throbber
24 June 2010
`EMA/CHMP/450053/2010
`
`Evaluation of Medicines for Human Use
`
`
`
`
`CHMP assessment report
`
`
`Ruconest
`
`
`
`International Nonproprietary Name: conestat alfa
`
`
`
`Procedure No. EMEA/H/C/001223
`
`
`
`
`
`Assessment Report as adopted by the CHMP with
`all information of a commercially confidential nature deleted.
`
`An agency of the European Union
`
` 7
`
` Westferry Circus ● Canary Wharf ● London E14 4HB ● United Kingdom
`Telephone +44 (0)20 7418 8400 Facsimile +44 (0)20 7523 7455
`E-mail info@ema.europa.eu Website www.ema.europa.eu
`
`
`
`European Medicines Agency, 2010. Reproduction is authorised provided the source is acknowledged.
`
`Page 1 of 57
`
`CSL EXHIBIT 1041
`
`

`

` Table of contents
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Page
`
`1. Background information on the procedure .............................................. 3
`1.1. Submission of the dossier.................................................................................... 3
`1.1.1. For new centralised dossiers orphan medicinal products ........................................ 3
`1.1.2. Information on paediatric requirements .............................................................. 3
`1.1.3. Information relating to orphan market exclusivity ................................................ 3
`1.1.4. Licensing status: ............................................................................................. 4
`1.2. Steps taken for the assessment of the product ....................................................... 4
`2. Scientific discussion ................................................................................ 4
`2.1. Introduction ...................................................................................................... 4
`2.2. Quality aspects .................................................................................................. 6
`2.2.1. Introduction ................................................................................................... 6
`2.2.2. Active substance ............................................................................................. 7
`2.2.3. Finished Product.............................................................................................. 9
`2.3. Non-clinical aspects .......................................................................................... 10
`2.3.1. Introduction ................................................................................................. 10
`2.3.2. Pharmacology ............................................................................................... 11
`2.3.3. Pharmacokinetics .......................................................................................... 11
`2.3.4. Toxicology.................................................................................................... 13
`2.3.5. Ecotoxicity/environmental risk assessment........................................................ 17
`2.3.6. Discussion on non-clinical aspects.................................................................... 17
`2.3.7. Conclusion on the non-clinical aspects .............................................................. 18
`2.4. Clinical aspects ................................................................................................ 18
`2.4.1. Introduction ................................................................................................. 18
`2.4.2. GCP............................................................................................................. 20
`2.4.3. Pharmacokinetics .......................................................................................... 20
`2.4.4. Pharmacodynamics........................................................................................ 22
`2.4.5. Discussion on clinical pharmacology ................................................................. 23
`2.4.6. Conclusions on clinical pharmacology ............................................................... 24
`2.4.7. Clinical efficacy ............................................................................................. 24
`2.4.8. Discussion on clinical efficacy .......................................................................... 41
`2.4.9. Conclusions on the clinical efficacy ................................................................... 43
`2.4.10. Clinical safety.............................................................................................. 43
`2.5. Discussion on clinical safety............................................................................... 48
`2.6. Conclusions on the clinical safety........................................................................ 49
`2.7. Pharmacovigilance............................................................................................ 49
`2.7.1. Detailed description of the pharmacovigilance system......................................... 49
`2.7.2. Risk management plan................................................................................... 49
`2.7.3. Benefit-risk balance ....................................................................................... 54
`2.7.4. Similarity with authorised orphan medicinal products.......................................... 57
`2.7.5. Recommendation........................................................................................... 57
`
`
`
`Page 2 of 57
`
`

