throbber
PublicationDate:August19,1994|doi:10.1021fh1<-1994~056?.ch0ll1
`
`
`
`Downloadedby174.471.174.222onMarch10,2015Ihttp:/l‘pub5_acs.org
`
`Chapter 1
`
`Formulation and Delivery of Proteins
`and Peptides
`
`Design and Development Strategies
`
`Jclfrey L. Clelaudl and Robert Longer‘
`
`‘Pharmaceutical Research and Development, Genentech, Inc., South San
`Francisco, CA 94080
`‘Department of Chemical Engineering, Massachusetts Institute
`of Technology, Cambridge, MA 02139
`
`The success of most peptide and protein drugs is dependent upon the
`delivery of the biologically active form to the site of action.
`In the
`design and development of formlllations to achieve this goal, the
`formulation scientist must consider
`the clinical
`indication,
`pharmacokinetics. toxicity. and physicochemical stability of the drug.
`The development of a stable formulation is a necessary step for each
`new protein or peptide therapeutic. The degradation pathways and
`their impact on stability should be systematically analyzed and
`competing degradation rates must be balanced to arrive at the most
`stable formulation possible. Several routes of administration should
`also be considered and future development of new formulations may
`expand the number of potential options. Formulations for each route
`of administration may be unique and,
`therefore, have special
`requirements.
`In the case of depot formulations, there are many
`potential matrices, each of which has distinct characteristics that affect
`its interactions with the drug and its behavior in vivo. The formulation
`characteristics may have a dramatic impact on the in viva stability of
`the drug as well as the pharmacokinetics and pharmacodynamics. The
`optimization of formulations. the routes of delivery, the design of
`depot systems, and the correlation between physicochernical stability
`and in viva behavior are discussed in detail with recent examples. For
`new biotechnology-derived drugs including nucleic acids (DNA
`vectors and antisense RNA) to reach commercialization, all of the
`issues involved in the design and development of a drug formulation
`must be considered at an early stage of the overall development
`process.
`
`Many aspects of biopharrnaceutical process development have been well studied over
`the past twenty years. Difficulties in fermentation. cell culture, and. to some extent,
`purification and recovery have largely been overcome and these process steps have
`been well characterized for the production of many protein pharmaceuticals.
`However, one important field lags behind these others in its development. The design
`and production of protein and peptide drug formulations is not well developed and
`many of the mechanisms for stabilization and delivery of these drugs have not been
`
`D09’?-6156.’94r'D567—lI]01$U8.l]JlU
`© 1994 Alnerican Chemjml Society
`
`CFAD Exhibit 1045
`1045
`CFAD v. NPS
`CFAD V. NPS
`IPR2015-01093
`IPR2015—O1093
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, .I., et aI..
`ACS Symposium Series: American Chemical Society: Washington, DC‘, I994.
`1
`
`CFAD Exhibit 1024
`
`

`
`2
`
`FORMUI.ATION AND DELIVERY OF PROTEINS AND PEPTIDES
`
`determined. In many cases, companies may initially neglect formulation and stability
`issues, resolving to simply store proteins or peptides in phosphate buffered saline or
`other solutions that have not been optimized for stabilizing the drug. Several
`unknowns still exist when developing a stable dosage form for peptides and proteins.
`Each molecule has its own unique physical and chemical properties which determine
`its in vitro stability. The formulation scientist must also be concerned about the in
`vivo stability of the drug. Thus, the development of successful formulations is
`dependent upon the ability to study both the in vitro and in vivo characteristics of the
`drug as well as its intended application.
`
`Effect of Formulation Design and Delivery on Drug Development
`
`As shown in Figure 1, a formulation scientist is confronted with a complex decision
`in choosing a formulation for delivery of a therapeutic protein or peptide. In the
`literature, the most common discussions of protein and peptide formulations focus on
`the physicochemical stability of these molecules. Indeed, the properties of the drug
`molecule are critical in determining the appropriate formulation for successful
`delivery and stability. The vast majority of the literature on protein and peptide
`formulations describes the degradation pathways for the drug. Many degradation
`pathways have been well characterized and, in some cases, degradation may often be
`predicted from the primary sequence of the protein or peptide (see 1 for examples).
