REVIEW
Formulation and Stability of Cytokine Therapeutics 1 1 ¨ ¨ TIINA LIPIAINEN, MARIKKI PELTONIEMI,1 SANJAY SARKHEL,1 TEIJO YRJONEN, HEIKKI VUORELA,1 ARTO URTTI,1,2 ANNE JUPPO1 1 2
University of Helsinki, Faculty of Pharmacy, Helsinki, Finland University of Eastern Finland, School of Pharmacy, Kuopio, Finland
Received 30 June 2014; revised 29 September 2014; accepted 13 October 2014 Published online in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.24243 ABSTRACT: Cytokines are messenger proteins that regulate the proliferation and differentiation of cells and control immune responses. Interferons, interleukins, and growth factors have applications in cancer, autoimmune, and viral disease treatment. The cytokines are susceptible to chemical and physical instability. This article reviews the structure and stability issues of clinically used cytokines, as well as formulation strategies for improved stability. Some general aspects for identifying most probable stability concerns, selecting excipients, and developing stable cytokine formulations are presented. The vast group of cytokines offers possibilities for new biopharmaceuticals. C 2014 Wiley The formulation approaches of the current cytokine products could facilitate development of new biopharmaceuticals. Periodicals, Inc. and the American Pharmacists Association J Pharm Sci Keywords: biopharmaceuticals characterization; biosimilar; excipients; injectables; lyophilization; oxidation; protein aggregation; protein formulation; protein structure; solid state
INTRODUCTION Cytokines are small secreted proteins that enable receptormediated communication between cells and act as key modulators of immune responses by controlling growth, differentiation, and activation of various cell types. Hundreds of cytokines have been identified1 and some of these proteins are currently used in therapeutic products, mostly for treating cancer, autoimmune, and viral diseases (Table 1). The biological mechanisms and clinical use of cytokines have been well reviewed,1–5 but aspects of formulation and stability have been less widely studied. Here, we review cytokine therapeutics marketed in the United States and in Europe, aspects related to their structural instability, and the role of formulation excipients toward improving stability. There are decades between the market introduction of the first and the most recent cytokine products. Formulation strategies have evolved during that time and some of the first-generation products have been reformulated. The main difference to the older approaches is the current preference of albumin-free formulations. In addition, cytokines with conjugated polyethylene glycol (PEG) chains have been introduced to market. These pegylated proteins have greatly increased halflives compared with the unmodified cytokines, allowing more convenient administration regimes. Cytokines bind to the cell surface cytokine receptors with high affinity, making them high-potency molecules. Thus, cytokines are generally administered at low doses and they have a narrow therapeutic index. For this reason, structural stabilAbbreviations used: CHO, Chinese hamster ovary; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colonystimulating factor; EDTA, ethylenediaminetetraacetic acid; EMA, European Medicines Agency; Fimea, Finnish Medicines Agency; HSA, human serum albumin; MAH, marketing authorization holder; PEG, polyethylene glycol; SDS, sodium dodecyl sulfate. ¨ Correspondence to: Tiina Lipiainen (Telephone: +358-294159346; Fax: +358294159138; E-mail:
[email protected]) Journal of Pharmaceutical Sciences C 2014 Wiley Periodicals, Inc. and the American Pharmacists Association
ity is a critical factor in ensuring biological function and efficacy. Low concentrations of active ingredient cause difficulties characteristic to cytokine products. Analysis of the cytokine protein structure and stability becomes challenging, especially if the product additionally contains albumin as a stabilizing excipient. Another important result is loss of protein because of adsorption on container or other surfaces. Most cytokines have a helical bundle fold, generally associated with marked hydrophobicity.6 Hydrophobicity can lead to problems related to solubility, tendency to aggregate and adsorption on surfaces, and consequent challenges in manufacturing and long-term storage. Physical and chemical instabilities may also lead to the formation of immunogenic degradation products. Oxidation is a major chemical degradation pathway for cytokines.7–13 Cytokine aggregates, especially those formed by oxidized proteins, have been linked to immunogenicity.9,10 Immunogenicity of therapeutic proteins is a topic of concern that has gained wide interest lately.14–17 Many of the cytokine products are intended for long-term use, and many patients eventually develop an antibody response to the protein drug.15 Antibodies formed against therapeutic proteins may cause serious adverse effects if they cross-react with endogenous proteins.18 Protein aggregates are a key risk factor for immunogenicity.16 Therefore, the hydrophobicity and aggregation tendency of cytokines is a matter of concern, and a major aim in formulation development is to inhibit unfolding and aggregation. In this review, we have covered the available information on structural characteristics (Table 2) and degradation pathways of interferon-alpha (IFN-"), interferonbeta (IFN-$), interferon-gamma (IFN-(), interleukin-2 (IL-2), IL-11, granulocyte colony-stimulating factor (GCSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF). As a result, we attempt to identify common properties within these molecules and formulation strategies, and to provide a rationale for developing stable cytokine products. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
1
2
REVIEW
Table 1. Cytokine IFN-"
IFN-$
IFN-(
IL-2
IL-11
Cytokine Products Approved for Clinical Use in the United States and Europe145–147 Trade Name (Company)
Active Ingredient
Production Host
Indication
Year of Authorization (Responsible Agency)
IntronA (US: Schering, EU: Merck Sharp and Dohmea )
IFN-"-2b
E. coli
Roferon-A (Roche)
IFN-"-2a
E. coli
PegIntron (EU: Mercka ,US: Schering) ViraferonPeg (Mercka ) Sylatron (Schering) Pegasys (Roche)
Peginterferon-"-2b
E. coli
Multiple myelomab Chronic myelogenous leukaemiab Chronic hepatitis B Carcinoid tumorb Hairy cell leukemia Follicular lymphoma Malignant melanoma Chronic hepatitis C Condylomata acuminatac AIDS-related Kaposi’s sarcomac Hairy cell leukemia AIDS-related Kaposi’s sarcomab Chronic myelogenous leukaemia Cutaneous T cell lymphomab Chronic hepatitis Bb Chronic hepatitis C Follicular lymphomab Renal cancerb Malignant melanomab Chronic hepatitis C
Peginterferon-"-2b
E. coli
Chronic hepatitis C
2000 (EMA)
Peginterferon-"-2b Peginterferon-"-2a
E. coli E. coli
Infergen (US: Intermune Pharms) Alferon N Injection (Interferon Sciences)
Interferon-alfacon-1
E. coli
Melanoma Chronic hepatitis B Chronic hepatitis C Chronic hepatitis C
2011 (FDA) 2002 (EMA) 2002 (FDA) 1997 (FDA)
IFN-"-n3
Human leucocytes
Condylomata acuminata
1989 (FDA)
Betaseron (US)/Betaferon (EU) (Bayer) Extavia (Novartis)
IFN-$-1b
E. coli
Multiple sclerosis
1993 (FDA) 1995 (EMA)
IFN-$-1a
CHO cells
Multiple sclerosis
Avonex (Biogen Idec)
IFN-$-1a
CHO cells
Multiple sclerosis
Rebif (US: Serono, EU: Merck Serono Europe) Actimmune (US) (Intermune/Vidara Ther.) /Imukin (EU) (Boehringer Ingelheim) Proleukin (EU: Novartis, US: Chiron) Ontak (Eisai) Neumega (Wyeth Pharms Inc.)
IFN-$-1a
CHO cells
Multiple sclerosis
2008 (EMA) 2009 (FDA) 1996 (FDA) 1997 (EMA) 1998 (EMA) 2002 (FDA)
IFN-(-1b
E. coli
Chronic granulomatous disease Osteopetrosis
1994 (Fimea) 1999 (FDA)
Aldesleukin (rhIL-2)
E. coli
Renal cell carcinoma Melanomac
1992 (FDA) 1992 (Fimea)
Denileukin diftitox Oprelvekin (rhIL-11)
E. coli E. coli
T-cell lymphoma Chemotherapy-induced thrombocytopenia
1999 (FDA) 1997 (FDA)
1986 (FDA) 2000 (EMA)
1986 (FDA) 2001 (Fimea, Finnish Medicines Agency)
2000 (EMA) 2001 (FDA)
Continued
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
DOI 10.1002/jps.24243
REVIEW
Table 1. Cytokine G-CSF
GM-CSF
3
Continued Trade Name (Company)
Active Ingredient
Production Host
Indication
Year of Authorization (Responsible Agency)
Neupogen (Amgen)
Filgrastim (recombinant methionyl hG-CSF)
E. coli
Neutropenia Hematopoietic stem cell transplantation Cancer
1991 (FDA) 1991 (Fimea)
Biograstim (MAH: AbZ-Pharma; Batch release: Merckle Biotec) Ratiograstim (MAH: Ratiopharm; Batch release: Merckle Biotec) Tevagrastim (MAH: TEVA Generics; Batch release: Teva Pharma) Filgrastim Hexal (MAH:Hexal; Batch release: Sandoz) Zarzio (Sandoz) Nivestim (Hospira) Grastofil (Apotex Europe) Granocyte (ChugaiSanofi-Aventis)
Filgrastim Biosimilar, reference drug Neupogen
E. coli
Neutropenia Hematopoietic stem cell transplantation Cancer
2008 (EMA)
Lenograstim (rhG-CSF)
CHO cells
Neulasta (Amgen)
Pegfilgrastim (pegylated r-metHuG-CSF) Lipegfilgrastim (glycol-pegylated r-metHuG-CSF)
E. coli
Sargramostim (rhGM-CSF)
Lonquex (MAH: Teva; Batch release TEVA and Merckle Biotec) Leukine (Berlex/Sanofi)
2008 (EMA)
2008 (EMA)
2009 (EMA)
Neutropenia
2009 (EMA) 2010 (EMA) 2013 (EMA)
Neutropenia Hematopoietic stem cell transplantation Cancer Neutropenia Cancer
1994 (Fimea)
E. coli
Neutropenia
2013 (EMA)
S. cerevisiae
Bone marrow transplantation Cancer
1991 (FDA)
2002 (FDA) 2002 (EMA)
a
Marketing authorization transferred from Schering-Plough Europe in 2011–2012. Indication approved by EMA or Fimea only. Indication approved by FDA only. MAH, marketing authorization holder. b c
Pharmaceutical Protein Formulations As for proteins in general, structural integrity of a cytokine is critical for its biological function. Changes in the native structure often lead to loss of activity, as the cytokine no longer binds its receptor in a proper manner, and may cause harmful immunogenic reactions. Proteins are prone to alterations in their tertiary fold (physical instabilities, e.g., unfolding and aggregation) and/or modifications in their primary structures (chemical degradation, e.g., oxidation, deamidation, disulfide scrambling, and proteolysis).19–22 The risk of such changes is pronounced when the environmental conditions are different from the physiological conditions where the protein has evolved to function. Furthermore, the product protein concentration generally differs markedly from physiological conditions, bringing about additional stress to the protein. Product manufacturing and processing steps including purification, filtration, and filling, DOI 10.1002/jps.24243
as well as transportation, storage, and administration expose the protein to various stress factors.23 Shear forces, contacts with various interfaces, and temperature variations increase the risk of protein degradation.24 The magnitude of this risk can be greatly mitigated by efficient formulation.25–30 When formulating a pharmaceutical protein, the choice of excipients, ionic strength, and pH need to be optimized for a stable and safe product. The storage conditions and shelf-life of a product depends on the stability of the formulation. Product shelf-lives may reflect business decisions rather than absolute protein stability, though. Stability studies to extend product shelf-life can be time-consuming and expensive and might not give any further value for the company. Pharmaceutical protein formulation development is often based on previous experience and on the process of trial-anderror.21 One of the first steps of development is the choice of an ideal buffer system to maintain optimal pH. Commonly Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
165–166
165
166
143 (x2)
133
178 174
127
IFN-"
IFN-"2
IFN-$
IFN-(
IL-2
IL-11 G-CSF
GM-CSF
177 174 (rhG-CSFc ) 175 (r-methG-CSFd ) 127
132
140
166 (IFN-$-1a) 165 (IFN-$-1b)
166 (IFN-"-con) 165–166 (IFN-"-n3) 165
Product
15–23
19.0 ∼20
15–18
30–50
∼23
19–21
17.5–22.0
Natural
19.0 ∼20 (rhG-CSF) 18.8 (r-metHuG-CSF) 19.5, 16.8, and 15.5
15.3
16.5 (x2)
19.4 (IFN-"-con) 16.0–27.0 (IFN-"-n3) 19.2 (IFN-"-2a) IFN-"-2b 22.5 (IFN-$-1a) 18.5 (IFN-$-1b)
Product
Molecular Weight (kDa)
Yes (six sites)
No Yes (one site)
No Yes (rhG-CSF) No (r-methG-CSF) Yes
b
No
No
Yes/Noa (two sites) Yes (one site)
Yes (IFN-$-1a) No (IFN-$-1b)
No
No
Product
Yes
Yes (one site)
Most subtypes not (one site)
Natural
Glycosylation
IFN-( has two glycosylation sites on each monomer, and of these 0, 1, or 2 may be occupied by a carbohydrate chain. 3D structure has not been verified. c Lenograstim. d Filgrastim.
a
Natural
Number of Amino Acids
Summary of Cytokine Protein Characteristics
Cytokine
Table 2.
