12-29
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 the management of the risks and benefits to health related to 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), VELCADE*, Bortézomib, 3,5 mg/fiole, poudre lyophilisée pour injection, Janssen-Ortho Inc., No contrôle de la présentation 090084
Foreword
Health Canada's Summary Basis of Decision (SBD) documents outline the scientific and regulatory considerations that factor into Health Canada regulatory decisions related to drugs and medical devices. SBDs are written in technical language for stakeholders interested in product-specific Health Canada decisions, and are a direct reflection of observations detailed within the evaluation reports. As such, SBDs are intended to complement and not duplicate information provided within the Product Monograph.
Readers are encouraged to consult the 'Reader's Guide to the Summary Basis of Decision - Drugs' to assist with interpretation of terms and acronyms referred to herein. In addition, a brief overview of the drug submission review process is provided in the Fact Sheet entitled 'How Drugs are Reviewed in Canada'. This Fact Sheet describes the factors considered by Health Canada during the review and authorization process of a drug submission. Readers should also consult the 'Summary Basis of Decision Initiative - Frequently Asked Questions' document.
The SBD reflects the information available to Health Canada regulators at the time a decision has been rendered. Subsequent submissions reviewed for additional uses will not be captured under Phase I of the SBD implementation strategy. For up-to-date information on a particular product, readers should refer to the most recent Product Monograph for a product. Health Canada provides information related to post-market warnings or advisories as a result of adverse events (AE).
For further information on a particular product, readers may also access websites of other regulatory jurisdictions. The information received in support of a Canadian drug submission may not be identical to that received by other jurisdictions.
Other Policies and Guidance
Readers should consult the Health Canada website for other drug policies and guidance documents. In particular, readers may wish to refer to the 'Management of Drug Submissions Guidance'.
1 Product and submission information
Brand name:
Manufacturer/sponsor:
Medicinal ingredient:
International non-proprietary Name:
Strength:
Dosage form:
Route of administration:
Drug identification number(DIN):
- 02262452
Therapeutic Classification:
Non-medicinal ingredients:
Submission type and control no:
Date of Submission:
Date of authorization:
*Trademark of Millennium Pharmaceuticals, Inc., all trademark rights used under license.
2 Notice of decision
On January 27, 2005, Health Canada issued a Notice of Compliance under the Notice of Compliance with Conditions (NOC/c) Policy to Janssen-Ortho Inc. for the anti-neoplastic agent Velcade. The product was authorised under the NOC/c Policy on the basis of the promising nature of the clinical evidence, and the need for a confirmatory study to verify the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions.
Velcade contains the active ingredient bortezomib which is a selective, reversible proteasome inhibitor. Proteasome plays an essential role in maintaining homeostasis within the cells and disruption of these normal cellular processes can lead to cell death. Experiments have demonstrated that bortezomib is cytotoxic to a variety of cancer cell types. Bortezomib has reduced tumour growth in vivo in many preclinical tumour models, including multiple myeloma.
The safety and efficacy of Velcade were evaluated in clinical trials involving 202 patients, 183 with relapsed and refractory myeloma, who had received at least 2 prior lines of treatment and who were progressing on their most recent treatment. The effectiveness was based on response rates. The overall response rate was 28%. There were no controlled trials demonstrating a clinical benefit such as an improvement in survival.
Velcade is indicated for the treatment of multiple myeloma patients who have relapsed following front-line therapy and are refractory to their most recent therapy. Treatment must be initiated and administered under the supervision of a qualified healthcare professional who is experienced in the use of antineoplastic therapy.
Velcade is supplied in 10 mL vials containing 3.5 mg of bortezomib as a mannitol boronic ester. The drug is contraindicated in patients with hypersensitivity to bortezomib, boron or to any of the excipients.
The recommended starting dose of bortezomib is 1.3 mg/m2 body surface area administered as a bolus intravenous injection twice weekly for two weeks (Days 1, 4, 8, and 11) followed by a 10-day rest period (Days 12-21). This 3-week period is considered a treatment cycle. At least 72 hours should elapse between consecutive doses of Velcade* administered as an intravenous injection.
Detailed conditions for the use of Velcade are described in the Product Monograph.
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
Materials used in the manufacture of the drug substance (bortezomib) were considered to be suitable and/or meet standards appropriate for their intended use. The manufacturing process was considered to be adequately controlled within justified limits.
Characterisation
Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be either within International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) established limits and/or were qualified from batch analysis and therefore considered to be acceptable.
Control of Drug Substance
Validation reports were satisfactory for all analytical procedures used for in-process and release testing of the drug substance, and to justify the specification of the drug substance.
Data from batch analyses were reviewed and considered to be acceptable according to the specification of the drug substance.
Stability
Based upon the real-time and accelerated stability data submitted, the proposed shelf-life, storage and shipping conditions for the drug substance were supported and considered to be satisfactory.
3.1.2 Drug Product
Description and Composition
Velcade is supplied in individually cartoned 10 mL vials containing 3.5 mg of bortezomib as a mannitol boronic ester, as a white to off-white cake or powder.
Pharmaceutical Development
Changes to the manufacturing process and formulation made throughout the development of the product were considered acceptable upon review. Several studies which justified the type and proposed concentrations of excipients to be used in the drug product were found to be acceptable.
Manufacturing Process and Process Controls
The manufacturing process was considered to be adequately controlled within justified limits.
Control of Drug Product
Validation reports were satisfactory for all analytical procedures used for in-process and release testing of the drug product, and to justify the specification of the drug product.
Data from final batch analyses were reviewed and considered to be acceptable according to the specification of the drug product.
Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be either within ICH established limits and/or were qualified from batch analysis and therefore considered to be acceptable.
Stability
Based upon the real-time and accelerated stability study data submitted, the proposed 24-month shelf-life at 15-30°C for Velcade was considered to be satisfactory.
When reconstituted as directed, Velcade may be stored at 25°C. Reconstituted Velcade should be administered within eight hours of preparation. The reconstituted material may be stored in the original vial and/or the syringe prior to administration. The product may be stored for up to three hours in a syringe, however, total storage time for the reconstituted material must not exceed eight hours when exposed to normal indoor lighting.
3.1.3 Facilities and Equipment
The design, operations, and controls of the facility and equipment met Good Manufacturing Practice (GMP) requirements, and are considered to be suitable for the activities and products manufactured at the site.
3.1.4 Adventitious Agents Safety Evaluation
N/A
3.1.5 Summary and Conclusion
The design, operations, and controls of the facility and equipment met Good Manufacturing Practice (GMP) requirements, and are considered to be suitable for the activities and products manufactured at the site.
3.2 Non-Clinical Basis for Decision
3.2.1 Pharmacodynamics
In vitro and in vivo studies evaluated the characterization of bortezomib (Velcade) activity on proteasome, as well as its effects on tumour growth in a wide variety of cancer cell lines and tumour models, including multiple myeloma.
