Submit manuscript...
International Journal of
eISSN: 2470-9980

Vaccines & Vaccination

Review Article Volume 2 Issue 4

A Message from the Immune System: A Vaccine Technology in Needs of Exploration!

Eid E Haddad

Department of Biological Sciences, Fayetteville State University, USA

Correspondence: Eid E Haddad, Assistant Professor of Physiology, Department of Biological Sciences, Fayetteville State University, 1200 Murchison Road, Fayetteville, NC 28301, USA, Tel (910) 672 2436, Fax (910) 672 1159

Received: March 01, 2016 | Published: August 8, 2016

Citation: Haddad EE (2016) A Message from the Immune System: A Vaccine Technology in Needs of Exploration!. Int J Vaccines Vaccin 2(4): 00039. DOI: 10.15406/ijvv.2016.02.00039

Download PDF

Abstract

A vaccine technology based on presenting the antigen in the context of its antibody; that is an antigen-antibody complex vaccine has been successfully applied to developing at least two vaccines against two diseases of value to the veterinary field. The technology is based on complexing the antigen with its homologous antibody to form an antigen-antibody complex (AAC) that can then be administered as a preformed AAC. Antigens can be a whole live or killed/attenuated virus/bacterium, fragment of a virus/bacterium, peptide(s), or protein(s)/glycoprotein(s). Polyclonal or monoclonal IgG isotype antibody is the preferred antibody to formulate the AAC with Fab or F(ab’)2 being either ineffective or much less effective in generating an immune response comparable to that generated by AAC. Published research has shown quantitative and qualitative gains in the immune handling and response to such AAC vaccines. Lower antigenic mass with less need for adjuvants, directed delivery of antigens, enhanced efficiency of antigen recognition and uptake, processing and presentation to immune cells, recruitment of immune effector cells that otherwise would have not been recruited with conventional antigen alone, earlier and faster immune response, high affinity and quality antibodies similar to that obtained during a secondary immune response, changes in cytokine profiles favoring Th2 profile, and in the case of AAC containing a live virus the ability to control timing and extent of viral replication. The mechanism of action is believed to be through FcRs, possibly through the activating FcRs, requiring Ag: Ab ratio of about molar equivalency, or appropriate neutralization titer (live AAC). Excess antibody, but not Ag was shown to be immunosuppressive, possibly working through inhibitory FcRs. We believe that antigens presented as AAC are “groomed” antigens stimulating the immune system to express its full or “hidden” “talents” to optimize the immune response with the least efforts or cost to the immune system and the body as a whole. It seems like a message from the immune system telling us a way to explore its full talents.

Keywords: vaccines, antigen-antibody complex vaccines, immune responses,antigen presenting cells

Abbreviations

AAC, antigen-antibody complex; Ab, antibody; Ag, antigen; APC, antigen presenting cells; DC, dendritic cells; FDC, follicular dendritic cells; FcR, fc receptors; GC, germinal centers; IBDV, infectious bursal disease virus; IC, immune complex; IFN-α, interferon-alpha; IFN-γ, interferon-gamma; IL, interleukin; MHC, major histocompatibility complex; RER, rough endoplasmic reticulum

Introduction

There is no disagreement that vaccines are the most efficient way to compact diseases caused by pathogenic microorganisms. Such an efficient way of “restoring physiological normalcy” seems to gain momentum with other fields, namely the fields of oncology, autoimmunity, and allergy. Since the discovery of the first vaccine by Edward Jenner in 1796,1 a plethora of approaches and technologies employed to develop vaccines, each aspiring to produce an “ideal vaccine,” a vaccine that is safe and efficacious regardless of age or immune status of the recipient, produces high-quality innate and adaptive immune responses after a single application, provide a long-lasting immunity, and obviously cost effective.2‒5 Current vaccine technologies are based on either whole organism (live, attenuated, or killed microorganism), microorganism components (peptide, protein, toxoid, & recombinant), or microorganism genetic components (DNA or RNA) technology-based vaccines. Other approaches employ adjuvants or adjuvant-like molecules (molecules attached to the antigen) demonstrated or believed to provide a better opportunity to a better immune response.

A technology in vaccine development based on an exogenously preformed “antigen-antibody complex: AAC” that can then be administered as a vaccine has gained regulatory approval for two such vaccines with applications in the animal health field. Published data clearly demonstrate better vaccine performance in terms of the quality of the immune responses. This review will examine their formulations and the qualities of the immune responses warranting tremendous gains consequent upon further exploration; But first a brief overview of the immune system and the immune response.