Two
One (+ one free Cys. In rhIL-2, Cys is substituted by Ser.) 0 Two (+ one free Cys)
One (+ one free Cys. In IFN-$-1b, Cys is substituted by Ser.) 0
Two
Two
Number of Disulfide Bridges
Helical bundle
Helical bundle Helical bundle
Dimer of two helical bundles Helical bundle
Helical bundleb Helical bundle
Helical bundle, also $-sheet content
∼6 ∼6 ∼9 ∼10
∼11 ∼6 ∼5
∼7
Overall Fold
Isoelectric Point
4 REVIEW
DOI 10.1002/jps.24243
REVIEW
used buffers include phosphate (pKa = 7.21), acetate (pKa = 4.76), citrate (pKa = 6.39), histidine (pKa = 6.0), and Tris(hydroxymethyl)aminomethane (pKa = 8.10) in pH ranges of 4–7.21 Pain perception associated with injection may vary with buffer choice. Subcutaneous injection of a citrate-buffered solution was found more painful than a histidine-containing solution, for example.31 Often a liquid formulation is desired. In case long-term stability cannot be achieved by liquid formulation, the formulation may be dried by lyophilization. In the dry solid state, degradation reactions are generally slowed down, facilitating storage at higher temperatures. This is because of greatly reduced water content and molecular mobility compared with solutions. However, the drying process and storage in the dry state cause stress to the protein. Therefore, additional stability-enhancing excipients are usually required. As a general rule, minimum number of excipients necessary should be used, but specific needs relating to inhibition of protein degradation and administration route requirements often necessitate the inclusion of many excipients. The most commonly used excipients include protein stabilizers (e.g., sugars, polyols, polymers, and amino acids), albumin, surfactants (e.g., polysorbates and poloxamers), bulking agents for dry formulations (e.g., mannitol and glycine), antioxidants (e.g., methionine), chelating agents [ethylenediaminetetraacetic acid (EDTA) and citric acid], tonicity modifiers (e.g., NaCl and sucrose), and preservatives for multidose products (e.g., benzyl alcohol, phenol, metacresol, and chlorbutanol).21,26,27,29,30 Formulations of therapeutic cytokine products currently on the market are presented in Table 2. The roles of specific excipients in the formulations and stability of each cytokine are discussed in the following sections. Interferon Alpha Interferon-alpha was the first recombinant cytokine approved for therapeutic use (IntronA and Roferon-A in 1986). It has antiviral, antiproliferative, and immunoregulatory properties, mechanisms of which are still not completely understood. Several reviews have covered this topic of interest.32–34 The diverse biological activities of IFN-" render therapeutic applications in cancer and viral disease treatment (Table 1). Interferon-alpha is a family of multiple protein species.34 Fourteen genes encode for different interferon alphas harboring post-translational modifications. They are composed of 165 or 166 amino acids and contain two conserved disulfide bonds. Some IFN-"s are glycosylated and their molecular weights vary in the range of 17.5–22 kDa. Alferon, a product manufactured from pooled human leukocytes, contains a mixture of interferon subtypes (at least 14) as active ingredient labeled as IFN-"-n3.35 The product is claimed to have reduced immunogenicity compared with the other interferon products, probably because of the presence of natural glycosylation and multiple subtypes instead of only one.35 However, there are safety concerns related to natural source materials because of the risk of blood-borne diseases in plasma-derived products.3 All other products contain recombinant proteins, either IFN-"2 or interferon alfacon-1. The latter is a non-naturally occurring protein, with a synthetic consensus sequence derived by scanning several natural IFN-" subtype sequences and selecting the most frequently observed amino acid in each corresponding position. Interferon alfacon-1 possibly has higher activity than the natural interferons.36 DOI 10.1002/jps.24243
5
Interferon-alpha-2 is the most widely used and the best characterized IFN-". It contains 165 amino acids and occurs naturally in three subtypes (2a, 2b, and 2c). IFN-"-2a is the active ingredient in products marketed by Roche (Roferon-A and Pegasys), whereas the Schering/Merck products (IntronA, PegIntron ViraferonPeg, and Sylatron) contain IFN-"-2b as the active component. These two variants only differ by one amino acid: position 23 is occupied by lysine in IFN-"-2a and by arginine in IFN-"-2b. The variants have similar structure and biological activity. In both, the three-dimensional structure is formed by a cluster of five "-helices.37,38 Although topographically similar to other helical bundle proteins, the fifth helix (also present in IFN-$) is a unique feature, distinguishing the IFN-fold. Klaus et al.37 have suggested that IFN-"-2a might have more stability in the monomeric form than IFN-"-2b when stressed at high concentration and low ionic strength. Two pegylated forms of IFN-" that are less prone to proteolytic degradation and renal clearance have been developed to improve half-life and efficacy. This allows reduced dosing frequency, from three times a week to once a week. However, pegylation has also raised concerns. PEG-linked proteins with molecular weights less than 70 kDa have resulted in renal accumulation in rats because the PEG molecules cannot be degraded in the lysosomes.39 Furthermore, there has been discussion about the immunogenicity of the PEG moiety itself; however, the conclusions are controversial.40,41 Pegasys contains IFN-"-2a conjugated with a branched 40 kDa PEG moiety, characterized as a mixture of nine positional isomers.42 Pegylated IFN-"-2b is the active ingredient in the identical formulations PegIntron and ViraferonPeg. PegIntron comprises 14 different monopegylated variants (positional isomers), as well as dipegylated and unpegylated (<5%) forms of IFN-"-2a.43 The PEG moiety in PegIntron/ViraferonPeg is a linear 12 kDa polymer. The average molecular weight of the conjugated protein is about half lower as compared with Pegasys (31 kDa vs. 63 kDa). The biological activities and serum half-lives of these two modified interferons are not identical. Sylatron is approved for melanoma treatment by US FDA with a formulation identical to PegIntron except for a higher Peg-IFN-"-2a concentration. The natural human IFN-"2 is glycosylated (O-linked sugar chain at Thr-106) with variation in the sugar composition leading to size heterogeneity of 19–21 kDa.44 However, the therapeutically used recombinant IFN-"s are nonglycosylated and less hydrophilic than the natural protein. The lack of glycosylation does not cause a dramatic reduction in solubility, though, as is the case for recombinant IFN-$.44 The proteins are described water soluble in the manufacturers’ documentation. Interestingly, and somewhat unexpectedly (compared with IFN-$), the glycosylation results in slightly decreased thermal stability of IFN-"-2b (melting temperature decreases from about 65◦ C–66◦ C to 64◦ C).45 Interferon-alpha is acid labile and undergoes structural changes and unfolding below pH 4.46,47 IFN-"-2b is reported to have maximal conformational stability at pH 7, where it may be self-associated as dimers or even higher-order oligomers.46,48 Self-association might favor formation of aggregates and for this reason ways to suppress it have been evaluated. Conjugation to PEG moieties or albumin, or lowering the pH to 4, and increasing ionic strength (e.g., 400 mM NaCl) could change the self-association behavior.48 The marketed products containing IFN-"-2b, IFN-alfacon, or IFN-"-n3 are formulated in pH 7 phosphate buffer. Apparently, the possible self-association does Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
6
REVIEW
not adversely affect biological activity of IFN-". Different from the other IFN-" products, the IFN-"-2a formulations have acetate buffer and lower pH (Roferon pH 547,49 and Pegasys pH 6). The acetate-containing products are only available as liquid products. Dry product development would require change of buffer system, as acetate is volatile and partially sublimes during lyophilization.25 Loss of acetate results in altered buffer composition and pH shift in the lyophilized product. Interferon-alpha-2a adsorbs to borosilicate glass surfaces, causing loss of protein and possible induction of denaturation especially at low protein concentrations.7 Potential surface adsorption has also been reported for dilute Pegasys samples.42 Polysorbate 80 inhibits adsorption by competing with the protein for the hydrophobic surfaces and interfaces,7,50 and it is included in nearly all IFN-" formulations (at 0.05–0.2 mg/mL). Polysorbate-free IFN-" formulations contain albumin that is known to compete with proteins for surface adsorption sites.51 In fact, polysorbate and albumin seem to be interchangeable in aqueous IFN-" solutions.52 Infergen is a rare cytokine product without surfactant or albumin. This is noteworthy because the IFN concentration in Infergen is low (30 :g/mL) and loss of protein because of surface adsorption is expected. Perhaps the modified sequence of IFN-alfacon results in different surface adsorption tendency compared with the natural sequence IFN-"s. In addition to glass surfaces, contact with chlorobutyl stoppers and the process of sealing the ampoules by heating may cause degradation. This was reported for IFN-"-2b in phosphate buffer, where adsorption and chemical instability (appearance of degradation impurities in HPLC) was observed.7 Improved stability was achieved in citrate or sodium-citrate phosphate buffer, or by adding EDTA or polysorbate 80 in the formulation. EDTA and citric acid act as chelators that bind metal ions.28 Redox-active transition metal ions such as Fe, Cu, Mn, and Cr have the potential to initiate chemical degradation, oxidation in particular.28 These metal ions are often found in pharmaceutical preparations, originated from impurities in excipients (especially in sugars and polymers), or from storage and processing containers (e.g., stainless steel manufacturing vessels).22 Phosphate is not generally named as a metal-chelating excipient for protein formulations, but it might prevent oxidative damage of proteins by binding iron.53 Interferon-alpha contains amino acid residues that are susceptible to oxidation and deamidation reactions. Formation of methionine sulfoxide was suggested as one of the degradation products induced by IFN-"-2b contact with chlorobutyl stoppers.7 Various methionine sulfoxide forms were also observed in another IFN-"-2b stability study; however, deamidation products could not be detected.54 Furthermore, an oxidized form was identified in bulk IFN-"-2b solution, where methionine sulfoxide was present at about 3%. However, it was concluded not to impair the biological activity.8 Even though the oxidation products might not directly affect activity, oxidation is clearly a concern. Oxidized IFN-"-2b is prone to aggregate and in particular, metal-catalyzed oxidation might result in the formation of highly immunogenic aggregates.9 To mitigate the risk of oxidation and immunogenicity from aggregates, EDTA has been included in the IntronA solution formulation. It is the only cytokine formulation with EDTA. Surprisingly, no metal chelators are included in the other IFN-" products and no literature is available on IFN-"-2a oxidation. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
All liquid IFN-" formulations contain sodium chloride as tonicity modifier, whereas the two powder formulations have glycine and sucrose for that purpose (Table 3). Sucrose also acts as a lyoprotectant, providing stability to the dried PEGIFN product. In the dry powder formulation of IntronA, the bulking agent glycine is combined with albumin, which also has stability-enhancing roles as lyoprotectant and cryoprotectant. The multidose products comprise m-cresol, benzyl alcohol, or phenol as antimicrobial preservatives. Infergen represents one of the simplest formulations of all cytokine products: sodium chloride in phosphate buffer. The pegylated IFN products have shelf-lives of 3 years: the longest among all cytokine formulations. Interestingly, a liquid formulation PegIntron was not introduced to the market because of depegylation on storage in this form.55 In contrast, Pegasys is formulated as a solution and it showed stability problems because of PEG-IFN oxidation.56 Degradation of PEG-IFN via oxidation was inhibited by addition of benzyl alcohol,56 although benzyl alcohol is not commonly known to possess antioxidant properties. Interferon Beta Interferon beta belongs to the same category of Type I interferons, binds to the same receptor complex, and activates similar, though not identical, antiviral, antiproliferative, and immunomodulatory effects as IFN-".34 Products containing rhIFN-$ as active substance are used to treat patients with multiple sclerosis. Unlike IFN-", human IFN-$ is only encoded by one gene. IFN-$ is a glycoprotein composed of 166 amino acid residues with a molecular weight of about 23 kDa.57 It has one disulfide bridge, one free cysteine residue, and the tertiary structure is composed of five "-helices, the overall fold being similar to that of IFN-".57 The recombinant protein in Avonex and Rebif, IFN$-1a, is produced in Chinese hamster ovary (CHO) cells. It has an identical sequence and similar glycosylation pattern as the natural protein. On the contrary, the active ingredient in Betaseron, Betaferon, and Extavia, IFN-$-1b, is not glycosylated and has a lower molecular weight of 19 kDa. Furthermore, the sequence of IFN-$-1b has been modified by deleting a methionine residue (Met-1) and substituting cysteine (Cys-17) to serine, for enhanced stability. The free cysteine residue in the native protein is buried but close to the surface, and will readily participate in disulfide scrambling in an event of unfolding, leading to heavy aggregation.57 Despite the high costs of protein production in mammalian cell lines (in this case CHO cells) compared with bacterial fermentation, it has clear advantages with IFN-$. The N-linked carbohydrate complex attached at Asn-80 shows some variation in its composition, but it plays a key role as its removal dramatically reduces solubility of the protein.58 Importantly, Runkel et al.59 reported that the specific activity of IFN-$-1b is 10 times lower compared with IFN-$-1a. The reduction in activity is caused by the lack of glycosylation and the resulting decrease in solubility, not by the sequence modifications.59 They accounted for 60% of IFN-$-1b in Betaseron being lost because of aggregation when exposed to physiological pH. There is a high number of hydrophobic side chains located on the protein surface close to the glycosylation site and the carbohydrate complex exhibits an unusually high degree of order. Therefore, it is believed that the carbohydrates shield the side chains from solvent exposure and stabilize the folded state of the protein.60 DOI 10.1002/jps.24243
DOI 10.1002/jps.24243
Alferon
Infergen
Pegasys
Sylatron
PegIntron/ ViraferonPeg
Roferon-A
IntronA
IFN-α–2b (23–192 µg/mL) Dibasic sodium phosphate anhydrous (1.8 mg/mL) Monobasic sodium phosphate monohydrate (1.3 mg/mL) Edetate disodium (0.1 mg/mL) Sodium chloride (7.5 mg/mL) m -Cresol (1.5 mg/mL) Polysorbate 80 (0.1 mg/mL) Water for injections (ad 0.5–3.0 mL) Lyophilized powder c IFN-α-2b (38–192 µg) Glycine (20 mg) Sodium phosphate dibasic (2.3 mg) Sodium phosphate monobasic (0.55 mg) Human albumin (1.0 mg) Solvent (1.0 mL): water for injections IFN-α-2a (22 –110 µg/mL) Ammonium acetate (0.77 mg/mL) Sodium chloride (7.2 mg/mL) Benzyl alcohol (10 mg/mL) Polysorbate 80 (0.2 mg/mL) (Acetic acid, Sodium hydroxide) Water for injections (ad 0.5 mL) Lyophilized powder: Peg-IFN-α-2b (74–222 µg) Dibasic sodium phosphate anhydrous (1.11 mg) Monobasic sodium phosphate dihydrate (1.11 mg) Sucrose (59.2 mg) Polysorbate 80 (0.074 mg) Solvent (0.7 mL): water for injections Lyophilized powder: Peg-IFN-α-2b (296 –888 µg) Excipients identical to PegIntron. Peg-IFN-α-2a (135–360 µg/mL) Sodium chloride (8.0 mg/mL) Polysorbate 80 (0.05 mg/mL) Benzyl alcohol (10 mg/mL) Sodium acetate trihydrate (2.6 2 mg/mL) Acetic acid (0.05 mg/mL) Water for injections (ad 0.5 or 1.0 mL), pH 6 ± 0.5 Interferon alfacon-1 (30 µg/mL) Sodium chloride (5.9 mg/mL) Sodium phosphate (3.8 mg/mL) Water for injections (ad 0.3 or 0.5 mL), pH 7.0 ± 0.2 IFN-α-n3 (25 µg/mL) Phenol (3.3 mg/mL) Human albumin (1.0 mg/mL) Sodium chloride (8.0 mg/mL) Sodium phosphate dibasic (1.7 mg/mL) Potassium phosphate monobasic (0.20 mg/mL) Potassium chloride (0.20 mg/mL) Water for injections (ad 1.0 mL), pH 7.4
Formulation (Shaded Background for Liquid Products)
Vial (single use)
Vial
SC Three times a week or daily
IL Two time s a week
Vial, prefilled syringe, prefilled pen/autoinjector (single use )
Vial (single use)
SC Once a week
SC Once a week
Vial Prefilled pen (single use)
Prefilled syringe (single use)
SC Daily or three times a week
SC Once a week
Vial (single use )c
Vial (s ingle use b and multidose) Penb (multidose)
Container
IM, SC, IV, IL Daily or three times a week
IM, SC, IV, ILa Daily or three times a week
Route and Frequency of Administration
Cytokine Product Formulations, Shelf Lives and Special Precautions for Storage145–147
Trade name
Table 3.