The primary pharmacodynamic studies showed that bortezomib acted in the following manner:
- Inhibits proteasome activity1. Studies with isolated 20S proteasomes of rabbit reticulocytes and different proteasome isoforms showed that bortezomib inhibited proteasome activity and appeared to be selective for the subunit, 20S, over other proteases. The inhibition of 20S proteasome activity was reversible but the forward-rate constant for inhibition was much greater than the reverse rate; and under the conditions tested, the inhibition was relatively constant across the different proteasome preparations. Studies with rats and mice also demonstrated reversible inhibition, and in vivo anti-tumourogenic effects were seen in a number of mouse tumour models. Results from a rat study also showed that the duration of proteasome inhibition increased with multiple as compared to single administration.
- Induced apoptosis but had no effect on paraprotein levels independent of cell death when evaluated in vitro in a multiple myeloma cell line2.
- Showed no effect on hematopoietic stem cell function. Bortezomib treatment of mice did not affect the ability of surviving hematopoietic stem cells to form colonies in vitro and cells from bortezomib-treated animals were functional in a bone marrow transplant to completely repopulate peripheral blood cells.
- Retained cytotoxicity in vitro in the presence of efflux pumps that confer drug resistance to other chemotherapeutic agents. Was anti-proliferative and cytotoxic at low concentrations in a wide variety of cancer cell lines and freshly isolated cancer cells.
- Exerted additive anti-tumourogenic effects with radiation and other chemotherapeutic agents in in vivo models. The potential effect in drug resistant tumours and additive therapeutic benefits remain to be demonstrated clinically.
- Showed no significant affinity or effect on the receptors, ion channels and enzymes that were screened including the major receptors involved in heart rate and blood pressure regulation. In spite of bortezomib being highly selective for the proteasome, a single dose of bortezomib exerted significant effects on the heart rate and blood pressure in animals at low multiples of clinical dose and exposure. The potential risks identified throughout the preclinical studies are justifiable in light of the indication and clinical benefit. Ongoing and planned pre-clinical studies into the etiology of these changes are requested and are to be submitted for review to support any subsequent submissions.
No secondary pharmacodynamic studies were conducted with bortezomib.
1 The proteasome is a multicomponent system present in all cells and one of the key elements in protein and peptide degradation in pro and eukaryotic cells. Proteasome is believed to affect cell adhesion, chemotaxis, angiogenesis, and cell division, which can contribute to tumour growth; and 20S, a subunit of the proteasome, is required for the in vivo catalytic activity of the proteasome
2 Multiple myeloma is a B-cell malignancy of the plasma cell. Malignant plasma cells undergo clonal proliferation, producing a form of monoclonal immunoglobulin heavy and/or light-chain called paraprotein (M protein). The measurement of the paraprotein in the serum and/or urine is a marker for the tumour burden and also provides a mechanism to evaluate response to treatment.
3.2.2 Pharmacokinetics
Absorption
Absorption was not characterized extensively.
Distribution
The kinetic and metabolic profiles of bortezomib were similar in rats, monkeys and humans. Bortezomib was rapidly distributed after intravenous (IV) administration. In the animal studies, the highest tissue concentrations were initially in organs of excretion and metabolism, tissues related to endocrine and secretory functions, and regions of rapid dividing cells. Radioactivity was detected in the pituitary and choriod plexus suggesting that entry into these parts of the central nervous system is possible. Concentrations in tissues generally decreased with time with exception of the lymph nodes, spleen and thymus where the highest concentrations were noted at the last sampling time point in the monkey (144 hours) and 24 and 48 hours in the rat. Kinetic analysis of repeated-dose studies are consistent with accumulation occurring with repeated dosing using the clinical dosing regimen with steady state achieved in both rat and monkey. The kinetic data in human is limited but consistent with accumulation occurring in humans.
Metabolism
Bortezomib was extensively metabolized; more than 30 metabolites were identified. The principal metabolic pathway was cytochrome (CYP) mediated deboronation. Limited characterization of the effects of the deoboronated metabolite have been conducted and generally they were inactive as a proteasome inhibitor.
Bortezomib was not a potent inhibitor of CYP isoforms and it appears unlikely to affect the metabolic clearance of concomitant medication, however, the effect of potent inducers/inhibitors on the clearance of bortezomib was not assessed. With its extensive metabolism, it is important that the potential drug-drug interactions between bortezomib and potent inducers/inhibitors in particular CYP 3A4 be assessed. Warnings of these possible drug interactions are highlighted in the Product Monograph.
Elimination
Bortezomib was eliminated via the hepato-biliary system and renal system; the hepatic route predominant in the rat and the renal route in the monkey. Routes of elimination have not been characterized in humans but with the animal data suggesting that both hepatic and renal clearance are quantitatively important, the potential of effects of renal and hepatic insufficiency on bortezomib elimination needs to be highlighted and further characterized. Appropriate warnings are placed in the Product Monograph.
Transfer of bortezomib across the placenta and secretion in milk have not been determined. Even though this lack of information is acceptable given the indication sought, the lack of this data is reflected in the Product Monograph.
3.2.3 Toxicology
Acute and repeat-dose toxicity were assessed in rats and monkeys. These species are appropriate models to evaluate the toxicity of bortezomib and its metabolites since their metabolism is qualitatively similar to humans. The selection of species was acceptable, the studies were Good Laboratory Practice (GLP) compliant and the studies were conducted in accordance with ICH S4A guidelines.
Studies to evaluate genotoxicity and effects on embryo and fetal development were performed. Carcinogenicity studies, dedicated fertility and early embryonic development and pre-and postnatal development studies were not conducted. Exposure at the administered dose in gravid rabbit dams was also not determined. With the indication sought, it is acceptable not to characterize these effects.
Plasma drug levels were determined in the toxicity studies but with probable and yet undetermined increased exposure with repeated dosing occurring in humans as well as in other species. The administered dose in mg/m2 was used to compare results in animals to the clinical dose (1.3 mg/m2).
Acute Single-Dose Toxicity
After a single bolus IV dose, lethal dose was 1.8 mg/m2 and 3.0 mg/m2 in rat and monkey, respectively. In monkeys, the cause of morbidity appeared to be effects on blood pressure and heart rate. There was a narrow range between lethal and non-lethal doses after acute dose administration in all species evaluated (mice, rat, monkey and dog).
Repeat-Dose Toxicity
The studies were GLP compliant and conducted in accordance with ICH S4A guidelines.
The range between lethal and non-lethal doses after repeated-dose administration was narrow in studies with mice, rats, monkeys and dogs.
Results showed that a single dose of bortezomib exerted significant effects on heart rate and blood pressure in animals at low multiples of clinical dose and exposure. The indication and clinical benefits (see Section 3.4.1 Benefit/Risk Assessment) warrant the risk but as part of the conditions associated with the NOC/c, ongoing and planned preclinical studies into the etiology of these changes will be submitted for review.
