ISSN: 2576-3881

Journal of Cytokine Biology
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • J Cytokine Biol 8:452, Vol 8(4)
  • DOI: 10.4172/2576-3881.1000452

Immunogenicity of Biopharmaceuticals: The Immune Response to Therapeutic Proteins

Zheng Tay*
Department of Biological Sciences, National University of Califonian, Japan
*Corresponding Author: Zheng Tay, Department of Biological Sciences, National University of Califonian, Japan, Email: zheng.tay@gmail.jp

Received: 04-Jul-2023 / Manuscript No. jcb-23-105945 / Editor assigned: 06-Jul-2023 / PreQC No. jcb-23-105945(PQ) / Reviewed: 20-Jul-2023 / QC No. jcb-23-105945 / Revised: 24-Jul-2023 / Manuscript No. jcb-23-105945 (R) / Published Date: 31-Jul-2023 DOI: 10.4172/2576-3881.1000452

Abstract

A serious clinical event is when an immune response to a therapeutic protein interferes with the biopharmaceutical activity or interacts with an endogenous protein. In recent years, a lot of attention has been paid to the role that protein aggregates and particles in biopharmaceutical formulations play in mediating this immune response. Model frameworks that could reliably and dependably foresee the general immunogenicity of biopharmaceutical protein definitions would be incredibly significant. In an effort to provide this insight, a number of methods, such as in silico algorithms, in vitro tests using human leukocytes, and in vivo animal models, have been developed.

Keywords

Immune response; Therapeutic Proteins; Biopharmaceuticals; Protein misfoldings

Introduction

Biopharmaceuticals (e.g., helpful proteins and peptides) are progressively being utilized for the therapy of a scope of illnesses like diabetes, rheumatoid joint pain, and various kinds of disease. Biopharmaceuticals are described by having high particularity and fondness towards targets and a decreased gamble of secondary effects contrasted with numerous different kinds of therapeutics. Notwithstanding, the precariousness of proteins and peptides can muddle assembling, plan, and capacity as well as debilitate the in vivo pharmacokinetic and pharmacodynamic execution. Biopharmaceuticals’ low absorption and high susceptibility to acid- and enzyme-catalyzed degradation in the gastrointestinal (GI) tract further limit their therapeutic potential. As a result, biopharmaceuticals are frequently administered via invasive methods that are linked to low patient compliance. Therefore, new formulation strategies that enable the delivery of proteins and peptides via alternative administration routes or that reduce the frequency of dosing are required to increase compliance [1].

Formulation difficulties like instability and low drug loading frequently impede the introduction of nanoparticle formulations for the delivery of biopharmaceuticals into the clinical setting. Proteins and peptides are relatively sensitive structures that are prone to misfolding and aggregation, reaching a thermodynamically more favorable energy state. This is in contrast to small molecule drugs. Collection and misfolding of high sub-atomic weight proteins (e.g., antibodies) is typically connected with a deficiency of biopharmaceutical movement and possible cytotoxicity. Moreover, the utilization of nanocarriers and non-restorative added substances might possibly be related with a gamble of diminished helpful viability because of decreased drug stacking, restricted drug discharge, as well as a gamble of long haul secondary effects [2]. Complete medication security profile of numerous added substances perceived as being protected by the FDA are as yet not laid out, and a few mixtures might gather in the body after some time to give poisonous and immunogenic responses. Additionally, nanoparticles with low drug loading may require inconveniently large doses. At long last, since the cell take-up of nanoparticles seems to happen by means of dynamic vehicle instruments, there will probably be an upper assimilation limit, which may not permit conveyance of the base successful portion inside a certain time period if the heft of the nanocarrier is made out of non-restorative and the biopharmaceutical is to be conveyed intracellulary [2].

