Open access peer-reviewed chapter

Biobetters: IFN-α2b Variants with Reduced Immunogenicity for the Treatment of Human Viral Diseases

Written By

Eduardo F. Mufarrege, Lucía C. Peña and Marina Etcheverrigaray

Submitted: 19 May 2023 Reviewed: 29 May 2023 Published: 06 December 2023

DOI: 10.5772/intechopen.112006

From the Edited Volume

Antiviral Strategies in the Treatment of Human and Animal Viral Infections

Edited by Arli Aditya Parikesit

Chapter metrics overview

45 Chapter Downloads

View Full Metrics

Abstract

For more than three decades, IFN-α2b has been widely used for the treatment of multiple human viral infections such as chronic hepatitis B and C, and certain types of cancers. However, IFN-α2b can be immunogenic, and these undesired immune responses can lead to a decrease in therapeutic efficacy. In addition, IFN-α therapy has also been associated with the progression of certain autoimmune diseases. For these reasons, the development of new IFN-α2b versions with reduced (or even null) immunogenicity has become the focus of several investigations. The “de-immunization” strategies usually involve several steps starting with T cell epitope identification and mutation of those immunogenic residues using immuno-informatics tools. Then, further experimental validation through in vitro and in vivo experimental platforms is needed to confirm in silico predictions. In this chapter, we will review the main strategies addressed so far to develop more effective and safer IFN-based therapies.

Keywords

  • interferon alpha
  • antiviral therapy
  • immunogenicity
  • de-immunization
  • biobetters
  • in silico epitope prediction
  • in vitro assay
  • in vivo assay

1. Introduction

Interferons (IFN) were initially discovered in 1957 by Lindenmann and Isaacs while performing infection experiments on chicken cells with the influenza virus [1]. In this study, the investigators observed a factor secreted by the infected cells that induced an antiviral state in the cell itself and in neighboring cells. This and other further studies [2] established the basis for elucidating the precise mechanism of action of IFNs. These secreted factors were later named “interferon” because of their ability to interfere with viral infections.

IFNs constitute a family of species-specific cytokines secreted in an autocrine and paracrine manner by host cells in response to pathogens, mainly viruses, and microbial products such as endotoxins, pyrans, and double-stranded polyribonucleotides, among others. They are composed of polypeptides ranging in molecular weights from 20 to 100 kDa. Like most proteins, they are sensitive to proteolytic enzymes and heat, although relatively stable at low pH values [3]. Essentially, the main function of IFNs is to increase the transcription of hundreds of IFN-stimulated genes (ISGs) whose products play a key role in the innate and adaptive immune response to viral infections, producing profound metabolic changes [4].

Due to the antiviral and immunomodulatory function of IFN-α2b, this protein was developed as a recombinant version (rhIFN-α2b), which was first approved by the Food and Drug Administration (FDA) in June 1986 for the treatment of hairy cell leukemia. Then, in 1988, its use was also approved for the treatment of AIDS-related Kaposi’s Sarcoma and in 1991 and 1992 to treat chronic hepatitis B and C virus infections, respectively.

Since its approval as an antiviral agent more than three decades ago, the use of rhIFN-α2b in the clinic has grown markedly. However, some adverse effects can be observed during rhIFN-α2b-based therapy. Among the most common unwanted effects are flu-like symptoms such as fever, fatigue, myalgia, chills, and headache. Also, subacute and chronic side effects such as myelosuppression, neuropsychiatric effects, and immunogenicity events can occur [5]. Regarding product immunogenicity, the development of antidrug antibodies is the most frequently detected response in patients. As an example, more than 40% of patients with malignant intestinal tumors showed anti-rhIFN-α2b antibody formation [6]. In addition, a similar proportion of patients suffering from renal cell carcinoma showed the presence of neutralizing antibodies after being treated with this biologic [7, 8, 9]. These rhIFN-α2b-based therapy unwanted effects represented a major challenge in the biopharmaceutical industry and prompted the development of new rhIFN-α2b variants with reduced immunogenicity. In the present chapter, we will discuss the main strategies addressed to develop reduced immunogenicity (de-immunized) versions of the cytokine.

Another drawback related to rhIFN-α2b therapy is the protein’s short circulating half-life, which poses the need for repeated high doses to reach the therapeutic window. This can lead to the appearance of treatment adverse effects. In general, the higher the dose of rhIFN-α2b administered and the longer the treatment, the more frequent and severe adverse effects are observed. To circumvent this issue, numerous platforms based on glycoengineering and pegylation have been approached with encouraging results. Here, we will describe the strategies that led to more stable and safer rhIFN-α2b-based products.

Advertisement

2. Antiviral action of IFN-α

IFN-α triggers intracellular signaling cascades that activate multiple immune functions, affecting numerous cellular processes such as proliferation, differentiation, activation, migration, and apoptosis [10].

In the canonical signaling pathway, known as the JAK–STAT pathway (Janus kinase pathway, JAK, and the signal transducer and activator of transcription, STAT), the binding of IFN-α promotes the IFNAR heterodimeric receptor complex dimerization, which enables phosphorylation and activation of the Janus family kinases JAK1 and TYK2. Then, JAK1 and TYK2 recruit and activate STAT1 and STAT2 proteins, which allow their dimerization and consequently the recruitment of IFN regulatory factor nine (IRF9). These three proteins form the ternary ISGF3 complex, capable of translocating to the cell nucleus where they bind to IFN-stimulated response elements (ISREs) in the DNA and mediate the expression of different IFN-stimulated genes (ISGs) involved in the development of antiviral, proinflammatory, and immunomodulatory responses. Additionally, through the noncanonical signaling pathway, IFN-α binding induces the expression of ISGs from the activation of other STAT proteins (such as STAT 3, 5, and 6) or through the mitogen-activated kinases (MAPKs) or phosphatidylinositol 3-kinase (PI3K) pathway [10].

Thus, IFN-α mediates antiviral actions by inhibiting the virion cycle and promoting enzyme synthesis, which interferes with viral particle transport, viral DNA transcription, replication, or RNA translation [11, 12].

