Interactions of formulation excipients with proteins in solution and in the dried state

https://doi.org/10.1016/j.addr.2011.06.011Get rights and content

Abstract

A variety of excipients are used to stabilize proteins, suppress protein aggregation, reduce surface adsorption, or to simply provide physiological osmolality. The stabilizers encompass a wide variety of molecules including sugars, salts, polymers, surfactants, and amino acids, in particular arginine. The effects of these excipients on protein stability in solution are mainly caused by their interaction with the protein and the container surface, and most importantly with water. Some excipients stabilize proteins in solution by direct binding, while others use a number of fundamentally different mechanisms that involve indirect interactions. In the dry state, any effects that the excipients confer to proteins through their interactions with water are irrelevant, as water is no longer present. Rather, the excipients stabilize proteins through direct binding and their effects on the physical properties of the dried powder. This review will describe a number of mechanisms by which the excipients interact with proteins in solution and with various interfaces, and their effects on the physical properties of the dried protein structure, and explain how the various interaction forces are related to their observed effects on protein stability.

Introduction

Many proteins are structurally unstable in solution, and are susceptible to conformational changes due to various stresses encountered during purification, processing, and storage [1], [2], [3], [4], [5], [6], [7], [8]. These stresses include elevated temperature, exposure to extreme pH, shear strain, and surface adsorption, to name a few [5], [6]. Thus, protein-based pharmaceuticals have the potential to undergo physical degradation (e.g., unfolding, aggregation, and insoluble particulate formation) by a number of mechanisms, which can negatively impact both the efficacy and safety of the therapeutic product [7], [8]. The solvent environment of the protein plays a major role in determining its stability [9]. Numerous solvent additives, the so-called “osmolytes”, have been shown to enhance the stability of proteins and, as a consequence, reduce the aggregation of marginally stable proteins [10], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28]. In this case, protein unfolding precedes aggregation, and the structure-stabilizing co-solvents reduce aggregation by stabilizing the native structure. The lack of affinity for, or repulsive interaction with, the protein surface is the reason why these co-solvents stabilize the protein structure. Conversely, excipients such as arginine, surfactants, proteins, and polymers are often used to suppress protein aggregation without enhancing its stability [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38]. These additives exert their effects by weakly binding to the protein surface, or by competitively binding to the surface/interface that have the potential to destabilize the protein structure. Some of these excipients are also used to stabilize proteins in the dry state. However, in the absence of water, fundamentally different mechanisms are in effect, as any mechanism that involves excipient-water interactions will not play its part. This chapter summarizes the effects of additives that are used to mitigate protein aggregation and will discuss the mechanistic basis of their effects both in solution and in the dried state. In addition, the effects of additives on protein stability during freezing will also be discussed, as freezing is an intermediate processing step involved in lyophilization. It should be noted that as water is still present, yet is gradually removed during ice crystallization, the freezing process involves an interesting physical state that is described mainly by interaction forces that are present in solution.

Section snippets

Solution

A wide variety of protein stabilizing excipients is used for enhancing the stability of both pharmaceutical and reagent proteins and they are referred to as stabilizing co-solvents [9], [22], [23], [24]. These excipients have been reported to stabilize the structure of native proteins at moderate (0.1 M) to high concentrations (1 M). In fact, these compounds played a critical role at the dawn of classical enzymology and biochemistry of cellular proteins. Many proteins are inactivated when they

Solution

Hydrophilic polymers have often been used to stabilize proteins and enhance protein assembly [152], [153], [154]. Sasahara et al. [155] demonstrated that the stability of a protein against heat treatment was increased through the incorporation of dextran. Manning et al. [156] have studied the effects of polymeric excipients on the thermally-induced aggregation of low molecular weight urokinase, and found hydroxyl ethyl (HETA) starch, PEG4000, and gelatin to all be effective in stabilizing the

Solution

Surfactants are widely used to stabilize proteins, suppress aggregation and assist in protein refolding [217], [218]. Polysorbate 80 (polyoxyethylene sorbitan monooleate) and polysorbate 20 (polyoxyethylene sorbitan monolaurate) are two of the widely incorporated surfactants in marketed protein pharmaceuticals [176], [178], [219], and are typically used in the 0.0003–0.3% range [176]. The effects of surfactants and their interaction mechanism in aqueous solution is described in more detail