`

`1. Background information on the procedure
`
`1.1. Submission of the dossier
`
`The applicant Pharming Group N.V. submitted on 03 September 2009 an application for Marketing
`Authorisation to the European Medicines Agency for Ruconest, through the centralised procedure falling
`within the Article 3(1) and point 4 of Annex of Regulation (EC) No 726/2004. The eligibility to the
`centralised procedure was agreed upon by the Agency/CHMP on 27 April 2006.
`
`
`
`The legal basis for this application refers to:
`
`Article 8.3 of Directive 2001/83/EC, as amended - complete and independent application
`
`The application submitted is a complete dossier composed of administrative information, complete
`quality data, non-clinical and clinical data based on applicants’ own tests and studies
`
`
`
`The applicant applied for the following indication:
`
`Treatment of acute angioedema attacks in adults with hereditary angioedema (HAE) due to C1
`esterase inhibitor deficiency.
`
`1.1.1. For new centralised dossiers orphan medicinal products
`
`The applicant Pharming Group N.V. submitted on 03 September 2009 an application for Marketing
`Authorisation to the European Medicines Agency through the centralised procedure for Ruconest, which
`was designated as an orphan medicinal product EU/3/01/036 on 11 May 2001. Ruconest was
`designated as an orphan medicinal product in the following indication: Treatment of angioedema
`caused by C1 inhibitor deficiency. The calculated prevalence of this condition was approximately 2.1 in
`10,000 EU population.
`
`In connection with the review of the orphan designation criteria by the Committee on Orphan Medicinal
`Products (COMP) at its meeting of 7-8 September 2010, the Applicant requested the Commission to
`remove the product from the Community Register of Orphan Medicinal Products on 9 September 2010.
`
`1.1.2. Information on paediatric requirements
`
`Pursuant to Article 7, of Regulation (EC) No 1901/2006 the application included an Agency Decision
`P/132/2009 for the following condition:
`
`Hereditary angioedema
`
`on the agreement of a paediatric investigation plan (PIP).
`
`The PIP is not yet completed.
`
`1.1.3. Information relating to orphan market exclusivity
`
`1.1.3.1. Similarity
`
`Pursuant to Article 8 of Regulation (EC) No. 141/2000 and Article 3 of Commission Regulation (EC) No
`847/2000, the application contained a critical report addressing the possible similarity with authorised
`orphan medicinal products.
`
`Page 3 of 57
`
`

`

`1.1.3.2. Protocol assistance
`
`The applicant received Protocol Assistance from the CHMP on 21 November 2003. The Protocol
`Assistance pertained to quality, non-clinical and clinical aspects of the dossier.
`
`1.1.4. Licensing status:
`
`The product was not licensed in any country at the time of submission of the application.
`
`The Rapporteur and Co-Rapporteur appointed by the CHMP and the evaluation teams were:
`
`Rapporteur:
`
`Ian Hudson
`
` Co-Rapporteur:
`
`Kristina Dunder
`
`1.2. Steps taken for the assessment of the product
`
`•
`•
`•
`
`•
`
`•
`
`The application was received by the Agency on 03 September 2009.
`The procedure started on 23 September 2009.
`The Rapporteur's first Assessment Report was circulated to all CHMP members on 11 December
`2009. The Co-Rapporteur's first Assessment Report was circulated to all CHMP members on 11
`December 2009.
`• During the meeting on 18-21 January 2010, the CHMP agreed on the consolidated List of Questions
`to be sent to the applicant. The final consolidated List of Questions was sent to the applicant on
`21 January 2010.
`The applicant submitted the responses to the CHMP consolidated List of Questions on 18 March
`2010.
`The Rapporteurs circulated the Joint Assessment Report on the applicant’s responses to the List of
`Questions to all CHMP members on 30 April 2010.
`• During the CHMP meeting on 17-20 May 2010, the CHMP agreed on a list of outstanding issues to
`be addressed in writing by the applicant.
`The applicant submitted the responses to the CHMP consolidated List of Outstanding Issues on
`24 May 2010.
`The Rapporteurs circulated the Joint Assessment Report on the applicant’s responses to the List of
`Outstanding Issues to all CHMP members on 7 June 2010.
`• During the meeting on 21-24 June, the CHMP, in the light of the overall data submitted and the
`scientific discussion within the Committee, issued a positive opinion for granting a Marketing
`Authorisation to Ruconest on 24 June 2010. The applicant provided the letter of undertaking on the
`follow-up measures to be fulfilled post-authorisation on 23 June 2010.
`• On 9 September 2010 the applicant requested the Commission to remove the product from the
`Community Register of Orphan Medicinal Products.
`
`•
`
`•
`
`For new centralised dossiers orphan medicinal products
`
`The CHMP adopted a report on similarity of Ruconest with Firazyr on 21 January 2010.
`
`•
`
`Note: The product was previously known as Rhucin.
`
`
`
`2. Scientific discussion
`
`2.1. Introduction
`
`C1 inhibitor (C1INH), a serine proteinase inhibitor (serpin), is primarily synthesized in the liver and the
`normal range of C1INH activity in the general population is 0.7 to 1.3 U/mL (70 to 130%). The main
`function of C1INH is inhibition of several complement proteinases and contact-system proteinases.
`
`Hereditary angioneurotic oedema (HAE) is characterized by recurrent, often unpredictable, acute
`attacks of soft tissue swelling (angioedema). Acute angioedema attacks in HAE patients impair the
`quality of life, and can be fatal if the angioedema swelling occurs in the throat. An untreated attack can
`persist for up to five days. Attacks of oedema of the gastrointestinal tract are associated with severe
`
`Page 4 of 57
`
`