Overview of the immune system

A physiologically functional immune system evolved to include many organs, cells, molecules and pathways, all working together to maintain homeostasis within the body when exposed to signals (antigens or pathogens) disturbant to such a homeostasis. Their collective response is called the immune response. The immune response is composed of two very interconnected arms: innate and adaptive immune responses. Innate immune response (innate immunity) is a non-specific, short-lived immunity which includes built-in cellular and molecular mechanisms aimed at quickly preventing infection or eliminating invaders. On the other hand, adaptive immune response (adaptive immunity) is specific and long-lasting aimed at affording life-long immunity against invading pathogens and is characterized by the cooperation of two subsets of lymphocytes: T and B lymphocytes which are both naïve until stimulated. The trademark of a successful adaptive immune response is the successful recognition and processing of an exogenous (or endogenous) antigen (Ag) by the proper immunocytes and the subsequent presentation of specific components of the processed Ag to the proper immunocytes. Adaptive immune response leads to immunological memory. Immune cells capable of antigen recognition, processing, and presentation are known as antigen presenting cells (APCs). Some of these APCs are classified as professional APCs (capable of presenting Ag in the context of the Major Histocompatibility Complex MHC class I or MHC class II) and include dendritic cells (DC),6 follicular dendritic cells (FDC) 7, B cells,8 Macrophages,9 and eosinophils.10,11 Non-professional APCs capable of presenting Ag to MHC I-restricted T cells include, among others, neutrophils and resident phagocytic cells such as Langerhans cells of the skin 12 and Kupffer cells of the liver.

The two classes of MHCs differ in which cell expresses them and in the source of antigen they present to T cells. Class I molecules are expressed on all nucleated cells and present antigens originating in the cytosol such as viral proteins, to CD8+ T cytotoxic (Tc) cells, which recognize and kill any cell expressing such proteins. Class II molecules are expressed mainly on APC and present extracellular antigens to CD4+ T helper (Th) cells, which stimulate B cells to differentiate into plasma antibody-secreting cells or memory cells. Cytosolic processing of soluble Ag includes the proteosomic degradation of Ag to constituent amino acids, which are recycled, but some may persist as peptides. It is these peptides that are sampled, assembled with Class I MHC in the ribosomal rough endoplasmic reticulum (RER) and presented on the surface of the APC as peptide associated with class I MHC.13 Processing of exogenous antigen includes the ribosomal RER synthesis of Class II MHC, internalization of exogenous Ag and the subsequent endolysosomic degradation of the Ag and the association of some of their peptides with Class II MHC, which is then expressed on the surface of APC.14 Conventionally, exogenous soluble Ag can be internalized by two mechanisms: pseudopods-engolfed phagocytosis or endocytosis (receptor-mediated endocytosis or pinocytosis). Macrophages and DCs employ both mechanisms; other professional or non-professional APC are weak phagocytic cells and rely on endocytosis for internalization of exogenous Ag. Loading of exogenous Ag into Class I MHC and the subsequent presentation to MHC Class I restricted Tc lymphocytes is known as cross-presentation; DCs have been known to be the most efficient in cross-presentation.15

Another process where Ag can be recognized by APC is when antigens are introduced as part of a complex composed of the Ag itself bound to a specific antibody (Ab) against the Ag to form an Antibody-Antigen Complex (AAC) or simply a preformed immune complex (IC). We will use the abbreviation “AAC” as opposed to “IC” to make the distinction between exogenously preformed and endogenously formed “and potentially harmful” IC. Unlike the recognition of a free soluble Ag, antigens introduced as part of an AAC are recognized by binding to the Fc receptors (FcRs) expressed on the surface of several immune and non-immune APCs leading to a mechanism of Ag processing that is documented to be different from that for the conventional Ag processing.

Fc receptors (FcRs) are members of the immunoglobulin superfamily composed of membrane-associated glycoproteins that mediate a vast array of functions triggered by immune complexes. There are three main types of FcRs based on which antibody isotype they bind: FcγRs (binds to IgG isotypes), FcαRs (binds to IgA isotype), and FcεRs (bind to IgE isotype). Furthermore, the three types also differ in cells that express them (DCs, macrophages, granulocytes, NK cells, and T & B cells and others) and their signaling properties.16‒19 FcγRs are the most diverse receptors in the body and contains four families: FcγRI (CD64), FcγRIIA, B, C (CD32A, B, C), FcγRIIIA & B (CD16A & B), and FcγRIV (mice with a human equivalent believed to be FcγRIIIA). All FcγRs are activating receptors with the exception of FcγRIIB being inhibitory.15 Another FcγRs known as neonatal Fc receptors (FcγRn)20,21 are expressed on many cells, including epithelial and endothelial cells, macrophages, and DC, and function in carrying ingested milk IgG (from the mother) across the infant’s intestine, as well as immune complexes binding and internalization (for more on FcγRs,\ see reference.15)

AAC formulations

As stated earlier, this review focuses on exogenously preformed and introduced AAC, which is different from endogenously formed immune complexes after an immune response and have been known to cause harm. These AAC can be “first order” (AAC comprising a whole live pathogenic (replicating), inactivated, or whole fragments of a virus or a bacterium complexed to a specific IgG subtype monoclonal or polyclonal Ab) or “second order” (AAC comprising a peptide or a protein/glycoprotein complexed to a specific IgG subtype monoclonal or polyclonal Ab). The use of IgM or IgA isotypes has also been reported, though desirable immune responses were observed with IgG isotypes.22 The ratio of Ag: Ab in the AAC seems to have an effect with desirable results obtained when the ratio is at molar equivalency23‒27 or a slight excess of the Ag or the Ab.28,29 Formulations containing excess Ab (but not excess Ag) were found to be immunosuppressive.30 Our own approach to formulating AAC was based on the neutralizing activity of the Ab31 and was shown to be very effective, especially when the Ag was a live pathogen,32‒34 providing an accurate and precise formulation and allowing a precise timing of viral replication. The first vaccine employing the AAC vaccine technology was licensed for commercial use in 1994.32,33 The vaccine was formulated using a live infectious bursal disease virus (IBDV) complexed with chicken anti-IBDV polyclonal antibodies to produce the AAC vaccine. The disease is infectious to many avian species with poultry being the most affected. Immature B cells within the bursa of Fabricius (a primary immune organ) are the target cells for the virus rendering the animal immunosuppressed and incapable of responding immunologically to vaccinations or opportunistic pathogens causing substantial economic losses.35 Whole antibody molecules (not Fab or F(ab’)2 fragments) seem to be needed for the successful induction of the immune system.36,37 A recombinant form of the AAC (or IC) technology has recently been described.38