After repeated dose administration in both rats and monkeys, bortezomib induced multi-target organ toxicity. The effects in the animals were similar to some side effects observed in clinical trials (see Section 3.3.3 Clinical Safety). In animals, the repeated dosing was associated with peripheral neuropathy; decreases in red cell measurements and platelet counts; and changes in lymphoid tissues/organs, gastrointestinal tract, liver, kidney, testes and ovary. The hematopoietic, gastrointestinal and lymphoid system lesions are considered to be contributing factors to the debilitated state and early death and lethality observed in animal models in the repeated-dose studies.
Bortezomib lethality occurred after multiple cycles (twice weekly for 2 weeks, 10 days off) at 0.9 mg/m2 in both rats and monkeys, i.e. lower than the proposed clinical dose. The clinical benefit in the indication sought warrants the risk. However, with the effects generally dose-related, it is important that the warning concerning dose reductions and/or cessation of treatment with signs of toxicity be prominent in the Product Monograph.
In the clinical trials, bortezomib was associated with sensory peripheral neuropathy. Peripheral neurotoxicity in the monkey suggests that the incidence increases with dose and/or duration of treatment. Also, the changes seem to persist after discontinuation of treatment.
The effects of bortezomib on the central nervous system (CNS) were a concern. The pharmacokinetic studies detected small amounts of bortezomib in the choriod plexus and pituitary. Additional sampling of nervous tissue in the chronic rat and monkey studies were performed and results showed that the distribution into the CNS was restricted in the animals and CNS toxicity was not noticed. Postmarket surveillance with respect to CNS effects will be of paramount importance. The clinical benefit in resistant and refractory multiple myeloma outweighs this theoretical risk.
Genotoxicity
The battery of tests conducted to assess the genetic toxicity of bortezomib conformed to ICH guidance S2B and included GLP compliant valid studies to assess bacterial mutagenicity, a mammalian test for chromosome damage and an in vivo assay for chromosomal effects. Test results showed that bortezomib was not mutagenic but induced clastogenic effects in vitro in mammalian cells. The latter was due to the interruption of the cell cycle related to proteasome inhibition.
Carcinogenicity
Carcinogenicity studies were not conducted and are not required with the current indication (ICH guideline S1A).
Reproductive and Development Toxicity
Effects on embryo and fetal development studies were assessed in gravid rats and rabbits using daily IV injections of bortezomib during the period of organogenesis in accordance with ICH guideline S5A. There were no dedicated studies conducted to assess effects on fertility and early embryonic development or pre- and postnatal development. Considering the indication sought, it is acceptable not to characterize these effects.
A 6-month toxicity study in rats showed degenerative effects in the ovary at ≥0.3 mg/m2 and degenerative changes in the testes at 0.9/1.2 mg/m2. With the results on ovary and testes observed in rat, the Product Label needs to reflect warnings concerning reduced fertility. Bortezomib was not teratogenic in rats or rabbits but was embryo lethal in rabbits. Due to toxicity to the dams, bortezomib was evaluated at subclinical doses. Warnings in the Product Monograph about avoiding pregnancy are warranted.
Local Tolerance
The reconstituted clinical formulation was not irritating after IV and subcutaneous administration but did induce irritation after perivascular and intramuscular administration.
3.2.4 Non-clinical Issues/Recommendations
Single acute-dose studies showed that the effects of bortezomib on blood pressure and heart rate appeared to be the cause of morbidity. Repeat-dose studies below therapeutic doses showed that bortezomib induced multi-target organ toxicity without completely reversing after withdrawal. Effects in animals were shown to be similar to the side effects observed in the clinical trials. Repeated dosing in animals was associated with peripheral neuropathy, decreased red blood cell and platelet counts, and changes in lymphoid tissues/organs, gastrointestinal tract, liver, kidney, testes and ovary. The effects were generally dose-related. Warnings concerning dose reductions and/or cessation of treatment with signs of toxicity are prominent in the Product Monograph.
Given the high acute toxicity of bortezomib in all species in the preclinical studies, which appears causally related to dose-related changes in the cardiovascular system, and the high bortezomib-related incidence of cardiovascular events in clinical trials, the sponsor will be requested to submit additional cardiovascular studies in dogs to support subsequent applications. This will include an assessment of the time course and potential etiology of the cardiovascular changes, including sufficient monitoring of electrolytes, acid/base balance, physiologic and hemodynamic changes during the first 24 hours after administration. The sponsor will also be requested to submit a neurological assessment study in rats (Irwin test) for evaluation.
Despite the outstanding issues mentioned above, the indication for Velcade is for the treatment of multiple myeloma patients who have relapsed following front-line therapy and are refractory to their most recent therapy; this is a life-threatening condition. The preclinical data does not raise any issues that should prevent authorization of Velcade for the proposed indication. Precautionary measures are stated in the Product Monograph.
3.3 Clinical basis for decision
3.3.1 Human Pharmacology
Pharmacodynamics
The inhibitory effects of bortezomib on proteasome were evaluated in three Phase 1 dose-escalation studies and two Phase 2 studies. The Phase 1 dose-escalation studies were conducted to define toxicity profiles and determine the appropriate dosage and schedule for Phase 2 development. The Phase 1 trials included 123 patients with advanced cancers, and IV bolus unit doses of bortezomib ranging from 0.13 mg/m2 to 2.0 mg/m2. The anti-tumour activity noted in these Phase 1 studies led to the initiation of two Phase 2 studies (M34100-025 and M34100-024) on patients with multiple myeloma3.
Proteasome inhibition across the Phase 1 and 2 studies showed similar mean maximum inhibition and inter-individual variability. The control 20S proteasome values before treatment were variable, therefore explaining the high variability of data obtained with low doses of bortezomib and those recorded before subsequent doses of bortezomib. According to the sponsor, the assay was robust for high levels of proteasome inhibition observed with clinically relevant doses of bortezomib; there was concurrence of dose-response relationships for maximum inhibition obtained across studies with consistency of results at different timepoints within treatment cycle and across different cycles and a close range of values of maximal inhibition across the studies. Nevertheless, a large variability of the levels of inhibition and its kinetic of recovery was observed.
Dose-response and a plasma concentration response relationship with proteasome inhibition exist, but are based on a limited number of patients with pharmacokinetic/ pharmacodynamic data. The time course of proteasome inhibition was characterized by partial recovery of proteasome activity inhibition within 6-24 hours followed by persistent variable low levels of inhibition up to the next scheduled dosing. The dose range where a high level of inhibition was observed was between 1.0 mg/m2 and 2.0 mg/m2. In patients with refractory multiple myeloma, the level of proteasome inhibition seemed to be unrelated to the observed response rate. Multiple analyses revealed no significant relationship between degree of 20S proteasome activity and response to treatment or maximum change from baseline in M-protein.