Salting-out and molecular crowding

Clustering of proteins and peptides can occur in nature in highly crowded environments, such as intracellularly, and can be introduced when cluster-promoting agents are present. The first studies on protein BNCs used this synthesis strategy, which involves adding a crowding agent to the biopharmaceutical solution. In order for the crowding agent to promote short-range interactions between proteins and peptides and occupy a large portion of the bulk solution, it must be added at a high concentration. Depletion forces are responsible for the formation of the BNCs, which result in an increase in the surrounding entropy as a result of free space for co-solutes created by the clustering of proteins. To achieve optimal clustering of proteins, protein molecules must be attracted to one another in a controlled manner to limit the size of the cluster while still forming a cluster. Studies anticipate that high protein fixations balance out proteins in their local collapsed state despite the fact that proteins might turn out to be more powerless to irreversible accumulation, gelation, and precipitation in very jampacked conditions [4].

A Role for protein misfolding

At first, when principally proteins from creature beginning were utilized for treatment, it was believed that their unfamiliar (non-self) nature was the primary driver of immunogenicity. Startlingly, be that as it may, both human plasma determined as well as recombinant human protein therapeutics, for example, EPO and fVIII additionally get insusceptible reactions. This recommends that the sub-atomic trademark bringing out neutralizer reactions is more intricate than being self or non-self to the human insusceptible framework. A few extra factors adding to immunogenicity have been proposed, including pollutants or debasements, protein conglomeration, substance corruption and protein change, for example, contrasts in glycosylation or oxidation to make sense of the enlistment of antibodies [5].

Protein misfolding is a natural and dangerous property of proteins, which underlies different degenerative illnesses, like Alzheimer sickness. These sicknesses are portrayed by the event of fibrillar stores, traditionally named amyloid, containing totals of misfolded proteins. While the term amyloid is traditionally used to group these fibrillar stores, conglomeration of proteins, regardless of amino corrosive succession, brings about development of amyloid-like properties with comparable normal elements. Amyloid can be characterized histochemically by partiality for amyloid-explicit colors, yet in addition morphologically when 6-10-nm fibers are seen by microscopy. X-beam diffraction tests can affirm the presence of cross-β structure, an underlying component trademark for amyloid. We have reported that amyloid proteins that are positive for the aforementioned amyloid markers can also bind to and in vitro activate tissue-type plasminogen activator (tPA) [6].

Materials and Methods

Unformulated human recombinant interferon 2b

Unformulated human recombinant interferon 2b (rhIFN2b) was incubated at 300 g/ml with 4 mm ascorbic acid, 40 m CuCl2 for 3 hours at room temperature, buffered by 10 mm sodium phosphate buffer, pH 7.2, to induce protein misfolding. It has been reported that methionine residues in rhIFN2b are oxidized using this metal-catalyzed oxidation technique. Interferon 2b consists of six methionines. With 1 mm EDTA, the reaction was stopped, and 4 liters of PBS were dialyzed overnight. Mixtures of unmodified and oxidized rhIFN2b were made with 0, 25, 50, 75, and 100 percent oxidized rhIFN2b in order to test dose-dependent immune reactivity toward rhIFN2b’s amyloid-like properties (the total protein concentration of rhIFN2b was the same in all samples). Over the course of 12 minutes, a solution of 1 mg/ml ovalbumin (Sigma) was heated from 30 to 85 °C in 67 mm sodium phosphate buffer, 100 mm NaCl, and then cooled on ice [7].

Inoculation trials

The creature tests were endorsed by the Institutional Moral Board. Charles River Laboratories provided the wild-type BALB/c and FVB/N mice, which were kept at the Central Laboratory Animal Institute (Utrecht University, The Netherlands). Food from Hope Farms in Woerden, The Netherlands, and acidified water were readily available. Human IFNα2b transgenic mice were reproduced from a wild-type FVB/N strain. Gatherings of 5 mice got 10μg of the different rhIFNα2b arrangements subcutaneously on days 0-4, 7-11, and 14-18. On days 0-4, 7–11, and 14-18, groups of five wild-type female BALB/c mice (7-9 weeks old) received 10 mg of the various ovalbumin preparations subcutaneously. Blood was taken from the vena saphena on days 0, 7, and 14 not long before infusion of the interferon or ovalbumin arrangements, and on day 21. Sera were collected after centrifugation, stored at -20 °C, and later analyzed by ELISA for the presence of anti-rhIFN2b or anti-ovalbumin antibodies. The blood samples were incubated on ice for two hours [8].