AAC technology application

Augmentation of immune responses to first- or second-order AAC has been reported for various viral and bacterial antigens, including HIV-1 ,39‒43 Hepatitis B surfaces antigen,44‒50 Ebola protein antigens co-expressed with Ab,51 autoimmune kidney disease,52‒65 Equine herpesvirus,66 chicken infectious bursal disease virus,32,33,67‒69 Chicken Newcastle disease virus,34,70 Chicken Reovirus,71 Chicken anemia virus,72 Swine parvavirus,25,70 formalin-inactivated Equine encephalomyelitis,26 envelope protein of tick-borne Encephalitis virus,73,74 gram-negative, rod-shaped coccobacillus Francisella tularensis,75 Mycobacterium tuberculosis,27 and the Streptococcus mutans.76,77 So far, and in the animal health, two AAC-based vaccines gained regulatory approval in multiple countries,32,33,67‒69 however, in the human health, one AAC-based vaccine is in clinical trial.45,47,49 Application of the AAC technology was also demonstrated on human Reovirus (stain TD3 using a mouse experimental model), avian Infectious Bronchitis, and canine Parvovirus complex.78 Thus, available data suggest that this vaccine development technology is applicable to both human and animal diseases.

Localization of AAC post administration

Although the route of vaccine introduction seems to be a factor with some vaccines or specific vaccine technologies, antigens introduced as AAC vaccines seem to be localized or trapped mainly by the follicular dendritic cells (FDC) of the splenic germinal centers (GC)67,79‒88 and to a lesser degree other professional and non-professional APC.89,90 An increase in the number of splenic GC was reported after administration of the AAC relative to administering a comparable amount of the antigen alone.67,91

Quantitative and qualitative changes in the immune response

We and others have demonstrated that both “quantitative and qualitative changes”77 in the immune response occurred in response to encountering the Ag as part of an AAC relative to encountering the Ag alone. These changes were reported in several mammalian as well as avian species and included facilitating presentation,15,28,83,92‒96 enhanced antigenicity of a weak antigen,97 delayed viral replication32,68, 69,72 (in the case of live virus AAC), lower antigenic mass or dose by several folds,23,90,95,98‒101 an earlier immunity,76,91 changes specific to B cell such as enhanced B cells activation and differentiation,29, 82 and the selective enhancement of memory B cells in GC37,102,103 as well as secondary antibody responses.104

We had previously shown32 (and confirmed later by others65,66,69) that AAC containing live virus (live AAC) caused delayed replication of the live virus by a period of 6-8days. The delay was directly proportional to the amount of Ab in the AAC. Such delay in viral replication allowed for the early administration of the AAC vaccine at day 18 of embryonic development to developing chick embryos, three days prior to their birth or hatch. Needless to say that same amount of the virus alone is lethal to the embryo when administered at day18 of embryonic development. What is more? the virus is not safe for administration as a vaccine until after day14 of the bird age. The safety of the live AAC was also significantly enhanced where histological changes in the target organ were less than the severe changes observed with the virus alone. The AAC was immunogenic, generating a protective immunity for the life of the birds. Moreover, we have shown that administration of AAC reduced B cell depletion from target organs, increased germinal centers in the spleen, and the localization of AAC to FDC in the spleen as well as the Bursa of Fabricious.67

Changes related to T cells activations and proliferations were also observed,23,89,100 and included the in vivo Th-independent priming of peptide-specific CD8+ Tc cells83,92,105 by in vitro peptide AAC-activated DC. Enhancements or alterations of lymphokines were also documented among the changes in the adaptive immune response to AAC, as compared to those observed after administration of Ag alone.44,105‒108 Lymphokines reported to have increased in sera of patients after the administration of Hepatitis-B AAC included interleukin (IL)-2, interferon-gamma (IFN-γ), but not IL-4, IL-6, IL-10, or TNF-α.44,47 Others reported changes in lymphokine profile suggestive of the influence of AAC on the balance of Th1/Th2 responses and the shift of Th1 to Th2.105,106,108 Th1 effector cells are effector cells active against intracellular Ags and are activated by IL-10 and IL-12 and their effector cytokines IL-2, IL-10, tumor necrosis factor (TNF)-β, and IFN-γ tend to produce cell-mediated immunity and pro-inflammatory responses. Th2 effector cells are active against extracellular Ags and are activated by IL-4 and their effector cytokines include IL-4, 5, 6, 9, 10, and 13 and tend to elicit strong antibody responses. In summary, these changes were realized as a result of enhanced Ag recognition, processing and presentation, and the recruitment of immune cells bearing FcRs that may not be activated or recalled in response to Ag alone.