Storage at 2°C – 8°C Do not freeze Do not shake
Storage at 2°C – 8°C Do not freeze. Do not shake Protect from light
3 years (vials and syringes) or 2 years (pens) at 2°C – 8°C. 24 h at room temperature Do not freeze. Do not shake. Protect from light
Storage at 25°C 24 h after reconstitution (2°C – 8°C) Do not freeze. Do not shake
3 years (2°C – 8°C or 25°C)e 24 h after reconstitution (2°C – 8°C) Do not freeze Do not shake (pens)
2 years (2°C –8°C) 28 days at or below 25°C Do not freeze Protect from light
3 years d (2°C – 8°C) 24 h after reconstitution (2°C –8°C) Do not freeze
Continued
Single dose vials:18 months (2 °C – 8°C) 7 days at or below 25°C Multidose vials:2 years 28 days after opening (2°C – 8°C) 7 days at or below 25°C Multidose pens:15 months 27 days after opening (2°C – 8°C) Do not freeze
Shelf Life and Precautions
REVIEW
7
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
Neumega
Ontak
Proleukin
Actimmune/ Imukin
Rebif
Avonex
Betaferon/ Betaseron/ Extavia f
Lyophilized powder: IFN-β-1b (300 µg) Human albumin (15 mg ) Mannitol (15 mg) Solvent (1.2 mL): Sodium chloride (5.4 mg/mL) Water for injections Lyophilized powder: IFN-β-1a (33 µg) HSA (16.5 mg) Sodium phosphate dibasic (6.3 mg) Sodium phosphate monobasic (1.3 mg) Sodium chloride (6.4 mg) Solvent (1.0 mL): water for injections, pH 7.3 (after reconstitution) IFN-β-1a (60 µg/mL) Sodium acetate trihydrate (1.6 mg/mL) Acetic acid, glacial (0.5 mg/mL) Arginine hydrochloride (32 mg/mL) Polysorbate 20 (0.05 mg/mL) Water for injections (ad 0.5 mL), pH 4.8 US: IFN-β-1a (44 or 88 µg/mL) HSA (4 or 8 mg/mL) Mannitol (55 mg/mL) Sodium acetate (0.8 mg/mL) Water for injections (ad 0.2 or 0.5 mL) EU: IFN-β-1a (44 or 88 µg/mL) Mannitol (45 or 55 mg/mL) Poloxamer 188 (0.5 mg/mL) L-methionine (0.1 mg/mL) Benzyl alcohol (5 mg/mL) Sodium acetate Acetic acid and NaOH for pH adjustment Water for injections (ad 0.2– 1.5 mL), pH 4 ± 0.5 IFN-γ-1b (200 µg/mL) Mannitol (40 mg/mL) Sodium succinate (0.72 mg/mL) Polysorbate 20 (0.1 mg/mL) Water for injection (ad 1.0 mL) Lyophilized powder Aldesleukin (rhIL-2) (1.3 mg) Mannitol (60 mg) SDS (0.22 mg) Sodium phosphate monobasic (0.20 mg) S odium phosphate dibasic (1.1 mg) Solvent (1.2 mL):water for injections, pH 7.5 ± 0.3(after reconstitution) Denileukin diftitox (150 µg/mL) Citric acid ( 20 mM) EDTA (0.05 mM) Polysorbate 20 (<1%) Water for injection (ad 2.0 mL), pH 6.9 –7.2 Lyophilized powder Oprelvekin (rhIL-11) (5.0 mg) Glycine (23 mg) Dibasic sodium phosphate heptahydrate (1.6 mg) Monobasic sodium phosphate monohydrate (0.55 mg) Solvent (1.0 mL): water for injections (ad 1.0 mL), pH 7.0
Formulation (Shaded Background for Liquid Products)
Continued
Trade name
Table 3.
Vial (single use)
IV Daily during short treatment cycles separated by rest periods.
Vial (single use)
Vial (single use)
SC or IV Daily during short treatment cycles separated by rest periods.
SC Daily
Vial (single use)
Prefilled syringe (single use), Prefilled pen (single use) , Prefilled cartridge for electronic injection device or manual pen-injector device (multidose)
Storage at 2°C –8°C 3 h after reconstitution Do not freeze. Protect from light. Do not shake reconstituted solution.
Store frozen (at or below –10°C)
2 years (2°C– 8°C) 24 h after reconstitution (2°C– 8°C) 48 h after dilution (2°C– 8°C) Do not freeze Protect from light Do not dilute with NaCl solution
3 years (2°C– 8°C) 12 h below 25°C Do not freeze Do not shake
18 months (2°C– 8°C) g 4 days below 25°C Multidose products: 28 days after opening Do not freeze Protect from light
Storage at 2°C– 8°C 30 days below 25°C Do not freeze
Prefilled syringe, autoinjector (single use)
Continued
Vial (single use)
3 years (2°C– 8°C) 7 days at 15°C– 30°C Do not freeze Protect from light
2 years (EU: below 25°C, US: at 2°C–8°C, 30 days at 25°C) 6 h after reconstitution (2°C– 8°C) Do not freeze
Prefilled syringe. Prefilled pen/autoinjector (single use)
2 years (below 25°C) 3 h after reconstitution (2°C– 8°C) Do not freeze
Shelf Life and Precautions
Vial (single use)
Container
SC Three times a week
SC Three times a week
IM Once a week
SC Every other day
Route and Frequency of Administration
8 REVIEW
DOI 10.1002/jps.24243
DOI 10.1002/jps.24243
SC or IV Daily
SC or IV Daily
Vial (single use)
Vial (single use)
Prefilled syringe (single use)
Prefilled syringe (single use)
SC Once after each chemotherapy cycle
SC Once after each chemotherapy cycle
P refilled syringe (single use)
Prefilled syringe (single use)
Prefilled syringe (single use)
Vial Prefilled syringe (single use)
Container
SC or IV Daily
SC or IV Daily
SC or IV Daily
SC or IV Daily
Route and Frequency of Administration
(2°C –8°C) Do not freeze 6 h (without preservative) 20 days at 2°C –8°C (0.9% benzyl alcohol) after reconstitution Do not shake
(2°C –8°C) Do not freeze Do not shake 20 days (2 °C –8°C) after vial entering
2 years (below 30°C) 24 h after reconstitution (2°C –8°C) Do not freeze
2 years (2°C –8°C) 3 days below 25°C Do not freeze Protect from light
3 years (2°C –8°C) 72 h at room temperature Do not freeze. One -time freezing does not cause harm. Do not shake. Protect from light .
Same as Neupogen
36 months (2°C –8°C). 72 h at RT. 24 h after dilution. One-time freezing not harmful. Protect from light. Do not dilute with saline
Same as Neupogen
30 months (2°C –8°C) 24 h at room temperature. 24 h after diluti on. Do not shake. One -time freezing does not cause harm. Do not dilute with NaCl solution.
Shelf Life and Precautions
IL, intralesional; IM, intramuscular; IV, intravenous; RT, room temperature; SC, subcutaneous; WFI, water for injections. a Recommended routes of administration are different in the US and Europe. FDA label states that solution for injection is not recommended for IV administration, instead the lyophilized product should be used for this purpose. In contrast, the European product information documents instruct to administer the liquid product via IV or SC routes. b Only available in Europe. c Only available in the US. d Lyophilized Intron A product currently only authorized by FDA. In a European public assessment report (EPAR) summary, a powder product containing HSA was stated to have a shelf life of 3 years (2◦ C–8◦ C). e Available as single-use vials and prefilled pens. The European product information document recommends storage of both product types at 2◦ C–8◦ C (document revised 5/2010), but an FDA label (revised 6/2013) states that vials should be stored at room temperature (at 25◦ C, excursions permitted to 15◦ C–30◦ C). f Brand name is Betaseron in the US and Betaferon in Europe. Extavia is marketed by Novartis but manufactured by Bayer and has identical composition and pharmaceutical form as Betaseron/Betaferon. g The original Rebif formulation in Europe contained HSA and had a shelf-life of 24 months. h Neupogen Novum, 0.5 mL prefilled syringe filgrastim product licensed in Europe, has an additional strength of 0.96 mg/mL.
Leukine
Granocyte
Lonquex
Neulasta
Nivestim and Grastofil
Filgrastim Hexal/ Zarzio
Biograstim/ Ratiograstim/ Tevagrastim
Neupogen
Filgrastim (r-methG-CSF) (300 or 600 µg/mL)h Acetate (0.59 mg/mL) Sorbitol (50 mg/mL) Polysorbate 80 (0.04 mg/mL) Sodium (0,035 mg/mL) Water for injections (ad 0.5–1.6 mL) Filgrastim (600 µg/mL) Biosimilar, same composition except slight differences in polysorbate content and in pH value compared to Neupogen Filgrastim (600 and 960 µg/mL) Quantitatively identical composition to Neupogen, except that buffer system is glutamate instead of acetate Filgrastim 600 and 960 µg/mL Biosimilars, same composition as Neupogen Pegfilgrastim (pegylated r-methG-CSF) (10 mg/mL protein) Acetate (0.58 mg/mL) Sorbitol (US: 50 mg/mL; EU: 10 mg/mL) Polysorbate 20 (US: 0.03 mg/mL) Sodium (0.03 mg/mL) Water for injections (ad 0.6 mL), pH 4 Lipegfilgrastim (10 mg/mL protein) Acetic acid Sodium hydroxide Polysorbate 20 Sorbitol (30 mg) Water for injections (ad 0.6 mL) Lyophilized powder Lenograstim (rhG-CSF) (105 µg or 263 µg) Arginine (10 mg) Phenylalanine (10 mg) Methionine (1.0 mg) Mannitol (25 mg) Polysorbate 20 (0.1 mg) Hydrochloric acid Solvent (1.0 mL): water for injections, pH 6.5 after reconstitution Sargramostim (rhGM-CSF) (500 µg/mL) Mannitol (40 mg/mL) Sucrose (10 mg/mL) Tromethamine (1.2 mg/mL) Benzyl alcohol (11 mg/mL) Water for injections (ad 1.0 mL), pH 6.7–7.7 Lyophilized powder Sargramostim (rhGM-CSF) (250 µg) Mannitol (40 mg) Sucrose (10 mg) Tromethamine (1.2 mg) Solvent (1.0 mL): WFI or 0.9% benzyl alcohol pH 7.1–7.7
Formulation (Shaded Background for Liquid Products)
Continued
Trade name
Table 3.
