throbber
104
`
`Michael 1. Akers et al.
`
`development scientist must keep in mind that the scale of the process may
`influence results. For example, it is commonly assumed that development
`of the freeze-drying cycle changes greatly on going from lab scale to pilot
`scale to production scale. Ideally, experiments can be performed at pilot
`scale using equipment representative of the production process. In
`addition, the sequence of steps in the process, acceptable environmental
`conditions (temperature, humidity, air, etc.) for the unit operation, and
`acceptable excipient ranges should be evaluated.
`For solutions, the following process conditions may impact product
`quality:
`
`1. Agitation (rate, duration): High rates may result in undesired
`foaming, denaturation, aggregation, or oxidation. Kim et al. (1994)
`reported on the effect of high shear force on the IX helix-to-fJ sheet
`conversion of insulinotropin resulting in a reduced solubility of the
`protein. Thus, agitation or mixing rates will affect the conformation
`and solubility of this protein, thus requiring special control of these
`rates.
`2. Compounding sequence: Order of addition of excipients and drug
`substance must be determined. Normally, excipients are added and
`dissolved before drug substance is added (Harwood et al., 1993).
`This allows sufficient time for excipients to dissolve while minimiz(cid:173)
`ing time in solution for unstable drug substances.
`3. pH adjustments: Prolonged exposure to acid or basic pHs can cause
`protein degradation.
`4. Filtration: A membrane is selected which offers low protein binding
`as confirmed by protein assays on the solutions before and after
`filtration to detect any losses (Hawker and Hawker, 1975). This is
`discussed in detail in Chapter 5.
`5. Filtration and filling: The temperature may need to be controlled to
`prevent chemical degradation during filling. Rates of filtration and
`filling should be considered to prevent shearing of the protein,
`although this possibility is remote. Effects of filtration and filling
`rates were studied for human growth hormone (Hsu et al., 1988;
`Pikal et al., 1991 b). Shear forces encountered during processing were
`found not to cause aggregation of human growth hormone.
`However, aggregation of proteins during filtration and filling can
`be caused by interactions of the protein with polymeric hydrophobic
`surfaces, such as those composing sterilizing filters and process
`tubing for filling equipment, suspected to be the cause of human
`growth hormone aggregation during processing (Hsu et aI., 1988).
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 60 of 144
`
`

`

`Formulation Development of Protein Dosage Forms
`
`105
`
`Insulin aggregation and fibrillation can be caused by interaction of
`the protein in solution with plastics (Thurow and Geisen, 1984) (see
`previous discussion on insulin binding to plastic surfaces).
`6. Freeze-drying: Denaturation of a protein solution may occur
`because of pH shifts or ionic strength changes during freezing (Orii
`and Morita, 1977). The freezing rate, concentration, endpoint pH,
`and time and temperature of holding prior to lyophilization may
`affect the chemical and physical stability of the product (see Chapter
`6).
`7. Environmental conditions: Sparging of nitrogen into solution to
`remove dissolved gases or use of a nitrogen overlay to replace the air
`headspace during filling to retard the oxidation of oxygen-labile
`formulation may be required (Brown and Leeson, 1969).
`8. Materials compatibility: During the manufacture, the drug is
`exposed to various materials such as stainless steel, filters, tubing,
`and pump diaphragms. It is
`important to ensure
`that the
`formulation components are compatible with these materials.
`9. Time and temperature: Throughout manufacture, critical holding
`times and temperatures need to be established, not only for protein
`stability protection purposes, but also to assure microbiological
`control, particularly to prevent endotoxin contamination.
`
`See Chapter 7 on quality assurance and quality control for additional
`details on this topic.
`
`12.3. Clinical Trial Supplies
`
`As the transfer of a process from laboratory scale into the clinical
`phase occurs, the development scientist needs to be involved during the
`early production of clinical trial supplies as well as during any subsequent
`formulation and process changes. The development scientist can obtain
`valuable insight into the conditions of manufacturing as well as provide
`key
`input
`to
`the manufacturing groups about
`the
`rationale for
`formulation and process decisions. These lots also provide an opportunity
`to collect information under larger scale manufacturing conditions than
`in laboratory-scale experiments. In-process testing should be routinely
`conducted during the manufacture of clinical supplies. The data are
`used to evaluate the process and lead to process improvements, which
`ultimately support the transfer into the manufacturing phase at full
`scale.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 61 of 144
`
`