The regimen selected for the Phase 2 studies was based on preclinical pharmacodynamic data and results of the Phase 1 studies. The studies demonstrated a lack of correlation between proteasome inhibition and toxicity. Evaluation of dose-limiting toxicities was based on clinical observation, for dose-limiting toxicity (DLT) and maximum tolerated dose (MTD). The MTD of bortezomib ranged from 1.04 mg/m2 to 1.6 mg/m2 depending on the dosing schedule. Intermittent administration resulted in high but transient inhibition of proteasome activity and was better tolerated than sustained inhibition, with return towards the pre-treatment level and recovery of proteasome activity between doses and cycle, minimizing toxicity. The twice-weekly dosing regimen was based on return to baseline (approximating 72 hours) and the supporting data that the earlier rest period after 2 weeks of twice-weekly dosing was better tolerated. However, return to baseline of proteasome activity was not a prerequisite for continuing treatment.
No formal studies to assess the effects of age, gender, race, impaired renal or hepatic function and concomitant medications on the pharmacokinetics and duration of 20S proteasome inhibition of bortezomib have been reported.
Early disposition kinetics of bortezomib in a subset of patients with reduced creatinine clearance indicate that disposition is not affected by variation in creatinine clearance (from 31 mL/min to 169 mL/min, which is normal renal function to moderate impairment) but no patients had severe renal impairment (creatinine clearance of <30 mL/min). The potential effect of moderate to severe renal impairment on inhibition of 20S proteasome activity by bortezomib and terminal elimination were not evaluated in this study. Phase 2 multiple myeloma enrolled 10 patients with a creatinine clearance between 10 mL/min and ≤30 mL/min but the levels were not measured at peak and not studied over many cycles.
3 Several distinct mechanisms have been proposed in the anti-neoplastic effects of bortezomib in multiple myeloma: inhibition of cell growth signalling pathways and induction of apoptosis; inhibition of nuclear factor-κβ activation with inhibition of cellular adhesion molecules, angiogenic factors and IL-6 production as well as reduction of adherence of myeloma cells to bone marrow stromal cells. Multiple myeloma cells over-express IL-6 which have been shown to have a major growth and anti-apoptotic effect in multiple myeloma cells.
Pharmacokinetics
Only limited data is available for the pharmacokinetic characterization of bortezomib at the recommended dose in multiple myeloma patients. However, the submitted data is considered sufficient to assess the benefit/risk assessment for Velcade in the treatment of relapsed and refractory multiple myeloma.
Results from the plasma concentration-time profiles of bortezomib showed rapid and extensive distribution from the systemic circulation; the initial distribution phase had an estimated half-life of 30 minutes followed by an elimination half-life between 9 and 15 hours. Following multiple doses of bortezomib, systemic exposure (AUC) doubled and this was associated with decreased clearance. The terminal elimination half-life increased approximately two fold. Similar results were seen in the animal studies.
The kinetic profile of bortezomib was not extensively characterized. The sponsor would be required to submit additional studies to support a submission for expanded use.
The submitted data is insufficient for the following reasons:
- Data was limited with very few patients as monotherapy. Sampling was also limited in these patients coupled with samples below the limit of quantification for the assay.
- Kinetics were evaluated at a narrow dose range with few conclusions drawn on dose proportionality.
- High interindividual variability was reported with no clear kinetic reason identified (rapid distribution with infrequent sampling times has been proposed but not confirmed).
- Accumulation was observed after 3 doses in cycle 1 in patients treated with borterzomib and gemcitabine in combination, with no data as a single agent or multiple cycles. Data was limited but consistently showed accumulation of borterzomib in humans. The decrease in clearance after repeated doses could possibly result in accumulation and toxicity over time if no treatment adjustments are made.
- Elimination routes were not studied in humans. Animal studies provided data for hepatic, urinary and biliary excretion routes, suggesting both hepatic and renal clearance as being quantitativily important. There is insufficient clinical data available for analysis of liver impairment or moderate to severe renal impairment on the terminal disposition kinetics of borterzomib.
- No interaction studies (CYP 3A4 and CYP 2C19 inducers and inhibitors) were conducted.
- The effects of gender, age or race on the pharmacokinetics have not been investigated.
- Transfer of borterzomib across the placenta and secretions in bile have not been determined.
3.3.2 Clinical Efficacy
Two Phase 2 multicentre, open-label, non-comparative studies (M34100-025 and M34100-024) used bortezomib alone or in combination with dexamethasone to establish the efficacy and the safety of bortezomib in relapsed (and refractory, in Study M34100-025) multiple myeloma, for a maximum duration of 8 cycles.
The pivotal Study M34100-025 enrolled 202 patients, 91% of which had relapsed and refractory multiple myeloma, with at least two prior lines of therapy at a dose and schedule of 1.3 mg/m2 administered IV twice weekly for two weeks, followed by a 10 day rest period, for a maximum of 8 cycles of treatment. Supporting efficacy and safety data was provided from Study M34100-024 which randomized 54 patients relapsed after front-line therapy to one of two doses of bortezomib, 1.0 mg/m2 or 1.3 mg/m2 administered on the same schedule as in Study M34100-025. Dexamethasone was allowed in both studies for patients not responding to bortezomib alone. The primary objective in these Phase 2 studies was overall response rate to treatment with bortezomib alone using response as a surrogate for survival, with secondary endpoints being time to progression (TTP) and survival.
Efficacy measurements included serial electrophoretic analysis of the serum proteins to determine the amounts of immunoglobulins in the serum4. Other measurements included urine electrophoresis and immunologic typing of the M-component, and analysis of the bone marrow biopsy and aspirate, laboratory data and skeletal survey. The primary objective was to determine overall response rate. The secondary objectives and efficacy analyses included time to event parameters, time to response, duration of response, time to progression, survival, clinical benefit parameters (improvement in non M-protein immunoglobulins, renal function, haemoglobin, performance status, quality of life, reduction of myeloma paraprotein levels). The efficacy assessment and determination of disease response was based on criteria developed by Blade criteria5, with all data of response to treatment reviewed by the Independent Review Committee (IRC).
Five patients of 193 patients from the ITT population (patients treated with any amount of study drug) were excluded from the efficacy analyses because of inadequate prior therapy. Of the 188 evaluable patients, complete responses6 (CR) occurred in 5 (2.7%) and partial responses7 (PR) in 47 (25%) of the patients. Response was rapid at 38 days, and consistent across different response categories.
Supporting data was found in Study M34100-024; the paraprotein response to all-treatment showed 50% of the patients at 1.3 mg/m2 (41% in 1.0 mg/m2) had ≥50% reduction of M-protein levels. These results provided evidence for activity at the 1.0 mg/m2 dose level and supports continued use of bortezomib at this dose for patients reduced from a higher dose secondary to adverse events. In Study M34100-025, results also showed that response rates to bortezomib were independent of dose reductions8. From the group of subjects that were administered bortezomib alone, 32 (42%) of the 76 patients with dose reductions for adverse events had a response to treatment.