`Once the formulation scientist has found a set of conditions that provide extensive
`stability (>2 year shelf-life), the formulated drug is tested in animal models for
`toxicity and pharmacokinetics. In many cases, this. testing phase does not occur until
`the drug has moved from research into development. At this stage, many problems
`can occur including poor bioavailability due to the instability of the drug in vivo,
`rapid clearance, or the distribution of the drug in the body. Furthermore, an attempt is
`often made to resolve these difficulties by administering excess drug to achieve the
`desired biological effect. However, excessive drug doses often lead to toxicity
`problems. By this stage, the development of the drug has reached a critical decision
`point. The tendency in most organizations is to reconsider the development of the
`drug, sometimes resulting in the 'death' of the development project. However •. the
`formulation scientist has the unique opportunity to work with the scientists in
`pharmacokinetics and toxicology to 'save' the development of the drug. By altering
`the formulation or the route of delivery, a drug can often have another opportunity to
`reach the stage of an Investigational New Drug (IND) filing. Unfortunately, the
`formulation scientist may not become involved until the drug has already encountered
`difficulties in animal studies. Thus, it is essential for the formulation scientist to work
`closely with the discovery research team, the pharmacokinetics department, and the
`toxicology department prior to the decision to move the drug into full scale
`development.
`
`After all the difficulties are resolved in the early development stages, many protein
`and peptide drugs can still encounter problems in the clinic. The major clinical
`hurdles may be similar to those observed in the pre-IND animal studies. However,
`the company may have filed an IND for a therapeutic indication that will encounter
`complex formulation and delivery problems. The route and frequency of
`administration and the bioactivity or potency of the drug in humans are critical issues
`that are often not addressed in the pre-IND animal studies. If difficulties in delivery
`or potency of the drug arise during clinical trials, the formulation scientist along with
`others on the development team must reconsider the design of both the drug
`formulation and the clinical plan. These pitfalls may often be avoided by testing the
`drug in a suitable animal model, if available, and an extensive analysis of the patient
`population including a marketing survey of the end users (physicians, nurses, and/or
`patients). By establishing early in the development stage (e.g. between research and
`Phase I clinical trials) the best route and formulation for the drug, the potential for a
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`2
`
`

`
`1. CLELAND AND I.ANGER Design and Development Strategies
`
`3
`
`Physicochemical
`Properties of Drug
`
`•In Vivo
`and
`In Vitro
`Stability
`
`• _Biological
`Function
`•Potency
`
`Phannacokinetics
`& Toxicity
`
`__ .._,
`
`Clinical Indications
`(Acute/Chronic)
`
`Figure 1: Key factors influencing the design of drug formulations and delivery. The
`physicochemical properties of the drug can affect the pharmacokinetics and toxicity
`as well as the clinical indication. The in vitro and in vivo stability of the drug
`determines its fate upon administration. The potential clinical utility of the drug is
`dependent upon the drug characteristics, biological function, and potency. To obtain
`the desired pharmocological response, a drug must be administered with a stable
`formulation. The design of a delivery system must also consider the clinical
`indication, pharmacokinetics, pharmacodynamics, toxicology, and drug properties.
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`3
`
`

`
`4
`
`FORMUIATION AND DELIVERY OF PROTEINS AND PEPTIDES
`
`clinically successful product and, ultimately, a marketed product increases
`dramatically.
`
`The best route for delivery of a protein or peptide drug is often not investigated
`during the research stage or early in development. The protein or peptide is
`commonly administered systemically through an intravenous (i.v.) injection in initial
`animal testing. Thus, for indications that require a high local dose of the drug at the
`target site, high drug doses are required by i.v. injections. Due to toxicity problems,
`the efficacious dose may not be reached via i.v. administration. More recently,
`alternative routes of delivery have been studied. In particular, the therapeutic protein,
`recombinant human deoxyribonuclease I (rhDNAse), must be delivered directly to the
`lung of cystic fibrosis patients to degrade the DNA in the mucus. rhDNAse delivered
`systemically would clearly have little effect on the target site. While this example is
`an obvious candidate for an alternate delivery route (aerosol delivery of rhDNAse),
`many other proteins and peptides may also benefit from alternative routes of delivery
`for therapeutic or clinical reasons. It is therefore essential to investigate the site of
`action and assess any side effects before choosing a route of administration.
