Neutralizing antibody

Last updated

Neutralizing antibodies
Antibody.svg
Standard antibody representation
Properties
Protein TypeImmunoglobin
FunctionNeutralization of antigens
ProductionB cells [1] [2]

A neutralizing antibody (NAb) is an antibody that defends a cell from a pathogen or infectious particle by neutralizing any effect it has biologically. Neutralization renders the particle no longer infectious or pathogenic. [3] [4] Neutralizing antibodies are part of the humoral response of the adaptive immune system against viruses, intracellular bacteria and microbial toxin. By binding specifically to surface structures (antigen) on an infectious particle, neutralizing antibodies prevent the particle from interacting with its host cells it might infect and destroy.

Contents

Mechanism

Covering a pathogen's antigen in antibodies make the pathogen less infectious and less pathogenic. In the image on the right, virus entry to the cell is prevented by neutralizing antibodies binding to the virus. NAb esquema.jpg
Covering a pathogen's antigen in antibodies make the pathogen less infectious and less pathogenic. In the image on the right, virus entry to the cell is prevented by neutralizing antibodies binding to the virus.

In order to enter cells, pathogens, such as circulating viral particles or extracellular bacteria, use molecules on their surfaces to interact with the cell surface receptors of their target cell which allows them to enter the cell and start their replication cycle. [5] Neutralizing antibodies can inhibit infectivity by binding to the pathogen and blocking the molecules needed for cell entry. This can be due to the antibodies statically interfering with the pathogens, or toxins attaching to host cell receptors. In case of a viral infection, NAbs can bind to glycoproteins of enveloped viruses or capsid proteins of non-enveloped viruses. Furthermore, neutralizing antibodies can act by preventing particles from undergoing structural changes often needed for successful cell entry. For example, neutralizing antibodies can prevent conformational changes of viral proteins that mediate the membrane fusion needed for entry into the host cell. [5] In some cases, the virus is unable to infect even after the antibody dissociates. The pathogen-antibody complex is eventually taken up and degraded by macrophages. [6]

Neutralizing antibodies are also important in neutralizing the toxic effects of bacterial toxins. An example of a neutralizing antibody is diphtheria antitoxin, which can neutralize the biological effects of diphtheria toxin. [7] Neutralizing antibodies are not effective against extracellular bacteria, as the binding of antibodies does not prevent bacteria from replicating. Here, the immune system uses other functions of antibodies, like opsonisation and complement activation, to kill the bacteria. [8]

Difference between neutralizing antibodies and binding antibodies

Not all antibodies that bind to a pathogenic particle are neutralizing. Non-neutralizing antibodies, or binding antibodies, bind specifically to the pathogen, but do not interfere with their infectivity. That might be because they do not bind to the right region. Non-neutralizing antibodies can be important to flag the particle for immune cells, signaling that it has been targeted, after which the particle is processed and consequently destroyed by recruited immune cells. [9] Neutralizing antibodies on the other hand can neutralize the biological effects of the antigen without a need for immune cells. In some cases, non-neutralizing antibodies, or an insufficient amount of neutralizing antibodies binding to viral particles, can be utilized by some species of virus to facilitate uptake into their host cells. This mechanism is known as antibody-dependent enhancement. [10] It has been observed for Dengue virus and Zika virus. [11]

Production

Antibodies are produced and secreted by B cells. When B cells are produced in the bone marrow, the genes that encode the antibodies undergo random genetic recombination (V(D)J recombination), which results in every mature B cell producing antibodies that differ in their amino acid sequence in the antigen-binding region. Therefore, every B cell produces antibodies that bind specifically to different antigens. [12] A strong diversity in the antibody repertoire allows the immune system to recognize a plethora of pathogens which can come in all different forms and sizes. During an infection only antibodies that bind to the pathogenic antigen with high affinity are produced. This is achieved by clonal selection of a single B cell clone: B cells are recruited to the site of infection by sensing interferons that are released by the infected cells as part of the innate immune response. B cells display B-cell receptors on their cell surface, which is just the antibody anchored to the cell membrane. When the B-cell receptor binds to its cognate antigen with high affinity, an intracellular signalling cascade is triggered. In addition to binding to an antigen, B cells need to be stimulated by cytokines produced by T helper cells as part of the cellular response of the immune system against the pathogen. Once a B cell is fully activated, it rapidly proliferates and differentiates into plasma cells. Plasma cells then secrete the antigen-specific antibody in large quantities. [13] After a first encounter of the antigen by vaccination or natural infection, immunological memory allows for a more rapid production of neutralizing antibodies following the next exposure to the virus.