`

`pain similar to acute abdominal syndromes and may cause nausea, vomiting, diarrhoea, ascites and
`symptoms of hypovolemia.
`
`Two types of congenital functional C11NH deficiency (phenotypic variants) can be distinguished (HAE
`Type I and HAE Type II). Both types are autosomal dominant disorders and the levels of functional
`C11NH in plasma are below 50% of normal
`levels. The median plasma level of C11NH activity in
`patients with HAE is about 0.2 U/mL, or 20% of the level found in healthy individuals
`
`Key inflammatory mediators regulated by C1INH of concern for patients with HAE include activated
`proteases of the complement system such as C1r, C15 and Mannan Binding protein (MBP)-associated
`proteinases (MASPs), and factor XIIa, factor XIa and kallikrein of the contact system (Figure 1). Over
`activity of these inflammatory proteases is thought to lead to the generation of vasoactive peptides
`such as bradykinin that mediate angioedema attacks.
`
`Negatively
`
`charged
`
`
`Contact system
`
`surface
`
`FXII —’ FXIIa
`
`Complement system
`
`Prekallikrein ‘ C1
`
` C42
`Kallikrein
`c1 rs L.
`
`
`Wen_ ’
`
`l
`BK2R
`
`W
`
`
`Fibrinolytic system
`
`Plasmin
`
`Figure 1. Schematic picture showing the C1INH mechanism of action.
`
`The current available treatments of acute attacks of HAE include:
`
`. Human C11NH preparations, which are purified and pasteurized concentrates from pooled
`human plasma (one approved in most EU member states via the Mutual Recognition
`Procedure, one is approved in the Netherlands);
`Icatibant (approved through the centralised procedure), which acts as a selective competitive
`antagonist at the bradykinin type 2 (32) receptor.
`
`.
`
`Conestat alfa, the active substance of Ruconest, a recombinant human component 1 (C1) esterase
`inhibitor (rhC1INH),
`is the recombinant analogue of human C1 esterase inhibitor (C1INH), and is
`obtained from the milk of rabbits expressing the gene encoding for human C11NH. The transgenic
`rabbits, which are genetically modified organisms (GMO), are maintained in specific pathogen free
`enclosed housings. The product, however,
`is not a GMO. The availability of a non-blood product
`derived C11NH for treatment of acute attacks of HAE provides a further treatment option for patients.
`
`The applicant Pharming Group N.V. submitted a complete and independent application for Marketing
`Authorisation to the European Medicines Agency for Ruconest (previously known as Rhucin) for
`treatment of acute angioedema attacks in adults with hereditary angioedema (HAE) due to C1 esterase
`inhibitor deficiency.
`
`Ruconest was designated as an orphan medicinal product in the EU with the following orphan indication:
`treatment of angioedema caused by C1 inhibitor deficiency. HAE was considered as chronically
`debilitating conditions, characterised by acute and repetitive attacks, which might be life-threatening.
`The calculated prevalence of this condition at the time or orphan medicinal product designation was
`
`CHMP assessment repout
`EMA/CHMP/450053/2010
`
`Page 5 0f 57
`
`Page 5/57
`
`Page 5 of 57
`
`