`

`106
`
`Michael J. Akers et al.
`
`13. QUALITY CONSIDERATIONS DURING FORMULATION
`DEVELOPMENT
`
`13.1. Early Product Assessment
`
`All known physical and chemical properties of a protein should be
`viewed in light of how these properties will affect final quality of the
`finished dosage form. The development scientist should build quality into
`the process so that it can be validated and data exist to support "worst(cid:173)
`case" limits (e.g., extremes in pH, excipient levels, time/temperature limits).
`Process design should be evaluated to assure control of quality parameters.
`
`13.2. Documentation
`
`An essential element of validation is establishing thorough documenta(cid:173)
`tion that a process will consistently meet its predetermined specifications
`and quality attributes. Ongoing documentation is essential during formula(cid:173)
`tion development. Development history reports are required during
`preapproval inspections and are generally a high-level overview describing
`the history of the drug product from preliminary studies to the commercial
`formulation and the process submitted in the regulatory document. There
`may be one report or several to describe the various steps of development
`(e.g., fermentation, granule isolation, drug substance, drug product,
`methods development). It is as important to document what did not work
`as what worked and why decisions were made. Where necessary, bioequi(cid:173)
`valence of lots used in clinical trials should be demonstrated. A method
`history report should contain information on regulatory commitments
`during the development process and give the history of each method.
`Rationale for specifications, reference standards, and cleaning methods
`should be provided. For product development reports, lot rejections,
`deviations, and resolution of the issues need to be covered. Typical contents
`of a parenteral drug product development history report include the
`following:
`
`• Preformulation data
`• Selection of excipients supported by preformulation studies
`• Antimicrobial characteristics of the product
`• Rationale and basis for packaging component choices
`• Description of container/closure integrity studies
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 62 of 144
`
`

`

`Formulation Development of Protein Dosage Forms
`
`107
`
`• Overview of the manufacturing process with a description of each
`unit operation, acceptance criteria for all critical steps of the
`process, and a history of results for each step
`• Scale-up studies
`• Stability overview, including product use studies
`
`Development history reports include, but are not limited to, method
`histories, formulation, primary packaging, process development, and control
`strategy.
`Technical reports prepared on an ongoing basis aid in compiling the
`development history reports and are key in the information transfer to the
`manufacturing sites. Typical information contained in these periodic reports
`are the rationale for decisions, description of what worked and what did not
`work, and information for solving manufacturing problems.
`
`13.3. Stability Studies
`
`Stability testing of protein and peptide dosage forms should follow
`the ICH guidelines (U.S. FDA, 1996; ICH, 1995). Table XVI summarizes
`the requirements of this guideline. Three batches or more of the final
`product in the final container/closure system that represents the finished
`product manufacturing scale must be put on stability testing. The batches
`should use different lots of bulk material. A minimum of 6 months of data
`at desired storage conditions must be available at the time of submission
`(less than 6 months of data for products that will have less than 6 months
`dating). Product expiration dating
`is based on real-time data, not
`extrapolated from accelerated stability studies. If different volumes and/
`or strengths of the same formulation are to be tested, a matrix system or
`bracketing may be permitted. Details of matrixing and bracketing are
`found in the guideline.
`Stability testing must use methods that are stability-indicating and
`validated. Methods should monitor changes in potency, purity, and other
`product characteristics* as a function of time and storage conditions as
`defined in the stability protocol. Data on final .product in containers
`maintained in an inverted or horizontal position and all different container/
`closure combinations must be obtained. Multiple dose containers must have
`data to support stability during simulated use, for example, repeated
`
`·Visual appearance, visible particulates in solutions. pH, moisture level of powders, sterility
`testing or container/closure integrity, degradation, if any. of additives.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 63 of 144
`
`