Also, IFN-α elicits a powerful immunomodulatory action aiming to promote an effective antiviral immune response. For instance, IFN-α induces the activation of macrophages and NK cells, enhancing their cytotoxic activity, which helps promote the development of adaptive immune responses by increasing the expression of major histocompatibility complex (MHC) class l and ll molecules. As a result, this enhances the cytotoxic T cell-mediated killing efficiency of infected cells. Furthermore, IFN-α favors cell sequestration in lymph nodes, which promotes their activation and acts directly on T and B cells, guiding the progression of the antiviral response [13].

In addition, IFN-α inhibits cell proliferation through the regulation of different genes involved in the cell cycle, such as c-myc oncogenes, cyclin D3, phosphatase CDC25A [14], and cyclin-dependent kinase CDK-2 [15], or through the activation of numerous proapoptotic genes and proteins [16].

Advertisement

3. Immunogenicity of rhIFN-α2b in the clinic

rhIFN-α2b therapy can lead to the formation of antidrug antibodies. These antibodies can have varied consequences on the patient and the therapy efficacy. For example, these antibodies can bind to the biologic without having a significant impact on its biological effect or, on the contrary, neutralize its function or affect its pharmacokinetic properties [17, 18]. Regarding the nature of the antibody response, there are cases where rhIFN-α2b treatment resulted in the appearance of neutralizing IgG1 antibodies [19]. However, there is also evidence demonstrating the emergence of anti-rhIFN-α2b IgM antibodies [20]. Although the persistence and affinity of these antibodies are lower than the IgG antibody-mediated response, the consequences are potentially more severe due to their multivalent nature and the capacity to engage complement molecules and promote Fc-receptor cross-linking [20].

rhIFN-α2b immunogenicity was also evidenced by the appearance or exacerbation of certain autoimmune diseases. This is the case of autoimmune thyroiditis associated with rhIFN-α2b therapy. Initially, the occurrence of this disease was attributed to impurities in the biologic [21, 22]. However, the persistence of this disease after interruption of treatment and replacement with a product of higher purity showed that the observed autoimmunity was a direct consequence of the rhIFN-α2b proinflammatory capacity [23, 24]. The product’s proinflammatory properties could explain the immunogenicity events observed in the clinic. There is growing evidence showing that the presence of T cell epitopes in the molecule can facilitate a proinflammatory context and also contribute to the development of antidrug antibodies [25, 26, 27, 28]. Specifically, three regions enriched in T cell epitopes were identified in the rhIFN-α2b sequence, of which the region located at the c-terminal end of the molecule was found to be the most immunogenic [29].

The relevance of rhIFN-α2b immunogenicity in the clinic highlights the many efforts made to mitigate the effect of this unwanted drug attribute during treatment. The most promising strategies developed so far will be discussed in detail below.

Advertisement

4. Strategies aimed at reducing rhIFN-α2b immunogenicity

Because of the risk to patient health and therapy efficacy, several strategies are now available to reduce or mitigate the effects of rhIFN-α2b immunogenicity. Some of these involve modification of the protein, whereas others include coadministration of immunosuppressors.

Among the developments involving product modification, changes in the glycosylation pattern and the addition of polyethylene glycol (PEG) residues in the molecule deserve special mention. While the initial goal of these strategies was to increase the plasma half-life through an increase in apparent size and changes in the molecule’s overall charge, these modifications could also exert a masking effect on potentially immunogenic epitopes. More recently, the advent of immuno-informatics and in vitro and in vivo experimental platforms have further enabled the development of de-immunized versions of biologics. These new biotherapeutic variants had modified the most immunogenic residues, thus reducing the binding of the biologic-derived peptides to human MHC, and consequently, the activation of specific T and B cells.

We will now review these strategies, highlighting their main advantages and limitations.

4.1 Development of rhIFN-α2b variants by glycoengineering

Glycoengineering is a modification strategy that is based on the linkage of glycan residues to the protein structure through covalent bonds [30]. This technology allows optimizing several macromolecular properties, such as increased solubility in aqueous solvents, thermal stability, as well as proteolytic and chemical degradation. In addition, the attachment of glycans can also lead to changes in specificity, affinity, and protein biological activity. Furthermore, there are studies showing that the presence of glycans could reduce the formation of aggregates and improve polypeptide folding and final protein conformation [31, 32, 33, 34].

Protein glycosylation can take place in a variety of ways, on the side chains of the amino acids that constitute the polypeptide chain. However, the main forms of sugar linkage are through binding to the N atoms of the asparagine (Asn) side chain or the oxygen atom of the amino acids serine and threonine. In the first case, glycosylation is called N-glycosylation, while in the second it is known as O-glycosylation.

4.1.1 Development of N and O-glycosylated versions of rhIFN-α2b

rhIFN-α2b is a low molecular mass molecule and, therefore, susceptible to plasma clearance once it reaches the blood circulation. Indeed, the protein is nearly undetectable after 1-day post subcutaneous administration [35]. As a result, multiple doses of the product are required to reach the therapeutic window. For this reason, several approaches have been addressed to increase the apparent molecular size and, consequently, the stability, half-life, and in vivo biological activity of the molecule. Among the most widely used strategies, the incorporation of amino acid residues that allows the addition of specific glycans during posttranslational protein processing stands out [31, 36]. This not only increases cytokine size but also the overall protein charge, properties that are closely linked to renal clearance [37]. An additional effect is exerted by the sialic acid that decorates the glycans attached to the protein. This sugar residue restricts the binding of the cytokine to the liver receptors and consequently prolongs the half-life and in vivo biological activity [38, 39].

An interesting study using glycoengineering as a strategy to increase plasma half-life was addressed by Ceaglio et al. [40]. The authors modified the rhIFN-α2b sequence to add N-glycosylation sites to the molecule. These mutations were selected so as not to negatively impact the structure/function of the cytokine. Thus, the new hyperglycosylated version, known as 4 N-IN, evidenced a marked increase in plasma half-life and a significant improvement in systemic clearance in animal experiments. However, the in vitro antiviral activity showed a significant decrease as compared to the original molecule [40].

Similarly, another hyperglycosylated rhIFN-α2b was recently developed and referred to as GMOP-IFN. This new variant consisted of the addition at the N-terminal end of the cytokine of a peptide tag derived from the human granulocyte and macrophage colony-stimulating factor (GM-CSF), which contained additional O-glycosylation sites. As a result, the therapeutic protein was endowed with a higher sialic acid content and larger apparent molecular size. These properties allowed improved pharmacokinetic parameters in rats and increased thermal and plasma stability. In addition, unlike what was observed for 4 N-IFN, GMOP-IFN evidenced an antiviral activity similar to that of the wild-type molecule [41].

However, we further demonstrated using T cell proliferation and cytokine profiling assays that 4 N-IFN and GMOP-IFN were immunogenic. Moreover, through an immuno-informatics toolkit, we not only identified the potentially immunogenic amino acids and regions for both molecules but also the specific mutations that allow for disruption in the binding of IFN-derived peptides to the most prevalent human MHC (HLA, human leukocyte antigen) molecules in the world population [42, 43]. The development of these new variants will be discussed in detail below.

4.2 Use of pegylation for the development of rhIFN-α2b versions with enhanced stability

In addition to glycoengineering, another successful strategy to increase the circulating half-life of proteins for human therapeutic use is through the addition of polyethylene glycol (PEG) molecules. One case that exemplifies this is PEGIntron® produced by Schering Corporation. PEGIntron is a pegylated version of rhIFN-α2b which, due to the addition of a small PEG structure (approximately 12 kDa), exhibits increased residual biological activity. However, PEGIntron achieves only a modest improvement in circulating half-life over the original protein [44, 45, 46, 47]. In this product, almost half of the PEG residues added to the molecule are exclusively bound to histidine and show high instability in water. This could explain, at least partially, the limited improvement in the stability of the product with respect to the nonpegylated molecule [44, 46].

To circumvent this drawback, another product based on rhIFN-α2b was developed by Roche and includes the addition of a 40 kDa molecule, called PEGASYS®. This product is more heterogeneous and consists of different isoforms with varying biological potency [47, 48]. As a consequence of the addition of a larger PEG molecule, the plasma half-life is increased. However, this also results in a lower biological activity, which ranges between 1 and 7% with respect to the unmodified cytokine [47].

A remarkable improvement in rhIFN-α2b pegylation technology was addressed by Rosendahl et al. [49]. In this study, the authors demonstrated that the addition of only one cysteine in the rhIFN-α2b sequence followed by pegylation at that residue allowed the development of a homogeneous mono-pegylated product. Among the cysteine residues whose mutation does not lead to a significantly increased immunogenicity, mutein Q5C showed the most encouraging results. In fact, pharmacokinetic studies in rats inoculated subcutaneously with the product showed a plasma half-life 20 to 40 times higher than that of the unmodified protein [49].

In addition to facilitating an increased rhIFN-α2b in vivo stability, pegylation is frequently associated with reducing product immunogenicity [50, 51, 52]. Despite this, the presence of neutralizing anti-rhIFN-α2b antibodies has been detected in patients with chronic hepatitis C treated with pegylated rhIFN-α2b. Moreover, these antibodies caused a lack of response to IFN-based therapy, and consequently, these patients failed viral clearance [53].

4.3 Development of de-immunized therapeutic protein variants

As previously mentioned, although the development of improved versions of rhIFN-α2b with increased plasma half-life proved challenging, the results obtained were encouraging. Moreover, some of the rhIFN-α2b pegylated versions are commercially available on the market. However, the immunogenicity risk in the clinic still prevails [53]. As a result, this raises the need for reduced immunogenicity versions of the product. An interesting strategy is the socalled de-immunization for functional therapeutics (DeFT) [54]. The biotherapeutic de-immunization process involves several stages that will be discussed below. Finally, the de-immunization process of human rhIFN-α2b will be addressed, highlighting the challenges and achievements so far.

4.3.1 In silico analysis

The de-immunization process usually begins with the identification of potentially immunogenic protein regions. To accomplish this task, several in silico platforms are now available [55]. Usually, this identification involves testing the binding of potentially immunogenic regions of the molecule to the most relevant HLA class II alleles in the world population [56]. The results obtained from this analysis are summarized as an immunogenicity score that allows inferring the probability of binding of that peptide sequence to one or more HLA class II alleles. This information can be used to establish which HLA alleles/patients are most susceptible to developing an unwanted immune response against the therapeutic product.

Also, the computational study carries out an iterative process for the identification of changes or mutations that leads to greater disruption in peptide-HLA binding. The selection of which mutations are more appropriate should also be made taking into account the available protein structural information, to avoid significant disruptions in the molecule structure/function. This information can be obtained from a literature search, if available, or through protein molecular modeling.

4.3.2 In vitro studies

Although the results achieved through in silico analysis allow reducing the number and complexity of experimental assays, they are still predictions and consequently must be validated through in vitro studies.

For this purpose, peptides defined as potentially immunogenic are synthesized, including the “original” sequences, that is, those derived from the wild-type protein and the “modified” ones, which include the mutations defined from the in silico and structural study.

Then, the synthesized peptides must be tested using binding assays to specific HLA molecules. These assays allow estimating the binding affinity of the original and modified peptides to multiple HLA molecules and inferring which mutations actually produce a significant disruption in the antigenicity of the tested peptide. One of the most widely used assays consists of a competitive binding assay [57] that allows assaying various selected alleles in order to cover a wide repertoire of HLA pockets [56]. This experiment establishes competition between original and modified untagged peptides at different concentrations and a biotinylated standard peptide for binding to HLA class II molecules. The complexes formed are then captured on multi-well plates using immobilized anti-HLA class II antibodies. Assay development is performed by incubating the immuno-complexes with a streptavidin-enzyme conjugate and subsequent treatment with the enzyme substrate.

This in vitro binding assay provides valuable information that allows the identification of specific changes in the molecule that lead to a disruption of the interaction between the antigenic peptide and the HLA molecule. However, it is still an artificial assay that does not consider critical issues of antigenic protein processing. For example, peptides analyzed in binding assays are designed based on the clustering of highly immunogenic epitopes and do not take into account that such peptides are formed by the action of proteases (lysosomal cathepsins). As a result, the peptides actually formed inside the endosome and then loaded onto HLA molecules may differ from those designed on the basis of epitope content. Moreover, while binding assays provide information on the antigenicity of therapeutic protein-derived peptides, they do not establish whether those peptides will ultimately be recognized by the T cell receptor (TCR). Consequently, a peptide could be highly antigenic but not necessarily immunogenic. To overcome these limitations, ex vivo assays are now available and will be described below.

4.3.3 Ex vivo assays

Considering the relevance of achieving clinical trials with effective and safe biotherapeutics, it is necessary to rely on preclinical assays to anticipate the risks in the clinic. As a result, in the last decades, much effort has been devoted to the development of experimental platforms based on cell cultures (bioassays). These bioassays engage immune system mediators that could detect the product and consequently trigger an unwanted response in the patient.

One of the most commonly used assays involves the utilization of human peripheral blood mononuclear cells (PBMCs). PBMC samples typically engage monocytes, B cells, T cells, NK cells, and to a lesser extent dendritic cells. Therefore, these cell cultures include antigen-presenting cells (APCs) and T cells, which are the cellular mediators engaged in developing an immune response against immunogenic proteins. Wullner and collaborators [58] developed an experimental strategy based on the utilization of PBMC samples in which the therapeutic product is used to successively “challenge” the cell cultures. Initially, the APCs present in the PBMC sample internalize the product, process it at the lysosomal level, and the resulting peptides are presented in the context of HLA molecules. Thus, if the biotherapeutic contains T cell epitopes, repeated stimulation with the product will lead to activation, proliferation, and differentiation of specific naϊve T cells into a specific T helper (Th) profile. In particular, the most frequently observed effector Th profiles are Th1, Th2, and Th17, by ELISA and/or ELISPot assays.

Due to the HLA allelic diversity in the world population and the potential differences in the elicited immune response, this assay should be performed using PBMC samples from donors expressing different HLA alleles. In particular, it has been reported that HLA-DRB1 alleles are the most frequently involved in antigenic presentation and, therefore, required for T cell-mediated responses [59, 60].

Although this experimental strategy allows predicting with good accuracy the risk of immunogenicity of certain biologics in preclinical stages [27], it has a limitation associated with the direct effect that some biotherapeutics may have on T cells and, consequently, on the results derived from this assay. For example, IFN-α exerts a potent antiproliferative action on naϊve T cells. Therefore, this experimental approach cannot be used to analyze the immunogenicity of rhIFN-α2b-based products.

To circumvent this issue, an interesting approach derived from this bioassay includes an initial step based on the development of immature dendritic cells as antigen-presenting cells [60]. Immature dendritic cells can be obtained in vitro from the differentiation of monocytes isolated from PBMC samples. Differentiation can be achieved by culturing monocytes with GM-CSF and IL-4 for 6 days [61]. The generation of dendritic cells can then be confirmed by microscopic inspection and flow cytometry by analyzing CD11 and CD14 cell marker expression. Immature dendritic cells are cocultured with the biotherapeutic and then maturation of the Ag pulsed-DCs is induced with human tumor necrosis factor (TNF) or LPS [42]. During the maturation process, there is an increase in the expression of costimulatory molecules CD80 and CD86 required for T cell activation. Finally, mature dendritic cells presenting biologic-derived peptides are cocultured with autologous T cells and the specific cell activation is analyzed as mentioned above.

In vitro and ex vivo assays provide valuable information on biologic immunogenicity risks and allow validation of results derived from in silico predictions [27]. However, on occasion, it is also necessary to confirm these data through in vivo assays.

4.3.4 In vivo studies

Initially, toxicity and immunogenicity tests of biological products were carried out using “wild-type” animals (mainly mice). However, these animal models exhibit species-specific characteristics. In particular, the results achieved when studying the immune response developed by these animals may differ considerably from those in humans.

The advent of molecular biology and genetic engineering techniques has allowed the development of animals with particular characteristics of interest for this type of study. Currently, the most commonly used models for immunogenicity analysis are therapeutic protein-tolerant animals and transgenic animals expressing HLA molecules.

4.3.4.1 Mice immune tolerant to human proteins

Human protein-tolerant mice are engineered to express the therapeutic protein of interest. Hence, during cell ontogeny, tolerance processes allow the maintenance of immune homeostasis toward that specific protein.

These mouse strains are useful for evaluating the presence of neo-epitopes as well as subtle changes in product attributes such as formulation.

Several tolerant mouse strains are available. Among them are human tissue plasminogen activator (tPA)-tolerant murine models [62], mice producing human preproinsulin [63], and human IFN-α [64] and IFN-β [65] tolerant animals. Currently, other tolerant murine models that are of great utility for in vivo analysis of different biologic formulations include human growth hormone (GH)-producing mice [66] and human IgG-tolerant mice [67].

Although human protein-tolerant mouse strains have proven useful, they have some limitations. These mice express the murine antigenic processing and presentation system components and, consequently, the biologic-derived peptides that are exhibited by APCs and the immune response developed are still mouse-specific. Therefore, this may sometimes lead to results that do not necessarily correlate with those addressed during human clinical trials. In addition, this strategy raises the need to develop and validate a tolerant mouse strain for each therapeutic protein under study.

These issues could be addressed through the use of transgenic animals expressing HLA molecules, or combinations of these mice with tolerant animals (if available).

4.3.4.2 Mice expressing HLA molecules

In addition to mice tolerant to human therapeutic proteins, genetic engineering strategies have allowed the development of animals that have had the murine antigenic presentation system knocked out and express specific HLA molecules instead. In humans, numerous HLA genes have been identified and classified into three classes. HLA class I comprises the A, B, and C genes; HLA class II contains the DR, DQ, and DP genes; while HLA class III includes complement molecules. Class I and II molecules are involved in the selection processes of T cells in the thymus. Thus, HLA class I is responsible for the negative and positive selection of CD8 T cells, whereas HLA class II is involved in the selection processes of CD4 T cells [68]. Therefore, HLA transgenic mice exhibit an antigenic presentation system analogous to the human one (at least in those specific HLA molecules). Thus, peptides derived from antigenic proteins, including therapeutic proteins, will be identical to those presented by humans expressing those HLA molecules. In addition, due to the selection processes in the thymus mentioned above, these animals possess T cells endowed with TCRs capable of recognizing peptides presented in the context of these HLA molecules. As a result, these animals develop an immune response that mimics the human immune response. The close correlation of T cell-mediated responses observed in patients and in animals expressing HLA molecules represents clear evidence of the utility of this in vivo strategy [69].

These transgenic mice strains can be used for multiple purposes, including, in addition to the analysis of the immunogenicity of biological products, autoimmune diseases, vaccine development, certain infectious diseases, and oncology studies. In the immunogenicity analysis of therapeutic proteins, HLA-DR3 and HLA-DR4 mice have proven to be particularly useful [54, 70].

Despite the high correlation that in vivo studies involving transgenic animals may show, their execution and the number of animals required must be strongly justified. For this reason, before reaching in vivo assays, it is recommended to carry out all available in silico, in vitro, and ex vivo stages of the biologic immunogenicity analysis.

4.3.5 De-immunization of human rhIFN-α2b

To de-immunize rhIFN-α2b a multi-step strategy was approached [42]. First, the most immunogenic regions of the protein were identified by using the immuno-computational algorithms of EpiVax Inc. Then, mutations leading to a substantial disruption of IFN-derived peptide binding were selected from a group of eight HLA class II archetypes that combined exhibited a coverage of more than 90% of the allelic diversity of the world population. In particular, the DRB1 alleles analyzed in this study were *0101, *0301, *0401, *0701, *0801, *1101, *1301, and *1501.

The results were validated by in vitro binding assays following the experimental method described above and the same eight HLA alleles used for the in silico analysis. Most of the computational predictions were experimentally confirmed, which highlights the accuracy of the computational method used in this study. Then, with the knowledge of the most relevant changes to be introduced in the rhIFN-α2b sequence and an exhaustive search in the literature of the most relevant residues for protein structure/function, we developed two variants containing these mutations in different combinations. Both muteins evidenced a dramatic decrease in ex vivo immunogenicity assays using human PBMC samples [56]. Moreover, a recent study demonstrated that whereas rhIFN-α2b induced a robust neutralizing antibody response in HLA-DRB1 transgenic animals, both rhIFN-α2b variants induced a markedly lower response. In addition, this study also revealed that these biobetters showed improved stability against several physical factors such as heat stress, repeated freeze/thaw cycles, and low pH [70].

However, both muteins exhibited only a residual in vitro antiviral biological activity of 28.4 and 16.9%. These results indicate that despite selecting mutations while avoiding introducing changes in critical residues of the molecule, a reduction of in vitro antiviral activity was evident [56]. To address this issue, five of the mutations identified as key in reducing rhIFN-α2b immunogenicity were introduced into a hyperglycosylated variant of the cytokine that exhibits improved plasma stability with respect to the unmodified protein. Thus, the new rhIFN-α2b variant retained not only the improved pharmacokinetic properties of the original molecule but also reduced immunogenicity, and a high residual antiviral activity (72%) [43].

Advertisement

5. Conclusion

For more than 30 years, rhIFN-α2b has been used for the treatment of chronic and emerging viral infections. However, rhIFN-α2b is potentially immunogenic, which may compromise the efficacy and safety of rhIFN-α2b-based therapy. In addition, rhIFN-α2b is a low molecular size cytokine, which correlates with its reduced plasma half-life in patients.

For these reasons, several strategies have been addressed to reduce the impact of protein immunogenicity as well as increase the product’s plasma stability.

Glycoengineering and protein pegylation have been proposed as strategies aimed at increasing the apparent macromolecule size and, consequently, reducing the rhIFN-α2b clearance rate. The results obtained so far have been very encouraging, with remarkable pharmacokinetic parameters for these biobetters. In addition, both approaches would allow the addition of glycans or large PEG residues that would generate a shielding effect of protein epitopes. However, neither of these strategies has allowed the development of products that significantly ameliorate the negative impact associated with product immunogenicity.

In contrast, de-immunization for functional therapeutics (DeFT) of rhIFN-α2b has emerged as a promising alternative for markedly reducing product immunogenicity. The success of this strategy is based on the careful selection of the target residues, which will rely on the impact of such a change on the antigenicity/immunogenicity as well as on protein structure/function.

The individual or combined use of these strategies will enable the development of more effective and safer rhIFN-α2b-based antiviral therapies.

Advertisement

Acknowledgments

This work was supported by the Consejo Nacional de Investigaciones Científícas y Técnicas (CONICET, Argentina); Agencia Nacional de Promoción Científica y Tecnológica; and Universidad Nacional del Litoral. EFM and ME are members of CONICET; LCP is a fellow of the same institution.

References

  1. 1. Isaacs A, Lindenmann J. Virus interference. I. the interferon. Proceedings of the Royal Society of London. 1957;147:258-267. DOI: 10.1098/rspb.1957.0048
  2. 2. Nagano Y, Kojima Y. Inhibition de I’infection vaccinale par le virus homologue. Comptes Rendus des Seances de la Societe de Biologie et de Ses Filiales. 1958;152:1627-1630
  3. 3. Gugliotta A, Ceaglio N, Raud B, Forno G, Mauro L, Kratje R, et al. Glycosylation and antiproliferative activity of hyperglycosylated IFN-α2 potentiate HEK293 cells as biofactories. European Journal of Pharmaceutics and Biopharmaceutics. 2017;112:119-131. DOI: 10.1016/j.ejpb.2016.11.012
  4. 4. Raniga K, Liang C. Interferons: Reprogramming the metabolic network against viral infection. Viruses. 2018;10:1-21. DOI: 10.3390/v10010036
  5. 5. Burdick LM, Somani N, Somani AK. Type I IFNs and their role in the development of autoimmune diseases. Expert Opinion on Drug Safety. 2009;8:459-472. DOI: 10.1517/14740330903066726
  6. 6. Rönnblom LE, Janson ET, Perers A, Oberg KE, Alm GV. Characterization of antiinterferon-alpha antibodies appearing during recombinant interferon-alpha 2atreatment. Clinical & Experimental Immunology. 1992;89:330-335. DOI: 10.1111/j.1365-2249.1992.tb06957.x
  7. 7. Quesada JR, Rios A, Swanson D, Trown P, Gutterman JU. Antitumor activity of recombinant-derived interferon alpha in metastatic renal cell carcinoma. Journal of Clinical Oncology. 1985;3:1522-1528. DOI: 10.1200/JCO.1985.3.11.1522
  8. 8. Itri LM, Sherman MI, Palleroni AV, Evans LM, Tran LL, Campion M, et al. Incidence and clinical significance of neutralizing antibodies in patients receiving recombinant interferon-alpha 2a. Journal of Interferon & Cytokine Research. 1989;9:9-15
  9. 9. Wussow PV, Jakschies D, Freund M, Hehlmann R, Brockhaus F, Hochkeppel H, et al. Treatment of anti-recombinant interferon-alpha 2 antibodypositive CML patients with natural interferon-alpha. British Journal of Haematology. 1991;78:210-216. DOI: 10.1111/j.1365-2141.1991.tb04418.x
  10. 10. Chen K, Liu C. Regulation of type I interferon signaling in immunity and inflammation: A comprehensive review. Journal of Autoimmunity. 2017;83:1-11. DOI: 10.1016/j.jaut.2017.03.008
  11. 11. Goodbourn S, Didcock L, Randall RE. Interferons: Cell signalling, immune modulation, antiviral responses and virus countermeasures. Journal of General Virology. 2000;81:2341-2364. DOI: 10.1099/0022-1317-81-10-2341
  12. 12. Samuel CE. Antiviral actions of interferons. Clinical Microbiology Reviews. 2001;14:778-809. DOI: 10.1128/CMR.14.4.778-809.2001
  13. 13. McNab F, Mayer-Barber K, Sher A, Wack A, O’Garra A. Type I interferons in infectious disease. Nature Reviews Immunology. 2015;15:87-103. DOI: 10.1038/nri3787
  14. 14. Wang H, Hu H, Zhang K. Overview of interferon: Characteristics, signaling and anti-cancer effect. Archives of Biotechnology and Biomedicine. 2017;1:1-16. DOI: 10.29328/journal.hjb.1001001
  15. 15. Zhang KR, Kumar R. Interferon-α inhibits cyclin E- and cyclin D1-dependent CDK-2 kinase activity associated with RB protein and E2F in Daudi cells. Biochemical and Biophysical Research Communications. 1994;200:522-528. DOI: 10.1006/bbrc.1994.1479
  16. 16. Pestka S, Krause CD, Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunological Reviews. 2004;202:8-32. DOI: 10.1111/j.0105-2896.2004.00204.x
  17. 17. Deisenhammer F. Neutralizing antibodies to interferon-beta and other immunological treatments for multiple sclerosis: Prevalence and impact on outcomes. CNS Drugs. 2009;23:379-396. DOI: 10.2165/00023210-200923050-00003
  18. 18. Cohen BA, Oger J, Gagnon A, Giovannoni G. The implications of immunogenicity for protein-based multiple sclerosis therapies. Journal of the Neurological Sciences. 2008;275:7-17. DOI: 10.1016/j.jns.2008.08.003
  19. 19. van Schouwenburg PA, van de Stadt LA, de Jong RN, van Buren EE, Kruithof S, de Groot E. Adalimumab elicits a restricted anti-idiotypic antibody response in autoimmune patients resulting in functional neutralization. Annals of Rheumatic Diseases. 2013;72:104-109. DOI: 10.1136/annrheumdis-2012-201445
  20. 20. von Wussow P, Jakschies D, Freund M, Deicher H. Humoral response to recombinant interferon-alpha 2b in patients receiving recombinant interferon-alpha 2b therapy. Journal of Interferon & Cytokine Research. 1989;9:25-31
  21. 21. Vial T, Descotes J. Clinical toxicity of the interferons. Drug Safety. 1994;10:115-150. DOI: 10.2165/00002018-199410020-00003
  22. 22. De Somer P, Edy VG, Billiau A. Interferon-induced skin reactivity in man. Lancet. 1997;310:47-49. DOI: 10.1016/s0140-6736(77)90055-1
  23. 23. Oppenheim Y, Ban Y, Tomer Y. Interferon induced autoimmune thyroid disease (AITD): A model for human autoimmunity. Autoimmunity Reviews. 2004;3:388-393. DOI: 10.1016/j.autrev.2004.03.003
  24. 24. Rönnblom LE, Alm GV, Oberg KE. Possible induction of systemic lupus erythematosus by interferon-alpha treatment in a patient with a malignant carcinoid tumour. Journal of Internal Medicine. 1990;227:207-210. DOI: 10.1111/j.1365-2796.1990.tb00144.x
  25. 25. Clark EA, Ledbetter JA. How B and T cells talk to each other. Nature. 1994;367:425-428. DOI: 10.1038/367425a0
  26. 26. Klaus GG, Choi MS, Lam EW, Johnson-Léger C, Cliff J. CD40: A pivotal receptor in the determination of life/death decisions in B cells. International Reviews of Immunology. 1997;15:5-31. DOI: 10.3109/08830189709068169
  27. 27. Koren E, De Groot AS, Jawa V, Beck KD, Boone T, Rivera D, et al. Clinical validation of the in silico prediction of immunogenicity of a human recombinant therapeutic protein. Clinical immunology. 2007;124:26-32. DOI: 10.1016/j.clim.2007.03.544
  28. 28. Reveille JD. The genetic basis of autoantibody production. Autoimmunity Reviews. 2006;5:389-398. DOI: 10.1016/j.autrev.2005.10.012
  29. 29. Jones TD, Hanlon M, Smith BJ, Heise CT, Nayee PD, Sanders DA, et al. The development of a modified human IFN-alpha2b linked to the fc portion of human IgG1 as a novel potential therapeutic for the treatment of hepatitis C virus infection. Journal of Interferon & Cytokine Research. 2004;24:560-572. DOI: 10.1089/jir.2004.24.560
  30. 30. Spiro RG. Protein glycosylation: Nature, distribution, enzymatic formation, and disease implications of glycopeptide bonds. Glycobiology. 2002;12:43-56. DOI: 10.1093/glycob/12.4.43R
  31. 31. Sinclair AM, Elliott S. Glycoengineering: The effect of glycosylation on the properties of therapeutic proteins. Journal of Pharmaceutical Sciences. 2005;94:1626-1635. DOI: 10.1002/jps.20319
  32. 32. Beck A, Reichert JM. Marketing approval of mogamulizumab: A triumph for glyco-engineering. MAbs. 2012;4:419-425. DOI: 10.4161/mabs.20996
  33. 33. Dicker M, Strasser R. Using glyco-engineering to produce therapeutic proteins. Expert Opinion on Biological Therapy. 2015;15:1501-1516. DOI: 10.1517/14712598.2015.1069271
  34. 34. Varki A. Biological roles of glycans. Glycobiology. 2017;27:3-49. DOI: 10.1093/glycob/cww086
  35. 35. Chatelut E, Rostaing L, Grégoire N, Payen J, Pujol A, Izopet J, et al. A pharmacokinetic model for alpha interferon administered subcutaneously. British Journal of Clinical Pharmacology. 1999;47:365-371. DOI: 10.1046/j.1365-2125.1999.00912.x
  36. 36. Elliot S, Lorenzini T, Asher S, Aoki K, Brankow D, Buck L, et al. Enhancement of therapeutic protein in vivo activities through glycoengineering. Nature Biotechnology. 2003;21:414-421. DOI: 10.1038/nbt799
  37. 37. Gooche CF, Gramer MJ, Andersen DC, Bahr FB, Rasmussen JR. The oligosaccharides of glycoproteins: Factors affecting their synthesis and their influence on glycoprotein properties. Biotechnology. 1992;9:1347-1355. DOI: 10.1038/nbt1291-1347
  38. 38. Morell AG, Gregoriadis G, Scheinberg H. The role of sialic acid in determining the survival of glycoproteins in the circulation. Journal of Biological Chemistry. 1971;246:1461-1467
  39. 39. Koury MJ. Sugar coating extends half-life and improves effectiveness of cytokine hormones. Trends in Biotechnology. 2003;21:462-464. DOI: 10.1016/j.tibtech.2003.09.002
  40. 40. Ceaglio N, Etcheverrigaray M, Kratje R, Oggero M. Novel long-lasting interferon alpha derivatives designed by glycoengineering. Biochimie. 2008;90:437-449. DOI: 10.1016/j.biochi.2007.10.013
  41. 41. Sales MM, Kratje R, Oggero M, Ceaglio N. Bifunctional GM-CSF-derived peptides as tools for O-glycoengineering and protein tagging. Journal of Biotechnology. 2021;327:18-27. DOI: 10.1016/j.jbiotec.2020.12.016
  42. 42. Mufarrege EF, Giorgetti SI, Etcheverrigaray M, Terry F, Martin W, De Groot AS. De-immunized and functional therapeutic (DeFT) versions of a long lasting recombinant alpha interferon for antiviral therapy. Clinical Immunology. 2017;176:31-41. DOI: 10.1016/j.clim.2017.01.003
  43. 43. Giorgetti SI, Etcheverrigaray M, Terry F, Martin W, De Groot AS, Ceaglio N, et al. Development of highly stable and de-immunized versions of recombinant alpha interferon: Promising candidates for the treatment of chronic and emerging viral diseases. Clinical Immunology. 2021;233:108888. DOI: 10.1016/j.clim.2021.108888
  44. 44. Wang YS, Youngster S, Bausch J, Zhang R, McNemar C, Wyss DF. Identification of the major positional isomer of PEGylated interferon alpha-2b. Biochemistry. 2004;39:10634-10640. DOI: 10.1021/bi000617t
  45. 45. Gilbert CW, Park-Cho M. Interferon Polymer Conjugates. U.S. Patent 5,951,974. Alexandria, VA, USA: United States Patent and Trademark Office; 1999
  46. 46. Pedder SCJ. Pegylation of interferon alpha: Structural and pharmacokinetic properties. Seminars in Liver Disease. 2003;23:19-22. DOI: 10.1055/s-2003-41635
  47. 47. Grace MJ, Lee S, Bradshaw S, Chapman J, Spond J, Cox S, et al. Ite of PEGylation and polyethylene glycol molecule size attenuate interferon-R antiviral and antiproliferative activities through the JAK/STAT signaling pathway. Journal of Biological Chemistry. 2005;280:6327-6336. DOI: 10.1074/jbc.M412134200
  48. 48. Dhalluin C, Ross A, Leuthold L, Foser S, Gsell B, Muller F, et al. Structural and biophysical characterization of the 40 kDa PEG-interferon-alpha 2a and its individual positional isomers. Bioconjugate Chemistry. 2005;16:504-517. DOI: 10.1021/bc049781+
  49. 49. Rosendahl MS, Doherty DH, Smith DJ, Carlson SJ, Chlipala EA, Cox GN. A long-acting, highly potent interferon R-2 conjugate created using site-specific PEGylation. Bioconjugate Chemistry. 2005;16:200-207. DOI: 10.1021/bc049713n
  50. 50. Harris JM, Martin NE, Modi M. Pegylation: A novel process for modifying pharmacokinetics. Clinical Pharmacokinetics. 2001;40:539-551. DOI: 10.2165/00003088-200140070-00005
  51. 51. Karnam US. Pegylated interferons. Clinical Liver Disease. 2003;7:139-148. DOI: 10.1016/s1089-3261(02)00072-7
  52. 52. Luxon BA, Grace M, Brassard D, Bordens R. Pegylated interferons for the treatment of chronic hepatitis C infection. Clinical Therapeutics. 2002;24:1363-1383. DOI: 10.1016/s0149-2918(02)80042-x
  53. 53. Jorns C, Holzinger D, Thimme R, Spangenberg HC, Weidmann M, Rasenack J, et al. Rapid and simple detection of IFN-neutralizing antibodies in chronic hepatitis C non-responsive to IFN-alpha. Journal of Medical Virology. 2006;78(1):74-82. DOI: 10.1002/jmv.20506
  54. 54. Moise L, Song C, Martin WD, Tassone R, De Groot AS, Scott DW. Effect of HLA DR epitope de-immunization of factor VIII in vitro and in vivo. Clinical immunology. 2012;142:320-331. DOI: 10.1016/j.clim.2011.11.010
  55. 55. De Groot AS, Martin W. Reducing risk, improving outcomes: Bioengineering less immunogenic protein therapeutics. Clinical Immunology. 2009;131:189-201. DOI: 10.1016/j.clim.2009.01.009
  56. 56. Sidney SJ, Kondo A, Guercio MF, Appella E, Hoffman S, Kubo RT, et al. Several common HLA-DR types share largely overlapping peptide binding repertoires. Journal of immunology. 1998;160:3363-3373
  57. 57. Steere AC, Klitz W, Drouin EE, Falk BA, Kwok WW, Nepom GT, et al. Antibiotic-refractory Lyme arthritis is associated with HLA-DR molecules that bind a Borrelia burgdorferi peptide. Journal of Experimental Medicine. 2006;203:961-971. DOI: 10.1084/jem.20052471
  58. 58. Wullner D, Zhou L, Bramhall E, Kuck A, Goletz TJ, Swanson S, et al. Considerations for optimization and validation of an in vitro PBMC derived T cell assay for immunogenicity prediction of biotherapeutics. Clinical Immunology. 2010;137(1):5-14. DOI: 10.1016/j.clim.2010.06.018
  59. 59. Mazor R, Vassall AN, Eberle JA, Beers R, Weldon JE, Venzon DJ, et al. Identification and elimination of an immunodominant T-cell epitope in recombinant immunotoxins based on pseudomonas exotoxin a. Proceedings of the National Academy of Sciences. 2012;109:3597-3603. DOI: 10.1073/pnas.1218138109
  60. 60. Jaber A, Baker M. Assessment of the immunogenicity of different interferon beta-1a formulations using ex vivo T-cell assays. Journal of Pharmaceutical and Biomedical Analysis. 2007;43:1256-1261. DOI: 10.1016/j.jpba.2006.10.023
  61. 61. Elkord E, Williams PE, Kynaston H, Rowbottom AW. Human monocyte isolation methods influence cytokine production from in vitro generated dendritic cells. Immunology. 2005;114:204-212. DOI: 10.1111/j.1365-2567.2004.02076.x
  62. 62. Stewart TA, Hollingshead PG, Pitts SL, Chang R, Martin LE, Oakley H. Transgenic mice as a model to test the immunogenicity of proteins altered by site-specific mutagenesis. Molecular Biology in Medicine. 1989;6:275-281
  63. 63. Whiteley PJ, Lake JP, Selden RF, Kapp JA. Tolerance induced by physiological levels of secreted proteins in transgenic mice expressing human insulin. Journal of Clinical Investigation. 1989;84:1550-1554. DOI: 10.1172/JCI114331
  64. 64. Braun A, Kwee L, Labow M, Alsenz J. Protein aggregates seem to play a key role among the parameters influencing the antigenicity of interferon alpha in normal and trangenic mice. Pharmaceutical Research. 1997;14:1472-1478. DOI: 10.1023/a:1012193326789
  65. 65. Hermeling S, Jiskoot W, Crommelin D, Bornæs C, Schellekens H. Development of a transgenic mouse model immune tolerant for human interferon beta. Pharmaceutical Research. 2005;22:847-851. DOI: 10.1007/s11095-005-4578-z
  66. 66. Lee HJ, Riley G, Johnson O. In vivo characterization of sustained-release formulations of human growth hormone. Journal of Pharmacology and Experimental Therapeutics. 1997;281:1431-1439
  67. 67. Filipe V, Jiskoot W, Hafid Basmeleh A, Halim A, Schellekens H, Brinks V. mAbs immunogenicity of different stressed IgG monoclonal antibody formulations in immune tolerant transgenic mice. MAbs. 2012;4(6):740-752. DOI: 10.4161/mabs.22066
  68. 68. Mangalam AK, Rajagopalan G, Taneja V, David CS. HLA Class II Transgenic mice mimic human inflammatory diseases. Advances in Immunology. 2008;97, 65-147. DOI: 10.1016/S0065-2776(08)00002-3
  69. 69. Taneja V. Cytokines pre-determined by genetic factors are involved in pathogenesis of rheumatoid arthritis. Cytokine. 2015;75(221):216-221. DOI: 10.1016/j.cyto.2014.11.028
  70. 70. Mufarrege EF, Peña LC, Etcheverrigaray M, De Groot AS, Martin W. Specific sequence mutations in a long-lasting rhIFN-α2b version reduce in vitro and in vivo immunogenicity and increase in vitro protein stability. Heliyon. 2023;9(3):e14670. DOI: 10.1016/j.heliyon.2023.e14670

Written By

Eduardo F. Mufarrege, Lucía C. Peña and Marina Etcheverrigaray

Submitted: 19 May 2023 Reviewed: 29 May 2023 Published: 06 December 2023