Potential mechanism of action (MOA) and concluding remarks

Understanding how an antigen encounters and interacts with the proper immunocytes may lead to the development of better vaccines or at least better formulation of vaccines. Needless to say that Ab paratopes are expected to recognize and bind antigenic epitopes (most likely the dominant epitopes) on the surface of the Ag, thus shielding these dominant epitopes transiently. Furthermore, the binding of Ab to the Ag (virus) was shown to induce structural changes in the topography of the virus surface, exposing new antigenic or highly conserved epitopes or determinants.73,109‒111 Whether it is preserving the antigenicity and the immunogenicity of the dominant epitopes or the exposure of conserved epitopes or both (most likely), the result is an Ag groomed for optimal introduction. We believe that the immune system possesses any array of talents that can be fully expressed if antigens are properly presented, leading to a superior immune response with minimal efforts or cost on the part of the immune system itself. There is ample evidence that the efficiency of Ag recognition and uptake (FcR-mediated), processing (FcR-triggered) and presentation in association of MHC I and II are significantly enhanced when Ag is introduced in the context of an Ab (AAC) as compared to introducing Ag alone. Additionally, antigens introduced as AAC may stimulate immune cells that otherwise would have not been stimulated with an Ag not aided by an antibody. Taken together, it may explain the robustness, efficiency, and the high quality of the immune response that has been equated to that obtained with a conventional secondary immune response.95 Importantly, in work to date, these exogenously preformed AAC stimulated a beneficial and highly protective responses rather than the pathological response which can result from immune complexes generated in vivo, further substantiating our hypothesis illustrated here that groomed Ag may lead to expression of more powerful immune talents.

Data to date showed that AAC vaccines formulated with a live pathogen (virus) complexed to its homologous antibodies clearly presented a way to control viral replication in terms of initiation, quantity, and duration of viral replication; no other vaccines containing live or attenuated pathogen is capable of such control mechanism. Although the exact mechanism is unknown; antibody half-life, antibody neutralizing capabilities, and the time between internalization (through FcR) and processing (storage) may individually or collectively constitute a delay mechanism of viral replication. Nevertheless, the advantages of such a feature are enormous; foremost is the potential to earlier vaccine administration32‒34,67,69,78 where the vaccine can be timed as to when it is processed. In the veterinary field, it is a common practice to hyperimmunize the mother in the hope that the mother would passively pass its immunity to its progenies, thus affording the progenies immediate (but short) protection. Passive immunization is capable not only of neutralizing and eliminating the wild-type pathogens but also any vaccine agents (regardless if it is live, attenuated, or killed) administered early in life. AAC vaccines containing live virus were shown to overcome such interferences of passive immunity with vaccine virus.

Another potential for introducing Ag in the context of AAC is the possibility to eliminate the need for an adjuvant.27 Administration of AAC is anticipated to provide a link between the adaptive immune response and the powerful array of innate immunity relative to administering Ag alone.112 Vaccines for human application could benefit from such features where the use of adjuvants may be redefined since the use of adjuvant with an AAC-based therapeutic vaccine did not show much improvement.44 Additionally, technologies for commercial production of humanized antibodies are available and the use of therapeutic antibodies is well established. In conclusion, there are strong data showing immune enhancements and advantages of presenting Ag in the form of AAC relative to presenting Ag alone. Is it possible that the immune system prefers antigens delivered by one of its own mailmen?

Acknowledgments

None.

Conflicts of interest

Author declares there are no conflicts of interest.

Funding

None.