Solution

The effects of amino acids in general were described earlier in Section 2. Here, the focus is placed on one specific amino acid, arginine. Arginine is not a protein-stabilizing excipient, but is highly effective in suppressing protein aggregation. Due to this effect and its safety in humans, arginine is frequently used for enhancing the shelf life of proteins. The aggregation-suppressing effect of arginine was accidentally observed by Rudolph and Fischer [256] during their attempt to prevent

Overall discussion on mechanism

The mechanism of each class of excipients for their effects on protein stability, solubility, and aggregation in both liquid and lyophilized formulations has been described above based on their interactions with proteins. In liquid formulations, there are primarily two different modes of interactions present between excipients and proteins, or container surfaces. Those that enhance protein stability demonstrate an unfavorable interaction with the protein. Fig. 9 demonstrates how such an

Conclusion

We have shown here the effects of four classes of co-solvents (excipients), i.e., protein-stabilizers, polymers, surfactants, and arginine on the formulation and stability of proteins in solution and dry state. The efficacy of these excipients in conferring stability to proteins has been approached from the mechanistic point of view, highlighting the various interaction forces present under different protein environmental conditions. Typical protein formulation contains several components, and

References (290)

  • B.-L. Chen et al.

    Strategies to suppress aggregation of recombinant keratinocyte growth factor during liquid formulation development

    J. Pharm. Sci.

    (1994)
  • T. Arakawa et al.

    Protection of bovine serum albumin from aggregation by Tween 80

    J. Pharm. Sci.

    (2000)
  • T. Arakawa et al.

    Stabilizing effects of caprylate and acetyltryptophanate on heat-induced aggregation of bovine serum albumin

    Biochim. Biophys. Acta.

    (2000)
  • T. Arakawa et al.

    The effects of arginine on refolding of aggregated proteins: not facilitate refolding, but suppress aggregation

    Biochem. Biophys. Res. Commun.

    (2003)
  • T. Arakawa et al.

    Suppression of protein interactions by arginine: a proposed mechanism of the arginine effects

    Biophys. Chem.

    (2007)
  • T. Arakawa et al.

    Protein precipitation and denaturation by dimethyl sulfoxide

    Biophys. Chem.

    (2007)
  • A. Tiwari et al.

    Stabilization of yeast hexokinase A by polyol osmolytes: correlation with the physicochemical properties of aqueous solutions

    Biophys. Chem.

    (2006)
  • S. Ohtake et al.

    Trehalose: current use and future applications

    J. Pharm. Sci.

    (2011)
  • A.G. Welch et al.

    Non-A, non-B hepatitis from intravenous immunoglobulin

    Lancet

    (1983)
  • T.K. Rosengart et al.

    Heparin protects heparin-binding growth factor-I from proteolytic inactivation in vitro

    Biochem. Biophys. Res. Commun.

    (1988)
  • T. Kajio et al.

    Stabilization of basic fibroblast growth factor with dextran sulfate

    FEBS Lett.

    (1992)
  • D.B. Volkin et al.

    Physical stabilization of acidic fibroblast growth factor by polyanions

    Arch. Biochem. Biophys.

    (1993)
  • D.B. Volkin et al.

    Sucralfate and soluble sucrose octasulfate bind and stabilize acidic fibroblast growth factor

    Biochim. Biophys. Acta.

    (1993)
  • J. Wen et al.

    Characterization of keratinocyte growth factor binding to heparin and dextran sulfate

    Arch. Biochem. Biophys.

    (1996)
  • B.S. Chang et al.

    Physical factors affecting the storage stability of freeze-dried interleukin-1 receptor antagonist: glass transition and protein conformation

    Arch. Biochem. Biophys.

    (1996)
  • S.J. Prestrelski et al.

    Dehydration-induced conformational transitions in proteins and their inhibition by stabilizers

    Biophys. J.

    (1993)
  • L. Kreilgaard et al.