`
`In addition, when companies are developing competitive products, the future sales of
`the product may rest upon the superior formulation and delivery of the drug,
`assuming that the efficacy of the competing products are similar. For example, many
`existing therapeutic proteins such as human growth hormone and insulin are
`administered chronically requiring daily injections. Competitors with superior drug
`formulations that release a sustained level of the protein and, thus, require less
`frequent injections would dominate the market. An example of competing products is
`the development of sustained release formulations for a luteini~ing hormone-releasing
`hormone (LHRH) agonists. Takeda Pharmaceuticals developed an LHRH agonist
`(leuprolide acetate) - polylactide-coglycolide formulation that could be administered
`monthly and provided a continuous sustained therapeutic level of LHRH for one
`month (2-5). This product, Lupron Depot®, had a ¥57 billion (-$570 million) market
`in 1992 for prostate cancer, precocious puberty and endometriosis indications and
`competition from other types of LHRH agonist formulations, including daily
`injections and daily nasal delivery, have been insignificant (6). Similar competitive
`products also consist of controlled release systems using polylactide-coglycolide with
`different LHRH agonists (goserelin acetate, Zoladex®, 7, triptorelin, Decapeptyl®,
`8). Overall, the clinical administration, patient compliance, pharmacokinetics,
`toxicity, and physicochemical properties of the drug must be considered to
`successfully develop a pharmaceutical protein or peptide drug.
`
`Formulation Development Considerations
`
`While development of novel delivery routes or systems is often necessarY,, the first
`step in development of any protein or peptide drug formulation involves the, complete
`characterization of the drug properties and its stability in different formulations.
`Typically, a formulation scientist will begin by considering the physicochemical
`properties of the protein such as the isoelectric point, molecular weight, glyeosylation
`or other post-translational modification, and overall amino acid composition. These
`properties along with any known behavior of the drug in different solutions (e.g.
`different buffers, cofactors, etc.) as well as its in vivo behavior should guide the
`choice of formulation components for testing in the initial screen of candidate
`formulations. The potential candidate formulations are composed of U. S. ·Food and
`Drug Administration (FDA) approved buffer components, excipients,' and any
`required cofactors (e.g. metal ions). Often, the first choice of candidate formulations
`is based upon the previous experience of the formulation scientist with other proteins
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`4
`
`

`
`1. CLELAND AND IANGER Design and Develop111£111 Strategies
`
`5
`
`or peptides and, in many cases, a simple phosphate buffered saline solution may be
`one of the initial candidates.
`
`A simplified approach to formulation development may proceed through the steps
`depicted in Figure 2. After obtaining all the available background information, one
`often evaluates several parameters in the initial screen of candidate formulations.
`One parameter that impacts all the major degradation pathways is the solution pH.
`Thus, the initial formulations also assess the pH dependence of the degradation
`reactions and the mechanism for degradation can often be determined from the pH
`dependence (9). The formulation scientist must quickly analyze the stability of the
`protein in each solution. Rapid screening methods usually involve the use of
`accelerated stability at elevated temperatures (e.g. 40° C; see references 10-13 for
`discussions of elevated temperature studies). Unfortunately, the FDA will only
`accept real time stability data for shelf life and accelerated stability studies may only
`serve as a tool for formulation screening and stability issues related to shipping or
`storage at room temperature. The degradation of the protein for both accelerated and
`real time studies is then followed by assays developed for analysis of degradation
`products (see reference 14 for detailed review). The most common degradation
`pathways for proteins and peptides are listed in Table I. Several recent reviews have
`analyzed these pathways as well as potential methods to prevent degradation (11, 15-
`18). In each case, the amount of degradation must be minimized to achieve greater
`than or equal to 90% of the original drug composition after 2 years (e.g. t 90 ~ 2
`years). The FDA usually requires that a pharmaceutical product is not more than 10%
`degraded and the company must demonstrate that the degradation products do not
`have any adverse effects on the safety or efficacy of the drug. Many proteins and
`peptides can degrade extensively without effecting either their safety or efficacy. For
`example, 70% deamidated recombinant human growth hormone (rhGH) is fully
`bioactive and non-immunogenic, but this extent of degradation is not acceptable by
`regulatory agency standards for a therapeutic protein (J 9). The effect of degradation
`on the safety and efficacy of a protein or peptide is difficult to ascertain without
`extensive testing. Thus, the more conservative standards of the FDA and other
`regulatory agencies may often provide a less expensive alternative if a stable
`formulation(> 2 year shelf-life) can be developed.
`
`To fulfill the regulatory requirements for a stable formulation, the scientist must
`consider all of the major degradation routes and the potential conditions for
`optimization. In the case of aggregation, the addition of surfactants or sugars can
`prevent denaturation events that lead to irreversible aggregation. If the deamidation
`rate is the dominant degradation route, the use of amine buffers such as Tris,
`ammonium, or imidazole may slow the deamidation. Alternatively, a reduction in pH
`will also decrease the deamidation rate, but the reduced pH may also lead to cleavage
`or cyclization at Asp-X residues where Xis usually a residue with a small side chain
`(e.g. Gly or Ser) and this degradation has been observed in several proteins (J).
`Proteins with Asp-X degradation must then be placed in a higher pH buffer to avoid
`cleavage or cyclization. High pH conditions (> pH 8) will however catalyze
`oxidation, thiol disulfide exchange, and p-elimination reactions. These degradation
`pathways may be inhibited by the addition of free radical and thiol scavengers such as
`methionine. In addition, the method used to prevent one type of degradation may
`influence another degradation pathway. For example, by adding surfactants or other
`polymers to prevent aggregation, the residual peroxide in the surfactant may cause a
`more rapid oxidation (20). In some cases, the formulation pH must be reduced to
`decrease the rate of deamidation. Reducing the pH may also alter the solubility of the
`protein since many proteins have isoelectric points at or near the optimal pH (pH 5-6)
`for minimizing the deamidation rate. For each protein formulation, all the
`degradation pathways must be evaluated and often a balance must be achieved
`between the different degradation pathways.
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`5
`
`

`
`6
`
`FORMULATION AND DELIVERY OF PROTEINS AND PEPTIDES
`
`Protein or Peptide Drug
`Candidate lndentified
`
`hysicochemical Properties
`(pl, MW, composition,
`solution behavior, etc.)
`
`In Vivo Information
`(site of action, clinical
`application, toxicity,
`pharmacokinetics)
`
`Choose initial formulations
`and perform elevated T
`stability studies (liquid).
`
`Determine major degradation routes.
`
`Ai:mi:ation
`- Add surfactants
`- Alter buffer conditions
`or add other excipients
`to increase solubility
`
`Oxidation
`- Remove headspace 02
`- Remove excipients with
`oxidizing agents
`- Add antioxidants
`
`Deamidation
`- Change buffer to reduce rate
`Tris> NH3 > imidazole - C032- > H2P04"
`Slow rate -----------> Fast rate
`- Reduce pH to 4-5.
`Thiol Disulfide Exchani:e and a Elimination
`
`Aspartate Reactions
`- H Asp-Pro cleavage, raise pH.
`- H Asp-X cyclization, raise pH.
`
`- Reduce pH, remove metals, remove oxidizing
`excipients, add thiol scavengers or antioxidants.
`
`Balance Competing Degradation Pathways
`or Consider Lyophilization
`
`I Lyophilizationl
`Aggregation
`Oxidation
`- Reduce 0 2
`- Lyoprotectants
`- Cryprotectants
`- Surface area
`- Surface area
`- Residual moisture
`
`Screen next round of formulations
`1------~..t - Accelerated stability at elevated T
`
`No
`
`Figure 2: Simplified process diagram for formulation development (See Table I and
`text for detailed discussion).
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`6
`
`

`
`oo-
`'-'O
`~o
`"'.i::
`u u
`ro
`.
`. r--
`"''D
`.Don
`:lo
`::::: ""'
`0..'
`.&°' --.i::
`.. °'
`' _.,,.
`on <2 --ON
`
`NO
`
`0 ...-; _o
`.i:: ':"':
`U·-a o
`~"Cl
`"""' o~
`
`N -
`N
`Na\
`
`r--
`
`..
`
`~~
`,...: ~
`!<I'.