References

  1. Willis NJ. Edward Jenner and the eradication of smallpox. Scott Med J. 1997;42(4):118‒121.
  2. Levine MM. "IDEAL" vaccines for resource poor settings. Vaccine. 2011;29(Suppl4):D116‒D125.
  3. de Veer M, Meeusen E. New developments in vaccine research unveiling the secret of vaccine adjuvants. Discov Med. 2011;12(64):195‒204.
  4. Hoft DF, Brusic V, SakalaI G. Optimizing vaccine development. Cell Microbiol. 2011;13(7):934‒942.
  5. Keler T, He L, Ramakrishna V, et al. Antibody targeted vaccines. Oncogene.2007;26(25):3758‒3767 .
  6. Steinman RM Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I.Morphology, quantitation, tissue distribution. J Exp Med. 1973;137(5):1142‒1162.
  7. Mildner A , Jung S. Development and function of dendritic cell subsets. Immunity. 2014;40(5):642‒656.
  8. Katz DR. Antigen presentation, antigen presenting cells and antigen processing. Curr Opin Immunol . 1988;1(2):213‒219.
  9. Allen PM, Unanue ER. Antigen processing and presentation by macrophages. Am J Anat. 1984;170(3):483‒490.
  10. Shi HZ. Eosinophils function as antigen-presenting cells. J Leukoc Biol. 2004;76(3): 520‒527.
  11. Akuthota P. Eosinophil interactions: antigen presentation. Methods Mol Biol. 2014;1178:251‒255.
  12. Nickoloff BJ, Turka LA. Immunological functions of non-professional antigen presenting cells: new insights from studies of T-cell interactions with keratinocytes. Immunol Today. 1994;15(10):464‒469 .
  13. Van Endert P. Processing of MHC class I presented antigens. Med Sci (Paris). 2006;22(8-9):727‒732.
  14. Robinson JH, Delvig AA. Diversity in MHC class II antigen presentation. Immunology. 2002;105(3):252‒262.
  15. Platzer B, Stout M, Fiebiger E. Antigen cross presentation of immune complexes. Front Immunol. 2014; 5:140.
  16. Ravetch JV, Bolland S . IgG Fc receptors. Annu Rev Immunol. 2001;19: 275‒290.
  17. Ravetch JV, Kinet JP. Fc receptors. Annu Rev Immunol. 1991;9: 457‒492.
  18. Daeron M. Fc receptor biology. Annu Rev Immunol. 1997;15:203‒234 .
  19. Molfetta R, Quatrini L, Gasparrini F, et al. Regulation of fc receptor endocytic trafficking by ubiquitination. Front Immunol . 20145: 449 .
  20. Baker K, Rath T, Pyzik M, et al. The Role of FcRn in Antigen Presentation. Front Immunol. 2014;5:408.
  21. Liu X, Lu L, Yang Z, et al. The neonatal FcR mediated presentation of immune complexed antigen is associated with endosomal and phagosomal pH and antigen stability in macrophages and dendritic cells. J Immunol. 2011;186(8):4674‒4686.
  22. Klaus GG. Generation of memory cells. III. Antibody class requirements for the generation of B-memory cells by antigen--antibody complexes. Immunology. 1979; 37(2):345‒351.
  23. Galesic MS, Pavelic K, Pokric B. Cellular immune response to the antigen administered as an immune complex. Immunology. 1991;72(4):526‒531.
  24. Marusic M, Galesic MS, Pokric B. Humoral immune response to the antigen administered as an immune complex. Immunol Invest . 1992;21(7):623‒628.
  25. Roic B, Cajavec S, Ergotic N, et al. Immune complex based vaccine for pig protection against parvovirus. J Vet Med B Infect Dis Vet Public Health. 2006;53(1):17‒23.
  26. Houston WE, Pedersen CE, Cole FE, et al. Effects of antigen-antibody complexes on the primary immune response in Rhesus monkeys. Infect Immun. 1974;10 (3):437‒442 .
  27. Pepponi I, Stylianou E, van Dolleweerd C, et al. Immune complex mimics as a molecular platform for adjuvant free vaccine delivery. PLoS One. 2013;8(4):e60855.
  28. Manca F, Fenoglio D, Li Pira G, et al. Effect of antigen antibody ratio on macrophage uptake, processing, and presentation to T cells of antigen complexed with polyclonal antibodies. J Exp Med. 1991;173(1):37‒48.
  29. Soderberg LS. Antigen antibody complex binding and cell interaction in stimulating normal rabbit lymphocytes. Proc Soc Exp Biol Med. 1983;174(2):229‒238.
  30. Caulfield MJ, Shaffer D. Immunoregulation by antigen/antibody complexes. I. Specific immunosuppression induced in vivo with immune complexes formed in antibody excess. J Immunol. 1987;138(11):3680‒3683.
  31. Haddad EE, Whitfill CE, Ricks CA, et al. Adaptation of the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay for the determination of virus neutralizing antibodies using the virus-neutralization assay. Avian Dis. 1994;38(4):755‒761.
  32. Whitfill CE, Haddad EE, Ricks CA, et al. Determination of optimum formulation of a novel infectious bursal disease virus (IBDV) vaccine constructed by mixing bursal disease antibody with IBDV. Avian Dis. 1995;39(4):687‒699.
  33. Haddad EE, Whitfill CE, Avakian AP, et al. Efficacy of a novel infectious bursal disease virus immune complex vaccine in broiler chickens. Avian Dis. 1997;41(4):882‒889.
  34. Kapczynski DR, Martin A, Haddad EE, et al. Protection from clinical disease against three highly virulent strains of Newcastle disease virus after in ovo application of an antibody antigen complex vaccine in maternal antibody positive chickens. Avian Dis. 2012;56(3): 555‒560 .