Virus evasion of neutralizing antibodies

Viruses use a variety of mechanisms to evade neutralizing antibodies. [14]  Viral genomes mutate at a high rate. Mutations that allow viruses to evade a neutralizing antibody will be selected for, and hence prevail. Conversely, antibodies also simultaneously evolve by affinity maturation during the course of an immune response, thereby improving recognition of viral particles. Conserved parts of viral proteins that play a central role in viral function are less likely to evolve over time, and therefore are more vulnerable to antibody binding. However, viruses have evolved certain mechanisms to hinder steric access of an antibody to these regions, making binding difficult. [14] Viruses with a low density of surface structural proteins are more difficult for antibodies to bind to. [14] Some viral glycoproteins are heavily glycosylated by N- and O- linked glycans, creating a so-called glycan shield, which may decrease antibody binding affinity and facilitate evasion of neutralizing antibodies. [14] HIV-1, the cause of human AIDS, uses both of these mechanisms. [15] [16]

Medical uses of neutralizing antibodies

Neutralizing antibodies are used for passive immunisation, and can be used for patients even if they do not have a healthy immune system. In the early 20th century, infected patients were injected with antiserum, which is the blood serum of a previously infected and recovered patient containing polyclonal antibodies against the infectious agent. This showed that antibodies could be used as an effective treatment for viral infections and toxins. [17] Antiserum is a very crude therapy, because antibodies in the plasma are not purified or standardized and the blood plasma could be rejected by the donor. [18] As it relies on the donation from recovered patients it cannot be easily scaled up. However, serum therapy is today still used as the first line of defence during an outbreak as it can relatively quickly obtained. [19] [20] Serum therapy was shown to reduce mortality in patients during the 2009 swine flu pandemic [21] and the Western African Ebola virus epidemic. [22] It is also being tested as possible treatment for COVID-19. [23] [24] Immunoglobulin therapy, which uses a mixture of antibodies obtained from healthy people, is given to immunodeficient or immunosuppressed patients to fight off infections.

For a more specific and robust treatment, purified polyclonal or monoclonal antibodies (mAb) can be used. Polyclonal antibodies are collection of antibodies that target the same pathogen but bind to different epitopes. Polyclonal antibodies are obtained from human donors or animals that have been exposed to the antigen. The antigen injected into the animal donors can be designed in such a way to preferably produce neutralizing antibodies. [25] Polyclonal antibodies have been used as treatment for cytomegalovirus (CMV), hepatitis b virus (HBV), rabies virus, measles virus, and respiratory syncytial virus (RSV). [18] Diphtheria antitoxin contains polyclonal antibodies against the diphtheria toxin. [26] By treating with antibodies binding multiple epitopes, the treatment is still effective even if the virus mutates and one of the epitopes changes in structure. However, because of the nature of the production, treatment with polyclonal antibodies has batch to batch variation and low antibody titers. [25] Monoclonal antibodies, on the other hand, all bind the same epitope with high specificity. They can be produced with the Hybridoma technology, which allows the production of mAbs in large quantities. [17] mAbs against infections stop working when virus mutates the epitope that is targeted by the mAbs or multiple strain are circulating. Example of drugs that use monoclonal antibodies include ZMapp against Ebola [27] and Palivizumab against RSV. [28] Many mABs against other infections are in clinical trials. [17]

Neutralizing antibodies also play a role in active immunisation by vaccination. By understanding the binding sites and structure of neutralizing antibodies in a natural immune response a vaccine can be rationally designed such that it stimulates the immune system to produce neutralizing antibodies and not binding antibodies. [29] [30] Introducing a weakened form of a virus through vaccination allows for the production of neutralizing antibodies by B cells. After a second exposure, the neutralizing antibody response is more rapid due to the existence of memory B cells that produce antibodies specific to the virus. [31] An effective vaccine induces the production of antibodies that are able to neutralize the majority of variants of a virus, although virus mutation resulting in antibody evasion may require vaccines to be updated in response. [31] Some viruses evolve faster than others, which can require the need for vaccines to be updated in response. A well known example is the vaccine for the influenza virus, which must be updated annually to account for the recent circulating strains of the virus. [14]