`-~ "°' ro
`.nO
`"Cl " " '0
`"Cl·-
`~~
`"2 ;.::::
`;s: .D
`::l
`0
`o~
`
`1. CLELAND AND LANGER Design and Development Strategies
`
`7
`
`Table I: Common Degradation Routes for Proteins and Peptides in Aqueous
`Solutions a
`
`Degradation Route Region Effected/Results
`
`Major Factors
`
`Aggregation
`
`Whole protein; reversible or
`irreversible self-association
`
`Shear, surface area,
`surfactants, pH, T,
`buffers, ionic strength
`
`Deamidation
`
`Asn or Gln; acidic product,
`isoform, or hydrolysis
`
`pH, T, buffers
`ionic strength
`
`Isomerization/
`Cyclization
`
`Asn-X, Asp-X (X= Gly or Ser);
`basic product
`
`pH, T, buffers
`ionic strength
`
`Cleavage
`
`Oxidation
`
`Asp-X; fragments
`(proteolysis also possible from
`trace proteases)
`
`Met, Cys, His, Trp, Tyr;
`oxidized forms
`
`pH, T, buffers
`
`Oxygen {ions, radicals,
`peroxide), light, pH, T,
`buffers, metals,
`{surfactants), free radical
`scavengers
`
`Thiol Disulfide
`Exchange
`
`Cys; mixed disulfides:
`intermolecular or intramolecular
`
`pH, T, buffers, metals,
`thiol scavengers
`
`~ Elimination
`
`Cys; dehydroalanine, free
`thiol
`
`pH, T, buffers, oxygen
`{ions, radicals, peroxide),
`metals
`
`a This table lists degradation pathways commonly observed for proteins and peptides.
`However, this list is not comprehensive and many of these degradation routes may
`occur independently or in combination with one another.
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`7
`
`

`
`8
`
`FORMULATION AND DELIVERY OF PROTEINS AND PEPTIDES
`
`The formulation scientist also has the option of developing a solid formulation such
`as a lyophilized powder. The removal of excess water from the formulation
`minimizes the degradation rates for deamidation and hydrolysis. The residual
`moisture in a solid protein or peptide formulation can contribute to the physical
`stability of the protein by preventing its denaturation and subsequent aggregation
`upon reconstitution. Recent studies on lyophilization of proteins have shown that in
`the absence of excipients, proteins require some residual water, usually referred to as
`bound water, for stability (21). In the presence of excipients such as sugars, the
`amount of remaining water may often be reduced to levels below the hydration layer
`(22). Two different theories are currently used to explain the excipient stabilization
`of proteins and peptides in the lyophilized state. The first theory is based upon the
`observed differences in crystallinity for each excipient in the dry state. The major
`differences in each excipient have been correlated to their glass transition temperature
`(23). This theory neglects the specific interactions between excipients during drying
`and in the final dried state. The second theory, often referred to as the water
`replacement hypothesis, contends that some excipients can substitute for water in the
`dried state and thereby provide stabilization (24, 25). However, this theory also has
`its faults since many excipients with similar hydrogen bonding characteristics provide
`different degrees of stabilization (e.g. mannitol versus trehalose). Recent work by
`Carpenter and coworkers has demonstrated that there may be two distinct
`mechanisms for excipient stabilization. These mechanisms include protection during
`freezing (cryoprotectants) and drying (lyoprotectants) (26). The specific interactions
`between proteins and excipients with these properties have not yet been determined,
`but it is probable that an understanding of these interactions will lead to more rational
`design of solid protein and peptide formulations. While these studies focus on the
`prevention of denaturation and aggregation of proteins in the lyophilized state, other
`degradation routes are also critical for solid protein and peptide formulations. For
`example, residual oxygen head space in a vial containing the solid formulation can
`affect the rate of oxidation (22). Therefore, the degradation issues for lyophilized
`formulations are comparable to liquid formulations, but deamidation and hydrolysis
`rates are usually slower in the solid state.
`
`Once the decision has been made to narrow the formulation candidates or proceed
`with a lyophilized formulation, another round of screening at elevated temperatures is
`usually performed. If these studies account for the potential differences between real
`time data at the actual storage conditions (usually 2-8° C) as described elsewhere (10-
`13), then the formulation scientist can select two or three final candidate formulations
`that should have a greater than 2 year shelf-life for real time stability studies.