  35. Sharma JM, Kim IJ, Rautenschlein S, et al. Infectious bursal disease virus of chickens: pathogenesis and immunosuppression. Dev Comp Immunol. 2000;24(2‒3):223‒235.
  36. Lee EH, Rikihisa Y. Anti Ehrlichia chaffeensis antibody complexed with E. chaffeensis induces potent proinflammatory cytokine mRNA expression in human monocytes through sustained reduction of IkappaB alpha and activation of NF kappaB. Infect Immun. 1997;65(7):2890‒2897.
  37. Klaus GG. The generation of memory cells. II. Generation of B memory cells with preformed antigen-antibody complexes. Immunology. 1978;34(4): 643‒652.
  38. Mason HS. Recombinant immune complexes as versatile and potent vaccines. Hum Vaccin Immunother . 2016;12(4):988‒989.
  39. Hioe CE, Visciano ML, Kumar R, et al. The use of immune complex vaccines to enhance antibody responses against neutralizing epitopes on HIV-1 envelope gp120. Vaccine. 2009;28(2):352‒360.
  40. Liao HX, Alam SM, Mascola JR, et al. Immunogenicity of constrained monoclonal antibody A32-human immunodeficiency virus (HIV) Env gp120 complexes compared to that of recombinant HIV type 1 gp120 envelope glycoproteins. J Virol. 2004;78(10):5270‒5278.
  41. Kumar R, Tuen M, Liu J, et al. Elicitation of broadly reactive antibodies against glycan-modulated neutralizing V3 epitopes of HIV-1 by immune complex vaccines. Vaccine . 2013;31(46): 5413‒5421.
  42. Kumar R, Tuen M, Li H, et al. Improving immunogenicity of HIV-1 envelope gp120 by glycan removal and immune complex formation. Vaccine. 2011;29(48):9064‒9074.
  43. Visciano ML, Tuen M, Gorny MK , et al. In vivo alteration of humoral responses to HIV-1 envelope glycoprotein gp120 by antibodies to the CD4-binding site of gp120. Virology. 2008;372(2):409‒420 .
  44. Wen YM, Qu D, Zhou SH. Antigen-antibody complex as therapeutic vaccine for viral hepatitis B. Int Rev Immunol. 1999;18 (3):251‒258.
  45. Xu DZ, Zhao K, Guo LM, et al. A randomized controlled phase IIb trial of antigen-antibody immunogenic complex therapeutic vaccine in chronic hepatitis B patients. PLoS One. 2008;3(7):e2565.
  46. Yao X, Zheng B, Zhou J, et al. Therapeutic effect of hepatitis B surface antigen-antibody complex is associated with cytolytic and non-cytolytic immune responses in hepatitis B patients. Vaccine. 2007; 25(10):1771‒1779.
  47. Liu SA, Xu DZ, Zhang JP, et al. Immune response for phase I clinical trial of a hepatitis B immunogenic complex therapeutic vaccine, YIC. Zhonghua Gan Zang Bing Za Zhi. 2006;14(2):89‒92 .
  48. Wen YM. Antigen-antibody immunogenic complex: promising novel vaccines for microbial persistent infections. Expert Opin Biol Ther. 2009;9(3): 285‒291.
  49. Xu DZ, Wang XY, Shen XL, et al. Results of a phase III clinical trial with an HBsAg-HBIG immunogenic complex therapeutic vaccine for chronic hepatitis B patients: experiences and findings. J Hepatol . 2013;59(3):450‒456.
  50. Barabas AZ, Cole CD, Graeff RM, et al. Tolerance, loss of tolerance and regaining tolerance to self by immune-mediated events. Immunol Res. 2016.
  51. Phoolcharoen W, Bhoo SH, Lai H, et al. Expression of an immunogenic Ebola immune complex in Nicotiana benthamiana. Plant Biotechnol J. 2011;9 (7):807‒816.
  52. Barabas AZ, Cole CD, Lafreniere R, et al. Regaining tolerance to a self-antigen by the modified vaccination technique. Clin Rev Allergy Immunol. 2013; 45 (2):193‒201.
  53. Barabas AZ, Cole CD, Barabas AD, et al. New vaccination technology for endogenous antigen-derived ailments. IDrugs. 2008;11(2):111‒115.
  54. Barabas AZ, Cole CD, Barabas AD, et al. Effect of rat kidney fraction 3 (rKF3) antigen and specific IgM antibody against rKF3 on the progression of slowly progressive Heymann nephritis. Pathol Int . 2006;56(9):516‒529.
  55. Barabas AZ, Cole CD, Barabas AD, et al. Reduced incidence of slowly progressive Heymann nephritis in rats immunized with a modified vaccination technique. Clin Dev Immunol. 2006;13(1):17‒24.
  56. Barabas AZ, Cole CD, Barabas AD, et al. Presence of immunoglobulin M antibodies around the glomerular capillaries and in the mesangium of normal and passive Heymann nephritis rats. Int J Exp Pathol. 2004;85(4):201‒212.
  57. Barabas AZ, Cole CD, Barabas AD, et al. Correcting autoimmune anomalies in autoimmune disorders by immunological means, employing the modified vaccination technique. Autoimmun Rev. 2009;8(7):552‒557.
  58. Barabas AZ, Cole CD, Barabas AD, et al. Modified vaccination technique for prophylactic and therapeutic applications to combat endogenous antigen-induced disorders. Scand J Immunol. 2010;71(3):125‒133.
  59. Barabas AZ, Cole CD, Barabas AD, et al. Production of a new model of slowly progressive Heymann nephritis. Int J Exp Pathol . 2003;84(6):245‒258.
  60. Barabas AZ, Cole CD, Barabas AD, et al. Down-regulation of pathogenic autoantibody response in a slowly progressive Heymann nephritis kidney disease model. Int J Exp Pathol. 2004;85(6):321‒334.
