IFNA2 is a subtype of type I interferons (IFNs) critical for antiviral defense, immune modulation, and antitumor activity . It is secreted by virus-infected cells and signals through the ubiquitously expressed IFNAR1/IFNAR2 receptor complex . The mature protein comprises 165 amino acids and is encoded by the IFNA2 gene on chromosome 9 .
IFNA2 activates the JAK-STAT pathway via IFNAR1/IFNAR2 binding, inducing >300 interferon-stimulated genes (ISGs) . Key steps include:
Receptor Binding: High-affinity interaction with IFNAR2 and low-affinity interaction with IFNAR1 .
Signal Transduction: Phosphorylation of STAT1/STAT2 and formation of ISGF3 complex .
Biological Effects: Antiviral defense, immunomodulation, and inhibition of tumor cell proliferation .
IFNA2 has been used therapeutically but is increasingly replaced due to adverse effects :
Condition | Efficacy | Adverse Effects |
---|---|---|
Chronic Hepatitis C | Sustained virologic response (~30%) | Flu-like symptoms, depression, cytopenia |
Melanoma | Improved survival in adjuvant therapy | Fatigue, hepatotoxicity |
Hairy Cell Leukemia | High remission rates | Neurotoxicity, autoimmune reactions |
Feature | IFNα2a (Roferon-A) | IFNα2b (Intron-A) |
---|---|---|
Amino Acid 23 | Lysine | Arginine |
Pharmacokinetics | Shorter half-life | PEGylated forms available |
Clinical Use | HCV, cancer | Melanoma, hepatitis |
IFNA2 is produced in E. coli for clinical use :
Macrophages produce IFN-alpha, which possesses antiviral properties. This interferon stimulates the production of two key enzymes: protein kinase and oligoadenylate synthetase.
Recombinant Human Interferon-alpha 2c, produced in yeast, is a single, glycosylated polypeptide chain consisting of 165 amino acids with a molecular weight of 19.3kDa. The purification of IFNA2C is achieved through proprietary chromatographic techniques.
Sterile Filtered White lyophilized (freeze-dried) powder.
The product is lyophilized from a 0.2µm filtered solution concentrated in PBS with a pH of 7.4 and containing 0.02% Tween-20.
For reconstitution of the lyophilized Interferon-alpha 2c, it is recommended to use sterile 18 MΩ-cm H2O to achieve a concentration of at least 100µg/ml. This solution can then be further diluted into other aqueous solutions as needed.
While Lyophilized IFNA2C remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C for optimal long-term preservation. Once reconstituted, Interferon-alpha 2c should be stored at 4°C for a period of 2-7 days. For extended storage, it should be kept below -18°C. It is crucial to avoid repeated freeze-thaw cycles.
The purity is determined to be greater than 97.0% using the following methods:
(a) Analysis by RP-HPLC.
(b) Analysis by SDS-PAGE.
Activity is determined by measuring the inhibition of the cytopathic effect using a standardized assay.
Interferon alpha-2, IFN-alpha-2, Interferon alpha-A, LeIF A, IFNA2, IFNA2A, IFNA2B, IFNA2C.
Saccharomyces cerevisiae.
CDLPQTHSLG SRRTLMLLAQ MRRISLFSCL KDRRDFGFPQ EEFGNQFQKA ETIPVLHEMI QQIFNLFSTK DSSAAWDETL LDKFYTELYQ QLNDLEACVI QGVGVTETPL MKEDSILAVR KYFQRITLYL KEKKYSPCAW EVVRAEIMRS FSLSTNLQES LRSKE.
Human interferon alpha 2 (IFNA2) is a prototypic member of the type I interferon family and was the first highly active IFN subtype to be cloned in the early 1980s. It consists of 165 amino acids in its mature form, which is one amino acid shorter than all other human IFNα subtypes due to a missing aspartic acid at position 44. IFNA2 is non-glycosylated, making it particularly suitable for recombinant production in bacterial expression systems .
The protein's primary structure features several key domains that mediate receptor binding and biological activity. Unlike most other cytokines, IFNA2 has maintained remarkable conservation across human populations, suggesting essential physiological roles that prevent functional mutations .