4 Serum of patients with plasma cell tumours have an increased gamma globulin region with a sharp spike of the M (monoclonal) component. The amount of M-component in the serum is a reliable measure of tumour burden.
5 The Blade criteria are available from the following website: http://www.multiplemyeloma.org/treatments/3.08.02.table_1.html.
6 Complete response required 100% disappearance of the original monoclonal protein from blood and urine on at least 2 determinations at least 6 weeks apart by immunofixation and <5% plasma cells in the bone marrow on at least two determinations for a minimum of six weeks, stable bone disease and calcium.
7 Partial response required ≥50% reduction in serum myeloma protein and ≥90% reduction of urine myeloma protein on at least 2 occasions for a minimum of at least 6 weeks, stable bone disease and calcium.
8 During Phase 2 studies, protocol specified dose modifications for Grade 3 or 4 drug-related toxicities to re-start at a reduced dose of 1.0 mg/m2 upon resolution to Grade 1 or better. Further reduction from 1.0 mg/m2 to 0.7 mg/m2 were also permitted.
Subpopulations
In the Phase 2 studies, there was some evidence that response rates to bortezomib were likely to predict clinical benefit, with biological activity being shown by both objective data (reductions in measurable disease parameters including abnormal levels of serum paraprotein and % bone marrow plasma cells) as well as subjective data.
The final demonstration of efficacy relied on the anti-tumour activity results and response rates from Study M34100-025, with supportive data from M34100-024. Using each patient as their own control, there was a 2- to 4- fold increase in TTP on bortezomib alone compared to previous therapy, with a median survival of 16 months which is better than expected in this population.
Study M34100-024 was a low-powered study that assessed a different population (not refractory) than the one studied in the pivotal study, and assessed two doses (1.0 mg/m2 and 1.3 mg/m2) of bortezomib. Both the mean and median total dose, and duration of treatment were higher in the 1.0 mg/m2group. Nevertheless, the statistical summaries show differences consistent with dose response with bortezomib monotherapy (response rate and reduction of paraprotein in both dose groups). Although the small sample size precludes any definitive statements, there is some evidence for a dose effect with respect to both efficacy and safety. Therefore, the higher dose (1.3 mg/m2) was recommended as the optimal starting dose with a dose reduction to 1.0 mg/m2 for toxicities.
Additional clinical benefit analyses were performed in responding patients with relapsed and refractory multiple myeloma. These analyses were exploratory in nature; they were not prospectively defined in the protocol, and evaluating clinical benefit from the uncontrolled and open setting they were drawn from does not give valuable information. Nevertheless, responding patients (CR, PR) provided evidence of additional clinical benefit including improvement in haemoglobin with decline in transfusion support and improvement in levels of non-myeloma immunoglobulins. Clinical benefit analyses may be confounded by concomitant treatment and procedures, as there were no restrictions on use of erythropoietin or transfusion support during the study. The data was analyzed for clinical benefit in the treatment phase only, data on the duration of these responses in the follow-up period was not collected.
Tumour response cannot be seen as a surrogate marker for clinical benefit but there is support from the literature that a correlation exists between response and survival in relapsed multiple myeloma, and that Blade criteria are a surrogate reasonably likely to predict clinical benefit. The data support the proposal that durable complete remissions, as well as patients with partial response, in response to bortezomib, results in some clinical benefit both in terms of survival and in improvement of other complications of myeloma.
No adequate and well controlled trials have been presented and clinical benefit endpoints cannot be reliably measured without reference to a concurrently studied active comparator agent. Confounding factors in evaluation of safety and efficacy across dose groups included heterogeneity of patients based on types and number of front-line therapy administered, and dose group differences in baseline factors. Results of comparative trials are required to assess the association between response and duration of survival and the true clinical benefit of bortezomib in patients with relapsed and refractory multiple myeloma.
3.3.3 Clinical Safety
Three Phase 1 studies enrolled a total of 123 patients with advanced haematological and solid tumours. The patients were administered bortezomib in unit doses ranging from 0.13 mg/m2 to 2.0 mg/m2 in three different dosing regimens.
Data from the Phase 1 studies showed that 54% of the patients discontinued; 37% due to progressive disease which was lower in the >1.3 mg/m2 group then in the <0.7 mg/m2 group, and 21% due to adverse events or toxicity. A dose-relationship was seen with the incidence of adverse events leading to discontinuation; 7% at <0.7 mg/m2, 16% at 0.7 mg/m2-1.3 mg/m2 and 37% at >1.3 mg/m2 with the most common being fatigue (3%) and dyspnea (2%). Dose limiting toxicities (DLT) consisted of diarrhea and peripheral sensory neuropathy at 1.56 mg/m2 when administered at the same regimen as in the Phase 2 trials. Thrombocytopenia, nausea, constipation, diarrhea, vomiting, and headache all increased with dose. In 37% of the patients at least one serious adverse event was reported, mostly pyrexia at 5%, dyspnea and diarrhea at 4%, abdominal pain and hypotension non-organ specific (NOS) at 2%. The incidence of serious adverse events increased with dose. Serious adverse events consisted of respiratory failure, pneumonia, dehydration, portal vein thrombosis, sepsis and dehydration, increased bilirubin, infection and ileus with peripheral neuropathy, ascites, cases of pulmonary embolism, pericardial effusion, cardiac arrest and hypotension, abdominal pain and bowel obstruction, impaction, pleural effusion, peripheral neuropathy, acute renal failure, sepsis and deep venous thrombosis. There was a case of possible neurogenic postural hypotension for a patient with severe syncope and orthostatic hypotension. Another patient had severe generalized sensorimotor axonal polyneuropathy, small bowel obstruction and a case of severe respiratory distress that caused death after the study.
Two Phase 2 studies (M34100-024 and M34100-025) enrolled 256 patients with multiple myeloma, 28 were in the 1.0 mg/m2 dose group and 228 in the 1.3 mg/m2 dose group. All were assessed for safety.
The data from the Phase 2 studies showed consistency with the Phase 1 studies with regards to the target organs of toxicity, as well as, dose relationships with adverse events. The lower dose, 1.0 mg/m2, was more tolerable than the 1.3 mg/m2 dose; 39% of the patients at 1.3 mg/m2 completed the study compared to 67% in the 1.0 mg/m2 dose group, and 23% of the patients at 1.3 mg/m2 discontinued secondary to adverse events compared to 11% of the patients at 1.0 mg/m2. Increased exposure to bortezomib was associated with increased incidence rates of several of the most common reported events (constipation, pyrexia, dyspnea, dizziness, depression, anxiety, rigors, hypoesthesia and pain), with increased serious adverse events and discontinuations. The following adverse events were reported more frequently (≥10%) in the 1.3 mg/m2 dose group (N=228) than in the 1.0 mg/m2 group (N=28): nausea (64% vs 46%), diarrhea (51% vs 25%), constipation (43% vs 32%), thrombocytopenia (42% vs 32%), vomiting (36% versus 14%), anorexia (32% vs 11%), peripheral neuropathy NOS (30% vs 18%), neutropenia (23% vs 11%), and dehydration (18% versus 0%).