`

`2.1 per 10,000 EU population. The significant benefit at the time of designation was based on major
`contribution to patient care with regards to currently authorised medicinal products on the basis of a
`source of non-blood derived C1INH.
`
`In connection with the review of the orphan designation criteria by the Committee on Orphan Medicinal
`Products (COMP) at its meeting of 7-8 September 2010, the Applicant requested the Commission to
`remove the product from the Community Register of Orphan Medicinal Products on 9 September 2010.
`
`The applicant received Protocol Assistance from the CHMP on the quality, preclinical and clinical
`development. There are no guidelines on the evaluation of medicinal products for the treatment of
`acute HAE attacks. The primary and secondary endpoints used in the two RDCT 1205 and 1304 are
`mainly in line with the Protocol Assistance given by the CHMP.
`
`With regard to the paediatric development, the applicant has agreed to generate data in paediatric
`patients aged 2-18 years; these data are not yet available and will need to be provided post-
`authorisation. Studies in patients under 24 months are not requested.
`
`This is the second marketing authorisation application (MAA) submitted for this medicinal product. The
`first MAA was submitted in July 2006 and received a negative opinion on the basis of the limited clinical
`database and concerns regarding severe allergic reactions and the potential for immunogenicity
`following repeated administrations. Additional clinical data for the present application for the present
`MAA was submitted with two finalised placebo-controlled studies and data from the ongoing extension
`phases of the two open-label studies. The current data also specifically addresses immunogenicity,
`efficacy in laryngeal attacks, and the possibility of thrombogenic potential. Furthermore, a new dose
`regimen was proposed.
`
`The applicant initially applied for the following indication:
`“Ruconest is indicated for treatment of acute angioedema attacks in patients with hereditary
`angioedema (HAE) due to C1 esterase inhibitor deficiency”
`
`The finally approved indication is as follows:
`“Ruconest is indicated for treatment of acute angioedema attacks in adults with hereditary angioedema
`(HAE) due to C1 esterase inhibitor deficiency.”
`
`The product is administered by intravenous injection. The posology is one single dose of 50 U/kg body
`weight for adults up to 84 kg body weight or 4200 U for adults of 84 kg body weight and above.
`
`2.2. Quality aspects
`
`2.2.1. Introduction
`
`C1 inhibitor (C1INH) is a serine protease inhibitor belonging to the serpin superfamily. The active
`substance of Ruconest is a recombinant analogue of human C1INH (rhC1INH, INN: conestat alfa) that
`is purified from the milk of rabbits expressing the gene encoding for human C1INH. It is a plasma
`single-chain glycoprotein containing 478 amino acids with six sites of N-glycosylation and at least
`seven sites of O-glycosylation. It has a molecular mass of approximately 67,000 Da of which
`approximately 22% is due to oligosaccharides. The amino acid sequence has been provided and
`includes two disulphide bonds (between cys101- cys406 and cys108 - cys183).
`
`The drug product is presented as a powder for solution for injection. It is a sterile, non-pyrogenic,
`preservative-free, white to off-white lyophilized powder contained in a single-use type I, colourless
`sealed glass vial. The product is to be reconstituted with 14 mL sterile water for injections (WFI) before
`intravenous injection. Each vial contains 2100 U of rhC1INH (150 U/mL after reconstitution). The
`excipients used in the formulation are sodium citrate, sucrose and citric acid.
`
`Page 6 of 57
`
`