`

`108
`
`Michael J. Akers et al.
`
`Table XVI
`Summary of Guideline for Stability Testing of Biotechnological and Biological
`Productsa
`
`Selection of batches
`
`Guidance for stability studies for regulatory submissions
`
`A. Drug substance
`
`B. Intermediates
`
`C. Drug product
`
`D. Sample selection
`
`Stability-indicating
`profile
`
`A. Protocol
`
`B. Potency
`
`C. Purity/molecular
`characteristics
`
`At least three batches representative of production scale
`A minimum of 6 months of stability data at time of submission (unless
`storage period will be less than 6 months)
`Pilot-plant-scale batch data acceptable at submission as long as there is
`commitment to place first three manufacturing-scale batches into
`long-term stability program after approval
`Storage containers should represent actual holding containers to be
`used during manufacture; containers of reduced size acceptable,
`provided they are constructed of same material and use same type
`of container/closure system
`Identify intermediates and generate in-house data and process limits to
`assure final product stability
`At least three batches of final container product representative of final
`product at production scale
`Different batches of bulk material should be studied in final product
`A minimum of 6 months of data at time of submission (unless storage
`period will be less than 6 months)
`Product expiration dating based on real-time/real-temperature data
`Continuing stability updates should occur during review process
`Quality of final product must be representative of quality of material
`used in clinical studies
`Pilot-plant-scale batch data acceptable at submission as long as there is
`commitment to place first three manufacturing scale batches into
`long-term stability program after approval
`Matrix system and/or bracketing is acceptable when product consists
`of different fill volumes, units, or mass
`
`Guidance
`
`Include detailed protocol for assessment of stability of both bulk drug
`substance and drug product to support proposed storage conditions
`and expiration dating periods; include statistical methods, specifica(cid:173)
`tions, test intervals
`Potency must be compared to an appropriate reference standard
`Perform at appropriate intervals as defined in the protocol and report
`in units of biological activity calibrated against some recognized
`standard
`Purity should be assessed by more than one method
`Limits of acceptable degradation should be documented and justified,
`taking into account levels observed in material used in preclinical and
`clinical studies
`Usual methods include electrophoresis, high-resolution chromatrogra(cid:173)
`phy, and peptide mapping
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 64 of 144
`
`

`

`Formulation Development of Protein Dosage Forms
`
`109
`
`Stability-indicating
`profile
`
`D. Other product
`characteristics
`
`Storage conditions
`
`A. Temperature
`
`B. Humidity
`
`C. Accelerated and
`stress conditions
`
`D. Light
`
`E. Container/closure
`
`F. Stability after
`reconstitution of
`freeze-dried product
`Testing frequency
`:;;:; I year
`Shelf life
`Shelf life ~ I year
`
`Table XVI
`(continued)
`
`Guidance
`
`Visual appearance, visible particulates in ready-to-use or reconstituted
`solutions, pH, moisture level of solid products
`Sterility testing or alternatives (e.g., container/closure integrity) should
`be performed at a minimum initially and at the end of the proposed
`shelf life
`Additives such as stabilizers and preservatives that may degrade
`during the dating period of the product and might adversely
`affect product quality need to be monitored during stability
`
`Guidance
`
`Storage conditions for real-time/real-temperature stability studies may
`be confined to the proposed storage temperature
`As long as humidity-protected containers are used, stability tests at
`different relative humidities can be omitted
`Studies should be conducted on the drug substance and drug product
`under accelerated and stress conditions to provide useful support
`data for establishing the expiration data, be used for support of
`change in formulation or scaleup, assist in analytical method
`validation, or generate information that may help elucidate
`degradation profile of the drug; other benefits include data on
`accidental exposure to stress conditions, e.g., during transportation,
`determine what test parameters are best indicators of product
`stability, and give further insight into degradation patterns not seen
`under normal storage conditions
`Appropriate regulatory authories should be consulted on a case-by(cid:173)
`case basis
`Stability studies should include samples maintained in inverted or
`horizontal position for product to have maximum contact with
`closure
`Data should be supplied for all different container/closure combina(cid:173)
`tions that will be marketed
`Data must be generated that demonstrate that the closure used for
`multiple-dose application is capable of withstanding repeated
`insertions and withdrawals so that product retains full potency,
`purity, and quality
`Stability of product after reconstitution should be demonstrated for
`the conditions and maximum storage period specified
`
`Monthly testing for first 3 months; 3-month intervals thereafter
`Every 3 months during first year of storage, every 6 months during
`second year; annually thereafter
`
`"Summarized from FDA (1996). Also published in Federal Register on July 10, 1996 (61 FR 36466).
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 65 of 144
`
`