`Unfortunately, the formulation scientist is often asked to have a stable formulation
`within a very short time (< 6 months). Thus, the real time stability data is often
`limited, but these data are required for the IND filing since the FDA will not accept
`any accelerated stability data. Stability problems encountered later are treated as
`amendments during the clinical trials. Further changes after the IND filing may
`include scale-up of the manufacturing process including the formulation. The scale(cid:173)
`up of formulations may include new processing and storage containers, bulk filling
`equipment, and new delivery systems or vial confiqurations. These modifications
`may require another evaluation of the formulation with additional optimization.
`Hopefully, any necessary changes in the formulation already in clinical trials will not
`alter the in vivo characteristics of the drug (e.g. clearance, immunogenicity or
`potency).
`
`The Crucial Steps in Delivery
`
`While considering the alternatives for formulating a protein or peptide drug, the
`formulation scientist must also consider the route of administration. As mentioned
`
`In Fommlation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`8
`
`

`
`1. CLELAND AND LANGER Design and Development Strategies
`
`previously, the route of administration can often be a critical variable in determining
`the success of the final product. As shown in Table II, there are many potential routes
`for the delivery of protein or peptide drugs. For proteins, only the direct injection and
`pulmonary routes of delivery have been approved by the FDA. Due to their large
`size, susceptibility to proteolytic degradation and requirements for an intact tertiary
`structure, proteins are difficult to deliver by oral, topical, or transdermal routes. Oral
`delivery of peptides and proteins usually results in low bioavailability ( < 10%) and
`attempts to make stable prodrugs of peptides, which are more readily absorbed have
`provided some increase in the bioavailability (28). Unlike oral delivery, topical and
`transdermal delivery routes may be efficient enough for a localized treatment of skin
`disorders or diseases and, perhaps, for other localized therapies. Another method for
`protein delivery is the use of depot systems. Several companies are investigating
`their potential application. The depot systems offer the opportunity to localize the
`drug to the target site, reduce the frequency of injections, extend the half-life of the
`drug, and enhance its in vivo stability. Unfortunately, these systems also have
`inherent disadvantages that have not yet been overcome for their successful
`development into commercial products. A major disadvantage is often the use of
`harsh conditions (e.g. organic solvents or high temperature) to produce the depot,
`In addition, for a long term depot
`resulting in denaturation of the protein (29).
`formulation, the protein must be stabilized in an aqueous environment under
`physiological conditions at 37° C. Significant degradation has been observed for
`proteins stored under conditions analogous to those encountered in the depot (30, 31).
`For most of the novel delivery routes or depot formulations, the environment at the
`site of administration must be considered and stability studies of the drug in the same
`environment (e.g. serum) should be performed to assure that complete degradation
`does not occur before the drug reaches the desired site of action (32). Thus, the
`successful development of these formulations requires the initial development of
`protein formulations that are stable to both the process conditions and the in vivo
`environment.
`
`Design of Depot Systems for Delivery. After the development of a formulation that
`meets these criteria, the material for the depot system must be chosen. The material
`should be biodegradable, well characterized, and nontoxic. The depot should also
`not alter the pharmacological properties of the drug and should act only as an inert
`carrier. Several examples of materials tested for depot systems are listed in Table ill.
`While the materials derived from natural sources may often be more biocompatible,
`these materials may vary in their physical characteristics and can be difficult to obtain
`· in a highly purified form. Many of the natural materials are derived from
`recombinant or animal sources and may contain contaminants such as endotoxins. In
`addition, some natural materials such as collagen or other proteins may invoke an
`unwanted immune response. In contrast, the synthetic materials are usually well
`characterized, highly pure polymers. The synthetic polymers have different physical
`and chemical characteristics. The polyanhydrides, polyesters, polyiminocarbonates,
`and polycaprolactones require the use of organic solvents or high temperatures for
`drug formulation. These systems are however well characterized and can be made
`reproducibly. On the other hand, the hydrogels, polyamino acids, and
`polyphosphazenes can be used to encapsulated drugs in an aqueous environment
`without organic solvents or elevated temperatures. These polymers are usually less
`stable and additional development is required to completely characterize their
`properties for controlled release of proteins and peptides. Of the synthetic materials,
`only the polyesters, specifically polylactic-coglycolic acid (PLGA) and polylactic
`acid (PLA), are currently used in commercial depot systems, Lupron Depot®(2-5)
`and Zoladex® (58, 59). These products were recently approved as alternate
`formulations and the polymer matrix, PLGA, has been well characterized and has
`been used extensively in humans. These polymers have been used for over twenty
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`9
`
`

`
`10
`
`FORMULATION AND DELIVERY OF PROTEINS AND PEPTIDES
`
`Table II: Routes of Delivery for Proteins and Peptides
`
`Delivery Routes
`
`Formulation and Device
`Requirements
`
`Commercial
`Productsa
`
`Invasive
`
`Direct Injection
`
`Liquid or reconstituted solid, syringe
`
`intravenous (i. v.)