    Effects of additives on the stability of recombinant human factor XIII during freeze-drying and storage in the dried solid

    Arch. Biochem. Biophys.

    (1998)
  • J.F. Carpenter et al.

    Cryoprotection of phosphofructokinase with organic solutes: characterization of enhanced protection in the presence of divalent cations

    Arch. Biochem. Biophys.

    (1986)
  • J.F. Carpenter et al.

    Stabilization of phosphofructokinase with sugars during freeze-drying: characterization of enhanced protection in the presence of divalent cations

    Biochim. Biophys. Acta.

    (1987)
  • B. Chen et al.

    Influence of calcium ions on the structure and stability of recombinant human deoxyribonuclease I in the aqueous and lyophilized states

    J. Pharm. Sci.

    (1999)
  • H.R. Costantino et al.

    Fourier-transform infrared spectroscopic investigation of protein stability in the lyophilized form

    Biochim. Biophys. Acta.

    (1995)
  • S.N. Timasheff

    Control of protein stability and reactions by weakly interacting cosolvents: the simplicity of the complicated

    Adv. Protein Chem.

    (1998)
  • S.N. Timasheff

    Thermodynamic binding and site occupancy in the light of the Schellman exchange concept

    Biophys. Chem.

    (2002)
  • K.C. Aune et al.

    Thermodynamics of the denaturation of lysozyme by guanidine hydrochloride. II. Dependence on denaturant concentration at 25 degrees

    Biochemistry

    (1969)
  • R. Biltonen et al.

    Studies of the chymotrypsinogen family of proteins. VII. Thermodynamic analysis of transition I of alpha-chymotrypsin

    J. Am. Chem. Soc.

    (1969)
  • E.Y. Chi et al.

    Physical stability of proteins in aqueous solution: mechanism and driving forces in nonnative protein aggregation

    Pharm. Res.

    (2003)
  • M.E.M. Cromwell et al.

    Protein aggregation and bioprocessing

    AAPS J.

    (2006)
  • A.S. Rosenberg

    Effects of protein aggregates: an immunologic perspective

    AAPS J.

    (2006)
  • K. Brorson et al.

    Defining your product profile and maintaining control over it, Part 4. Product-related impurities: tackling aggregates

    Bioprocess Int.

    (2005)
  • T. Arakawa et al.

    Protein–solvent interactions in pharmaceutical formulations

    Pharm. Res.

    (1991)
  • J.F. Back et al.

    Increased thermal stability of proteins in the presence of sugars and polyols

    Biochemistry

    (1979)
  • S.Y. Gerlsma

    The effects of polyhydric and monohydric alcohols on the heat induced reversible denaturation of chymotrypsinogen A

    Eur. J. Biochem.

    (1970)
  • P.H. Von Hippel et al.

    The effect of ions on the kinetics of formation and the stability of the collagenfold

    Biochemistry

    (1962)
  • K. Gekko

    Calorimetric study on thermal denaturation of lysozyme in polyol–water mixtures

    J. Biochem.

    (1982)
  • K. Gekko et al.

    Increased thermal stability of collagen in the presence of sugars and polyols

    J. Biochem.

    (1983)
  • G.C. Na

    Interaction of calf skin collagen with glycerol: linked function analysis

    Biochemistry

    (1986)
  • K. Gekko et al.

    Mechanism of protein stabilization by glycerol: preferential hydration in glycerol–water mixtures

    Biochemistry

    (1981)
  • T. Arakawa et al.

    Stabilization of protein structure by sugars

    Biochemistry

    (1982)
  • B.-L. Chen et al.

    Stabilization of recombinant human keratinocyte growth factor by osmolytes and salts

    J. Pharm. Sci.

    (1996)
  • B.-L. Chen et al.

    Aggregation pathway of recombinant human keratinocyte growth factor and its stabilization

    Pharm. Res.

    (1994)
  • Cited by (297)

    View all citing articles on Scopus

    This review is part of the Advanced Drug Delivery Reviews theme issue on ”Formulating Biomolecules: Mechanistics Insights in Molecular Interactions”.

    View full text