`

`2.2.2. Active substance
`
`2.2.2.1. Manufacture
`
`Generation of the transgenic herd
`The genomic DNA fragment containing the C1 inhibitor gene and flanking regions was isolated from a
`P1 phage clone. The promoter regions are derived from the casein sequences since the caseins are the
`predominant milk proteins.
`
`Microinjection of DNA into a fertilized oocyte and transfer of the embryo into a foster mother led to the
`generation of a transgenic rabbit (Generation F0). This transgenic male animal was selected as the
`founder, establishing a transgenic line. The founder line was selected on the basis of expression level
`of C1INH in milk, gene copy number, site of integration and number of integration sites. The suitability
`of the selected line was determined by monitoring stability of expression throughout lactation, stability
`of transmission of the transgene, health and fertility of the rabbits Following breeding with a non-
`transgenic female, an F1 male was selected for genetic characterization and sperm collection to
`establish the Master Transgenic Bank (MTB). From the MTB, transgenic bucks were generated and
`genetically characterized. The selected bucks were then used to establish a Manufacturing Working
`Transgenic sperm Bank (MWTB).
`
`The development genetics has been fully discussed with relevant information provided about gene
`construction and identity, copy number, integration site and stability. Similarly, the information
`provided on the establishment, maintenance and pathogen safety of the transgenic line of rabbits is
`satisfactory. A two tier sperm bank has been established and production is limited to transgenic F4
`female New Zealand White rabbits, therefore preventing the possibility of genetic drift.
`
`Manufacture of milk starting material
`The manufacture of the milk starting material includes breeding, maintenance and milking of
`transgenic rabbits.
`
` A
`
` production rabbit colony is a group of rabbits of defined and tested genealogy housed in containment
`behind a biosecurity barrier.
`
`After a general health check, the rabbits are milked using a milking machine. Following collection and
`storage, the milk is skimmed by centrifugation and frozen before transfer to storage facilities. Milk
`from individual rabbits may be pooled prior to skimming.
`
`Besides maintenance of the rabbits as “closed” colonies behind a biosecurity barrier, a comprehensive
`health monitoring program for control of production and sentinel animals are used to control the safety
`of the raw material of the skimmed milk. The applied control procedures are considered acceptable.
`Pooling of thawed skimmed milk is adequately described and ensures a consistent starting material for
`the downstream purification. Control of the process is adequate, including in-process controls for the
`milking of the female rabbits as well as specifications for skimmed milk.
`
`Manufacture of formulated drug substance
`The formulated drug substance is manufactured and routinely controlled in compliance with GMP.
`
`The downstream processing of the milk consists of thawing of milk, pooling of milk bags, fat removal
`by centrifugation and a succession of filtration and chromatography steps as well as viral
`inactivation/removal steps. The drug substance
`is
`formulated using ultra-/diafiltration and
`subsequently filtered and filled in a bag for storage.
`
`Validation
`Three consecutive process validation runs at commercial scale have been performed and extended in
`process control tests have been presented for these batches and compared to data collected from pilot
`scale batches. These data have adequately validated the process and demonstrates process
`comparability to pilot scale manufacturing runs. A summary of all batches which have been used in
`pre-clinical and clinical trials has also been presented and comparability of (pre)-clinical product to
`commercial product has been demonstrated by validation data from the process runs and extended
`characterisation of the product.
`
`
`
`Page 7 of 57
`
`