REVIEW
9
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
10
REVIEW
In contrast, the corresponding sites in IFN-" (not close to its Olinked carbohydrate chain) are occupied by charged residues.60 Even though IFN-$-1a is much less hydrophobic than IFN$-1b, aggregation and surface adsorption seem to be concerns with both proteins. This is reflected by the inclusion of human serum albumin (HSA) in all original formulations. Both Betaseron (300 :g IFN-$-1b) and Avonex (30 :g IFN-$-1a) contain a high amount of albumin in the lyophilized formulations (15 mg HSA in both formulations, compared with, e.g., IntronA with 1 mg HSA). Albumin also serves other roles in lyophilized formulations (bulking agent, lyoprotectant, and cryoprotectant). The liquid Rebif (US) formulation has a lower content of albumin (Table 3). Recombinant production of albumin can alleviate the safety concerns related to plasma-derived albumin; however, analytical challenges posed by the presence of two proteins in the formulation remain. Thus, albumin-free formulations are more desirable, and reformulation cases where albumin has been replaced by surfactants are described in the following paragraphs. Betaseron was the first IFN-$ product to be approved. The powder formulation consists of IFN-$-1b, albumin, and mannitol; reconstitution is carried out in sodium chloride solution. It has 10-fold higher amount of active ingredient compared with other IFN-$ products. In a related case, an albumin-free formulation has been developed for a hydrophobic cytokine that resembles IFN-$-1b.61 In a study by Hawe and Friess,61 major stability concerns of solubility and surface adsorption could be mitigated without the addition of albumin. The solution pH was adjusted between 3.5–5.0 and low ionic strength, with the inclusion of polysorbate 20 or glycine in the formulations. Long-term stability (6 months) was afforded by a liquid formulation containing 4.5% mannitol and 0.02% polysorbate (pH 3.5–4.0) or a lyophilized formulation with 4% mannitol, 1% sucrose, and 0% or 0.005% polysorbate.61 Conditions of low ionic strength are achieved in Betaseron by including sodium chloride (tonicity modifier) in the reconstitution solution. Lyophilized Avonex formulation in sodium phosphate buffer contains IFN-$-1a, albumin, and sodium chloride. It has a much higher pH (7.3 for reconstituted solution) than the other IFN$-1a formulations (pH 3.5–4.8). IFN-$-1a has been stated to be more stable at physiological pH than IFN-$-1b59 ; however, IFN-$-1a has also been reported to form potentially immunogenic aggregates when formulated in sodium phosphate buffer and sodium chloride at pH 7.2.62 Phosphate-buffered saline can undergo large pH changes during freezing because of precipitation of the less-soluble buffer component (dibasic sodium phosphate), such as a decrease from pH 7.0 to 4.3–5.8 depending on the excipients present.63 Shift in formulation pH can deactivate proteins and for this reason phosphate buffer is not generally recommended for lyophilized protein formulations. However, in the case of Avonex, a pH decrease might be an advantage for protein stability (no data to support this hypothesis). Another interesting fact about Avonex formulation is the inclusion of sodium chloride. It is generally not recommended for lyophilized protein formulations to contain sodium chloride because of exposure to highly supersaturated salt solution during freezing.64 It is known that the unglycosylated IFN-$ aggregates at high ionic strength.61 Furthermore, the low eutectic melting temperature of sodium chloride water solution (−21◦ C) is not favorable for freeze-drying, because the product temperature must be kept low during the primary drying step, leading to a less efficient drying process.30,65 Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
A liquid Avonex formulation without albumin is currently marketed along with the lyophilized product.66 In this formulation, phosphate buffer has been replaced by acetate (pH 4.8). Reduced pH prevents IFN-$-1a aggregation.62 In addition, arginine and polysorbate 20 have been introduced and sodium chloride excluded from the formulation. The surfactant reduces adsorption and aggregation propensity, similar to the role played by albumin in the previous formulation. Arginine is a basic amino acid that can be used to preserve protein activity, especially when alternatives to the use of serum albumin are sought.67 Its importance as a protein formulation excipient is rapidly growing.50 It is known to increase protein solubility and stability via multiple molecular mechanisms, such as by disrupting protein–protein interactions and thus preventing aggregation (see reviews for more detailed discussion on protein– arginine interactions).27,50,68 In Avonex formulation, arginine hydrochloride also ensures formulation isotonicity along with the buffer components. Rebif is also available as two formulations. The original formulation (the one marketed in the US) is composed of albumin and mannitol in acetate buffer (pH 3.5–4.5). A new formulation, developed in conjunction with the introduction of a genetically engineered clone that supports enhanced expression and yield, was introduced in the European market in 2003.69 This formulation consists of mannitol, poloxamer 188, methionine, and benzyl alcohol in sodium acetate buffer (pH 3.5–4.5). The surfactant, poloxamer, serves to mimic the role of albumin in the formulation. However, the albumin-free, poloxamer-containing formulation has a shorter shelf life (18 months) as compared with the original albumin-containing product (24 months in EU). The difference in promised shelf life does not necessarily correlate with product stability but may be a mere company decision. Another Rebif formulation with poloxamer, lysine, and sodium acetate also showed promise in the prescreening phase of the development process.69 It is interesting to note the proposition of Jaber et al.69 that albumin protects IFN-$ against oxidation by acting as a scavenger, a property not commonly attributed to albumin. IFN-$-1a is vulnerable to oxidation and the oxidized form has a strong tendency to aggregate.10 Furthermore, oxidized and aggregated IFN-$-1a is potentially highly immunogenic.70 Antioxidant methionine is thus added to the formulation. However, the liquid formulation Avonex does not contain any excipient with known antioxidant properties. Uribeta is a biosimilar IFN-$ product marketed in Central and South America, with the same composition as Betaseron except that mannitol is replaced by glucose.71 Zheng et al.71 reported formation of potentially harmful glycated forms of both IFN-$ and HSA in Uribeta even when stored at +4◦ C and the extent of glycation increased when stored at room temperature. These results highlight the risk of using reducing sugars such as glucose in protein formulations. Interferon Gamma Interferon gamma is a proinflammatory cytokine, with powerful immunomodulatory properties and some antiviral activity.72 This cytokine is important in tumor surveillance and in immune responses against various types of infections, including intracellular bacterial infections.73,74 IFN-( is the only type II interferon; it has a unique receptor and differs structurally from other interferons.74 The biologically active form is a dimer composed of two identical 143 amino acid polypeptides. Each DOI 10.1002/jps.24243
REVIEW
monomer is composed of six "-helices and although the overall fold of the dimer does not resemble that of other interferons, five of the six helices have a folding topology similar to the type I IFN fold.75 IFN-( does not have any disulfide bonds and the dimer structure is stabilized by intertwining of the helices along the subunit interface.75 This kind of intimate linkage is unusual among globular proteins. Glycosylation is not essential for the biological activity of IFN-(. There are two glycosylation sites in each monomer and of the two sites either both, one or none, are occupied by an N-linked carbohydrate.72 Depending on the degree of glycosylation, a monomer has a molecular weight of 17, 20, or 25 kDa. The size heterogeneity is further increased by natural variation in the length of the carboxy-terminal. The mature dimers vary in the molecular weight range of 30–50 kDa.72 IFN-( activity is lost because of structural changes and dissociation of the dimer, when exposed to acidic conditions below pH 4.5, temperature above 50◦ C or high pressure.76,77 The monomers are prone to aggregate at certain conditions, such as combination of low pH and high ionic strength.76 Recombinant IFN-(-1b is approved for the treatment of chronic granulomatous disease, an inherited leukocyte disorder, and for osteopetrosis, a rare bone disease. The recombinant protein differs from the natural IFN-( in lacking glycosylation, having a shorter sequence of 140 residues with a methionine residue instead of a blocked pyroglutamate in the amino-terminus.73 The two marketed products, Actimmune and Imukin, are liquid products with identical compositions of IFN(-1b, mannitol, and polysorbate 20 in sodium succinate buffer. The formulation pH is 5.0 and a low ionic strength (5 mM sodium succinate) is critical for the cytokine stability. A study reported that acidification of a phosphate-buffered IFN-( formulation to pH 5.0 caused significant structural loss at high ionic strength.63 Feasibility of freeze-drying the product formulation has been investigated. A stable formulation could not be obtained by direct lyophilization as the monosodium form of succinic acid crystallized upon freezing and resulted in a pH drop and loss of activity.78 Replacing succinate with glycolate buffer in the marketed formulation retained activity after lyophilization and during storage in the solid state. Interestingly, Lam et al.,78 observed substantial deamidation in both succinate and glycolate formulations, but concluded that the chemical degradation had no effect on biological activity. There are no multiple-dose products available in the market, possibly because benzyl alcohol causes structural changes and aggregation of IFN-(-1b.79
Interleukin-2 Interleukin-2 stimulates T cells, natural killer cells, and antibody production by B cells. It is a key cytokine for immune response activation and also for immune tolerance and memory induction.80,81 Clinically, IL-2 is interesting because of its ability to enhance antitumor responses. It is a hydrophobic protein composed of 133 amino acids, that contains one disulfide bridge and one free cysteine residue, as well as a site for O-linked glycosylation.82 Because of variation in carbohydrate content, IL-2 is heterogeneous in size (15–18 kDa) and charge, but this does not seem to affect bioactivity.82 The overall structure is formed by a bundle of "-helices.83,84
DOI 10.1002/jps.24243
11
Aldesleukin (rhIL-2) is used for the treatment of renal cell carcinoma and melanoma. The recombinant protein differs from wild-type IL-2; it does not contain N-terminal alanine, it has serine in lieu of cysteine at position 125 and it is not glycosylated.85 Recombinant IL-2 is very hydrophobic and prone to aggregate. The aggregates have been held accountable for antidrug antibody formation.15 In addition, oxidation of IL-2 has been reported. EDTA and methionine are able to protect IL2 from oxidation.11 Glycine increases activity retention during storage, whereas polysorbate 80 may cause IL-2 degradation.11 Proleukin is a lyophilized formulation consisting of mannitol as stabilizer and bulking agent, sodium dodecyl sulfate (SDS; 0.02%) and sodium phosphate buffer, pH 7.5. SDS is an ionic surfactant that binds to proteins and solubilizes them. It is not commonly used in therapeutic protein products because the strong binding interactions often lead to denaturation of proteins. In fact, SDS is included in the Proleukin formulation to promote IL-2 aggregation.86 In case of aldesleukin, in vivo efficacy is improved when the cytokine is administered as aggregates, because monomers are rapidly cleared from the body.86 Proleukin contains noncovalently bound SDS/IL-2 microaggregates (27 interleukin molecules per aggregate, on average) that possibly have micellar or micelle-like structures.85,86 Thus, SDS does not have a conventional surfactant role in this formulation. Proleukin pH 7.5 is noteworthy as IL-2 is structurally more stable at pH 4 than at pH 7.87 Additionally, lyophilization with various buffers at pH 7 lead to extensive unfolding and aggregation of rIL-2. However, structural integrity was not compromised when freeze dried from solutions at pH 5 or below.88 Sucrose and other excipients were able to stabilize the structure at pH 7, but mannitol did not improve structure or activity retention.88 Furthermore, decreasing the pH to 4 lead to inhibition of surface adsorption in a liquid formulation of a similar IL-2 mutant (position 125 occupied by alanine instead of serine).11 Ontak, another product containing IL-2, is used to treat T cell lymphoma. The active ingredient, denileukin diftitox, is a recombinant fusion protein where fragments of diphtheria toxin are combined with IL-2.89 The cytokine serves to target the active ingredient to the cancerous T cells, allowing the toxin to be released in the diseased cells. Ontak is a liquid formulation with EDTA and polysorbate 20 in citric acid, pH 6.9–7.2, with recommended storage as frozen. Interleukin-11 Interleukin-11 is a thrombopoietic growth factor that stimulates megakaryocytopoiesis and platelet production, among multiple other biological activities.90–94 Recombinant IL-11 (oprelvekin; in Neumega) is used for the prevention of severe thrombocytopenia and reduction of the need for platelet transfusions after chemotherapy.95 Compared with the natural IL-11, the recombinant IL-11 (oprelvekin) lacks the amino-terminal proline and has 177 amino acids, one less than the natural protein.95 More than 50% of the polypeptide chain adopts "-helical conformation and a four-helix bundle tertiary fold resembling that of G-CSF has been suggested.91 However, the tertiary structure of IL-11 has not been validated. The helix-bundle structure is striking as IL11 has an exceptionally high content of proline residues (12%)
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
12
REVIEW
and lacks structure-stabilizing disulfide bridges.91 The cytokine also has an unusually high amount of leucine (23%), which has been suggested to account for considerable hydrophobicity.94 However, Czupryn et al.91 have hypothesized that the hydrophobic side chains are located on the helical interfaces and contribute to stabilize the bundle structure. Recombinant IL11 has also been described highly cationic, rich in arginine and other basic amino acids.96 In addition, the lack of glycosylation of the natural protein implicates that this secreted protein is adequately soluble. The cytokine has a methionine on its surface (Met58) that is critical for bioactivity.91 Oxidation of the methionine is therefore a potential concern of stability. Neumega is a lyophilized powder composed of rhIL-11 (5 mg), glycine, and sodium phosphate buffer (pH 7 after reconstitution). The lack of surfactant in the formulation is noteworthy. The protein concentration in Neumega is high for a cytokine formulation, which may provide an explanation for the lack of surfactant. Only a small proportion of the total protein is lost because of adsorption when the concentration is high. Polysorbates may be undesirable because peroxide impurities, often present in the raw materials, could impair the bioactivity by oxidizing the critical methionine residue. Glycine crystallizes during the freezing step of lyophilization and generally it needs to be combined with amorphous stabilizing agents that provide protection to the protein remaining in the amorphous phase.97 In Neumega, no such amorphous excipient is required. This may also be explained by the high protein concentration that can provide stability during lyophilization.30 Powders with low concentrations of rhIL-11 have been challenging to formulate because of adsorption on glass surfaces leading to significant activity loss.94 A formulation containing 1 :g/mL rhIL-11, 0.5% HSA, 0.1% trehalose, and 0.02% polysorbate 20 in potassium phosphate buffer (pH 7.4) inhibited surface adsorption and loss of bioactivity during long-term (6 months) storage.94 Both HSA and the surfactant were necessary for full recovery of activity; however, oxidation products were not analyzed. In another study, storage stability of rhIL-11 (3.9 mg/mL) was optimized by using mixtures of disaccharides (2.5% or 5% sucrose or trehalose) and hydroxyethyl starch (2.5% or 5%) in Tris buffer pH 7.0.98 The disaccharides inhibited unfolding during drying, whereas the polymer raised the glass transition temperature of the dried formulation. This resulted in improved storage stability compared with either disaccharide alone and allowed a faster and more economical drying process. Granulocyte Colony-Stimulating Factor Granulocyte colony-stimulating factor is a hematopoietic growth factor that stimulates the proliferation and differentiation of blood progenitor cells and activation of neutrophils.99 G-CSF products are blockbuster drugs, widely used to treat or prevent neutropenia, including cancer patients receiving chemotherapy. They are also used to mobilize blood progenitor cells for transplantation. There are four different recombinant forms of G-CSF in clinical use: lenograstim, filgrastim, pegfilgrastim, and lipegfilgrastim. Lenograstim (rhG-CSF; the active ingredient in Granocyte) is recombinant G-CSF produced in mammalian (CHO) cells. It has the same amino acid composition of 174 residues and glycosylation site (O-linked sugar chain at Thr-133) as the natural human G-CSF.100 Fil-
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