`

`110
`
`Michael J. Akers et at.
`
`punctures of the closure, effect of volume change due to repeated
`withdrawals on potency, purity, and quality.
`Stability testing intervals usually are every 3 months during the first
`year of storage, every 6 months during the second year, and annually
`thereafter.
`Although accelerated stability testing is not acceptable for direct
`assignment and support of expiration dates for finished protein products, it
`does have a role in developmental stability studies. ICH stability guidelines
`emphasize the importance of conducting stability studies under accelerated
`conditions, because such studies usually provide useful supporting data for
`establishing the expiration date. Additionally, accelerated stability studies
`provide information for short-term assessment of proposed formulation,
`packaging, or manufacturing changes, evaluate equivalence of materials
`from different bulk process methods, assist in the validation of analytical
`methods for the stability program, and can generate information which may
`help elucidate degradation profiles of the protein alone and in the final
`formulation.
`Accelerated stability studies also may be useful in determining whether
`accidental exposures to stress conditions during transportation of the final
`product are harmful to the product. The guidelines generally define
`accelerated stability test conditions at temperatures at least 15°C above
`the designated long-term storage temperature for the product.
`Typically, kinetic experiments are conducted at elevated temperatures.
`Estimates of rate constants at lower temperatures are then obtained by
`extrapolation of an Arrhenius plot. An assumption of this approach is that
`the kinetics does not change as a function of temperature. For proteins, it is
`important to determine the appropriate temperature range for these stability
`studies (Yoshioka et al., 1994). Ideally, it is desirable to obtain an
`approximately linear relationship to permit prediction of shelf life of the
`drug product. However, in cases where the results cannot be extrapolated,
`these data are still useful for determining the effects of short-term
`temperature excursions on the product and for specifying any special storage
`precautions that may be encountered during shipping and distribution.
`Shnek et al. (1998) described two automated physical stress tests for
`evaluating physical stability of insulin suspensions and solutions. One is a
`moderate stress test combining temperature cycling (25°C to 37°C) and
`resuspenion by agitation, and the other is a more considerable stress test
`combining high temperature (37°C) and extreme agitation (4 h daily). These
`tests provide more relevant physical stability predictions for using
`suspensions and solutions in pen-cartridge devices and determining effects
`of stressful conditions these protein products could encounter during
`shipping, distribution, and patient use.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 66 of 144
`
`

`

`Formulation Development of Protein Dosage Forms
`
`111
`
`13.4. Stability Studies Supporting Distribution of Protein Products
`
`Formulation development typically focuses on protein dosage form
`stability under well-controlled conditions. However, studies must be done to
`demonstrate that the final protein formulation in the final package produced by
`a validated process will retain its stability and other quality properties during
`distribution of the product throughout the world. These studies can be
`accomplished both by simulation in the laboratory and actual distribution of
`the product. During distribution, even if the product is to be held in controlled
`(e.g., refrigerated) conditions, aberrant situations can occur, such as due to
`transportation breakdown, mechanical failure, dropping or otherwise mis(cid:173)
`handling packages, and even overt violation of required handling procedures.
`Data should be available to aid in knowing what these extreme situations will do
`to the quality of the product. The effect of shear (i.e., agitation, mixing, and
`other mechanical forces experienced during processing and handling) and
`temperature extremes on protein stability during simulated and actual
`conditions should be evaluated.
`
`14. EXAMPLES OF FORMULATION PROBLEMS
`
`14.1. Aggregation
`
`Protein aggregation, as already emphasized in this chapter, is a major
`problem in developing stable protein solutions. * Acidic fibroblast growth
`factor (aFGF) is an example of a protein that aggregates readily when
`exposed to temperatures above refrigeration (Tsai et aI., 1993). To solve this
`problem, the authors described a four-part approach:
`
`1. Use a rapid screening procedure, based on turbidimetric measure(cid:173)
`ment, to identify solution conditions, polyanions, and common
`excipients that stabilize aFGF against heat-induced aggregation.
`2. Combine the most promising agents with aFGF and use circular
`dichroism and differential scanning calorimetry to quantitate
`stabilization effects against aggregation.
`3. Formulate the most promising agents with aFGF in solution and
`monitor protein stability at room temperature.
`
`*For more thorough treatment of protein formulation problems, the reader is referred to the
`two books edited by Wang and Pearlman (1993; Pearlman and Wang, 1996) on case histories
`involving stability and characterization of protein and peptide drugs.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 67 of 144
`
`