`

`Characterisation
`The product has been extensively characterised, and 87% of the amino acid backbone has been
`sequenced. The sites of the 2 disulphide bridges have been identified and 9 of the reported 13 sites of
`glycosylation have been identified. A quantitative assay for isoform analysis has been developed and
`will be implemented as a batch release assay. A quantitative specification will be set after the assay is
`validated and sufficient batch release data has been obtained. Alongside the characterisation data
`presented in the dossier, this extent of sequence identification is considered to be adequate.
`Monosaccharide composition has been demonstrated to be very consistent. Of the sialic acids, only
`NANA has been identified, the potentially immunogenic NGNA has not been found. The N-linked profile
`has been adopted by the Applicant as a drug substance batch release assay, but the O-linked assay
`has not. Instead, the applicant has successfully argued that rates of sialylation and (O-glycosylation)
`site occupancy are the most important parameters to monitor instead of the O-glycosylation profile and
`a site occupancy assay is currently being validated. Sialylation is already a batch release parameter.
`The Applicant has successfully demonstrated that the product has a very low level of host related
`impurities (milk proteins) and that this parameter is well controlled. Clinical assessment has confirmed
`a very low level of patient antibody formation to HRIs.
`
` Specification
`2.2.2.2.
`
`Specifications for the skimmed milk intermediate and the formulated bulk drug substance are generally
`considered adequate to ensure a good level of control. The analytical methods used are considered to
`be state-of-the-art and have been adequately validated. Each batch of active substance is tested for
`appearance, identity, purity, potency, quantity, excipients, general physicochemical properties and
`contaminants.
`
`The rhC1INH activity assay is based on the principle that C1s activity can be measured using a
`commercially available peptide. C1s cleaves the pNA part of the peptide, which absorbs at a
`wavelength of 405 nm. The amount of released pNA is directly proportional to the C1s activity.
`
`Consistency of the protein composition of the skimmed milk starting material is monitored, and a
`qualitative specification for this assay has been set and will be updated with quantitative specifications.
`To ensure blood proteins have not leaked across the blood/mammary barrier, a specific test has also
`been introduced to monitor skimmed milk. A specification for this assay will be set after 20 batches
`have been analysed.
`
`The applicant has committed to monitoring the O-glycan profile for all batches of drug substance, to
`complete validation of the O-glycan site occupancy assay and to introduce a specification for O-linked
`glycosylation.
`
` A
`
` new primary reference standard, derived from a commercial scale batch has been characterised and
`introduced. The procedure to introduce new reference standards has been provided. This reference
`standard is also used for drug product. It should be noted that International Standards for C1INH
`(plasma and concentrate) are being developed. The Applicant has committed to providing traceability
`to an international standard once established.
`
`Batch release data has been provided to demonstrate compliance of commercial product with the
`specifications.
`
` Stability
`2.2.2.3.
`
`Stability studies under real time, accelerated and stress conditions have been performed and the data
`provided support a drug substance shelf life of 3 years at -20°C.
`
`In accordance with EU GMP guidelines 1 , any confirmed out of specification result, or significant
`negative trend, should be reported to the Rapporteur and the EMA.
`
`
`
`
`1 6.32 of Vol. 4 Part I of the Rules Governing Medicinal Products in the European Union
`
`Page 8 of 57
`
`

`

`2.2.3. Finished Product
`
` Pharmaceutical Development
`2.2.3.1.
`
`Ruconest 2100 U powder for solution for injection is a sterile, non-pyrogenic, preservative-free, white
`to off-white lyophilized powder contained in a single-use type I, colourless sealed glass vial. The
`product is to be reconstituted with 14 mL sterile water for injections (WFI) before intravenous injection.
`The solvent (sterile WFI) for reconstitution is not supplied with the drug product.
`
`Each vial contains 2100 U of conestat alfa (150 U/mL after reconstitution). The excipients used in the
`formulation are sodium citrate, sucrose and citric acid.
`
` A
`
` justification of the formulation components has been provided, along with a description of how the
`formulation was developed. A full description of manufacturing process development has been provided
`which details changes to the process, closure system and fill volume, and relates changes to batch
`numbers and clinical studies.
`
`Initially, liquid formulations were investigated, however it was ultimately decided to have a lyophilised
`presentation as a liquid presentation was considered impractical.
`
`The same composition of drug product has been used throughout clinical development and this is the
`formulation intended for commercial batches.
`
` Adventitious agents
`2.2.3.2.
`
`The rabbit is not considered to be a TSE susceptible species and therefore TSE considerations for the
`rabbit milk are not deemed necessary. Adequate precautions to prevent contamination by TSE from
`alternative sources have been described.
`
`The animals are kept in SPF (specified pathogen free) conditions and are routinely monitored for
`evidence of viral contamination. The manufacturing process has been validated for the inactivation or
`removal of a panel of relevant or model viruses. The two specific virus inactivation/removal steps
`demonstrate excellent capacity for virus depletion with the three chromatography steps also
`contributing to viral safety. A risk assessment has been performed based on the worst case possible
`viral contamination (calculated from the LOD of the in vitro assays and quantity of starting material per
`dose) and validated virus removal capacity of the process.
`
`Overall, and taking all factors into consideration, it is considered that this product should not pose a
`risk to patients through adventitious agents.
`
` Manufacture of the product
`2.2.3.3.
`
`Manufacture of the drug product, including labelling and packaging and batch release, is carried out at
`GMP-qualified sites. Batch release is performed by Pharming Technologies B.V., The Netherlands.
`
`The manufacturing process for the drug product has been described in sufficient detail. Batches of drug
`substance are thawed, pooled, sterile filtered and filled aseptically into vials. The drug product is
`thereafter freeze-dried, capped, inspected, packaged and stored prior to shipment. No other
`components are added in the manufacturing process of the drug product.
`
`The container closure system consists of a type I, colorless glass vial, a siliconized chlorobutyl rubber
`stopper, and a flip-off seal of aluminum and colored plastic.
`
`Critical steps are identified and adequately controlled. The process has been appropriately validated.
`
` Product specification
`2.2.3.4.
`
`The drug product specification is generally relevant and justified, although several limits will be
`updated when 20 batches have been manufactured.
`
`Acceptable batch release data has been presented.
`
`
`Page 9 of 57
`
`