grastim (r-met-hG-CSF; the active ingredient in Neupogen and biosimilars) differs from the natural protein in lacking glycosylation and having one additional amino acid, the unprocessed N-terminal methionine. The recombinant G-CSFs are 19–20 kDa in size, contain two disulfide bonds, and one free cysteine residue, and possess similar biological activity as the endogenous protein.100,101 The three-dimensional structure is characterized as a four-helix bundle, having structural similarity with human growth hormone (hGH), GM-CSF, IFN-$, IL-2, and IL-4.102 Pegfilgrastim (Neulasta) and lipegfilgrastim (Lonquex) are pegylated forms of filgrastim, with increased elimination halflives. This facilitates a less frequent dosage regimen. Pegfilgrastim has a 20-kDa PEG molecule directly conjugated to the protein. The conjugation is different in lipegfilgrastim where the PEG chain is attached by a carbohydrate linker. The linker consists of glycine, N-acetylneuraminic acid, and N-acetylgalactosamine and it is attached to the unused O-glycosylation site, resulting in improved selectivity of pegylation.103,104 Recombinant met-G-CSF rapidly aggregates at neutral pH.105 The protein is however stable at acidic environment because of repulsive electrostatic protein–protein interactions at pH 3.5106 and even at pH 2.1.107 High ionic strength neutralizes the charge–charge repulsions and induces aggregation even at low pH.106 Sucrose, on the contrary, inhibits aggregation via nonspecific thermodynamic stabilization.105 Benzyl alcohol accelerates rhG-CSF aggregation; the effect is reduced by sucrose or reduction of pH to 3.5.108 Glycosylation contributes to GCSF stability by rendering the protein less sensitive to heat denaturation and aggregation, even at neutral pH.109 Filgrastim products are formulated at pH 4,12 whereas lenograstim formulation has a pH of 6.5 (after reconstitution). All methionine residues except for the additional N-terminal residue are critical for G-CSF biological activity. They are all susceptible to oxidization by peroxides.110 They are the only amino acid residues in G-CSF vulnerable to oxidation.12 Methionine oxidation is an important degradation pathway for filgrastim, possibly even the shelf-life-limiting factor.13 Free methionine is an effective antioxidant to prevent oxidation of rhG-CSF.111 Interestingly, liquid filgrastim products do not contain antioxidants, whereas the lenograstim dry powder formulation does have methionine. It could be possible that the amino-terminal methionine residue in filgrastim (not critical to activity), serves an antioxidant function. Chemical instability arising from deamidation was found to be the main degradation pathway for both Neupogen and a biosimilar product.112 The free cysteine residue could be replaced by serine without affecting activity or stability, to avoid intermolecular disulfide scrambling.101 This approach has been used to improve rIL2 stability. In G-CSF, the free sulfhydryl group appears to be well buried inside the protein and is rather unreactive. Therefore, cysteine substitution has not been utilized in the current products. The free cysteine residue could still account for dimer formation observed in stability studies. Because aggregation can be effectively inhibited at low pH and ionic strength, oxidation, and deamidation are the major pathways that need to be prevented. Polysorbate concentrations in the filgrastim products are relatively low (0.04 mg/mL); this reduces the risk of peroxide-induced
DOI 10.1002/jps.24243
REVIEW
oxidation. Light and elevated temperature accelerates peroxide formation in polysorbates and often polysorbates contain residual peroxides as impurities.113 Of the four types of G-CSF products, the pegylated proteins are similar, but the rest have significant differences affecting formulation strategies. All filgrastim products are liquid formulations containing buffer, sorbitol as stabilizer/tonicity modifier, and polysorbate 80. The originator product Neupogen and most of the biosimilars are formulated with acetate buffer but in Filgrastim Hexal/Zarzio glutamate buffer system has been used. In stability studies comparing Filgrastim Hexal/Zarzio to Neupogen, the biosimilar product formulation showed lower levels of oxidation and deamidation. The stability of the products was concluded to be comparable, though.112,114 It is noteworthy that Filgrastim Hexal and Zarzio have a longer shelf-life of 36 months, whereas for Neupogen and the other acetatecontaining products it is 30 months. However, the different shelf lives may reflect business decisions rather than protein stability. One-time freezing of the product does not adversely affect product stability that could be attributed to the low crystallization tendency of sorbitol upon freezing.97 In contrast, mannitolcontaining formulations such as Granocyte and Rebif must be discarded if accidentally frozen. The pegylated filgrastim products differ from the nonpegylated versions in having a much higher protein concentration. The Neulasta and Lonquex products contain 10 mg/mL protein, whereas the concentration range is 0.3–0.9 mg/mL in the filgrastim products. The formulations are otherwise similar to Neupogen (acetate buffer and sorbitol). The surfactant concentration required is lower compared with Neupogen, probably because of the reduced aggregation tendency because of pegylation.115 Polysorbate 20 is used instead of polysorbate 80 because of its availability from vegetable-derived source.115 It is difficult to rationalize the different sorbitol concentrations in the Neulasta product marketed in US (50 mg/mL) and in Europe (10 mg/mL). Higher sorbitol concentration reduces the amount of degradation products.115 The Granocyte formulation clearly differs from all the above described products. It is a lyophilized powder product with a lower cytokine concentration enabled by the higher activity of lenograstim compared with filgrastim.116 Lenograstim is less sensitive to degradation at neutral pH compared with filgrastim; therefore, the Granocyte formulation pH (after reconstitution) is higher (pH 6.5). It is somewhat counter-intuitive that even though the intrinsic stability of lenograstim is higher than that of filgrastim, and it is formulated as a dry product, the shelf-life of Granocyte is only 2 years, the shortest of all GCSF products. However, it can be stored at room temperature, whereas the others need to be refrigerated. The formulation contains several excipients: arginine, phenylalanine, methionine, mannitol, polysorbate 20, and hydrochloric acid. The complexity probably arises from reformulation challenges resulting from the removal of albumin from the formulation.117 In 1998, a simpler Granocyte formulation was marketed consisting of HSA (1 mg), mannitol (50 mg), and polysorbate 20 (0.1 mg) and sodium chloride in phosphate buffer.118 Removal of albumin apparently necessitated the introduction of an antioxidant (methionine). Mannitol, phenylalanine, and arginine contribute to the solid-state properties of Granocyte formulation. Mannitol is a commonly used bulking agent in freeze-dried products. Like glycine, it generally crystallizes in the process and results in an DOI 10.1002/jps.24243
13
excellent dried cake structure but does not stabilize the protein in the dry state. Additional amorphous stabilizers are needed. Phenylalanine is a hydrophobic amino acid not commonly used as excipient in protein formulations. It has been mentioned in a patent application on inhalable spray-dried four-helix bundle protein powders, along with other hydrophobic amino acids.119 It is believed to increase the glass transition temperature, decrease the surface tension of water, and inhibit aggregation by accumulating on particle surfaces where it protects the protein from the harsh process conditions. The inclusion of phenylalanine in Granocyte may have been influenced by the observations of Mattern et al.120 They found that arginine forms amorphous solids when freeze dried and that phenylalanine inhibited arginine crystallization in vacuum-dried powders by reducing the residual moisture content and increasing the dry product glass transition temperature.120 In the same study, GCSF was vacuum dried in combination with arginine, phenylalanine (1:1 Arg/Phe molar ratio), and a mineral acid (HCl or H3 PO4 ) resulting in effective stabilization of the protein during storage. Therefore, Granocyte seems to be a partially amorphous excipient system where crystalline mannitol provides cake structure, arginine functions as the amorphous stabilizer, and phenylalanine is included to inhibit arginine crystallization during storage. Granulocyte-Macrophage Colony-Stimulating Factor Granulocyte-macrophage colony-stimulating factor is an inflammatory mediator, stimulating proliferation and differentiation of hematopoietic stem cells, and activating granulocytes and macrophages.99 Leukine is an FDA-approved product containing recombinant GM-CSF (sargramostim), for chemotherapy-induced neutropenia or bone marrow transplantation. The indications are the same as for G-CSF products; however, adverse effects are more commonly associated with GM-CSF, making it a much less used product than G-CSF.99 Granulocyte-macrophage colony-stimulating factor is a glycoprotein that consists of 127 amino acid residues and forms a compact globular fold stabilized by "-helices, $-sheets, and two disulfide bridges.121 The $-sheet content is uncommon for bundle cytokines, but overall it has a very similar fold compared with G-CSF, IL-2, IFN-$, and hGH.102 Among the related structures, GM-CSF is the smallest.102 The molecular weight varies depending on the degree of glycosylation, and has been reported to be 14.7 kDa99,121 and around 23 kDa.81 Sargramostim in Leukine is produced in S. cerevisiae. It differs from the natural protein by a single substitution of arginine with leucine at position 23. Yeast cell expression produces glycosylated protein with both O- and N-linked oligosaccharides that are not identical to the ones present in the endogenous human protein. Variation in saccharide chains results in three molecular species (molecular weights of 19.5, 16.8, and 15.5 kDa). Leukine is available both as a liquid and a lyophilized powder product. Excipients in the liquid formulation are mannitol (stabilizer and isotonicity), sucrose (stabilizer and isotonicity), benzyl alcohol (preservative) buffered in tromethamine (Tris) at pH 6.7–7.7. The powder contains mannitol (bulking agent), sucrose (amorphous stabilizer), and tromethamine (reconstituted product pH 7.1–7.7). Tris buffer system remains amorphous during freezing.21 It is noteworthy that there is no surfactant in this formulation, even though the protein concentration is not very high (250–500 :g/mL). The Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
14
REVIEW
carbohydrate chains probably afford sufficient solubility to this cytokine.
Possible Upcoming Cytokine Pharmaceuticals Most of the cytokine products have been approved to market a number of years ago and new products are duly expected. Long-term treatment and frequent administrations, required with MS disease therapy, for example, is believed to result in an antibody response over time.122 This causes reduction in therapeutic efficacy and may lead to fatal immunogenic reactions. Several strategies to reduce administration frequency and immunogenicity are being explored. Simultaneously, patient compliance could be improved. Pegylation is one promising strategy; pegylated IFN-$-1a and IFN-$-1b formulations have been in clinical trials123,124 and a pegylated IFN-$ product (Plegridy) is in the filing stage.125 Albumin-interferon aggregates have also been proposed to be responsible for immunogenicity.126 Therefore, albumin-free formulations could provide reduced immunogenicity. On the contrary, albumin-IFN-" fusion protein (albinterferon), intended for a prolonged-action treatment has been developed. Albinterferon shows decreased aggregation tendency during agitation and storage.127 More cytokine biosimilars will also emerge in the near future. European Medicines Agency (EMA) has published a guideline on IFN-$ biosimilars in 2013 and Neulasta patent expires in 2015. Interleukin-2 has been at the forefront for cytokine delivery system development. It is commonly used to treat latestage cancers, in high-dose i.v. injections that cause severe, life-threatening side effects.81,128 Polymeric microsphere formulations, liposomes, pegylation, and pulmonary administration are some strategies evaluated to improve IL-2 therapy.81 Toxicity could be reduced by local delivery and/or by achieving lower plasma concentrations, whereas therapeutic efficacy may be improved by prolonging half-life and better reaching the target site. Sustained and local delivery of IL-2 might be achieved by injecting IL-2-loaded microspheres to the target site.81 New stabilizing excipients are studied for these purposes, such as choline dihydrogen phosphate intended for IL-2 liposomes or nanoparticles.128 In addition, future may show promise for combination products, such as a controlled-release microparticle formulation with a mixture of IL-2, transforming growth factor $ and rapamycin, implicated for treatment of autoimmune diseases.129 For rhIL-11, attempts to maximize the clinical potency and reduce the frequency of administration include developing a hyaluronic acid matrix for sustained release,130 pegylation,131 and site-directed mutagenesis.132 New expression systems are being developed, such as modified yeasts.133 For some cytokines, glycosylation is critical for stability and activity, but mammalian cell expression systems are costly and generally have low expression levels. Overexpression is difficult to achieve because high cytokine levels are toxic to the host cell. Further still, enhanced protein forms are being developed by hyperglycosylation, to provide higher solubility and half-life.134 Alternative manufacturing processes may be introduced. A dry formulation containing IFN-"-2a, trehalose, and mannitol was prepared by in situ precipitation by PEG followed by vacuum drying.135 It was said to have comparable stability against aggregation and oxidation than a similar lyophilized formulation. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
Many previously unknown cytokines have been discovered during the last decades. With greater understanding of the complex cytokine biology and improved genomics-related methods, cytokines provide a pool of new potential therapeutics. IFN-8 is the most promising candidate and it is in Phase 3 clinical trials (PEG-IFN-8).136 Interleukins IL-7 and IL-21 are already approved by EMA for rare disease indications. In addition, rhIL-21 (denenicokin) is in Phase 1 clinical trials.136 A few cytokines that previously failed in clinical trials (e.g., IL-12 and IL-10) are now being reinvestigated in light of new information and better delivery strategies.2 Cytokine-antibody fusion proteins are in company pipelines; IL-2,137 IL-12,137,138 and IL-10 (dekavil)139 are in Phase 1 clinical trials. There is also a long list of potential new indications, such as G-CSF for the treatment of noise-induced hearing loss140 and chronic obstructive pulmonary disease,141 rhIL-11 for HIV therapy,142 and IFN-"-2b for the treatment of viral conjunctivitis by extemporaneously prepared eye drop formulation.143 Because of their immunomodulatory activities, cytokines, especially IFN-( and GM-CSF have potential applications as vaccine adjuvants.73,99,144
DISCUSSION Cytokine therapeutics on the market include different subtypes of IFN-"s, IFN-$, IFN-(, IL-2, IL-11, G-CSF, and GM-CSF. Their protein characteristics are summarized in Table 2. They are all small proteins with molecular weight ranging between 15 and 20 kDa, except for IFN-(, which is a 30–50-kDa dimer. The cytokines have a helical bundle fold, generally associated with marked hydrophobicity.6 Therefore, challenges because of aggregation and surface adsorption are common with cytokines. However, the degree of hydrophobicity is quite different among the cytokines and production host affecting the degree of glycosylation can have significant impact, as in case for IFN-$. Residual impurities, which can be present in excipients, as well as product contact with packaging materials (e.g., glass surfaces and rubber stoppers) can cause protein degradation.7,113 Oxidation is another major degradation pathway for cytokines. To prevent loss of activity as well as formation of immunogenic species, attention must be paid to preventing aggregation, surface adsorption, and oxidation of cytokine proteins. The formulation excipients used in currently marketed cytokine products are listed in Table 4. The most common buffer systems are phosphate and acetate. Protein-stabilizing excipients include sucrose, mannitol, sorbitol, albumin, and amino acids. Surfactants (polysorbate 20/80, poloxamer, or SDS) or albumin are used in almost all cytokine products. The exceptions are Infergen (optimized sequence), Neumega (high protein concentration), and Leukine (highly glycosylated protein). As more therapeutic cytokine products are expected to be developed in the coming years, this review should serve as a useful guide for the formulation scientist. As a first, it is important to understand the structural properties of the cytokine and the possible degradation mechanisms that might exist. Most likely, the stability concerns relate to hydrophobicity and low solubility, whereas chemical instability arises from oxidation. Liquid formulations provide convenience of manufacture and use, and many of the cytokines in market are stable in solution. Therefore, development of a liquid formulation is a justified starting point. A liquid formulation requires an optimal buffer system and pH to ensure conformational stability. Dependency R
R
DOI 10.1002/jps.24243
REVIEW
Table 4.