`

`112
`
`Michael J. Akers et al.
`
`4. Test in vivo efficacy of the best formulations of aFGF using a wound
`healing animal model.
`
`This approach enabled the authors to learn that in addition to the
`well-known protection of aFGF by heparin, a surprisingly wide variety of
`polyanions (e.g., ATP, dextran sulfate, pentosan polysulfate, sulodexide,
`phytic acid, inositol hexasulfate, inorganic phosphate salts) stabilize aFGF
`against aggregation by increasing the temperature at which the protein
`unfolds by I5-30°C. Stabilization by polyanionic compounds occurs
`because these compounds enhance the structural integrity by binding to
`the protein. Common excipients also were studied for their stabilizing
`effects. Many were effective (e.g., glycine, glycerol, sucrose, dextrose,
`trehalose), but at relatively high concentrations. Their final formulation
`choices, all of which were equally capable of accelerating wound healing,
`contained either heparin, inositol hexasulfate, or sulfated p-cyclodextrin
`(with the latter two formulations also containing 1 % hydroxyethyl
`cellulose in phosphate buffer with 0.2 mM EDT A). This is an excellent
`paper from which to learn approaches for solving protein aggregation
`problems.
`
`14.2. Oxidation and Deamidation
`
`Human growth hormone is a protein that both in aqueous solution and
`in the solid state can undergo chemical decomposition both by oxidation
`(methionine 14 to methionine sulfoxide) and deamidation (asparagine at
`position 149) (Becker et ai., 1987). This protein is also prone to aggregation.
`Pikal et ai. (199Ia, b) studied how to overcome these stability problems.
`Variables that they studied included pH, levels of salts, type of lyoprotectant
`excipient, residual water content, and oxygen level in the vial headspace.
`They found that a combination of glycine and mannitol enhanced stability,
`largely because glycine in this combination can remain amorphous. In the
`amorphous state glycine serves as a lyoprotectant and as a "sink" for
`residual moisture, both effects serving to stabilize the protein against
`deamidation and aggregation. The level of phosphate buffer is important as
`an aid in the minimization of deamidation, as a pH value on either side of
`pH 7 increased chemical decomposition. Increases in levels of sodium
`chloride increased the rate of decomposition.
`Oxidation of human growth hormone is due to methionine oxidation.
`Oxidation occurred even when the air in the vial headspace was replaced
`with nitrogen (headspace < 1 % oxygen). Phosphate-buffered formulations
`containing glycine and mannitol as stabilizing excipients, although found to
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 68 of 144
`
`