Neutralizing antibodies may also assist the treatment of multiple sclerosis. [2] Although this type of antibody has the ability to fight retroviral infections, in some cases it attacks pharmaceuticals administered to the body which would otherwise treat multiple sclerosis. Recombinant protein drugs, especially those derived from animals, are commonly targeted by neutralizing antibodies. A few examples are Rebif, Betaseron and Avonex. [2]

Methods for detection and quantification of neutralizing antibodies

Neutralization assays are capable of being performed and measured in different ways, including the use of techniques such as plaque reduction (which compares counts of virus plaques in control wells with those in inoculated cultures), microneutralization (which is performed in microtiter plates filled with small amounts of sera), and colorimetric assays (which depend on biomarkers indicating metabolic inhibition of the virus). [32]

Broadly neutralizing antibodies

Most of the neutralizing antibodies produced by the immune system are very specific for a single virus strain due to affinity maturation by B cells. [13] Some pathogens with high genetic variability, such as HIV, constantly change their surface structure such that neutralizing antibodies with high specificity to the old strain can no longer bind to the new virus strain. This immune evasion strategy prevents the immune system from developing immunological memory against the pathogen. [33] Broadly neutralizing antibodies (bNAbs), on the other hand, have the special ability to bind and neutralize multiple strains of a virus species. [34]

bNAbs have been initially found in HIV patients. [35] However, they are quite rare: an in situ screening study showed that only 1% of all patients develop bNAbs against HIV. [36] bNABs can neutralize a wide range of virus strains by binding to conserved regions of the virus surface proteins that are unable to mutate because they are functionally essential for the virus replication. Most binding sites of bNAbs against HIV are on HIV's exposed surface antigen, the envelope (Env) protein (a trimer composed of gp120 and gp41 subunits). These site include the CD4 binding site or the gp41-gp120 interface. [37] Los Alamos National Laboratory's HIV Databases is a comprehensive resource that has a wealth of information about HIV sequences, bNAbs, and more. [38]

Additionally, bNAbs have been found for other viruses including influenza, [39] hepatitis C, [40] dengue [41] and West Nile virus. [42]

Research

Preliminary research is conducted to identify and test bNAbs against HIV-1. [43] bNAbs are used in research to rationally design vaccines to stimulate production of bNAbs and immunity against viruses. No antigen that triggers bNAb production in animal models or humans is known. [34]

See also

Related Research Articles

<span class="mw-page-title-main">Antigen</span> Molecule triggering an immune response (antibody production) in the host

In immunology, an antigen (Ag) is a molecule, moiety, foreign particulate matter, or an allergen, such as pollen, that can bind to a specific antibody or T-cell receptor. The presence of antigens in the body may trigger an immune response.

<span class="mw-page-title-main">Antiviral drug</span> Medication used to treat a viral infection

Antiviral drugs are a class of medication used for treating viral infections. Most antivirals target specific viruses, while a broad-spectrum antiviral is effective against a wide range of viruses. Antiviral drugs are a class of antimicrobials, a larger group which also includes antibiotic, antifungal and antiparasitic drugs, or antiviral drugs based on monoclonal antibodies. Most antivirals are considered relatively harmless to the host, and therefore can be used to treat infections. They should be distinguished from virucides, which are not medication but deactivate or destroy virus particles, either inside or outside the body. Natural virucides are produced by some plants such as eucalyptus and Australian tea trees.

<span class="mw-page-title-main">HIV vaccine development</span> In-progress vaccinations that may prevent or treat HIV infections

An HIV vaccine is a potential vaccine that could be either a preventive vaccine or a therapeutic vaccine, which means it would either protect individuals from being infected with HIV or treat HIV-infected individuals.

<span class="mw-page-title-main">Respiratory syncytial virus</span> Species of a virus

Respiratory syncytial virus (RSV), also called human respiratory syncytial virus (hRSV) and human orthopneumovirus, is a contagious virus that causes infections of the respiratory tract. It is a negative-sense, single-stranded RNA virus. Its name is derived from the large cells known as syncytia that form when infected cells fuse.