In Study M34100-025, where most patients were treated with the proposed dosage of 1.3 mg/m2 in the first cycles, the most common causes of discontinuation were: peripheral neuropathy (4%); thrombocytopenia (4%); diarrhea and disease progression (3%); and dehydration and syncope (2%). Results showed that 68% of patients had at least one Grade 3 adverse event with the most common being thrombocytopenia (28%); fatigue (12%); peripheral neuropathy (12%); and neutropenia (11%). The most common Grade 3 gastrointestinal adverse events were vomiting (8%), diarrhea (7%), and nausea (6%).9 At least one dose was held back in 64% of the patients because of an adverse event (thrombocytopenia 17%, neutropenia/decreased neutrophil count 14%, peripheral neuropathy 10%, and nausea 8%).
Grade 4 events occurred in 14% of the patients treated with 1.3 mg/m2 and serious adverse events in 50% (39% with 1.0 mg/m2). The most common Grade 4 events with the 1.3 mg/m2 dose were thrombocytopenia (3%) and neutropenia (3%). The most common serious events were pyrexia (7%), pneumonia (7%), diarrhea (6%), vomiting (5%), dehydration (5%) and nausea (4%), with adverse events leading to discontinuation in 18% of the patients (11% at 1.0 mg/m2). The most commonly reported adverse events leading to discontinuation were peripheral neuropathy (5%), thrombocytopenia (4%), diarrhea (2%) and fatigue (2%). It is important to note that 40-50% of all doses at 1.3 mg/m2 were held or reduced. Of the 98% that received a starting dose of 1.3 mg/m2, only 28% received this dose throughout the study, while 33% had dose reductions.
In both Phase 2 studies, 11 patients (5%) died within 20 days after the last bortezomib dose (or any time after if considered to be related to bortezomib). Most died of disease progression but two patients died of drug-related incidences; one of cardiopulmonary arrest 9 days after the last dose and the other of respiratory failure secondary to diaphragmatic failure due to myopathy 39 days after the last dose. There were no clinically relevant differences in Grade 3 or 4 events, serious adverse events, or discontinuations noted by age, gender, race, and body surface area. The incidence rates of Grade 3 or 4 events, serious adverse events or discontinuation due to adverse events was not higher among patients with higher levels of proteasome activity inhibition.
9 The criteria (CTCAE) for the grading of adverse events for oncology clinical trials are available from the National Cancer Institute website: http://ctep.cancer.gov/reporting/ctc.html.
Adverse Events
Note: The following results were taken from the two Phase 2 studies (M34100-024 and M34100-025) unless specified otherwise. The adverse events were separated by dosage (1.0 mg/m2 and 1.3 mg/m2) with the majority in the 1.3 mg/m2 dose group.
Gastrointestinal Events: Results showed 88% of the patients had at least one treatment-emergent adverse event (nausea, diarrhea, constipation, vomiting), 21% had Grade 3 or 4 events, and 13% of the patients (all were in the 1.3 mg/m2 dose group) had serious adverse events. The most common event was nausea (64%), diarrhea (51%), constipation (43%), vomiting (36%), dyspepsia (13%) and abdominal pain NOS (13%). The mechanism is unknown. These events had an early onset and continued for several cycles before resolution, with a trend towards increased incidence with dose. Cases of ileus were also noted.
Metabolism and Nutrition Disorders: Results showed 63% of the patients had a treatment-emergent adverse event in this category and 18% had Grade 3 or 4 events. The most common event was anorexia and decreased appetite NOS (43%), dehydration (18%, Grade 3 in 7%, serious in 5%), and cases of Grade 4 hypercalcemia and hyperuricemia.
Cardiac Disorders: The incidence of cardiac disorders was 20%; 4% of the patients had serious adverse events, 5% of the patients had Grade 3 or 4 events, including severe cardiac heart failure (acute development or exacerbation), pulmonary edema, amyloidosis, cardiac arrest and cardiorespiratory arrest, ventricular extrasystoles, pulmonary embolism, atrial fibrillation, peripheral embolism, bipedal edema, severe sinus and ventricular tachycardia, and ventricular arrhythmias.
Hypotension (orthostatic and postural): Results showed 12% of the patients had hypotension (NOS, orthostatic and postural) of which half had pre-existing hypertension. Grade 3 events occurred in 4% of the patients. The mechanism for hypotension is currently unknown but it could be associated with autonomic neuropathy. One third of the patients with hypotension also had concurrent peripheral neuropathy. Ten patients (4%) had a concurrent syncopal episode with their hypotension.
Syncope: Severe syncope was reported for 17 patients in the 1.3 mg/m2 dose group, 13 patients had Grade 3 events, 4 patients discontinued because of syncope, and 3 patients needed dose modifications, many without underlying cardiovascular reserve problems or dehydration.
Autonomic Neuropathy: One patient was diagnosed with autonomic neuropathy and another with dysautonomia. An association of peripheral neuropathy with syncope or dizziness was noted. Peripheral neuropathy was reported as a serious event in 6 patients, including 4 who had to be hospitalized for the event, of which 3 had concurrent syncope or dizziness. In view of this association and the incidence of hypotension, syncope, and gastrointestinal adverse effects with the mechanism of action being unknown; drug induced autonomic neuropathy is strongly suspected. Special precautions should be taken in patients with increased risk of hypotension (dehydration, antihypertensive medication, history of syncope). A cautionary statement on this issue has been incorporated in the Product Monograph, and the sponsor has planned a study to assess the possibility of autonomic neuropathy.
Nervous System Disorders: Results showed 73% of the patients had at least one treatment-emergent nervous system disorder; 25% had Grade 3 or 4 events (2 patients with Grade 4). The nervous system disorders included peripheral neuropathy NOS (30%), headache NOS (28%, 4% Grade 3), dizziness (21%), paresthesia (12%), hypoesthesia (11%), mental status changes and confusion, emotional disturbance, irritability, restlessness and mood swings, anxiety, insomnia, dizziness and syncope, and rarely abnormal gait, haemorrhagic stoke, and cranial palsy. Most cases of peripheral neuropathy were sensory but cases of sensorimotor were noted. A low incidence of seizures was also reported.