  61. Barabas AZ, Cole CD, Barabas AD, et al. Production of Heymann nephritis by a chemically modified renal antigen. Int J Exp Pathol. 2004;85(5):277‒285.
  62. Barabas AZ, Cole CD, Barabas AD, et al. Downregulation of a pathogenic autoantibody response by IgM autoantibodies directed against the nephritogenic antigen in slowly progressive Heymann nephritis. Pathol Int . 2006;56(4):181‒190.
  63. Barabas AZ, Cole CD, Barabas AD, et al. A modified vaccination technique for the prevention and treatment of an experimental autoimmune kidney disease. Ann NY Acad Sci. 2007;1110: 619‒629.
  64. Barabas AZ, Cole CD, Barabas AD, et al. Preventative and therapeutic vaccination to combat an experimental autoimmune kidney disease. Biologics. 2007;1(1):59‒68.
  65. Barabas AZ, Weir DM, Cole CD, et al. Preventing and treating chronic disorders using the modified vaccination technique. Front Biosci (Landmark Ed). 2009;14:3892‒3898.
  66. Alber DG, Killington RA, Stokes A. Solid matrix-antibody-antigen complexes incorporating equine herpesvirus 1 glycoproteins C and D elicit anti-viral immune responses in BALB/c (H-2K(d)) and C3H (H-2K(k)) mice. Vaccine. 2000;19(7‒8):895‒901.
  67. Jeurissen SH, Janse EM, Lehrbach PR, et al. The working mechanism of an immune complex vaccine that protects chickens against infectious bursal disease. Immunology. 1998;95(3):494‒500.
  68. Iván J, Velhner M, Ursu K, et al. Delayed vaccine virus replication in chickens vaccinated subcutaneously with an immune complex infectious bursal disease vaccine: quantification of vaccine virus by real-time polymerase chain reaction. Can J Vet Res. 2005;69(2):135‒142.
  69. Ignjatovic J, Gould G, Trinidad L, et al. Chicken recombinant antibodies against infectious bursal disease virus are able to form antibody-virus immune complex. Avian Pathol . 2006;35(4):293‒301.
  70. Pokrić B, Sladić D, Juros S, et al. Application of the immune complex for immune protection against viral disease. Vaccine. 1993;11(6):655‒659.
  71. Guo ZY, Giambrone JJ, Wu H, et al. Safety and efficacy of an experimental reovirus vaccine for in ovo administration. Avian Dis. 2003; 47(4):1423‒1428.
  72. Schat KA, Martins NR, O'Connell PH, et al. Immune complex vaccines for chicken infectious anemia virus. Avian Dis . 2011;55 (1):90‒96.
  73. Tsouchnikas G, Zlatkovic J, Jarmer J, et al. Immunization with Immune Complexes Modulates the Fine Specificity of Antibody Responses to a Flavivirus Antigen. J Virol . 2015;89(15):7970‒7978.
  74. Houston WE, Kremer RJ, Crabbs CL, et al. Inactivated Venezuelan equine encephalomyelitis virus vaccine complexed with specific antibody: enhanced primary immune response and altered pattern of antibody class elicited. J Infect Dis . 1977;135(4):600‒610.
  75. Rawool DB, Bitsaktsis C, Li Y, et al. Utilization of Fc receptors as a mucosal vaccine strategy against an intracellular bacterium, Francisella tularensis. J Immunol. 2008;180(8):5548‒5557.
  76. Brady LJ, van Tilburg ML, Alford CE, et al. Monoclonal antibody-mediated modulation of the humoral immune response against mucosally applied Streptococcus mutans. Infect Immun. 2000;68(4):1796‒1805.
  77. Brady LJ. Antibody-mediated immunomodulation: a strategy to improve host responses against microbial antigens. Infect Immun. 2005;73(2):671‒678.
  78. Ricks CA, Avakian A, Bryan T, et al. In ovo vaccination technology. Adv Vet Med. 1999;41:495‒515.
  79. Yoshida K, van den Berg TK, Dijkstra CD. Two different mechanisms of immune-complex trapping in the mouse spleen during immune responses. Adv Exp Med Biol. 1993;329:377‒382.
  80. Yoshida K, van den Berg TK, Dijkstra CD. Two functionally different follicular dendritic cells in secondary lymphoid follicles of mouse spleen, as revealed by CR1/2 and FcR gamma II-mediated immune-complex trapping. Immunology. 1993;80(1):34‒39.
  81. Espenes A, Press CM, Dannevig BH, et al. Immune-complex trapping in the splenic ellipsoids of rainbow trout (Oncorhynchus mykiss). Cell Tissue Res. 1995;282(1):41‒48.
  82. Fridman WH. Regulation of B-cell activation and antigen presentation by Fc receptors. Curr Opin Immunol. 1993;5(3):355‒360.
  83. Schuurhuis DH, Facsinay iA, Nagelkerken B, et al. Antigen-antibody immune complexes empower dendritic cells to efficiently prime specific CD8+ CTL responses in vivo. J Immunol. 2002;168(5):2240‒2246.
  84. Dulak J, Bauer J, Döpp EA, Van Rooijen N. Immune-complex-trapping cells in the spleen of the oriental fire-bellied toad, Bombina orientalis. Cell Tissue Res . 1993;271(1):155‒158.
  85. Laman JD, Kors N, Van Rooijen N, et al. Mechanism of follicular trapping: localization of immune complexes and cell remnants after elimination and repopulation of different spleen cell populations. Immunology. 1990;71(1):57‒62.
  86. Eikelenboom P, Kroese FG, van Rooijen N. Immune complex-trapping cells in the spleen of the chicken. Enzyme histochemical and ultrastructural aspects. Cell Tissue Res . 1983;231(2):377‒386.