<span class="mw-page-title-main">Seroconversion</span> Development of specific antibodies in the blood serum as a result of infection or immunization

In immunology, seroconversion is the development of specific antibodies in the blood serum as a result of infection or immunization, including vaccination. During infection or immunization, antigens enter the blood, and the immune system begins to produce antibodies in response. Before seroconversion, the antigen itself may or may not be detectable, but the antibody is absent. During seroconversion, the antibody is present but not yet detectable. After seroconversion, the antibody is detectable by standard techniques and remains detectable unless the individual seroreverts, in a phenomenon called seroreversion, or loss of antibody detectability, which can occur due to weakening of the immune system or decreasing antibody concentrations over time. Seroconversion refers the production of specific antibodies against specific antigens, meaning that a single infection could cause multiple waves of seroconversion against different antigens. Similarly, a single antigen could cause multiple waves of seroconversion with different classes of antibodies. For example, most antigens prompt seroconversion for the IgM class of antibodies first, and subsequently the IgG class.

<span class="mw-page-title-main">Adaptive immune system</span> Subsystem of the immune system

The adaptive immune system, also known as the acquired immune system, or specific immune system is a subsystem of the immune system that is composed of specialized, systemic cells and processes that eliminate pathogens or prevent their growth. The acquired immune system is one of the two main immunity strategies found in vertebrates.

<span class="mw-page-title-main">Original antigenic sin</span> Immune phenomenon

Original antigenic sin, also known as antigenic imprinting, the Hoskins effect, immunological imprinting, or primary addiction is the propensity of the immune system to preferentially use immunological memory based on a previous infection when a second slightly different version of that foreign pathogen is encountered. This leaves the immune system "trapped" by the first response it has made to each antigen, and unable to mount potentially more effective responses during subsequent infections. Antibodies or T-cells induced during infections with the first variant of the pathogen are subject to repertoire freeze, a form of original antigenic sin.

<span class="mw-page-title-main">Envelope glycoprotein GP120</span> Glycoprotein exposed on the surface of the HIV virus

Envelope glycoprotein GP120 is a glycoprotein exposed on the surface of the HIV envelope. It was discovered by Professors Tun-Hou Lee and Myron "Max" Essex of the Harvard School of Public Health in 1984. The 120 in its name comes from its molecular weight of 120 kDa. Gp120 is essential for virus entry into cells as it plays a vital role in attachment to specific cell surface receptors. These receptors are DC-SIGN, Heparan Sulfate Proteoglycan and a specific interaction with the CD4 receptor, particularly on helper T-cells. Binding to CD4 induces the start of a cascade of conformational changes in gp120 and gp41 that lead to the fusion of the viral membrane with the host cell membrane. Binding to CD4 is mainly electrostatic although there are van der Waals interactions and hydrogen bonds.

<span class="mw-page-title-main">Antibody-dependent cellular cytotoxicity</span> Cell-mediated killing of other cells mediated by antibodies

Antibody-dependent cellular cytotoxicity (ADCC), also referred to as antibody-dependent cell-mediated cytotoxicity, is a mechanism of cell-mediated immune defense whereby an effector cell of the immune system kills a target cell, whose membrane-surface antigens have been bound by specific antibodies. It is one of the mechanisms through which antibodies, as part of the humoral immune response, can act to limit and contain infection.

<span class="mw-page-title-main">Gp41</span> Subunit of the envelope protein complex of retroviruses

Gp41 also known as glycoprotein 41 is a subunit of the envelope protein complex of retroviruses, including human immunodeficiency virus (HIV). Gp41 is a transmembrane protein that contains several sites within its ectodomain that are required for infection of host cells. As a result of its importance in host cell infection, it has also received much attention as a potential target for HIV vaccines.

Antigenic variation or antigenic alteration refers to the mechanism by which an infectious agent such as a protozoan, bacterium or virus alters the proteins or carbohydrates on its surface and thus avoids a host immune response, making it one of the mechanisms of antigenic escape. It is related to phase variation. Antigenic variation not only enables the pathogen to avoid the immune response in its current host, but also allows re-infection of previously infected hosts. Immunity to re-infection is based on recognition of the antigens carried by the pathogen, which are "remembered" by the acquired immune response. If the pathogen's dominant antigen can be altered, the pathogen can then evade the host's acquired immune system. Antigenic variation can occur by altering a variety of surface molecules including proteins and carbohydrates. Antigenic variation can result from gene conversion, site-specific DNA inversions, hypermutation, or recombination of sequence cassettes. The result is that even a clonal population of pathogens expresses a heterogeneous phenotype. Many of the proteins known to show antigenic or phase variation are related to virulence.