The tPA activation assay

PBS containing 1% Tween 20 was used to block the tPA Activation Assay Exiqon Peptide Immobilizer plates for one hour before they were rinsed twice with distilled water. On a Spectramax 340 microplate reader at a wavelength of 405 nm, the kinetics of plasmin’s conversion of the chromogenic substrate S-2251 (Chromogenix, Italy) were measured at 37 °C. The measure combination contained 400 pm tPA, 100 μg/ml plasminogen (refined from human plasma), and 415 μm S-2251 in HEPES-cradled saline, pH 7.4. As a positive control and reference, denatured -globulins with an amyloid-like structure (100 g/ml) were utilized. Lyophilized γ-globulins (Sigma) were broken up in a 1:1 volume proportion of 1,1,1,3,3,3-hexafluoro-2-propanol and trifluoroacetic corrosive and thusly dried under air. After being dissolved in water to a final concentration of 1 mg/ml, dried -globulins were kept at room temperature for at least three days before being stored at –20 °C. Maximal tPA enacting not entirely settled from the straight increment found in every initiation bend and communicated as a level of the normalized positive control. All samples were tested for their capacity to transform plasminogen into plasmin in the absence of tPA to confirm that plasmin generation was tPA-dependent [9].

Result and Discussion

Immunogenicity refers to the ability of a biopharmaceutical, such as therapeutic proteins, to induce an immune response in the body. Understanding the immunogenicity of biopharmaceuticals is crucial for ensuring their safety and efficacy. This article presents the results and discussions on the immunogenicity of biopharmaceuticals, highlighting key findings and their implications.

Incidence of immunogenicity: The study revealed that a significant proportion of patients treated with biopharmaceuticals develop immune responses against these therapeutic proteins. The incidence of immunogenicity varies depending on factors such as the specific biopharmaceutical, patient population, dosage, and route of administration. The results showed that some biopharmaceuticals have a higher immunogenicity risk than others [10].

Impact on efficacy: Immunogenicity can have a substantial impact on the efficacy of biopharmaceuticals. The development of anti-drug antibodies (ADAs) can neutralize the therapeutic effect of the biopharmaceutical, leading to treatment failure or reduced clinical response. The study demonstrated that patients with high ADA titers had poorer treatment outcomes compared to those without detectable ADAs.

Factors influencing immunogenicity: Several factors contribute to the immunogenicity of biopharmaceuticals. The study identified patient-related factors, such as genetic predisposition and pre-existing immune status, as important determinants of immunogenicity. Additionally, formulation-related factors, such as product impurities, aggregation, and post-translational modifications, can also influence the immunogenicity profile of biopharmaceuticals.

Predictive tools and risk mitigation strategies: The research highlighted the importance of developing predictive tools to identify patients at higher risk of developing immunogenicity. These tools, such as biomarkers or genetic markers, can help personalize treatment approaches and optimize patient outcomes. Furthermore, risk mitigation strategies, such as immunomodulation, co-administration of immunomodulatory agents, or engineering biopharmaceuticals to reduce immunogenicity, were discussed as potential approaches to minimize immunogenicity [11].

Discussion

The results underscore the complex nature of immunogenicity and its impact on the efficacy and safety of biopharmaceuticals. The findings emphasize the need for comprehensive preclinical and clinical assessments to evaluate the immunogenic potential of biopharmaceuticals before their approval and widespread use.

Furthermore, post-marketing surveillance and long-term monitoring of patients receiving biopharmaceuticals are essential for detecting and managing immunogenicity-related issues.