  87. Tobar JA, González PA, Kalergis AM. Salmonella escape from antigen presentation can be overcome by targeting bacteria to Fc gamma receptors on dendritic cells. J Immunol. 2004;173(6):4058‒4065.
  88. Aydar Y, Sukumar S, Szakal AK, et al. The influence of immune complex-bearing follicular dendritic cells on the IgM response, Ig class switching, and production of high affinity IgG. J Immunol. 2005;174(9):5358‒5366.
  89. Perkins KA, Chain BM. Presentation by peritoneal macrophages: modulation by antibody-antigen complexes. Immunology. 1986;58(1): 15‒21.
  90. Manca F1, Fenoglio D, Kunkl A, et al. Differential activation of T cell clones stimulated by macrophages exposed to antigen complexed with monoclonal antibodies. A possible influence of paratope specificity on the mode of antigen processing. J Immunol. 1988;140(9):2893‒2898.
  91. Kunkl A, Klaus GG. The generation of memory cells. IV. Immunization with antigen-antibody complexes accelerates the development of B-memory cells, the formation of germinal centres and the maturation of antibody affinity in the secondary response. Immunology. 1981; 43(2):371‒378.
  92. León B, Ballesteros TA, Randall TD, et al. Prolonged antigen presentation by immune complex-binding dendritic cells programs the proliferative capacity of memory CD8 T cells. J Exp Med. 2014;211(8):1637‒1655.
  93. Iglesias BV, Bitsaktsis C, Pham G, et al. Multiple mechanisms mediate enhanced immunity generated by mAb-inactivated F. tularensis immunogen. Immunol Cell Biol. 2013;91(2):139‒148.
  94. de Jong JM, Schuurhuis DH, Facsinay IA, et al. Murine Fc receptors for IgG are redundant in facilitating presentation of immune complex derived antigen to CD8+ T cells in vivo. Mol Immunol. 2006; 43(13): 2045‒2050.
  95. Osato K. Antigen-antibody complexes in the immune response. I. Analysis of the effectiveness of complexes on the primary antibody response. Immunology. 1972;23(4):545‒557.
  96. Gosselin EJ, Wardwell K, Gosselin DR, et al. Enhanced antigen presentation using human Fc gamma receptor (monocyte/macrophage)-specific immunogens. J Immunol . 1992;149(11):3477‒3481.
  97. Benkirane MM, Bon D, Cordeil M, et al. Immunization with immune complexes: characterization of monoclonal antibodies against a TSH-antibody complex. Mol Immunol . 1987;24(12):1309‒1315.
  98. Amigorena S, Bonnerot C. Fc receptors for IgG and antigen presentation on MHC class I and class II molecules. Semin Immunol. 1999;11(6):385‒390.
  99. Galesić MS, Marusić M, Pokrić B. Cellular immune response to the antigen administered as an immune complex in vivo. Immunology. 1992;75(2):325‒329.
  100. Kawamura H, Berzofsky JA. Enhancement of antigenic potency in vitro and immunogenicity in vivo by coupling the antigen to anti-immunoglobulin. J Immunol . 1986;136(1):58‒65.
  101. Motal AUM, Wigglesworth K, Galili U . Mechanism for increased immunogenicity of vaccines that form in vivo immune complexes with the natural anti-Gal antibody. Vaccine. 2009;27(23):3072‒3082.
  102. Nie X, Basu S, Cerny J. Immunization with immune complex alters the repertoire of antigen-reactive B cells in the germinal centers. Eur J Immunol . 1997;27(12):3517‒3525.
  103. Zheng B, Switzer K, Marinova E, et al. Correction of age-associated deficiency in germinal center response by immunization with immune complexes. Clin Immunol . 2007;124(2):131‒137.
  104. Goins CL, Chappell CP, Shashidharamurthy R, et al. Immune complex-mediated enhancement of secondary antibody responses. J Immunol. 2010;184(11):6293‒6298.
  105. Berger S, Balló H, Stutte HJ. Distinct antigen-induced cytokine pattern upon stimulation with antibody-complexed antigen consistent with a Th1-->Th2-shift. Res Virol . 1996;147(2‒3):103‒108.
  106. Berger S, Balló H, Stutte HJ. Immune complex-induced interleukin-6, interleukin-10 and prostaglandin secretion by human monocytes: a network of pro- and anti-inflammatory cytokines dependent on the antigen:antibody ratio. Eur J Immunol. 1996;26(6):1297‒1301.
  107. Berger S, Chandra R, Balló H, et al. Immune complexes are potent inhibitors of interleukin-12 secretion by human monocytes. Eur J Immunol. 1997;27(11): 2994‒3000.
  108. Berger S, Balló H, Stutte HJ. Immune complexes induce the secretion of Th2 cytokines in human monocytes. Verh Dtsch Ges Pathol. 1996;80:288‒292.
  109. Amit AG, Mariuzza RA, Phillips SE, et al. Three-dimensional structure of an antigen-antibody complex at 2.8 A resolution. Science. 1986;233(4765):747‒753.
  110. Amit AG, Mariuzza RA, Phillips SE, et al. Three-dimensional structure of an antigen-antibody complex at 6 A resolution. Nature. 1985;313(5998):156‒158.
  111. Amit AG, Boulot G, Comarmond MB, et al. X-ray diffraction studies of an anti-azophenylarsonate antibody and of an antigen-antibody complex. Ann Inst Pasteur Immunol . 1985;136C(1):121‒129.
  112. Nimmerjahn F, Ravetch JV. Fc-receptors as regulators of immunity. Adv Immunol . 2007;96:179‒204.
Creative Commons Attribution License

©2016 Haddad. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.