Entry inhibitors, also known as fusion inhibitors, are a class of antiviral drugs that prevent a virus from entering a cell, for example, by blocking a receptor. Entry inhibitors are used to treat conditions such as HIV and hepatitis D.

A breakthrough infection is a case of illness in which a vaccinated individual becomes infected with the illness, because the vaccine has failed to provide complete immunity against the pathogen. Breakthrough infections have been identified in individuals immunized against a variety of diseases including mumps, varicella (Chickenpox), influenza, and COVID-19. The characteristics of the breakthrough infection are dependent on the virus itself. Often, infection of the vaccinated individual results in milder symptoms and shorter duration than if the infection were contracted naturally.

<span class="mw-page-title-main">Polyclonal B cell response</span> Immune response by adaptive immune system

Polyclonal B cell response is a natural mode of immune response exhibited by the adaptive immune system of mammals. It ensures that a single antigen is recognized and attacked through its overlapping parts, called epitopes, by multiple clones of B cell.

<span class="mw-page-title-main">Antibody-dependent enhancement</span> Antibodies rarely making an infection worse instead of better

Antibody-dependent enhancement (ADE), sometimes less precisely called immune enhancement or disease enhancement, is a phenomenon in which binding of a virus to suboptimal antibodies enhances its entry into host cells, followed by its replication. The suboptimal antibodies can result from natural infection or from vaccination. ADE may cause enhanced respiratory disease, but is not limited to respiratory disease. It has been observed in HIV, RSV virus and Dengue virus and is monitored for in vaccine development.

CD4 immunoadhesin is a recombinant fusion protein consisting of a combination of CD4 and the fragment crystallizable region, similarly known as immunoglobulin. It belongs to the antibody (Ig) gene family. CD4 is a surface receptor for human immunodeficiency virus (HIV). The CD4 immunoadhesin molecular fusion allow the protein to possess key functions from each independent subunit. The CD4 specific properties include the gp120-binding and HIV-blocking capabilities. Properties specific to immunoglobulin are the long plasma half-life and Fc receptor binding. The properties of the protein means that it has potential to be used in AIDS therapy as of 2017. Specifically, CD4 immunoadhesin plays a role in antibody-dependent cell-mediated cytotoxicity (ADCC) towards HIV-infected cells. While natural anti-gp120 antibodies exhibit a response towards uninfected CD4-expressing cells that have a soluble gp120 bound to the CD4 on the cell surface, CD4 immunoadhesin, however, will not exhibit a response. One of the most relevant of these possibilities is its ability to cross the placenta.

Long-term nonprogressors (LTNPs), are individuals infected with HIV, who maintain a CD4 count greater than 500 without antiretroviral therapy with a detectable viral load. Many of these patients have been HIV positive for 30 years without progressing to the point of needing to take medication in order not to develop AIDS. They have been the subject of a great deal of research, since an understanding of their ability to control HIV infection may lead to the development of immune therapies or a therapeutic vaccine. The classification "Long-term non-progressor" is not permanent, because some patients in this category have gone on to develop AIDS.

Broadly neutralizing HIV-1 antibodies (bNAbs) are neutralizing antibodies which neutralize multiple HIV-1 viral strains. bNAbs are unique in that they target conserved epitopes of the virus, meaning the virus may mutate, but the targeted epitopes will still exist. In contrast, non-bNAbs are specific for individual viral strains with unique epitopes. The discovery of bNAbs has led to an important area of research, namely, discovery of a vaccine, not only limited to HIV, but also other rapidly mutating viruses like influenza.

Intrastructural help (ISH) is where T and B cells cooperate to help or suppress an immune response gene. ISH has proven effective for the treatment of influenza, rabies related lyssavirus, hepatitis B, and the HIV virus. This process was used in 1979 to observe that T cells specific to the influenza virus could promote the stimulation of hemagglutinin specific B cells and elicit an effective humoral immune response. It was later applied to the lyssavirus and was shown to protect raccoons from lethal challenge. The ISH principle is especially beneficial because relatively invariable structural antigens can be used for the priming of T-cells to induce humoral immune response against variable surface antigens. Thus, the approach has also transferred well for the treatment of hepatitis B and HIV.