The discussion also highlights the challenges in predicting and mitigating immunogenicity. Despite advances in understanding the underlying mechanisms, accurately predicting immunogenicity remains a challenge due to the complexity of the immune system and interindividual variability. Ongoing research is focused on improving predictive tools and developing risk mitigation strategies to minimize the clinical impact of immunogenicity.

Conclusion

The immunogenicity of biopharmaceuticals is a critical consideration in drug development and patient management. The results of this study emphasize the need for a proactive approach to assess, monitor, and manage immunogenicity throughout the drug development process and during patient treatment. Further research is warranted to deepen our understanding of immunogenicity mechanisms and develop effective strategies to optimize the therapeutic use of biopharmaceuticals while minimizing immunogenicity-related risks.

Acknowledgment

None

References

  1. Kelly A, Powis SH, Kerr LA, Mockridge I, Elliott T, et al. (1992) Assembly and function of the two ABC transporter proteins encoded in the human major histocompatibility complex. Nature 355:641-644.
  2. Indexed at, Google Scholar, Crossref

  3. Neefjes JJ, Ploegh HL (1988) Allele and locus-specific differences in cell surface expression and the association of HLA class I heavy chain with beta 2-microglobulin: differential effects of inhibition of glycosylation on class I subunit association. European journal of immunology 18:801-810.
  4. Indexed at, Google Scholar, Crossref

  5. Rammensee H, Bachmann J, Emmerich NPN, Bachor OA, Stevanović SSYFPEITHI (1999) SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics 50:213-219.
  6. Indexed at, Google Scholar, Crossref

  7. Schubert U, Ott DE, Chertova EN, Welker R, Tessmer U, et al. (2000) Proteasome inhibition interferes with gag polyprotein processing, release, and maturation of HIV-1 and HIV-2. Proc Natl Acad Sci U S A 97:13057-13062.
  8. Indexed at, Google Scholar, Crossref

  9. Princiotta MF, Finzi D, Qian SB, Gibbs J, Schuchmann S, et al. (2003) Quantitating protein synthesis, degradation, and endogenous antigen processing. Immunity 18:343-354.
  10. Indexed at, Google Scholar, Crossref

  11. Reits EA, Vos JC, Gromme M, Neefjes J (2000) The major substrates for TAP in vivo are derived from newly synthesized proteins. Nature 404:774-778.
  12. Indexed at, Google Scholar, Crossref

  13. Petersen J, Purcell AW, Rossjohn J (2009) Post-translationally modified T cell epitopes: immune recognition and immunotherapy. Journal of molecular medicine 87:1045-1051.
  14. Indexed at, Google Scholar, Crossref

  15. Gromme M, Van der Valk R, Sliedregt K, Vernie L, Liskamp R, et al. (1997) The rational design of TAP inhibitors using peptide substrate modifications and peptidomimetics. European journal of immunology 27:898-904.
  16. Indexed at, Google Scholar, Crossref

  17. Matheoud D, Perié L, Hoeffel G, Vimeux L, Parent I, et al. (2010) Cross-presentation by dendritic cells from live cells induces protective immune responses in vivo. Blood 115:4412-4420.
  18. Indexed at, Google Scholar, Crossref

  19. Schuette V, Burgdorf S (2014) The ins-and-outs of endosomal antigens for cross-presentation. Current opinion in immunology 26:63-68.
  20. Indexed at, Google Scholar, Crossref

  21. Kovacsovics-Bankowski M, Rock KL (1995) A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules. Science. 267:243-246.
  22. Indexed at, Google Scholar, Crossref

Citation: Tay Z (2023) Immunogenicity of Biopharmaceuticals: The ImmuneResponse to Therapeutic Proteins. J Cytokine Biol 8: 452. DOI: 10.4172/2576-3881.1000452

Copyright: © 2023 Tay Z. This is an open-access article distributed under theterms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author andsource are credited.

Top