Humans possess 17 type I interferon subtypes, including 13 alpha variants, 1 beta, 1 omega, 1 epsilon, and 1 kappa. While all type I interferons bind to the same receptor complex comprising IFNAR1 and IFNAR2, they exhibit different binding affinities and biological potencies .
IFNA2 shows moderate binding affinity to the receptor components compared to other subtypes. Specifically, IFNβ demonstrates higher binding affinity and biological activity than most IFNα subtypes including IFNA2. Within the alpha interferon family, most subtypes (except IFNα1, which binds weakly) share similar affinity ranges and activity profiles .
The differential activities of type I interferons are determined by the stability of the IFN-receptor ternary complex, which depends on individual affinities to IFNAR1 and IFNAR2. This relationship between affinity and activity varies for different biological responses, explaining why different IFN subtypes exhibit varied potencies across different biological functions .
Several IFNA2 alleles have been identified, with IFNA2a and IFNA2b being the most well-documented variants. These alleles differ by a single amino acid substitution at position 23 (K/R), which is considered functionally neutral. IFNA2a and IFNA2b have been commercialized as pharmaceutical products (RoferonA and IntronA, respectively) for clinical applications .
Population genetics studies have revealed that the IFNA2 gene is under evolutionary constraints that prevent mutations, suggesting its essential role in human physiology. This conservation contrasts with some other cytokines that exhibit greater polymorphic variation, highlighting IFNA2's fundamental importance in immune defense mechanisms .
When investigating IFNA2 signaling pathways, researchers should employ multiple complementary approaches:
Receptor-ligand binding studies: Surface plasmon resonance (SPR) technology can quantify binding kinetics between IFNA2 and its receptors IFNAR1 and IFNAR2. This approach allows determination of association/dissociation constants that correlate with biological activities .
Signaling cascade analysis: Phospho-specific antibodies targeting JAK-STAT pathway components can track activation kinetics following IFNA2 treatment. Western blotting, phospho-flow cytometry, and high-content imaging provide temporal and spatial resolution of signaling events.
Transcriptional profiling: RNA-seq or microarray analysis enables comprehensive assessment of IFNA2-induced gene expression. This approach has been successfully employed to compare IFNA2 effects with other interferon subtypes and in different cellular contexts .
CRISPR-based genetic screens: Genome-wide or targeted CRISPR knockout/knockdown approaches can identify essential components of the IFNA2 signaling pathway and functional mediators of specific biological responses.
Systems biology approaches: Integration of protein-protein interaction data, phosphoproteomics, and transcriptomics can elucidate the complex network of molecular interactions driving IFNA2 responses.
Distinguishing IFNA2-specific effects from those of other type I interferons requires several strategic approaches:
Comparative dose-response studies: Carefully designed dose-response experiments with multiple interferon subtypes can reveal differences in potency and efficacy across various biological readouts. The slope of the dose-response curve often differs between interferons for different biological activities .
Receptor mutant systems: Cell lines expressing modified IFNAR1 or IFNAR2 with altered binding affinities for specific interferon subtypes can help delineate receptor requirements for different biological responses.
Neutralizing antibodies: Subtype-specific neutralizing antibodies can be employed to selectively block individual interferons in complex biological systems.
Chimeric interferons: Recombinant chimeric proteins containing domains from different interferon subtypes can help map structure-function relationships and identify regions responsible for subtype-specific activities.
Comparative transcriptomics: Analysis of gene expression signatures induced by different interferons can identify IFNA2-specific transcriptional programs. This approach revealed differential effects between bovine IFNT and human IFNA2 in a bovine endometrium model .
Recent research has established critical roles for IFNA2 and other type I interferons in cancer immunosurveillance:
Tumor rejection mechanisms: Studies in the last decade have demonstrated that type I interferons, including IFNA2, are essential in processes of immunosurveillance, tumor rejection, and regulation of metastasis spread .
Combination therapy rationales: IFNA2 has been shown to improve the efficacy of traditional anticancer treatments including radiotherapy, chemotherapy, and monoclonal antibody-based therapies. This synergistic effect provides mechanistic rationale for combination approaches .
Chronic myelogenous leukemia (CML) treatment: After being temporarily supplanted by tyrosine kinase inhibitors (TKIs), IFNA2 has re-emerged as a valuable therapeutic agent in CML. Clinical data suggest that IFNA2 pre-treated patients experienced delayed relapses after TKI discontinuation, and combination therapy with IFNA2 and TKIs significantly improves major molecular response rates .
Mechanisms of IFNA2 action in CML: Several proposed mechanisms explain IFNA2's benefit in CML treatment, including effects on leukemic stem cells that are not effectively targeted by TKIs alone. These findings have sparked renewed clinical investigation of combination therapies .
Researchers seeking to quantify IFNA2 activity should consider multiple complementary methods:
Antiviral protection assays: Classical bioassays measuring protection against viral cytopathic effects (e.g., vesicular stomatitis virus in WISH cells) remain valuable for functional quantification of IFNA2 activity .
Antiproliferative assays: Cell lines with high sensitivity to the antiproliferative effects of IFNA2 (e.g., Daudi, WISH) provide reliable readouts for biological activity assessment .
ELISA-based detection: Specific ELISA systems like the DuoSet ELISA allow precise quantification of IFNA2 protein levels in various biological samples .
Reporter cell systems: Engineered cell lines expressing luciferase or fluorescent proteins under the control of interferon-stimulated response elements (ISREs) enable rapid, sensitive detection of IFNA2 activity.
Phospho-STAT detection: Quantification of STAT1/STAT2 phosphorylation by flow cytometry or ELISA provides a proximal measure of IFNA2 receptor engagement and signaling.
The choice of assay should be guided by the specific research question, with consideration of sensitivity requirements, specificity needs, and the biological context being studied.
The divergent biological outcomes of IFNA2 signaling in antiviral versus antiproliferative responses reflect distinct downstream pathways:
Receptor engagement dynamics: The stability of the ternary complex formed by IFNA2 with IFNAR1 and IFNAR2 differentially affects antiviral versus antiproliferative activities. Studies have demonstrated that the slope of the linear affinity-activity correlation differs between these biological responses. For example, in WISH cells, the slope of the antiproliferative specific activity versus affinity is higher than the slope establishing the relationship between anti-VSV (antiviral) specific activity and the global affinity .
Gene expression programs: Antiviral responses primarily depend on rapid induction of genes encoding direct antiviral effectors (e.g., MX1, OAS1, PKR), while antiproliferative effects require sustained expression of cell cycle inhibitors and pro-apoptotic factors.
Temporal dynamics: Antiviral states can be established rapidly after brief IFNA2 exposure, whereas antiproliferative effects typically require prolonged receptor engagement and signaling.
Cell type specificity: Different cell types exhibit variable sensitivities to the antiviral versus antiproliferative effects of IFNA2, likely reflecting differences in the expression of key signaling components and effector molecules.
These mechanistic differences explain why certain IFNA2 variants or analogs might preferentially elicit one biological response over another, which has important implications for therapeutic applications.
IFNA2 has a rich history of clinical applications that has evolved significantly over decades:
Historical development: IFNA2 was the first IFN and the first cytokine to be produced and commercialized by the pharmaceutical industry. It was first approved for treating hairy cell leukemia in 1986 .
Approved indications: Since its initial approval, recombinant IFNA2 has been approved for numerous conditions including chronic viral hepatitis B (HBV), chronic viral hepatitis C (HCV), chronic myeloid leukemia (CML), Kaposi sarcoma, follicular lymphoma, renal cell carcinoma (RCC), melanoma, T cell lymphoma, multiple myeloma, and condylomata acuminata .
Evolution in HCV treatment: IFNA2 was the first available therapy for chronic HCV. Its efficacy was enhanced by combination with ribavirin, and later by the development of PEG-IFNA2, which showed improved efficacy. PEG-IFNA2/ribavirin became the standard of care until the development of directly acting antiviral agents (DAAs) .
Treatment displacement and repositioning: In many indications, IFNA2 has been replaced by more targeted therapies. For HCV, DAAs have largely replaced interferon-based regimens. Similarly, targeted therapies have supplanted IFNA2 in metastatic RCC (VEGF and mTOR inhibitors), melanoma (BRAF inhibitors), and multiple myeloma (thalidomide, lenalidomide, bortezomib) .
Resurgence in CML: Interestingly, IFNA2 has experienced a renaissance in CML treatment. Initially replaced by tyrosine kinase inhibitors (TKIs), IFNA2 has returned to clinical investigation after observations that IFNA2 pre-treated patients had delayed relapses after TKI discontinuation. Combination therapy with IFNA2 and TKIs shows promising improvements in molecular response rates .
Researchers have developed several strategies to mitigate IFNA2's toxicity while preserving its therapeutic efficacy:
PEGylation technology: The attachment of polyethylene glycol (PEG) molecules to IFNA2 created PEG-IFNA2, which demonstrates improved pharmacokinetics and reduced immunogenicity compared to unmodified IFNA2. This modification allows for less frequent dosing and has shown improved efficacy in conditions like HCV .
Targeted delivery systems: Various nanoparticle formulations, antibody-IFNA2 conjugates, and cell-specific targeting approaches aim to concentrate IFNA2 activity in desired tissues while reducing systemic exposure and adverse effects.
Combination therapy approaches: Lower doses of IFNA2 used in combination with other therapeutic agents (e.g., TKIs in CML) can maintain efficacy while reducing side effects. These approaches leverage mechanistic synergies between IFNA2 and companion therapeutics .
Modified dosing schedules: Alternative dosing regimens, including lower dose, higher frequency schedules or intermittent dosing protocols, can sometimes maintain efficacy while improving tolerability.
Engineered IFNA2 variants: Structure-guided protein engineering has produced IFNA2 variants with altered receptor binding properties that maintain desired therapeutic activities while reducing toxicity-associated signaling.
The major limitation of IFNA2 use in conditions like CML remains its adverse effects, which restrict its full clinical benefit . Ongoing research continues to refine these approaches to optimize the therapeutic index of IFNA2-based treatments.
Accurate quantification of IFNA2 levels and activity requires selecting appropriate methodologies based on the specific research context:
ELISA-based detection systems: Commercial DuoSet ELISA kits provide sensitive and specific detection of human IFNA2 protein in research samples. These systems typically employ optimized capture and detection antibody pairings with standardized protocols .
Bioactivity assays: Functional assays measuring antiviral protection or antiproliferative effects in indicator cell lines remain the gold standard for assessing biological activity. These assays often demonstrate higher sensitivity than immunological detection methods and provide information about functional capacity rather than mere protein presence.
Reporter gene assays: Engineered cell lines expressing reporter constructs (luciferase, GFP) under control of interferon-stimulated response elements (ISREs) offer rapid, sensitive detection of IFNA2 activity in biological samples.
RT-qPCR for interferon-stimulated genes (ISGs): Measuring expression of known ISGs (e.g., MX1, OAS1, IFIT1) in cells exposed to test samples provides an indirect but sensitive measure of IFNA2 bioactivity.
Receptor occupancy assays: Flow cytometry-based techniques can assess IFNAR1/2 receptor occupancy and downregulation, providing insights into local IFNA2 activity.
For clinical samples, researchers should consider potential confounding factors such as the presence of autoantibodies against interferons, interferon-binding proteins, or soluble receptor fragments that may interfere with detection or activity measurements.
Despite decades of research, several fundamental questions about IFNA2 remain unanswered:
Physiological specificity: Stricto sensu, nothing is known about functions unique to IFNA2 in human physiology. It remains unclear whether IFNA2 has specific roles distinct from other IFNα subtypes, as there is no known context where IFNA2 is specifically produced as an isolated subtype .
Evolutionary conservation: The conservation of IFNA2 despite the presence of multiple other IFNα subtypes suggests unique or specialized functions that have not been fully elucidated.
Cellular sources: The specific cellular sources of IFNA2 in various physiological and pathological contexts, and how these might differ from sources of other interferons, remain incompletely characterized.
Tissue-specific effects: Whether IFNA2 has distinct tissue-specific activities compared to other type I interferons, and the molecular basis for any such differences, requires further investigation.
Role in pregnancy and reproduction: Studies comparing bovine IFNT with human IFNA2 suggest potential roles in reproductive physiology that warrant further exploration in human contexts .
Immunoregulatory functions: Beyond direct antiviral effects, the role of IFNA2 in shaping adaptive immune responses and immunological memory remains an active area of investigation.
These knowledge gaps represent important opportunities for future research that may uncover new therapeutic applications or improve existing treatment strategies involving IFNA2.
Emerging technologies offer promising approaches to overcome current limitations of IFNA2 therapeutics:
Structure-guided protein engineering: Rational design approaches based on detailed understanding of IFNA2-receptor interactions can yield variants with enhanced specificity for desired biological activities (e.g., antiviral vs. antiproliferative) or reduced interaction with receptor components linked to adverse effects.
Cell-type targeting strategies: Fusion proteins combining IFNA2 with antibody fragments or ligands directed against cell-specific surface markers can concentrate activity in desired target tissues while reducing systemic effects.
Controlled release formulations: Advanced biomaterial platforms including hydrogels, nanoparticles, and implantable devices can provide sustained local delivery of IFNA2, potentially improving efficacy while reducing toxicity associated with peak-and-trough kinetics.
Gene therapy approaches: Viral vectors or non-viral delivery systems carrying the IFNA2 gene under control of inducible or tissue-specific promoters offer potential for localized, regulated production with reduced systemic exposure.
Combination with immune checkpoint inhibitors: Emerging evidence suggests synergistic activity between type I interferons and immune checkpoint blockade, potentially opening new therapeutic windows for IFNA2 in cancer immunotherapy.
These technological advances may revitalize interest in IFNA2-based therapeutics by addressing the toxicity concerns that have limited its use in many clinical contexts .
Interferon-alpha 2c (IFN-α2c) is a member of the type I interferon family, which plays a crucial role in the innate immune response against viral infections. Interferons are cytokines, proteins that are secreted by cells in response to various stimuli, including viral infections, and they have antiviral, antitumor, and immunomodulatory effects .
Interferons were first discovered in 1957 by Isaacs and Lindenmann, who observed that cells infected with a virus produced a substance that could inhibit viral replication in other cells . Interferons are classified into three types based on their receptor specificity: type I, type II, and type III. Type I interferons include multiple subtypes, such as IFN-α and IFN-β, with IFN-α being the most extensively studied .
IFN-α2c is one of the three acid-stable forms of IFN-α2, the others being IFN-α2a and IFN-α2b. These forms share a high degree of sequence identity, with IFN-α2b sharing 99.4% amino acid sequence identity with both IFN-α2a and IFN-α2c . IFN-α2c contains four highly conserved cysteine residues that form two disulfide bonds, which are essential for its biological activity .
Recombinant DNA technology has enabled the production of human interferons, including IFN-α2c, in various expression systems such as Escherichia coli (E. coli). This technology involves inserting the gene encoding IFN-α2c into a plasmid vector, which is then introduced into a host cell for protein expression . The recombinant protein is subsequently purified and formulated for therapeutic use.
IFN-α2c, like other interferons, is used in the treatment of various viral infections and cancers. It exerts its antiviral effects by inducing the expression of interferon-stimulated genes (ISGs) that inhibit viral replication and enhance the immune response . Additionally, IFN-α2c has antitumor properties, making it a valuable therapeutic agent in oncology .
One of the main challenges in the clinical application of interferons, including IFN-α2c, is their short half-life, which necessitates frequent dosing. Researchers are exploring strategies to extend the half-life of interferons, such as pegylation (attachment of polyethylene glycol) and fusion with other proteins . These approaches aim to improve the pharmacokinetic properties and therapeutic efficacy of interferons.