`

`Formulation Development of Protein Dosage Forms
`
`113
`
`be overall the most stable of all hGH formulations studied (Pikal et ai.,
`1991a), were also found to be the most sensitive to changes in the oxygen
`level of the vial headspace. Because these formulations are amorphous due
`to the effect of glycine in the presence of mannitol, any dissolution of
`headspace oxygen in the amorphous phase can significantly affect protein
`oxidation. It was assumed by the authors that most of the oxygen supply in
`the formulated vial with nitrogen headspace came from oxygen "trapped" in
`the amorphous phase of the product during freeze-drying, the trapped
`oxygen originally being dissolved in the solution filled into the vial.
`
`14.3. Binding to Glass
`
`Early identification of the properties of surface adsorption to solid
`surfaces such as glass is critical to the development of packaging and
`delivery systems for proteins. As discussed in earlier sections, various
`formulation additives can be added to minimize or inhibit adsorption.
`Johnston (1996) examined the effects of solvent additives which would
`minimize the degree of adsorption to various surfaces for a model protein,
`recombinant human granulocyte colony stimulating factor (rGH-CSF). For
`glass vials, he examined solutions containing the following additives: 0.5%
`w/w PluronicC!t) F-l27, 0.05% and 0.5% w/w PluronicCR) F-68, 0.5% v/v
`glycerin USP, and 0.005%, 0.05%, and 0.5% Tween 20. A concentration of
`
`Table XVII
`Adsorption of Bovine Serum Albumin (BSA) and Bovine Immunoglobulin G (IgG)
`to Commercial O.2-llm Microfiltration Capsuleso
`
`Membrane type and
`capsule manufacturer
`
`Protein concentration
`Ilg/ml; (wt%)
`
`BSA adsorption
`Ilg/cm2
`
`IgG adsorption
`Ilg/cm2
`
`Polyvinylidene difluoride,
`millipore
`
`Nylon, Pall
`
`Cellulose acetate, sartorius
`
`50 (0.0050)
`250 (0.025)
`1000 (0.10)
`5000 (0.50)
`50 (0.0050)
`250 (0.025)
`1000 (0.10)
`5000 (0.50)
`50 (0.0050)
`250 (0.025)
`1000 (0.10)
`5000 (0.50)
`
`2
`11
`56
`6
`31
`31
`102
`
`5
`17
`56
`
`1
`3
`
`108
`126
`
`1
`5
`
`"Reproduced with permission from Brose and Waibel (1996). Copyright 1996 Aster Publishing.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 69 of 144
`
`

`

`114
`
`Michael J. Akers et al.
`
`2 ng/ml rGH-CSF in DSW was used as a control. Each solvent additive was
`examined in triplicate at each concentration tested, and solutions were
`sampled at times 0, O.S, 1, 2, and 3 hr and assayed for rGH-CSF. A rapid
`adsorption was observed up to 1 hr, after which time a slower rate
`approaching steady state was observed, similar to adsorption reported for
`insulins and other proteins. Furthermore, viscometry was applied to
`estimate that the thickness of the adsorbed layer to glass was about I !lm.
`Tween 20 at a concentration of O.S% showed the most potential for
`inhibiting surface adsorption to parenteral glass vials.
`
`14.4. Binding to Filter Surfaces
`
`Proteins are known to bind to filter surfaces. Brose and Waibel (1996)
`reported on the adsorption of BSA and sheep IgG to three types of commercial
`filter surfaces, polyvinylidene difluoride (PVDF, Millipore), nylon (Ultipor
`N66, Pall), and cellulose acetate (CA, Sartorius). They filtered S-mg/ml
`solutions of proteins at pressures < 1 psi and measured protein concentration
`in the filtrate as a function of filtrate mass. The mass of adsorbed protein was
`determined by measuring the difference between theoretical (S mg/ml = O.S
`wt%) and actual protein weight percent. This mass was divided by the
`membrane surface area (SOO cm2) to obtain the amount of protein adsorbed
`per unit surface area. They found that both proteins adsorb to all filters, but
`the extent of adsorption is markedly affected by protein concentration and the
`type of filter (see Table XVII). Nylon filters adsorbed the most protein. BSA
`adsorption was linearly related to its concentration, with adsorption to nylon
`being twice as high as its adsorption to PVDF and CA membranes. IgG
`adsorption was linear, but very low to PVDF and CA filters, whereas its
`adsorption to nylon was extensive and nonlinear (it leveled off at higher
`concentrations). Other authors (Brose and Waibel, 1996) have also reported
`on protein adsorption on filter surfaces, so the phenomenon will occur, but the
`question is how much and how can the adsorption be minimized? The choice
`of filter, the concentration and type of protein being filtered, and the filtration
`conditions (pressure, rate, size of filter, etc.) all will affect the extent of protein
`adsorption to filters.
`
`REFERENCES
`
`Absolom, D. R., Zingg, W., and Neumann, A. W., 1987, Protein adsorption to
`polymer particles: Role of surface properties,J. Biomed. Mater. Res. 21:161-171.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1024, p. 70 of 144
`
`

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