Peripheral Sensory Neuropathy: In the Phase 1 study, peripheral sensory neuropathy was a dose-limiting toxicity. There was a linear increase in estimated probability as a function of cumulative dose. In the 1.3 mg/m2 dose group, 34% of the patients reported symptoms of neuropathy as "quite a bit" or "very much", and 18% had difficulty walking. In a neurological exam, 85% of the patients at 1.3 mg/m2 reported sensory or motor symptoms and 51% of the patients had functional impairment associated with the symptoms reported. The overall incidence of peripheral neuropathy as a serious adverse event was more prominent in patients with a baseline condition; the incidence of Grade 3 events was much lower if there was no peripheral neuropathy at baseline. Reversibility was only demonstrated in 14% of the patients who had more severe symptoms. Reversal of toxicity in most target organs was observed following recovery with exception of axonal degeneration of the sciatic nerve. It is unknown if the potential for similar results exist in other nerves and this safety concern has been incorporated in the Product Monograph. The mechanism of Velcade-induced peripheral neuropathy is unknown and the complete time course of this toxicity is not fully characterized. Careful risk/benefit assessment is necessary if there is pre-existing severe neuropathy.
Asthenic Conditions: Results showed that 65% of the patients had at least one asthenic condition; 18% had Grade 3 or 4 events, 52% had fatigue.
Blood and Lymphatic System Disorders: Results showed no evidence of cumulative bone marrow toxicity; myelosuppression was uncommon. The most common haematological toxicity was thrombocytopenia. Results showed that it was dose-related. Thrombocytopenia occurred early during the dosing period (days 1-11) and the platelet count returned to baseline during the rest period of the treatment cycle (days 12-21). Thrombocytopenia occurred in 65% of the patients at the 1.3 mg/m2 dose (46% at 1.0 mg/m2); 27% of the patients at the 1.3 mg/m2 dose had Grade 3 events (32% at 1.0 mg/m2), 3% had Grade 4 events, and 2% were classed as serious. Serious haemorrhagic complications were reported, both gastrointestinal and intracerebral. The possibility of these conditions has been clearly identified in the Product Monograph. Patients with risk factors for bleeding should be monitored carefully and prophylactic treatment should be considered in view of potential important clinical sequelae. Other common blood disorders were anemia (30% of the patients, Grade 3 in 9%) and neutropenia (23% of the patients, Grade 3 in 13%).
Infections: Results showed 61% of the patients had at least one treatment-emergent adverse event in this category; 13% had a Grade 3 or 4 event.
Pyrexia (>38): Results showed 36% of the patients had elevated body temperatures; 4% had Grade 3 or 4 events, 7% were classed as serious, 13% were from a clinically significant infection. Autonomic dysfunction cannot be excluded as one of the underlying mechanisms.
Musculoskeletal Disorders: Results showed 64% of the patients had at least one treatment-emergent adverse event in musculoskeletal and connective tissue disorders; 21% were Grade 3, 5% serious, 3% led to discontinuation. Common adverse events were arthralgia, pain in limb, bone pain, back pain, muscle cramps, and myalgia.
Electrolyte Imbalance: The mechanism for electrolyte imbalance is unknown. In the Phase 1 study, hyponatremia was a dose-limiting toxicity. Hyponatremia was in 8% of the patients; 5% had Grade 3. Hypokalemia was in 7% of the patients; 2% had Grade 3. Hyperkalemia was reported in 4% of the patients. Hypomagnesemia and hypophosphatemia were also reported.
Respiratory/Thoracic Disorders: Results showed 57% of the patients had at least one treatment-emergent adverse event in this category. The most common was dyspnea in 22% of the patients; 7% had Grade 3 or 4 events, 8 patients had severe or life-threatening, and 2 patients had respiratory failure. The incidence of dyspnea NOS increased with the age categories and differed by >10%. Coughing was also reported frequently (17%) and rare cases of hemoptysis and pleural effusion were observed.
Renal and Urinary Disorders: Results showed 23% of the patients had one treatment-emergent adverse event; 5% had a Grade 3 or 4 event, and 4% had renal failure NOS. Creatinine abnormalities with treatment were noted in 29% of the patients; Grade 3 in 3%, severe or life-threatening renal failure in 4 patients.
Immuno Complex-Mediated Reactions: One patient with multiple myeloma in the Phase 1 study at a dose of 1.04 mg/m2 had a immuno complex-mediated hypersensitivity reaction, a serum sickness reaction with rash and dyspnea as a serious adverse event. In the Phase 2 study, one patient had diffuse polyarthritis and rash, and one case of acute renal failure.
Tumour Lysis Syndrome: The patients at risk of tumour lysis syndrome are those with high tumour burden prior to treatment. These patients should be monitored closely and appropriate precautions taken.
Special Populations
Renal Impairment: The studies excluded patients with severe renal impairement, and the 6 patients that had creatinine clearances of ≤30mL/min in the 1.3 mg/m2 dose group experienced severe adverse events. Patients with mild to moderate renal impairment had similar rates of adverse events, but the incidence of serious adverse events increased with decreased renal function. The most common events were neutropenia, and renal and urinary disorders.
Hepatic Impairment: Conclusions on the safety profile of bortezomib in patients with liver impairment are not available as patients with abnormal liver function were excluded from the Phase 2 studies. Bortezomib is metabolized by the liver, therefore the clearance may be decreased in patients with liver function abnormalities. These patients should be treated with extreme caution and be monitored for toxicity. A cautionary statement on this issue has been incorporated in the Product Monograph.
Reproduction: Fertility studies were not performed. Animal toxicity studies with rats showed degenerative effects in the ovary and testes.
Pregnant Women and Nursing Mothers: Studies did not include pregnant or lactating women.
Pediatrics and Adolescents: The safety and effectiveness of bortezomib in children and adolescents have not been established.
Diabetic Patients: Diabetic patients on oral hypoglycemic agents experienced hypoglycemia and hyperglycemia on bortezomib. Close monitoring of glucose levels is therefore recommended.
Elderly Patients: Patients who were >65 years of age (35% of the patients) experienced a lower response rate and higher incidence of Grade 3-4 adverse events (74% ≤50 to 85% >65), mostly anorexia, dehydration, hypotension, cardiac disorders (congestive heart failure) and respiratory disorders (dyspnea).
Drug-Drug Interactions: Concomitant use with medications that are inhibitors or substrates of 2D6 and 3A4 isozymes were recorded, although no formal drug interactions studies were conducted. The inhibitors included amiodorane, cimetidine, and ritonavir and the substrates were amiodarone, codeine, dexamethasone, odansetron, haloperidol, lidocaine and propanolol. The incidence of Grade 3 or 4 adverse events was greater for patients receiving substrates (89% versus 78%) and patients receiving inhibitors of these isoenzymes had higher Grade 3, 4 or serious adverse events (89% versus 73% and 60% versus 45% for serious). Nausea, vomiting, anorexia, constipation, weakness, thrombocytopenia and neutropenia seem higher in patients receiving substrates and anorexia, insomnia and dyspepsia were more common if inhibitors of 2D6 3A4 were co-administered.