`subcutaneous (s.q.)
`intramuscular (i.m.)
`intracerebral vein (i.c.v.)
`
`Depot system
`
`Biodegradable polymers, liposomes
`permeable polymers (not degradable),
`microspheres, implants
`
`LHRH analogs (s.q. or i.m.)
`
`Noninvasive (see reference27 for review)
`
`Activase®
`Nutropin®
`RecombiVax®
`
`Lupron Depot®
`Zoladex®
`Decapeptyl®
`
`Pulmozyme®
`
`Pulmonary
`
`Oral
`
`Nasal
`
`Topical
`
`Transdermal
`
`Liquid or powder formulations,
`nebulizers, metered dose inhalers,
`dry powder inhalers
`
`Solid, emulsion, microparticulates,
`absorption enhancers
`
`Liquid, usually requires permeation
`enhancers
`
`Synarel®
`
`Emulsion, cream or paste (liposomes)
`
`Electrophoretic (iontophoresis ),
`electroporation, chemical permeation
`enhancers, prodrugs, ultrasonic
`
`Buccal, Rectal, Vaginal
`
`Gels, suppositories, bioadhesives, particles
`
`a Nutropin® (recombinant human growth hormone), Activase® (recombinant human
`tissue plasminogen activator), and Pulmozyme® (recombinant human deoxyribo(cid:173)
`nuclease I) are all products of Genentech, Inc. RecombiVax® (recombinant
`Hepatitis B surface antigen) is produced by Merck & Co. Lupron Depot®
`(leuprolide acetate - PLGA) is a product of Takeda Pharmaceuticals. Zoladex®
`(goserelin acetate - PLGA) is produced by the Imperial Chemical Industries, Ltd.
`Decapeptyl® is a manufactured by Debiopharm. Synarel® (nafarelin acetate) is
`made by Syntex Corporation.
`
`In Formulation and Delivery of Proteins and Peptides; Cleland, J., et al.;
`ACS Symposium Series; American Chemical Society: Washington, DC, 1994.
`10
`
`

`
`1. CLELAND AND LANGER Design and Development Strategies
`
`11
`
`Table ID. Biodegradable Materials for Controlled Delivery of Proteins or
`Peptides
`
`Materials
`
`Degradation Mechanism
`
`Reference a
`
`Natural
`
`Starch
`
`Alginate b
`
`Collagen
`(Gelatin)
`
`Amylase
`
`pH, Enzymes
`
`Collagenase
`
`Proteins
`(Cross-linked albumin)
`
`Enzymes
`
`Tricalcium phosphate c
`or calcium carbonate
`(hydroxyapatite)
`
`Dissolves over time
`
`Synthetic
`
`Hydro gels
`
`Polyanhydrides
`
`Polyesters
`(polylactides)
`
`Chemical or enzymatic hydrolysis,
`solubilization in aqueous media
`
`Hydrolysis
`
`Ester hydrolysis, esterases
`
`Poly (ortho esters)
`
`Ester hydrolysis, esterases
`
`Polyiminocarbonates
`
`Hydrolysis
`
`Polycaprolactones
`
`Polyamino acids
`
`Hydrolysis
`
`Enzymes
`
`Polyphosphazenes b
`
`Hydrolysis, dissolution
`
`33,34
`
`35,36
`
`37-39
`
`40,41
`
`42-44
`
`45
`
`46-48
`
`49,50
`
`49
`
`51,52
`
`53,54
`
`55
`
`56,57
`
`a Reviews describing

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