`

` Stability of the product
`2.2.3.5.
`
`Real-time and accelerated stability studies were performed on 4 batches of finished product. In
`addition, data from one batch stored under stress conditions was provided.
`
`Based on the data presented, a shelf-life for the drug product of 36 months at 25°C is considered
`justified.
`
`The applicant also performed studies on photostability and in-use stability of the reconstituted product.
`
`In accordance with EU GMP guidelines 2 , any confirmed out of specification result, or significant
`negative trend, should be reported to the Rapporteur and the EMEA.
`
` GMO
`2.2.3.6.
`
`The transgenic rabbits are considered to be genetically modified organisms (GMO). The manufacturer
`has been authorised by the authorities to handle transgenic rabbits in a contained environment and the
`rabbit housing areas are classified in accordance with GMO regulations. The product itself is not a GMO.
`
` Discussion on chemical, pharmaceutical and biological aspects
`2.2.3.7.
`
`Information on development, manufacture and control of the drug substance and drug product have
`been presented in a satisfactory manner. The results of tests carried out indicate satisfactory
`consistency and uniformity of important product quality characteristics, and these in turn lead to the
`conclusion that the product should have a satisfactory and uniform performance in the clinic.
`
`Conclusions on the chemical, pharmaceutical and biological aspects
`The quality of this product is considered to be acceptable when used in accordance with the conditions
`defined in the SPC. Physicochemical and biological aspects relevant to the uniform clinical performance
`of the product have been investigated and are controlled in a satisfactory way. Data has been
`presented to give reassurance on viral/TSE safety.
`
`At the time of the CHMP opinion, there were a number of minor unresolved quality issues having no
`impact on the Risk-benefit balance of the product. The applicant gave a Letter of Undertaking and
`committed to resolve these as Follow Up Measures after the opinion, within an agreed timeframe.
`
`2.3. Non-clinical aspects
`
`2.3.1. Introduction
`
`The non-clinical testing program was conducted to establish the safety of rhC1INH for short term use
`(< 7 days) in this chronically debilitating and potentially life-threatening disease. Studies presented
`covered in vitro pharmacology, in vivo safety pharmacology, pharmacokinetics, general toxicology with
`dosing of up to two weeks, teratology in rats and rabbits and local tolerance.
`
`Two different pharmaceutical presentations were developed: a liquid formulation of 25 mg/mL and a
`freeze-dried presentation. Liquid presentations were used in early non-clinical studies. However, from
`formulation studies it became apparent that a lyophilized formulation was preferred for stability
`reasons. Later studies were performed with the lyophilized presentations.
`
`Protocol Assistance from CHMP was obtained on the appropriateness of the proposed duration of
`2 weeks for the repeated dose toxicity studies in rats, to support product safety with regard to the
`intended clinical use of recombinant human C1 inhibitor (acute treatme

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket