throbber
mAbs
`
`ISSN: 1942-0862 (Print) 1942-0870 (Online) Journal homepage: https://www.tandfonline.com/loi/kmab20
`
`Developability studies before initiation of process
`development
`
`Xiaoyu Yang, Wei Xu, Svetlana Dukleska, Sabrina Benchaar, Selina Mengisen,
`Valentyn Antochshuk, Jason Cheung, Leslie Mann, Zulfia Babadjanova, Jason
`Rowand, Rico Gunawan, Alexander McCampbell, Maribel Beaumont, David
`Meininger, Daisy Richardson & Alexandre Ambrogelly
`
`To cite this article: Xiaoyu Yang, Wei Xu, Svetlana Dukleska, Sabrina Benchaar, Selina Mengisen,
`Valentyn Antochshuk, Jason Cheung, Leslie Mann, Zulfia Babadjanova, Jason Rowand, Rico
`Gunawan, Alexander McCampbell, Maribel Beaumont, David Meininger, Daisy Richardson &
`Alexandre Ambrogelly (2013) Developability studies before initiation of process development,
`mAbs, 5:5, 787-794, DOI: 10.4161/mabs.25269
`To link to this article: https://doi.org/10.4161/mabs.25269
`
`Copyright © 2013 Landes Bioscience
`
`View supplementary material
`
`Published online: 07 Jun 2013.
`
`Submit your article to this journal
`
`Article views: 2651
`
`Citing articles: 51 View citing articles
`
`Full Terms & Conditions of access and use can be found at
`https://www.tandfonline.com/action/journalInformation?journalCode=kmab20
`
`Lassen - Exhibit 1012, p. 1
`
`

`

` REPORT
`mAbs 5:5, 787–794; September/October 2013; © 2013 Landes Bioscience
`
`REPORT
`
`Developability studies before initiation
`of process development
`Improving manufacturability of monoclonal antibodies
`
`Xiaoyu Yang,1 Wei Xu,1 Svetlana Dukleska,1 Sabrina Benchaar,2 Selina Mengisen,1 Valentyn Antochshuk,3 Jason Cheung,3
`Leslie Mann,1 Zulfia Babadjanova,1 Jason Rowand,1 Rico Gunawan,1 Alexander McCampbell,4 Maribel Beaumont,2
`David Meininger,2 Daisy Richardson1 and Alexandre Ambrogelly1,*
`
`1BioProcess Development; Merck Research Laboratories; Union, NJ USA; 2Discovery Biologics; Merck Research Laboratories Palo Alto; Palo Alto, CA USA;
`3BioProcess Development; Merck Research Laboratories; Summit, NJ USA; 4Molecular biomarkers; Merck Research Laboratories; West Point, PA USA
`
`Keywords: developability, discovery, stability, analytics, process control, development, forced degradation, CQA, QbD
`
`Monoclonal antibodies constitute a robust class of therapeutic proteins. Their stability, resistance to stress conditions
`and high solubility have allowed the successful development and commercialization of over 40 antibody-based drugs.
`Although mAbs enjoy a relatively high probability of success compared with other therapeutic proteins, examples
`of projects that are suspended due to the instability of the molecule are not uncommon. Developability assessment
`studies have therefore been devised to identify early during process development problems associated with stability,
`solubility that is insufficient to meet expected dosing or sensitivity to stress. This set of experiments includes short-term
`stability studies at 2–8°C, 25°C and 40°C, freeze-thaw studies, limited forced degradation studies and determination of
`the viscosity of high concentration samples. We present here three case studies reflecting three typical outcomes: (1) no
`major or unexpected degradation is found and the study results are used to inform early identification of degradation
`pathways and potential critical quality attributes within the Quality by Design framework defined by US Food and Drug
`Administration guidance documents; (2) identification of specific degradation pathway(s) that do not affect potency of
`the molecule, with subsequent definition of proper process control and formulation strategies; and (3) identification of
`degradation that affects potency, resulting in program termination and reallocation of resources.
`
`Introduction
`
`Development of a therapeutic protein is a long and costly process
`that can take over a decade from discovery to commercialization.
`Although therapeutic biologics generally have a higher probabil-
`ity of success than their small molecule counterparts,1 the rate of
`attrition remains substantial. While decisions to terminate proj-
`ects can be based on the competitive landscape and commercial
`opportunities, projects are also terminated for technical reasons,
`including unsuitable safety profile, lack of efficacy in human,
`instability of the molecule or formulation, poor expression and
`purification issues. To reduce the risk associated with a given
`project, the sooner these technical challenges are identified, the
`sooner appropriate control strategies can be put in place. When
`irreducible challenges are identified, the project can be termi-
`nated and resources diverted to the development of a more prom-
`ising molecule. Derisking process development early enough is
`critical to success because challenges not identified early enough
`may lead to expensive and time-consuming remediation steps
`later in the development of a given program. Determination of
`the safety profile is usually performed during preclinical studies
`
`and Phase 1 clinical studies in human, which is well into the
`development path. Likewise, the true assessment of a molecule
`efficacy is obtained during Phase 2/3 clinical studies.
`In contrast with safety and efficacy assessments, a number
`of technical hurdles can be evaluated early during development
`programs. To identify technical challenges, we devised a compre-
`hensive set of developability experiments and applied the strategy
`to a number of monoclonal antibodies (mAbs) currently in our
`pipeline. This strategy includes a series of in silico and experi-
`mental approaches that we grouped under the Developability
`Assessment term. In silico work includes structure sequence
`analysis based on primary sequence alignment and molecular
`model; prediction of possible degradation pathways is based on
`prior experience and on published literature. This constitutes the
`first step toward establishing potential critical attributes within
`the Quality by Design (QbD) paradigm encouraged by the US
`Food and Drug Administration.2 The experimental part of the
`developability assessment includes short-term stability studies at
`various temperatures, freeze-thaw studies and limited forced deg-
`radation studies. This series of studies determine the biochemical
`(e.g., sensitivity to oxidation and deamidation) and biophysical
`
`*Correspondence to: Alexandre Ambrogelly; Email: alexandre.ambrogelly@merck.com
`Submitted: 04/28/13; Revised: 05/31/13; Accepted: 06/03/13
`http://dx.doi.org/10.4161/mabs.25269
`
`www.landesbioscience.com
`
`mAbs
`
`787
`
`Lassen - Exhibit 1012, p. 2
`
`

`

`Conditions
`
`Initial
`
`Table 1. Developability stability schedule
`t = 6
`t = 3
`t = 2
`t = 0.5
`t = 1
`mo
`mo
`mo
`mo
`mo
`/
`/
`/
`/
`X
`−80°C
`X
`X
`X
`X
`X
`2–8°C
`X
`X
`X
`X
`X
`25°C
`X
`X
`X
`X
`X
`40°C
`/ No samples staged at these conditions/timepoints. X Samples staged
`and analyzed at these conditions/timepoints. Details of the assays used
`during stability are reported in Table S1.
`
`X
`
`(e.g., unfolding and formation of large molecular weight spe-
`cies) stability profiles of the molecule of interest. Determination
`of maximum solubility and associated viscosity are also essential
`for molecules destined to be formulated at high concentrations
`for subcutaneous injections. Developability studies offer the first
`opportunity to assess the technical viability of a project at the
`industrial scale and function as a bridge between discovery and
`process development activities. We present in this report three
`case studies and include data generated during developability
`assessment of antibody molecules.
`
`Results
`
`General strategy for developability studies. Developability stud-
`ies are considered a derisking activity designed to provide under-
`standing of the colloidal properties and prevalent degradation
`pathways of a molecule early in the development process, thereby
`allowing the analytical strategy to be tailored to the molecule
`of interest. To be relevant, developability studies should be per-
`formed on material produced with a stable cell line of the same
`host as the one that will be used later for process development;
`when possible, use of the final stable clone is preferable.
`The most widespread host for expression of mAbs is the
`Chinese hamster ovary (CHO) cell line; however, a developabil-
`ity strategy can be applied to material produced by any expression
`system. The recombinant protein is produced via an upstream
`early cell culture process and purified by a downstream, 2–3 col-
`umns standard process. It is important to note that, at this stage
`of development, the formulation has not been optimized and the
`protein matrix might be suboptimal. Keeping these limitations in
`mind, developability studies are aimed at detecting gross changes
`in the protein that could hinder the stability or potency of the
`molecule.
`Sequence alignment and molecular modeling. Developability
`assessment starts with analysis of the sequence of the protein
`entering the development phase. Some of this work has likely been
`done in the discovery space. For example, potentially problematic
`residues such as methionine, asparagine or aspartic acid residues
`localized in exposed region of the mAb, including the comple-
`mentarity-determining regions (CDRs), may have been removed.
`Relevant knowledge and experience generated during the discov-
`ery phase, such as whether the molecule was generated through
`phage display or hybridoma cells, should be taken into account.
`The first examination of the sequence during developability
`
`assessment is therefore aimed at identifying potential hot spots
`for degradation that remain in the sequence. Sequence alignment
`can be performed by any software available on the market. To
`complete the analysis of the primary sequence, building a molec-
`ular model may help to visualize residues exposed to the solvent,
`which are susceptible to degradation. The software we currently
`use for this exercise is MOE (CCG, Montreal, Canada).
`Short-term research stability studies and freeze/thaw cycles. To
`gain a first impression about how the molecule will degrade,
`short-term stability studies are performed. Material is staged
`at −80°C, 2–8°C, 25°C and 40°C for up to 6 mo (Table 1).
`Samples are removed at different time points and analyzed by size
`exclusion (SEC) HPLC to detect the formation of high molecular
`weight species and by ion exchange (IEX) HPLC to measure the
`effect of stress on distribution of charges at the surface of the
`molecule due to the deamidation and isomerization of asparagine
`and aspartic acid residues. Samples are also analyzed by reducing
`and non-reducing sodium dodecyl sulfate capillary electrophore-
`sis (CE-SDS) to detect proteolytic cleavage of the heavy or light
`chains, peptide mapping to quantify the formation of post-trans-
`lational modifications formed during stress and a potency test,
`which is most commonly in a binding ELISA format in the very
`early stage of the program development.
`Propensity of the recombinant protein of interest to form
`aggregates is gauged during freeze/thaw cycles. The protein is
`subjected to five cycles. Aliquots are analyzed by SEC-HPLC,
`SEC-multi-angle laser light scattering and dynamic light scatter-
`ing at each of the five cycles.
`Limited stress conditions. To complete the initial assessment
`of the stability of the molecule, a series of stress conditions are
`applied (Table 2). These conditions include standard stresses
`suggested by regulatory agencies, such as exposure to low and
`high pHs, light and oxidative reagents.3 More emphasis may be
`put on some of these stress conditions if a potential degradation
`pathway has been identified by molecular modeling or during
`early experience with the material. The stresses are likely to trig-
`ger deamidation, isomerization, aggregation and oxidation of
`amino acid side chains. These degradation pathways are the most
`commonly described for mAbs.4 A direct measurement of the
`effect of the degradation on potency is also taken.
`Determination of viscosity and ability to concentrate formulated
`materials (surrogate for solubility studies). High concentrations of
`protein are usually required when mAb therapeutics are admin-
`istered via subcutaneous injection. To verify whether the drug
`candidate can achieve a target concentration, the protein is con-
`centrated to 100–200 mg/mL in standard buffers. Viscosity of
`the high concentration solutions is measured to evaluate purifica-
`tion and delivery device strategies.
`mAb A case study: No major or unexpected degradation.
`The first case study concerns an IgG1 antibody, termed mAb
`A. Sequence structure analysis of the CDRs did not reveal any
`particularly exposed asparagine or aspartic residues in an amino
`acid context prone to deamidation or isomerization (i.e., located
`directly upstream from a glycine, serine or proline residues).7
`CDRs did not contain exposed methionine or unpaired cys-
`teine residues. The short-term stability of early representative
`
`788
`
`mAbs
`
`Volume 5 Issue 5
`
`Lassen - Exhibit 1012, p. 3
`
`

`

`Table 2. Typical limited forced degradation conditions
`Tests
`Duration
`Condition
`Step
`UV, SEC, IEX, peptide mapping, potency
`48 hr
`Low pH
`Adjust pH with HCl to 3.0
`UV, SEC, IEX, peptide mapping, potency
`48 hr
`High pH
`Adjust pH with NaOH to 9.0
`UV, SEC, IEX, peptide mapping, potency
`N/A
`Light stress
`1X ICH (UV, Visible)
`UV, SEC, IEX, peptide mapping, potency
`2–5 h
`Methionine oxidation
`0.05% tBHP
`Measure A280, A320 and DLS before and after pH adjustment to pH 5.5
`N/A
`pH jump
`Coordinate with purification area
`tBHP, tert-Butyl hydroperoxide a reagent.5 ICH, International Conference on Harmonisation; details on guidance for Light stress can be found in ICH
`guideline Q1B.6 Details of the assays used during forced degradation are reported in Table S1.
`
`Figure 1. Overlay of IEX-HPLC chromatograms of mAb A samples stored for one month (A) and three
`months (B) at −80°C, 2–8°C, 25°C and 40°C. Overlay of IEX-HPLC chromatograms of mAb A samples chal-
`lenged with high and low pHs for 48 h at 40°C (C) and light induced (0.2xICH) stress (D).
`
`material was evaluated for up to
`three months at 2–8°C, 25°C and
`40°C via a battery of assays. IEX-
`HPLC profiles after one and three
`months are shown in Figure 1A and
`1B. Decrease of relative peak area of
`the main and basic species (eluting
`after the main) and increase of the
`relative peak area of the acidic vari-
`ants (eluting before the main) was
`noted for samples stored at 40°C. A
`decrease of basic variants is typical
`for mAbs; temperature accelerates
`the cyclisation of N-terminus gluta-
`mine into pyroglutamic acid, a post-
`translational modification common
`to many therapeutic and endogenous
`human mAbs.8 Likewise, an increase
`of acidic variants upon exposure to
`elevated temperature is a common
`occurrence that may reflect deami-
`dation of asparagine residue, isom-
`erization of aspartic residues into
`isoaspartic or structural changes
`affecting the overall surface charge distribution of the molecule.8
`In the present case, deamidation was only detected at conserved
`position 385 within the Fc portion of the mAb and did not
`exceed 10% (Table 3). Limited forced degradation studies at low
`and high pH (3.0 and 9.0) after 48 h at 40°C, resulted in pH
`dependant degradation routes: exposure to high pH resulted in
`an increase of acidic variant (and deamidation of position 385),
`while low pH resulted in the formation of basic species not iden-
`tified by peptide mapping (Fig. 1C, Table 3). Exposure to other
`forms of stress was performed and did not result in unexpected
`alterations of the primary, secondary or tertiary structure of the
`molecule. Exposure to UV/visible light following ICH guide-
`lines, for instance, resulted in the formation of a basic species
`reflecting oxidation of conserved Fc methionines 253 and 429
`(Fig. 1D, Table 3). Oxidation of the conserved Fc methionine
`residues under these conditions was expected and documented
`in the literature.4 While developability studies of mAb A did not
`identify major chemistry, manufacturing and control (CMC)
`challenges, the outcome of these studies provided information
`about primary degradation pathways and potential critical qual-
`ity attributes (CQA) for the molecule.
`
`5°C
`25°C
`40°C
`
`5°C
`25°C
`40°C
`
`4%
`4%
`4%
`
`5%
`5%
`6%
`
`8%
`8%
`8%
`
`Table 3. Relative amounts of PTMs in mAb A by LC-MS peptide mapping
`Storage Temperature
`Met 253 ox
`Met 429 ox
`N385D
`Initial
`7%
`4%
`4%
`1 mo time point
`8%
`8%
`9%
`3 mo time point
`4%
`8%
`5%
`7%
`4%
`21%
`Limited forced degradation
`14%
`8%
`4%
`pH 3.0/40°C
`7%
`8%
`4%
`pH 9.0/40°C
`5%
`22%
`17%
`Photostress
`6%
`7%
`4%
`Dark Ctrl
`Met253ox and Met429 ox refer to methionine oxidation position 253
`and 429, respectively. N385D refers to relative amount of aspartic acid
`at position 385. Photostress conditions were 0.2x ICH, Dark Ctrl was the
`corresponding control.
`
`www.landesbioscience.com
`
`mAbs
`
`789
`
`Lassen - Exhibit 1012, p. 4
`
`

`

`Figure 2. Overlay of IEX-HPLC chromatograms of mAb B samples stored for one month (A) and three months (B) at −80°C, 2–8°C, 25°C and 40°C. Evo-
`lution during stability studies of relative amount of deamidation at HCAsn55 (N55, blue trace) and mAb B potency (red trace) (C). Overlay of IEX-HPLC
`chromatograms of mAb B samples stored for one month at 40°C at pH 4.5 and pH 6.0 (D). Thermal unfolding profile ofsolution of mAb B (E).
`
`mAb B case study: Identification of degradation pathways
`informs appropriate control strategy. The second example of
`developability studies involves a S228P hinge-modified IgG4
`molecule, termed mAb B. Scanning of the primary sequence
`for exposed asparagine, aspartic acid, tyrosine, tryptophan and
`methionine residues did not predict the presence of particularly
`sensitive hot spots. Results from the short-term stability studies,
`however, show a collapsed IEX-HPLC profile after three months
`at 40°C and a significant increase of acidic variants at 25°C and
`essentially no changes when stored at 2–8°C or frozen (Fig. 2A and
`B). No increase of aggregates or fragments resulting from chemi-
`cal proteolysis was detected by SEC-HPLC (data not shown).
`Peptide mapping and LC-MS analysis indicated that deamida-
`tion of heavy chain Asn55 exceeded 50% and thus was largely
`responsible for the instability of the molecule (Table 4). Other
`expected post-translational modifications, such as oxidation of the
`
`conserved Fc methionine residues were also detected, albeit to low
`levels. Testing of relative potency by binding ELISA showed that,
`in spite of the chemical degradation, mAb B retained its complete
`ability to bind its target antigen (Table 4, Fig. 2C). Lowering the
`pH of the mAb B formulation from 6.0 to 4.8 reduced greatly the
`amount of deamidation after one month at 40°C (Fig. 2D). For
`this particular molecule, deamidation at Asn55 was recognized as
`a potential CQA. A proper control strategy involving monitoring
`deamidation during upstream and down stream process develop-
`ment and manufacturing was put in place. Efforts toward defin-
`ing an adequate formulation and storage conditions of the drug
`substance were made to ensure protein stability and the longest
`possible product shelf-life. mAb B’s desirable commercial drug
`product form is a highly concentrated solution for subcutaneous
`injection. Part of the developability exercise is to verify that the
`protein can remain in solution at concentrations above 100 mg/
`
`790
`
`mAbs
`
`Volume 5 Issue 5
`
`Lassen - Exhibit 1012, p. 5
`
`