15
Summary of Cytokine Formulation Excipients Product Name
Excipient Buffers/pH adjustment Phosphate
Acetate
Glutamate Succinate Tromethamine (TRIS) Hydrochloric acid No buffer Salts Sodium chloride
Solution
IntronA (IFN-") Infergen (IFN-") Alferon (IFN-")
Roferon-A (IFN-") Pegasys (PEG-IFN-") Avonex (IFN-$) Rebif (IFN-$) Neupogen (G-CSF) Biograstim/Ratiograstim/Tevagrastim (G-CSF) Nivestim (G-CSF) Neulasta (PEG-GSF) Lonquex (LiPEG-G-CSF) Filgrastim Hexal/Zarzio (G-CSF) Actimmune/Imukin (IFN-() Leukine (GM-CSF)
Lyophilized Powder
IntronA (IFN-") PegIntron/ViraferonPeg (PEG-IFN-") Sylatron (PEG-IFN-") Avonex (IFN-$) Proleukin (IL-2) Neumega (IL-11)
Leukine (GM-CSF) Granocyte (G-CSF) Betaferon/Betaseron/Extavia (IFN-$) Betaferon/Betaseron/Extavia (IFN-$) Avonex (IFN-$)
Potassium chloride
IntronA (IFN-") Roferon-A (IFN-") Pegasys (PEG-IFN-") Infergen (IFN-") Alferon (IFN-") Alferon (IFN-")
Sugars, Sugar Polyols Sucrose
Leukine (GM-CSF)
PegIntron/ViraferonPeg (PEG-IFN-") Sylatron (PEG-IFN-") Leukine (GM-CSF) Betaferon/Betaseron/Extavia (IFN-$) Proleukin (IL-2) Granocyte (G-CSF) Leukine (GM-CSF)
Mannitol
Rebif (IFN-$) Actimmune/Imukin (IFN-() Leukine (GM-CSF)
Sorbitol
Neupogen (G-CSF) Biograstim/Ratiograstim/Tevagrastim (G-CSF) Filgrastim Hexal/Zarzio (G-CSF) Nivestim (G-CSF) Neulasta (PEG-GSF) Lonquex (LiPEG-G-CSF)
Surfactants Polysorbate 80
Polysorbate 20
Poloxamer 188 SDS Albumin HSA
IntronA (IFN-") Roferon-A (IFN-") Pegasys (PEG-IFN-") Neupogen (G-CSF) Biograstim/Ratiograstim/Tevagrastim (G-CSF) Filgrastim Hexal/Zarzio (G-CSF) Nivestim (G-CSF) Avonex (IFN-$) Actimmune/Imukin (IFN-() Neulasta Neulasta (PEG-GSF) Lonquex (LiPEG-G-CSF) Rebif (IFN-$) (EU)
PegIntron/ViraferonPeg (PEG-IFN-") Sylatron (PEG-IFN-")
Granocyte (G-CSF)
Proleukin (IL-2) Alferon (IFN-") Rebif (IFN-$) (US)
IntronA (IFN-") Betaferon/Betaseron/Extavia (IFN-$) Avonex (IFN-$) Continued
DOI 10.1002/jps.24243
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
16
REVIEW
Table 4.
Continued Product Name
Excipient
Solution
Amino Acids Glycine Arginine Methionine Phenylalanine Chelating Agents Edetate disodium Preservatives Benzyl alcohol
M-cresol Phenol
Avonex (IFN-$) Rebif (IFN-$) (EU)
IntronA (IFN-") Neumega (IL-11) Granocyte (G-CSF) Granocyte (G-CSF) Granocyte (G-CSF)
IntronA (IFN-") Roferon-A (IFN-") Pegasys (PEG-IFN-") Rebif (IFN-$) (EU) Leukine (GM-CSF) IntronA (IFN-") Alferon (IFN-")
of structural stability on pH varies between cytokines. Some have increased stability in acid, whereas others have decreased stability.6 Optimal pH might be in the neutral or slightly acidic range, where phosphate (pKa = 7.21) and acetate (pKa = 4.76) buffers have proven to be good options. It is possible that pH adjustment is enough to alleviate hydrophobicity related concerns. A general rule in protein formulation development is to set the formulation pH at least 0.5 units above or below the protein isoelectric point.21 However, for example, Pegasys (a pegylated formulation) is stable even at the isoelectric point of IFN-". In cases where sodium chloride does not adversely affect stability, the formulation may be as simple as that in Infergen (Table 3). To achieve desired stability, additional stabilizers such as sucrose or mannitol may be required and the presence of a surfactant might prove helpful. For polysorbates, attention should be paid to peroxide impurities. To mitigate oxidationrelated instability, antioxidants such as methionine or a metal chelator such as EDTA could be useful agents in formulation. Albumin has several useful properties such as being a stabilizer, surfactant, and even antioxidant; however, its use in formulations can complicate the analytics (with two proteins in the formulation) and also raise concerns of HSA–cytokine aggregate-induced immunogenicity. In case a multidose product is developed and a preservative is needed, attention to the potential of benzyl alcohol causing cytokine degradation should be paid. M-cresol and phenol are other preservative options previously used in cytokine products. If liquid formulations do not achieve the desired long-term stability, dry formulations with suitable buffer system (e.g., phosphate or Tris) and a bulking agent (e.g., mannitol or glycine) can first be investigated. A surfactant might be needed in order to prevent aggregation during the manufacturing process and rehydration. Phosphate buffer is not generally recommended for lyophilized formulations because of the risk of pH shifts owing to crystallization of the less soluble dibasic salt component during freezing. Several lyophilized cytokine products containing phosphate buffer are in the market, though, and a pH drop could even prove to be advantageous as some cy-
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
Lyophilized Powder
tokines have improved stability under acidic conditions.6 Usually, an amorphous stabilizer is required for lyophilized protein formulations. However, stable cytokine products have been formulated without them (e.g., Proleukin and Neumega), and the formulation with minimum amount of excipients necessary should be selected. For proteins that otherwise degrade during lyophilization, excipients that remain in the amorphous state (e.g., sucrose or arginine in combination with phenylalanine) may provide the additional protection required.
CONCLUSION Cytokines have shared properties such as small size and helical bundle tertiary structure. They are often vulnerable to aggregation, surface adsorption, and oxidation. However, there is intrinsic variation between these molecules because of differences in amino acid sequences and in glycosylation patterns produced by different expression hosts. The significance of these variations on solubility and susceptibility to aggregation or chemical degradation is cytokine dependent. Consequently, the optimal formulation must be uniquely developed based on extensive understanding of individual stability characteristics of the cytokine of interest. However, advice for best first choices in cytokine formulation development can be adopted from this review.
REFERENCES 1. Commins SP, Borish L, Steinke JW. 2010. Immunologic messenger molecules: Cytokines, interferons, and chemokines. J Allergy Clin Immun 125:S53–S72. 2. Donnelly RP, Young HA, Rosenberg AS. 2009. An overview of cytokines and cytokine antagonists as therapeutic agents. Ann NY Acad Sci 1182:1–13. 3. Kozlowski S, Cherney B, Donnelly RP. 2009. Hurdles and leaps for protein therapeutics. Ann NY Acad Sci 1182:146–160. 4. Nelles M, Salerno V, Xu Y, Paige C. 2011. Cytokine immunotherapy. In Experimental and applied immunotherapy; Medin J, Fowler D, Eds. New York: Humana Press, pp 281–305.
DOI 10.1002/jps.24243
REVIEW
5. George PM, Badiger R, Alazawi W, Foster GR, Mitchell JA. 2012. Pharmacology and therapeutic potential of interferons. Pharmacol Ther 135:44–53. 6. Ricci MS, Brems DN. 2004. Common structural stability properties of 4-helical bundle cytokines: Possible physiological and pharmaceutical consequences. Curr Pharm Design 10:3901–3911. 7. Ruiz L, Reyes N, Aroche K, Tolosa V, Simanca V, Rogriguez T, Hardy E. 2005. Influence of packaging material on the liquid stability of interferon-alpha2b. J Pharm Pharm Sci 8:207–216. 8. Gitlin G, Tsarbopoulos A, Patel ST, Sydor W, Pramanik BN, Jacobs S, Westreich L, Mittelman S, Bausch JN. 1996. Isolation and characterization of a monomethioninesulfoxide variant of interferon alpha-2b. Pharm Res 13:762–769. 9. Hermeling S, Aranha L, Damen JM, Slijper M, Schellekens H, Crommelin DJ, Jiskoot W. 2005. Structural characterization and immunogenicity in wild-type and immune tolerant mice of degraded recombinant human interferon alpha2b. Pharm Res 22:1997–2006. 10. Torosantucci R, Sharov VS, van Beers M, Brinks V, Sch¨oneich C, Jiskoot W. 2013. Identification of oxidation sites and covalent crosslinks in metal catalyzed oxidized interferon beta-1a: Potential implications for protein aggregation and immunogenicity. Mol Pharm 10:2311– 2322. 11. Reyes N, Ruiz L, Aroche K, Geronimo H, Brito O, Hardy E. 2005. Stability of Ala 125 recombinant human interleukin-2 in solution. J Pharm Pharmacol 57:31–37. 12. Reubsaet JL, Beijnen JH, Bult A, Hop E, Scholten SD, Teeuwsen J, Underberg WJ. 1998. Oxidation of recombinant methionyl human granulocyte colony stimulating factor. J Pharm Biomed Anal 17:283– 289. 13. Holzmann J, Hausberger A, Rupprechter A, Toll H. 2013. Top-down MS for rapid methionine oxidation site assignment in filgrastim. Anal Bioanal Chem 405:6667–6674. 14. Jiskoot W, Randolph TW, Volkin DB, Middaugh CR, Schoneich C, Winter G, Friess W, Crommelin DJ, Carpenter JF. 2012. Protein instability and immunogenicity: Roadblocks to clinical application of injectable protein delivery systems for sustained release. J Pharm Sci 101:946–954. 15. Singh SK. 2011. Impact of product-related factors on immunogenicity of biotherapeutics. J Pharm Sci 100:354–387. 16. Sauerborn M, Brinks V, Jiskoot W, Schellekens H. 2010. Immunological mechanism underlying the immune response to recombinant human protein therapeutics. Trends Pharmacol Sci 31:53– 59. 17. Rosenberg AS. 2006. Effects of protein aggregates: An immunologic perspective. AAPS J 8:E501–E507. 18. Casadevall N, Eckardt K-U, Rossert J. 2005. Epoetin-induced autoimmune pure red cell aplasia. J Am Soc Nephrol 16:S67–S69. 19. Arakawa T, Prestrelski SJ, Kenney WC, Carpenter JF. 2001. Factors affecting short-term and long-term stabilities of proteins. Adv Drug Deliv Rev 46:307–326. 20. Manning MC, Chou DK, Murphy BM, Payne RW, Katayama DS. 2010. Stability of protein pharmaceuticals: An update. Pharm Res 27:544–575. 21. Schiffter HA. 2011. Pharmaceutical proteins—Structure, stability, and formulation. In Comprehensive biotechnology; Moo-Young M, Eds. 2nd ed. Burlington, Vermont: Academic Press, pp 521–541. 22. Topp EM, Zhang L, Zhao H, Payne RW, Evans GJ, Manning MC. 2010. Chemical instability in peptide and protein pharmaceuticals. In Formulation and process development strategies for manufacturing biopharmaceuticals; Jameel F, Hershenson S, Eds. Hoboken, New Jersey: John Wiley & Sons, Inc., pp 41–67. 23. Hawe A, Wiggenhorn M, van de Weert M, Garbe JH, Mahler HC, Jiskoot W. 2012. Forced degradation of therapeutic proteins. J Pharm Sci 101:895–913. 24. Wang W, Nema S, Teagarden D. 2010. Protein aggregation— Pathways and influencing factors. Int J Pharm 390:89–99. 25. Lai MC, Topp EM. 1999. Solid-state chemical stability of proteins and peptides. J Pharm Sci 88:489–500. DOI 10.1002/jps.24243
17