Conclusion
The safety experience in patients with multiple myeloma is limited and additional toxicities are likely to be observed upon wider use of this agent. Limitations of the safety database are based upon exclusion criteria in the trials: hepatic dysfunction, severe renal dysfunction, severely debilitated patients, recent myocardial infarction, hypercalcemia, pregnancy, lactation and pediatric age group. There were no formal pharmacokinetic or pharmacodynamic studies in these sub-groups of patients. The Warning and Precautions section of the Product Monograph provides guidance on the use of Velcade* in these special populations with the exception of those experiencing debilitation, myocardial infarction, and hypercalcemia. Additional precautions may be needed in these populations, as determined by the administering physician.
3.3.4 Issues Outstanding
Under the NOC/c policy, the following clinical commitments were required of the sponsor:
- Provide complete study reports of Study M34101-039, "An International, Multi-Center, Randomized, Open-label Study of Velcade* Versus High-Dose Dexamethasone in Patients with Relapsed or Refractory Multiple Myeloma.
- Include monitoring and follow-up of adverse events from Study M34101-039 as well as events noted in post-marketing data (cardiovascular, seizures) and major safety concerns identified (neurological, psychiatric, immunological).
3.4 Benefit/Risk Assessment and Recommendation
3.4.1 Benefit/Risk Assessment
The efficacy of bortezomib was evaluated in an open-label, single-arm study (M34100-025) with 202 patients, 91% with relapsed and refractory multiple myeloma, who had received at least two prior lines of treatment and who were progressing on their most recent treatment. The goal of treatment in multiple myeloma is to reduce paraprotein levels; greater reductions are associated with clinical benefits. In this study, benefits were seen with response rates, time to progression (TTP), and survival. TTP in response to bortezomib compared to TTP on last therapy (using patients as their own controls) revealed a 4-fold increase in response to bortezomib for responders whereas a reversal of pattern is typically observed with shortening of response duration with subsequent treatments. However, this analysis must be interpreted cautiously, being unplanned and not pre-specified by study protocol, and compared in certain cases to TTP on experimental therapy of unconfirmed efficacy. Study results also showed additional clinical benefits including improved haemoglobin, normalization of non-myeloma immunoglobulins and quality of life (QOL) in patients with complete and partial responses to bortezomib.
Information on survival and TTP in relapsed and refractory multiple myeloma patients is sparse. However, survival benefit resulting from responses is observed in a review of published randomized studies. Study M34100-025 also revealed an association between a higher response rate and increased survival; 80% of the responding patients were alive at 16 months while the median survival for non-responders was 8 months which is the expected survival for relapsed and refractory patients.
The safety of bortezomid therapy has been evaluated in 256 patients in Phase 2 clinical trials. Most adverse events were reversible and manageable, with fatigue, nausea, diarrhea, and constipation being the most common. Peripheral neuropathy (predominantly sensory, but including some cases of sensorimotor neuropathy) was dose-related, cumulative, and often irreversible (full reversibility was documented for only 14% of patients with severe symptoms). Some events of autonomic neuropathy were also reported.
The efficacy at the 1.3 mg/m2 dose was marginally better compared to the 1.0 mg/m2 dose but the difference was not statistically significant and it was difficult to quantify. When evaluating the 98% of the patients in the 1.3 mg/m2 dose group, only 28% kept receiving this dose throughout the study and 33% had dose reductions. In both studies (Study M34100-025 and Study M34100-024), the mean total dose administered across all patients was 80.0% of the expected dose in each treatment cycle, equivalent to 1.04 mg/m2 for a planned 1.30 mg/m2 dose.
Response rate was independent of the dose reductions. In Study M34100-025, of the 76 patients that were treated with bortezomib alone and had dose reductions for adverse events, 32 (42%) had a response to treatment. It is worthy to note that the mean and median total dose and duration of treatment were higher with the 1.0 mg/m2 dose in patients who required dose reductions to manage toxicity. The 1.3 mg/m2 dose clearly increased toxicity but no clear evidence for the response rate was evident. There is insufficient data on efficacy at 1.0 mg/m2, which is a non-measurable effect with no surrogate markers of efficacy (such as pharmacodynamic or kinetic measurements), but the dose is more tolerable. Additional clinical trials are encouraged to further explore the relative safety and efficacy of the 1.0 mg/m2 and 1.3 mg/m2 doses. The benefit claimed also needs to be confirmed by comparative trials to better assess the association between response and duration of survival and the clinical benefit of bortezomib in this population, considering the size of the benefit and the degree of homogeneity of the population of patients studied.
The risk of rare events not yet observed versus the risk of no treatment also has to be weighed. Bortezomib does exhibit some efficacy in patients with relapsed and refractory multiple myeloma but there are significant risks involved. Many of the serious adverse events have an unknown mechanism of action and therefore makes it difficult to identify the population at risk. Nevertheless, in view of the poor prognosis for patients with relapsed and refractory multiple myeloma who have failed at least two standard therapies, the benefit of treatment with bortezomib outweighs the risk in this patient population. The risk/benefit ratio does not justify this therapy taking precedence over existing standard therapies that carry much less risk as first and second line agents but bortezomib does seem to be acceptable as a third-line agent.
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 Velcade is favourable in the treatment of multiple myeloma patients who have relapsed following front-line therapy and are refractory to their most recent therapy.
This New Drug Submission (NDS) qualifies for authorization under the NOC/c policy. The NDS 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.
Consistent with the NOC/c policy, the sponsor has agreed to the following commitments:
- To provide complete study reports of Study M34101-039, An International, Multi-Center, Randomized, Open-label Study of Velcade Versus High-Dose Dexamethasone in Patients with Relapsed or Refractory Multiple Myeloma.
- To include monitoring and follow-up of adverse events from Study M34101-039 as well as events noted in post-marketing data (cardiovascular, seizures) and major safety concerns identified (neurological, psychiatric, immunological).
4 Submission Milestones
Submission Milestones: Velcade
Submission Milestone | Date |
---|---|
Acceptance for advanced consideration under the NOC/c Policy: | 2004-02-20 |
Screening | |
Screening Acceptance Letter issued: | 2004-03-17 |
Review | |
Quality Evaluation complete: | 2004-11-18 |
Clinical Evaluation complete: | 2004-11-05 |
Labelling review complete: | 2004-12-10 |
NOC/c-QN issued: | 2004-12-13 |
Response filed: | 2004-12-21 |
NOC/c issued by Director General: | 2005-01-27 |
Related Drug Products
Product name | DIN | Company name | Active ingredient(s) & strength |
---|---|---|---|
VELCADE | 02262452 | JANSSEN INC | BORTEZOMIB (BORTEZOMIB MANNITOL BORONIC ESTER) 3.5 MG / VIAL |