`

`−80°C
`5°C
`25°C
`40°C
`
`3.7%
`3.4%
`3.8%
`4.1%
`
`100%
`100%
`103%
`101%
`
`113%
`108%
`94%
`
`M250ox M426ox
`N55D
`1 mo time point
`21.6%
`10.1%
`21.7%
`10.0%
`23.9%
`10.8%
`36.4%
`11.7%
`3 mo time point
`3.1%
`21.1%
`14.7%
`5°C
`2.7%
`28.1%
`16.7%
`25°C
`3.5%
`55.6%
`18.6%
`40°C
`N55D refers to relative amount of aspartic acid at position 55 on the
`heavy chain. Met248ox and Met250ox refer to methionine oxidation
`positions 248 and 250. Potency by binding ELISA is expressed as % of
`reference standard.
`
`Table 4. Potency and Relative amounts of PTMs in mAb B by LC-MS
`peptide mapping
`Storage Temperature
`
`Potency
`
`mL and such a solution has viscosity that would allow develop-
`ment of prefilled syringe image and autoinjector devices. mAb
`B was successfully concentrated to 190 mg/mL and remained in
`solution. At these concentrations, viscosity remained under 19 cP,
`which allows development of purification processes and prefilled
`syringe image (Table 5). Additionally, mAb B has demonstrated
`good thermodynamic stability (onset of the first transition above
`50°C) and thermal profile with three unfolding transitions at
`63, 68 and 80°C, respectively (Fig. 2E, Table 5), which is typi-
`cal for multidomain proteins. Developability studies of mAb B
`highlighted primary degradation pathways and potential criti-
`cal attributes. It also identified CMC issues addressable with an
`appropriate manufacturing and analytical control strategy.
`mAb C case study: Identification of degradation pathways
`leads to project termination and reallocation of resources. The
`third case study concerns an IgG2 molecule, termed mAb C.
`Analysis of the primary sequence of the molecule showed the pres-
`ence of an asparagine-glycine (Asn33-Gly34) sequence in one of
`the CDRs of the antibody light chain. Analysis of the structural
`context of these residues in the molecular model showed that
`Asn33 is exposed to the solvent and located in a flexible loop. The
`Asn-Gly sequence has been documented as a site particularly sen-
`sitive to facile deamidation to aspartic acid and isomerization to
`isoaspartic acid.7 IEX-HPLC chromatograms of mAb C samples
`showed a series of three charge variant peaks eluting at about 8, 12
`and 16 min (Fig. 3A and B). These peaks were first believed to be
`associated with the three different hinge region disulfide isomers
`commonly found in IgG2s with kappa light chain.9 Isolation and
`characterization of the charge variants demonstrated, however,
`that the two acidic variants (eluting at 8 and 12 min) were mAb
`C molecules with one or both of the Asn33 deamidated (data
`not shown). Analysis by IEX-HPLC of the short-term stability
`at one and three months time points showed a rapid increase of
`acidic variants when the molecule was stored at 25°C and 40°C
`(Fig. 3A and B). Under the same storage conditions and at the
`same time points, the molecule showed only minimal increase of
`high molecular weight species by SEC-HPLC (data not shown).
`Consistent with the increase of the acidic variants peaks, analysis
`of the stability samples by peptide mapping showed dramatic for-
`mation of aspartic acid and isoaspartic acid at light chain position
`33 (Table 6). Deamidation of light chain Asn33 reached 92.6%
`at 40°C after three months and correlated with mAb C loss of
`potency (Table 6, Fig. 3C). Other post-translational modifica-
`tions were also detected: oxidation of the conserved methionine
`248 in the Fc domain and limited cyclization of the glutamic acid
`residue located at heavy chain position 1 (Table 6). These later
`modifications are well-documented and shown to have no effect
`on the potency of mAbs.4,8 Asn33 deamidation can be controlled
`by optimizing the formulation; lowering of the pH reduced deam-
`idation at elevated temperature over time (Fig. 3D). However,
`stabilization of the molecule in vitro is not sufficient. Ex vivo and
`in vivo rhesus monkey plasma stability studies indicated that deg-
`radation of mAb C drug candidate occurs in plasma (Fig. 3E
`and F). Titration of mAb C by anti-kappa light chain and anti-
`gen ligand-capture ELISAs, in serum samples drawn from rhe-
`sus monkeys, showed a time-dependent separation of the PK and
`
`Table 5. Viscosity and thermal melt of mAb B
`Thermal transitions (°C)
`Onset
`Tm1
`Tm2
`Tm3
`
`Concentration
`
`Viscosity (cP)
`
`19
`190
`2
`60
`Tm1, Tm2, Tm3 refer to melting temperature transition 1, 2, 3 shown in
`thermal melt curves (Fig. 2E).
`
`68
`
`80
`
`56
`
`63
`
`antigen binding curves, suggesting a loss of ligand binding capac-
`ity consistent with our in vitro observations (Fig. 3F). To remain
`viable as a product candidate, mAb C had to be reengineered by
`replacing Asn33 by an alternative amino acid without altering
`affinity for the target antigen. Developability studies of mAb C
`led to the identification of major CMC challenges that could be
`addressed only via re-engineering of the molecule.
`
`Discussion
`
`Developability studies are an important link between drug dis-
`covery and process development activities. After expression of a
`proposed therapeutic biologic in the expected host organism, early
`assessment of the molecule’s stability is the first occasion to gather
`knowledge on it to determine potential CQAs as encouraged by
`the QbD paradigm. We presented three cases of developability
`studies, one for each of the typical therapeutic mAb frameworks,
`that correspond essentially to three scenarios encountered during
`developability studies. For the first example, short-term stabili-
`ties, freeze-thaw and limited forced degradation studies did not
`uncover unexpected sources of concerns about the stability of the
`molecule. In this particular case, developability studies showed
`that the molecule’s degradation pathways conformed to those
`of standard IgGs. The second case study provides an example
`of a therapeutic mAb undergoing degradation without affecting
`potency. In this case, a proper analytical strategy is put in place
`to monitor the degradation detected. Proper process controls are
`also instituted to limit the degradation during manufacturing.
`
`www.landesbioscience.com
`
`mAbs
`
`791
`
`Lassen - Exhibit 1012, p. 6
`
`

`

`Figure 3. Overlay of IEX-HPLC chromatograms of mAb C samples stored for one month (A) and three months (B) at −80°C, 2–8°C, 25°C and 40°C.
`Evolution during stability studies of relative amount of deamidation at HCAsn33 (N33, blue trace) and mAb B potency (red trace) (C). The bar graph
`represents the relative area of acidic variants after one month at 40°C in six different formulations (D). Deamidation at Asn33 in rhesus monkey serum
`stability studies (E): TIC of the doubly charged ion of peptide 25–39 at day 0, 7, 14 and 21: The peak at ~33–34 min is the early eluting non deamidated
`parent peptide (Asn33), the two peaks eluting at 34–36 and 35–37 min are the deamidated product peptides (iso-Asp33 and Asp33, respectively). At
`all-time points the ratio of deamidated product peptides IsoAsp/Asp is the same, ~1:5 throughout the whole experiment. Titration of mAb C in rhesus
`monkeys serum by an anti-kappa light chain (PK curve) and antigen ligand-capture (Antigen curve) ELISAs after second injection (F).
`
`Lastly, formulation studies are put in place to ensure that the deg-
`radation does not occur during storage and does not compromise
`the shelf-life of the drug product. The last case study corresponds
`to the case where stability studies detect a major problem with
`the molecule’s stability and efficacy, leading to termination of the
`project and reallocation of resources.
`
`Materials and Methods
`
`Materials. mAb A, mAb B and mAb C are humanized IgG1,
`IgG4 and IgG2, respectively. All molecules have λ light chains.
`
`These Merck proprietary mAbs are expressed in CHO cells and
`purified using three chromatographic steps (protein A, anion
`exchange and cation exchange). Formulations included histi-
`dine, acetate and typical excipients such as sucrose and PS80;
`pHs ranged from 5.0 to 6.0.
`Ion exchange HPLC (IEX-HPLC). Ion exchange HPLC for
`mAbs A, B and C is run on the Dionex WCX-10 column and
`uses either a citrate or Mes-based mobile phase A and a phos-
`phate buffer with or without added NaCl as mobile phase B.
`Gradient is adjusted to suit the needs of each molecule.
`
`792
`
`mAbs
`
`Volume 5 Issue 5
`
`Lassen - Exhibit 1012, p. 7
`
`

`

`5°C
`25°C
`40°C
`
`3.4%
`5.2%
`12.4%
`
`30.3%
`44.1%
`64.5%
`
`Table 6. Potency and Relative amounts of PTMs in mAb C by LC-MS peptide mapping
`Storage Temperature
`N33D
`N33isoD
`N33D+N33isoD
`Initial
`3.5%
`29.2%
`32.6%
`1 mo time point
`33.6%
`49.2%
`76.9%
`3 mo time point
`96%
`1.5%
`3.2%
`30.1%
`26.6%
`3.5%
`5°C
`28%
`2.2%
`5.8%
`62.6%
`56.2%
`6.4%
`25°C
`5%
`6.1%
`21.7%
`92.6%
`68.5%
`24.1%
`40°C
`N33D and N33IsoD refer to relative amount of aspartic acid and isoaspartic acid at position 33, respectively. Total deamidation at position 33 is indi-
`cated as the sum of (N33D and N33IsoD). Met248ox refers to methionine oxidation position 248. HC pE1 refers to the proportion of pyroglutamic acid
`at HC position1. Potency by binding ELISA is expressed as % of control (Initial, indicated as N/A).
`
`Met248 ox
`3.2%
`
`HC pE1
`2.2%
`
`1.4%
`1.5%
`2.4%
`
`2.5%
`2.9%
`5.2%
`
`Potency
`N/A
`
`108%
`101%
`20%
`
`Peptide mapping. A typical peptide mapping procedure was
`followed. Samples were diluted in reducing buffer (50 mM TRIS-
`HCl at pH 8.0, 6 M guanidine-HCl, 5 mM EDTA and 20 mM
`DTT) and were incubated at 56°C for 30 min. Each sample was
`cooled at room temperature for 5 min prior to alkylation with 50
`mM iodoacetamide at room temperature for 30 min in the dark.
`Each sample was then buffer-exchanged to digestion buffer con-
`taining 50 mM Tris buffer at pH 8.0, 2 M urea and treated with
`an aliquot of trypsin. The mixture was incubated at 37°C. The
`digestion was quenched by the addition of 20% trifluoroacetic
`acid (TFA). Agilent 6538 Q-TOF or Xevo G2 Q-TOF system
`mass spectrometers were set up with Agilent 1290 Infinity HPLC
`system or Waters Acquity UPLC. The samples were loaded on
`a C18 column held at 75°C. Mobile phase A was 0.05% TFA
`in water and mobile phase B was 0.05% TFA in acetonitrile.
`Gradient is adjusted to suit the needs of each molecule Peptide
`mapping analysis was performed by using Agilent MassHunter
`Qualitative Analysis software.
`Quantitation of mAb C deamidated peptide in plasma by
`multiple reaction monitoring. mAb C was spiked in rhesus
`monkey plasma and incubated for up to 21 days at 37°C. Blank
`plasma was prepared and incubated along with the spiked sam-
`ples serving as a negative control for the rhesus monkey plasma’s
`IgG2. mAb C was subsequently affinity-purified from the bio-
`matrices using a goat anti-human IgG-heavy and light chain
`monkey adsorbed antibody conjugated to an agarose based Affi-
`Gel hydrazine gel, by coupling the sugar residues of the antibody
`to the hydrazine groups on the gel. The eluates were digested
`by trypsin followed by endoproteinase V8 to obtain a reason-
`ably sized peptide amenable to mass spectrometry analysis. The
`digests were separated by reverse phase chromatography, using
`a very shallow gradient necessary to separate the peptides con-
`taining Asn33, isoAsp33 and Asp33. The separated peptides
`were analyzed online by multiple reaction monitoring (MRM)
`targeted analysis on a TSQ Vantage Thermo mass spectrometer.
`Areas under the chromatogram were subsequently extracted for
`each species using Skyline software (MacCoss lab, Department
`of Genome Sciences, UW) as a platform or Xcalibur (Thermo
`Scientific) and plotted against time.
`
`Potency by direct binding ELISA. Typical direct binding
`ELISA was used. The 96-well ELISA plates were coated with the
`appropriate antigen for 1 h at 25°C for mAb B or overnight at 4°C
`for mAb C. Plates were washed with TBS (mAb B) or PBS (mAb
`C) containing 0.05% Tween 20 and blocked with a blocking buf-
`fer for 1 h at 25°C. For the primary incubations, various con-
`centrations of mAbs were added into the pre-coated plates. The
`amount of bound mAb was measured by either alkaline phospha-
`tase (mAb B) or horseradish peroxidase (mAb C) labeled anti-
`human IgG. The enzymatic reaction was developed with either
`PhosphoGlo AP substrate (mAb B) or Pico SuperSignal (mAb
`C). The EC50 vales were determined by nonlinear regression.
`Surrogate solubility. While not strictly a maximum solubil-
`ity study determination, viscosity measurements in conjunction
`with protein concentration experiments in standard buffers are
`meant to evaluate the feasibility of 100–200 mg/mL solutions.
`Buffer exchanges were performed at 5°C in 50 kDa Amicon®
`Ultra – 15 centrifugal filter units, with sample to buffer volume
`ratio of 1:1 and three volume exchanges. mAb C concentration
`was determined by UV absorption at 280 nm using a calculated
`extinction coefficient.
`Viscosity. Viscosity measurements were performed at 30°C
`using mVROC instrument.
`Thermal melts (DSC). Differential scanning calorimetry
`(DSC) was performed with Microcal VP-DSC. 400 μL of each
`sample and matching buffer were placed in a 96-well plate, and
`the thermograms were acquired between 10 and 90°C at a scan
`rate of 90°C/h, with 5 min pre-scan equilibration. The data was
`processed by subtracting from each sample thermogram, a cor-
`responding placebo–placebo scan, fitting a bas

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