26. Jorgensen L, Hostrup S, Moeller EH, Grohganz H. 2009. Recent trends in stabilising peptides and proteins in pharmaceutical formulation—Considerations in the choice of excipients. Expert Opin Drug Deliv 6:1219–1230. 27. Kamerzell TJ, Esfandiary R, Joshi SB, Middaugh CR, Volkin DB. 2011. Protein–excipient interactions: Mechanisms and biophysical characterization applied to protein formulation development. Adv Drug Deliv Rev 63:1118–1159. 28. Ohtake S, Wang W. 2014. Protein and peptide formulation development. In Biological drug products: Development and strategies; Wang W, Singh M, Eds. Hoboken, New Jersey: John Wiley & Sons, Inc. p 323–366. 29. Wang W. 1999. Instability, stabilization, and formulation of liquid protein pharmaceuticals. Int J Pharm 185:129–188. 30. Wang W. 2000. Lyophilization and development of solid protein pharmaceuticals. Int J Pharm 203:1–60. 31. Laursen T, Hansen B, Fisker S. 2006. Pain perception after subcutaneous injections of media containing different buffers. Basic Clin Pharmacol 98:218–221. 32. Brassard DL, Grace MJ, Bordens RW. 2002. Interferon-alpha as an immunotherapeutic protein. J Leukocyte Biol 71:565–581. 33. Fuchs SY. 2013. Hope and fear for interferon: The receptor-centric outlook on the future of interferon therapy. J Interf Cytok Res 33:211– 225. 34. Bekisz J, Schmeisser H, Hernandez J, Goldman ND, Zoon KC. 2004. Mini review human interferons alpha, beta and omega. Growth Factors 22:243–251. 35. Alferon product website. Hemispherx Biopharma Inc. Accessed June 28, 2014, at: http://www.hemispherx.net/content/products/. 36. Blatt LM, Davis JM, Klein SB, Taylor MW. 1996. The biologic activity and molecular characterization of a novel synthetic interferonalpha species, consensus interferon. J Interf Cytok Res 16:489– 499. 37. Klaus W, Gsell B, Labhardt AM, Wipf B, Senn H. 1997. The threedimensional high resolution structure of human interferon alpha-2a determined by heteronuclear NMR spectroscopy in solution. J Mol Biol 274:661–675. 38. Radhakrishnan R, Walter LJ, Hruza A, Reichert P, Trotta PP, Nagabhushan TL, Walter MR. 1996. Zinc mediated dimer of human interferon-alpha 2b revealed by X-ray crystallography. Structure 4:1453–1463. 39. Bendele A, Seely J, Richey C, Sennello G, Shopp G. 1998. Short communication: Renal tubular vacuolation in animals treated with polyethylene-glycol-conjugated proteins. Toxicol Sci 42:152–157. 40. Garay RP, El-Gewely R, Armstrong JK, Garratty G, Richette P. 2012. Antibodies against polyethylene glycol in healthy subjects and in patients treated with PEG-conjugated agents. Expert Opin Drug Deliv 9:1319–1323. 41. Schellekens H, Hennink WE, Brinks V. 2013. The immunogenicity of polyethylene glycol: Facts and fiction. Pharm Res 30:1729–1734. 42. Foser S, Schacher A, Weyer KA, Brugger D, Dietel E, Marti S, Schreitmuller T. 2003. Isolation, structural characterization, and antiviral activity of positional isomers of monopegylated interferon alpha2a (PEGASYS). Protein Expres Purif 30:78–87. 43. Wang YS, Youngster S, Grace M, Bausch J, Bordens R, Wyss DF. 2002. Structural and biological characterization of pegylated recombinant interferon alpha-2b and its therapeutic implications. Adv Drug Deliv Rev 54:547–570. 44. Adolf GR, Kalsner I, Ahorn H, Maurer-Fogy I, Cantell K. 1991. Natural human interferon-alpha 2 is O-glycosylated. Biochem J 276:511– 518. 45. Johnston MJ, Frahm G, Li X, Durocher Y, Hefford MA. 2011. O-linked glycosylation leads to decreased thermal stability of interferon alpha 2b as measured by two orthogonal techniques. Pharm Res 28:1661–1667. 46. Beldarrain A, Cruz Y, Cruz O, Navarro M, Gil M. 2001. Purification and conformational properties of a human interferon alpha2b produced in Escherichia coli. Biotechnol Appl Biochem 33:173–182. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
18
REVIEW
47. Sharma VK, Kalonia DS. 2003. Temperature- and pH-induced multiple partially unfolded states of recombinant human interferonalpha2a: Possible implications in protein stability. Pharm Res 20:1721– 1729. 48. Li Y, Stafford WF, Hesselberg M, Hayes D, Wu Z, Byrne M. 2012. Characterization of the self-association of human interferon-alpha2b, albinterferon-alpha2b, and pegasys. J Pharm Sci 101:68–80. 49. Gross G, Del Terzo S, Kumar SK. 1998. Stabilized interferon alpha solutions. Patent US5762923 A. 50. Ohtake S, Kita Y, Arakawa T. 2011. Interactions of formulation excipients with proteins in solution and in the dried state. Adv Drug Deliv Rev 63:1053–1073. 51. Jeyachandran YL, Mielczarski E, Rai B, Mielczarski JA. 2009. Quantitative and qualitative evaluation of adsorption/desorption of bovine serum albumin on hydrophilic and hydrophobic surfaces. Langmuir 25:11614–11620. 52. Ruiz L, Reyes N, Duany L, Franco A, Aroche K, Hardy Rando E. 2003. Long-term stabilization of recombinant human interferon alpha 2b in aqueous solution without serum albumin. Int J Pharm 264:57–72. 53. Graf E, Mahoney JR, Bryant RG, Eaton JW. 1984. Iron-catalyzed hydroxyl radical formation. Stringent requirement for free iron coordination site. J Biol Chem 259:3620–3624. 54. Cindric M, Galic N, Vuletic M, Klaric M, Drevenkar V. 2006. Evaluation of recombinant human interferon alpha-2b structure and stability by in-gel tryptic digestion, H/D exchange and mass spectrometry. J Pharm Biomed Anal 40:781–787. 55. European Medicines Agency. 2005. PegIntron: EPAR— Scientific discussion. Accessed June 28, 2014, at: http://www. ema . europa . eu / docs / en GB / document library/EPAR - Scientific Discussion/human/000280/WC500039385.pdf. 56. European Medicines Agency. 2005. Pegasys: EPAR— Scientific discussion. Accessed June 28, 2014: http://www.ema. europa.eu/docs/en GB/document library/EPAR - Scientific Discussion/ human/000395/WC500039192.pdf. 57. Karpusas M, Nolte M, Benton CB, Meier W, Lipscomb WN, Goelz S. 1997. The crystal structure of human interferon beta at 2.2-A resolution. Proc Natl Acad Sci USA 94:11813–11818. 58. Conradt HS, Egge H, Peter-Katalinic J, Reiser W, Siklosi T, Schaper K. 1987. Structure of the carbohydrate moiety of human interferon-beta secreted by a recombinant Chinese hamster ovary cell line. J Biol Chem 262:14600–14605. 59. Runkel L, Meier W, Pepinsky RB, Karpusas M, Whitty A, Kimball K, Brickelmaier M, Muldowney C, Jones W, Goelz SE. 1998. Structural and functional differences between glycosylated and non-glycosylated forms of human interferon-beta (IFN-beta). Pharm Res 15:641–649. 60. Karpusas M, Whitty A, Runkel L, Hochman P. 1998. The structure of human interferon-beta: Implications for activity. Cell Mol Life Sci 54:1203–1216. 61. Hawe A, Friess W. 2008. Development of HSA-free formulations for a hydrophobic cytokine with improved stability. Eur J Pharm Biopharm 68:169–182. 62. van Beers MM, Sauerborn M, Gilli F, Brinks V, Schellekens H, Jiskoot W. 2010. Aggregated recombinant human interferon beta induces antibodies but no memory in immune-tolerant transgenic mice. Pharm Res 27:1812–1824. 63. Webb SD, Cleland JL, Carpenter JF, Randolph TW. 2002. A new mechanism for decreasing aggregation of recombinant human interferon-gamma by a surfactant: Slowed dissolution of lyophilized formulations in a solution containing 0.03% polysorbate 20. J Pharm Sci 91:543–558. 64. Costantino HR. 2004. Excipients for use in lyophilized pharmaceutical peptide, protein, and other bioproducts. In Lyophilization of biopharmaceuticals; Costantino HR, Pikal MJ, Eds. Arlington, Virginia: AAPS Press, pp 139–228. 65. Kasper JC, Friess W. 2011. The freezing step in lyophilization: Physico-chemical fundamentals, freezing methods and consequences on process performance and quality attributes of biopharmaceuticals. Eur J Pharm Biopharm 78:248–263. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
66. Phillips JT, Rice G, Frohman E, Vande Gaer L, Scott T, Haas J, Eggenberger E, Freedman MS, Stuart W, Cunha L, Jacobs L, Oger J, Arnold D, Murray TJ, DiBiase M, Jethwa V, Goelz S. 2004. A multicenter, open-label, phase II study of the immunogenicity and safety of a new prefilled syringe (liquid) formulation of Avonex in patients with multiple sclerosis. Clin Ther 26:511–521. 67. Kim HJ, Shin CH, Kim CW. 2009. Stabilization of glycoprotein liquid formulation using arginine: A study with lactoferrin as a model protein. Biosci Biotechnol Biochem 73:61–66. 68. Arakawa T, Tsumoto K. 2003. The effects of arginine on refolding of aggregated proteins: Not facilitate refolding, but suppress aggregation. Biochem Biophys Res Commun 304:148–152. 69. Jaber A, Driebergen R, Giovannoni G, Schellekens H, Simsarian J, Antonelli M. 2007. The Rebif new formulation story: It’s not trials and error. Drugs R&D 8:335–348. 70. van Beers MM, Sauerborn M, Gilli F, Brinks V, Schellekens H, Jiskoot W. 2011. Oxidized and aggregated recombinant human interferon beta is immunogenic in human interferon beta transgenic mice. Pharm Res 28:2393–2402. 71. Zheng X, Wu SL, Hancock WS. 2006. Glycation of interferon-beta-1b and human serum albumin in a lyophilized glucose formulation. Part III: Application of proteomic analysis to the manufacture of biological drugs. Int J Pharm 322:136–145. 72. Farrar MA, Schreiber RD. 1993. The molecular cell biology of interferon-gamma and its receptor. Annu Rev Immunol 11:571–611. 73. Miller CH, Maher SG, Young HA. 2009. Clinical use of interferongamma. Ann NY Acad Sci 1182:69–79. 74. Schroder K, Hertzog PJ, Ravasi T, Hume DA. 2004. Interferongamma: An overview of signals, mechanisms and functions. J Leukocyte Biol 75:163–189. 75. Ealick SE, Cook WJ, Vijay-Kumar S, Carson M, Nagabhushan TL, Trotta PP, Bugg CE. 1991. Three-dimensional structure of recombinant human interferon-gamma. Science 252:698–702. 76. Arakawa T, Hsu YR, Yphantis DA. 1987. Acid unfolding and selfassociation of recombinant Escherichia coli derived human interferon gamma. Biochemistry 26:5428–5432. 77. Goossens K, Haelewyn J, Meersman F, De Ley M, Heremans K. 2003. Pressure- and temperature-induced unfolding and aggregation of recombinant human interferon-gamma: A Fourier transform infrared spectroscopy study. Biochem J 370:529–535. 78. Lam XM, Costantino HR, Overcashier DE, Nguyen TH, Hsu CC. 1996. Replacing succinate with glycolate buffer improves the stability of lyophilized interferon-(. Int J Pharm 142:85–95. 79. Lam XM, Patapoff TW, Nguyen TH. 1997. The effect of benzyl alcohol on recombinant human interferon-gamma. Pharm Res 14:725–729. 80. Malek TR. 2008. The biology of interleukin-2. Annu Rev Immunol 26:453–479. 81. Shaker MA, Younes HM. 2009. Interleukin-2: Evaluation of routes of administration and current delivery systems in cancer therapy. J Pharm Sci 98:2268–2298. 82. Robb RJ, Kutny RM, Panico M, Morris HR, Chowdhry V. 1984. Amino acid sequence and post-translational modification of human interleukin 2. Proc Natl Acad Sci USA 81:6486–6490. 83. Bazan JF. 1992. Unraveling the structure of IL-2. Science 257:410– 413. 84. Brandhuber BJ, Boone T, Kenney WC, McKay DB. 1987. Threedimensional structure of interleukin-2. Science 238:1707–1709. 85. ProleukinR (aldesleukin) label. 2011. Prometheus Laboratories Inc. Available from Drugs@FDA, the database for FDA-approved drug products. Accessed June 28, 2014, at: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm 86. Hora M. 2006. Preparing aldesleukin for pharmaceutical use. Patent EP1688146 (A1). 87. Ricci MS, Sarkar CA, Fallon EM, Lauffenburger DA, Brems DN. 2003. pH dependence of structural stability of interleukin-2 and granulocyte colony-stimulating factor. Protein Sci 5:1030–1038. 88. Prestrelski SJ, Pikal KA, Arakawa T. 1995. Optimization of lyophilization conditions for recombinant human interleukin-2 by DOI 10.1002/jps.24243
REVIEW
dried-state conformational analysis using Fourier-transform infrared spectroscopy. Pharm Res 12:1250–1259. 89. OntakR (denileukin difttox) label. 2011. Eisai Inc. Available from Drugs@FDA, the database for FDAapproved drug products. Accessed June 28, 2014, at: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm 90. Du XX, Williams DA. 1994. Interleukin-11: A multifunctional growth factor derived from the hematopoietic microenvironment. Blood 83:2023–2030. 91. Czupryn MJ, McCoy JM, Scoble HA. 1995. Structure–function relationships in human interleukin-11: Identification of regions involved in activity by chemical modification and site-directed mutagenesis. J Biol Chem 270:978–985. 92. Du X, Williams DA. 1997. Interleukin-11: Review of molecular, cell biology, and clinical use. Blood 89:3897–3908. 93. Adams VR, Brenner TL. 1999. Oprelvekin (NeumegaR ). J Oncol Pharm Pract 5:117–124. 94. Page C, Dawson P, Woollacott D, Thorpe R, Mire-Sluis A. 2000. Development of a lyophilization formulation that preserves the biological activity of the platelet-inducing cytokine interleukin-11 at low concentrations. J Pharm Pharmacol 52:19–26. 95. NeumegaR (oprelvekin) label. 2009. Wyeth Pharmaceuticals Inc. Available from Drugs@FDA, the database for FDA-approved drug products. Accessed June 28, 2014, at: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm 96. Takagi A, Masuda H, Takakura Y, Hashida M. 1995. Disposition characteristics of recombinant human interleukin-11 after a bolus intravenous administration in mice. J Pharmacol Exp Ther 275:537–543. 97. Gokarn YR, Kosky A, Kras E, McAuley A, Remmele RL Jr. 2006. Excipients for protein drugs. In Excipient development for pharmaceutical, biotechnology, and drug delivery systems; Katdare A, Chaubal MV, Eds. New York: Informa Healthcare USA, Inc., pp 291–332. 98. Garzon-Rodriguez W, Koval RL, Chongprasert S, Krishnan S, Randolph TW, Warne NW, Carpenter JF. 2004. Optimizing storage stability of lyophilized recombinant human interleukin-11 with disaccharide/hydroxyethyl starch mixtures. J Pharm Sci 93:684–696. 99. Wadhwa M, Thorpe R. 2008. Haematopoietic growth factors and their therapeutic use. Thromb Haemost 99:863–873. 100. Kubota N, Orita T, Hattori K, Oh-eda M, Ochi N, Yamazaki T. 1990. Structural characterization of natural and recombinant human granulocyte colony-stimulating factors. J Biochem 107:486–492. 101. Wingfield P, Benedict R, Turcatti G, Allet B, Mermod JJ, DeLamarter J, Simona MG, Rose K. 1988. Characterization of recombinantderived granulocyte-colony stimulating factor (G-CSF). Biochem J 256:213–218. 102. Hill CP, Osslund TD, Eisenberg D. 1993. The structure of granulocyte-colony-stimulating factor and its relationship to other growth factors. Proc Natl Acad Sci USA 90:5167–5171. 103. Lonquex Product Information. 2014. Sicor Biotech UAB, Teva Pharmaceuticals Europe. Available from the European Medicines Agency database of medicines. Accessed June 28, 2014, at: http://www.ema.europa.eu/ema/ ¨ 104. Mahlert F, Schmidt K, Allgaier H, Liu P, Muller U, Shen WD. 2013. Rational development of lipegfilgrastim, a novel long-acting granulocyte colony-stimulating factor, using glycopegylation technology. Blood 122:4853–4853. 105. Krishnan S, Chi EY, Webb JN, Chang BS, Shan D, Goldenberg M, Manning MC, Randolph TW, Carpenter JF. 2002. Aggregation of granulocyte colony stimulating factor under physiological conditions: Characterization and thermodynamic inhibition. Biochemistry 41:6422–6431. 106. Chi EY, Krishnan S, Kendrick BS, Chang BS, Carpenter JF, Randolph TW. 2003. Roles of conformational stability and colloidal stability in the aggregation of recombinant human granulocyte colonystimulating factor. Protein Sci 12:903–913. 107. Yamazaki K, Murayama K, Ishikawa R, Ozaki Y. 2005. An infrared spectroscopy study of acid stability and thermal unfolding process of granulocyte-colony stimulating factor. J Biochem 137:265–271. DOI 10.1002/jps.24243
19
108. Thirumangalathu R, Krishnan S, Brems DN, Randolph TW, Carpenter JF. 2006. Effects of pH, temperature, and sucrose on benzyl alcohol-induced aggregation of recombinant human granulocyte colony stimulating factor. J Pharm Sci 95:1480–1497. 109. Oh-eda M, Hasegawa M, Hattori K, Kuboniwa H, Kojima T, Orita T, Tomonou K, Yamazaki T, Ochi N. 1990. O-linked sugar chain of human granulocyte colony-stimulating factor protects it against polymerization and denaturation allowing it to retain its biological activity. J Biol Chem 265:11432–11435. 110. Lu HS, Fausset PR, Narhi LO, Horan T, Shinagawa K, Shimamoto G, Boone TC. 1999. Chemical modification and site-directed mutagenesis of methionine residues in recombinant human granulocyte colony-stimulating factor: Effect on stability and biological activity. Arch Biochem Biophys 362:1–11. 111. Yin J, Chu JW, Ricci MS, Brems DN, Wang DI, Trout BL. 2004. Effects of antioxidants on the hydrogen peroxide-mediated oxidation of methionine residues in granulocyte colony-stimulating factor and human parathyroid hormone fragment 13–34. Pharm Res 21:2377– 2383. 112. S¨orgel F, Lerch H, Lauber T. 2010. Physicochemical and biologic comparability of a biosimilar granulocyte colony-stimulating factor with its reference product. Biodrugs 24:347–357. 113. Ha E, Wang W, Wang YJ. 2002. Peroxide formation in polysorbate 80 and protein stability. J Pharm Sci 91:2252–2264. 114. Committee for Medicinal Products for Human Use (CHMP) assessment report for Zarzio. 2008. European Medicines Agency. EMEA/CHMP/651339/2008. Accessed June 28, 2014, at: http: //www.ema.europa.eu/docs/en GB/document library/EPAR - Public assessment report/human/000917/WC500046528.pdf 115. Piedmonte DM, Treuheit MJ. 2008. Formulation of Neulasta (pegfilgrastim). Adv Drug Deliv Rev 60:50–58. 116. Martin-Christin F. 2001. Granulocyte colony stimulating factors: How different are they? How to make a decision? Anti-cancer drug 12:185–191. 117. Fournier-Broggini E. 2001. Stability studies of a new formulation of glycosylated rHuG-CSF (lenograstim), stable at room temperature for 2 years. Bone Marrow Transpl 27:S164-S164. 118. Granocyte (lenograstim) data sheet. 1998. Chugai Pharmaceuticals Co, Ltd, AMRAD Pharmaceuticals Pty Ltd. MEDSAFE, New Zealand Medicines and Medical Devices Safety Authority. Accessed June 28, 2014, at: http://www.medsafe.govt.nz/profs/datasheet/g/Granocyteinj.htm. 119. Stevenson C, Hastedt JE, Lehrman SR, Chiang H-S, Bennett DB, Lesikar D, Yang B, Gong D, Cabot K. 2002. Inhaleable spray dried 4-helix bundle protein powders having minimized aggregation. International patent WO2002011695 A2. 120. Mattern M, Winter G, Kohnert U, Lee G. 1999. Formulation of proteins in vacuum-dried glasses. II. Process and storage stability in sugar-free amino acid systems. Pharm Dev Technol 4:199–208. 121. Wingfield P, Graber P, Moonen P, Craig S, Pain RH. 1988. The conformation and stability of recombinant-derived granulocytemacrophage colony stimulating factors. Eur J Biochem 173:65– 72. 122. Bertolotto A, Deisenhammer F, Gallo P, Solberg Sorensen P. 2004. Immunogenicity of interferon beta: Differences among products. J Neurol 251:II15–II24. 123. Basu A, Yang K, Wang M, Liu S, Chintala R, Palm T, Zhao H, Peng P, Wu D, Zhang Z, Hua J, Hsieh MC, Zhou J, Petti G, Li X, Janjua A, Mendez M, Liu J, Longley C, Zhang Z, Mehlig M, Borowski V, Viswanathan M, Filpula D. 2006. Structure-function engineering of interferon-beta-1b for improving stability, solubility, potency, immunogenicity, and pharmacokinetic properties by site-selective monoPEGylation. Bioconjugate Chem 17:618–630. 124. Hu X, Miller L, Richman S, Hitchman S, Glick G, Liu S, Zhu Y, Crossman M, Nestorov I, Gronke RS, Baker DP, Rogge M, Subramanyam M, Davar G. 2012. A novel PEGylated interferon beta-1a for multiple sclerosis: Safety, pharmacology, and biology. J Clin Pharmacol 52:798–808. Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
20
REVIEW
125. Biogen Idec product pipeline. 2014. Accessed June 28, 2014, at: http://www.biogenidec.com/research product pipeline.aspx?ID = 5778. 126. Braun A, Kwee L, Labow MA, Alsenz J. 1997. Protein aggregates seem to play a key role among the parameters influencing the antigenicity of interferon alpha (IFN-alpha) in normal and transgenic mice. Pharm Res 14:1472–1478. 127. Chou DK, Krishnamurthy R, Manning MC, Randolph TW, Carpenter JF. 2012. Physical stability of albinterferon-alpha(2b) in aqueous solution: Effects of conformational stability and colloidal stability on aggregation. J Pharm Sci 101:2702–2719. 128. Foureau D, Vrikkis R, Jones C, Weaver K, MacFarlane D, Salo J, McKillop I, Elliott G. 2012. In vitro assessment of choline dihydrogen phosphate (CDHP) as a vehicle for recombinant human interleukin-2 (rhIL-2). Cel Mol Bioeng 5:390–401. 129. Jhunjhunwala S, Balmert SC, Raimondi G, Dons E, Nichols EE, Thomson AW, Little SR. 2012. Controlled release formulations of IL2, TGF-beta1 and rapamycin for the induction of regulatory T cells. J Control Release 159:78–84. 130. Takagi A, Yamashita N, Yoshioka T, Takaishi Y, Nakanishi K, Takemura S, Maeda A, Saito K, Takakura Y, Hashida M. 2006. Incorporation into a biodegradable hyaluronic acid matrix enhances in vivo efficacy of recombinant human interleukin 11 (rhIL11). J Control Release 115:134–139. 131. Takagi A, Yamashita N, Yoshioka T, Takaishi Y, Sano K, Yamaguchi H, Maeda A, Saito K, Takakura Y, Hashida M. 2007. Enhanced pharmacological activity of recombinant human interleukin-11 (rhIL11) by chemical modification with polyethylene glycol. J Control Release 119:271–278. 132. Jung Y, Ahn H, Kim D-S, Hwang YR, Ho S-H, Kim J-M, Kim S, Ma S, Kim S. 2011. Improvement of biological and pharmacokinetic features of human interleukin-11 by site-directed mutagenesis. Biochem Biophys Res Commun 405:399–404. 133. Gasmi N, Lassoued R, Ayed A, Treton B, Chevret D, Nicaud JM, Kallel H. 2012. Production and characterization of human granulocytemacrophage colony-stimulating factor (hGM-CSF) expressed in the oleaginous yeast Yarrowia lipolytica. Appl Microbiol Biotechnol 96:89– 101. 134. Ceaglio N, Etcheverrigaray M, Kratje R, Oggero M. 2010. Influence of carbohydrates on the stability and structure of a hyperglycosylated human interferon alpha mutein. Biochimie 92:971– 978.
Lipi¨ainen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
135. Kumar V, Sharma VK, Kalonia DS. 2009. In situ precipitation and vacuum drying of interferon alpha-2a: Development of a singlestep process for obtaining dry, stable protein formulation. Int J Pharm 366:88–98. 136. Bristol-Myers Squibb pipeline. 2014. Accessed June 28, 2014, at: http://www.bms.com/research/pipeline/Pages/default.aspx. 137. Merck Serono pipeline. 2014. Accessed June 28, 2014, at: http:// www.merckserono.com/en/research development/pipeline/pipeline.html. 138. Roche pipeline. 2014. Accessed June 28, 2014, at: http:// www.roche.com/research and development/who we are how we work/ pipeline.htm. 139. Pfizer pipeline. 2014. Accessed June 28, 2014, at: http:// www.pfizer.com/research/science and technology/product pipeline. 140. Shi ZT, Lin Y, Wang J, Wu J, Wang RF, Chen FQ, Mi WJ, Qiu JH. 2013. G-CSF attenuates noise-induced hearing loss. Neurosci Lett 6:102–106. 141. Fortunato G, Vidal DTA, Klein W, Neto A, Angrizani A, Vasconcelos JF, Kaneto C, Souza BSdF, Ribeiro-dos-Santos R, Soares MBP, Macambira SG. 2013. Recovery of pulmonary structure and exercise capacity by treatment with granulocyte-colony stimulating factor (GCSF) in a mouse model of emphysema. Pulm Pharmacol Ther 27:144– 149. 142. Favors SE, Curd LM, Gregg RK. 2012. Use of the antiinflammatory cytokine interleukin-11 to reverse HIV-1gp120 repression of a natural killer cell line. Cell Immunol 276:1–5. 143. Ruiz L, Rodriguez I, Baez R, Aldana R. 2007. Stability of an extemporaneously prepared recombinant human interferon alfa-2b eye drop formulation. Am J Health Syst Pharm 64:1716–1719. 144. Francisco-Cruz A, Aguilar-Santelises M, Ramos-Espinosa O, Mata-Espinosa D, Marquina-Castillo B, Barrios-Payan J, HernandezPando R. 2014. Granulocyte-macrophage colony-stimulating factor: Not just another haematopoietic growth factor. Med Oncol 31:774. 145. Label information. Drugs@FDA. Database for FDA Approved Drug Products. FDA Center for Drug Evaluation and Research. 2014. Accessed June 28, 2014, at: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm. 146. Product information. Search for medicines. Database of medicines. European Medicines Agency. 2014. Accessed June 28, 2014, at: http://www.ema.europa.eu/ema/. 147. Information on medicinal products. FimeaWeb search. Finnish Medicines Agency. 2014. Accessed June 28, 2014, at: http://www.fimea.fi/laaketieto/laakehaku.
DOI 10.1002/jps.24243