Passive antibody therapy, also called serum therapy, is a subtype of passive immunotherapy that administers antibodies to target and kill pathogens or cancer cells. It is designed to draw support from foreign antibodies that are donated from a person, extracted from animals, or made in the laboratory to elicit an immune response instead of relying on the innate immune system to fight disease. It has a long history from the 18th century for treating infectious diseases and is now a common cancer treatment. The mechanism of actions include: antagonistic and agonistic reaction, complement-dependent cytotoxicity (CDC), and antibody-dependent cellular cytotoxicity (ADCC).

References

  1. Mike Recher; Karl S Lang; Lukas Hunziker; Stefan Freigang; Bruno Eschli; Nicola L Harris; Alexander Navarini; Beatrice M Senn; Katja Fink; Marius Lötscher; Lars Hangartner; Raphaël Zellweger; Martin Hersberger; Alexandre Theocharides; Hans Hengartner; Rolf M Zinkernagel (8 August 2004). "Deliberate removal of T cell help improves virus-neutralizing antibody production". Nature Immunology. 5 (9): 934–942. doi:10.1038/ni1102. PMID   15300247. S2CID   1351951.
  2. 1 2 3 Stachowiak, Julie (15 August 2008). "Neutralizing Antibodies and Disease-Modifying Therapies for Multiple Sclerosis". About.com. Retrieved 13 June 2009.
  3. "Neutralizing antibody". Biology-Online. 2008. Retrieved 4 July 2009.
  4. Klasse, P. J. (9 September 2014). "Neutralization of Virus Infectivity by Antibodies: Old Problems in New Perspectives". Advances in Biology. 2014: 1–24. doi: 10.1155/2014/157895 . PMC   4835181 . PMID   27099867.
  5. 1 2 Principles of Virology, Volume 1: Molecular Biology (4th ed.). ASM Press. 2015. p. 31. ISBN   978-1555819330.
  6. Principles of Virology, Volume 2: Pathogenesis and Control (4th ed.). ASM Press. 2015. p. 125. ISBN   978-1-555-81951-4.
  7. Treffers, Henry P. (2014). "Neutralizing antibody". AccessScience. McGraw-Hill. doi:10.1036/1097-8542.450600.
  8. Janeway's immunobiology (8th ed.). Garland Science. 2012. p. 388. ISBN   978-0-8153-4243-4.
  9. Schmaljohn, AL (July 2013). "Protective antiviral antibodies that lack neutralizing activity: precedents and evolution of concepts". Current HIV Research. 11 (5): 345–53. doi:10.2174/1570162x113116660057. PMID   24191933.
  10. Tirado, SM; Yoon, KJ (2003). "Antibody-dependent enhancement of virus infection and disease". Viral Immunology. 16 (1): 69–86. doi:10.1089/088282403763635465. PMID   12725690.
  11. Dejnirattisai, Wanwisa; Supasa, Piyada; Wongwiwat, Wiyada; Rouvinski, Alexander; Barba-Spaeth, Giovanna; Duangchinda, Thaneeya; Sakuntabhai, Anavaj; Cao-Lormeau, Van-Mai; Malasit, Prida; Rey, Felix A; Mongkolsapaya, Juthathip; Screaton, Gavin R (23 June 2016). "Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus". Nature Immunology. 17 (9): 1102–1108. doi:10.1038/ni.3515. PMC   4994874 . PMID   27339099.
  12. Jung, David; Alt, Frederick W (January 2004). "Unraveling V(D)J Recombination". Cell. 116 (2): 299–311. doi: 10.1016/S0092-8674(04)00039-X . PMID   14744439.
  13. 1 2 Janeway's immunobiology (8th ed.). Garland Science. 2012. pp. 389–404. ISBN   978-0-8153-4243-4.
  14. 1 2 3 4 5 VanBlargan, Laura A.; Goo, Leslie; Pierson, Theodore C. (2016). "Deconstructing the Antiviral Neutralizing-Antibody Response: Implications for Vaccine Development and Immunity". Microbiology and Molecular Biology Reviews. 80 (4): 989–1010. doi:10.1128/MMBR.00024-15. ISSN   1092-2172. PMC   5116878 . PMID   27784796.
  15. Crispin, Max; Ward, Andrew B.; Wilson, Ian A. (20 May 2018). "Structure and Immune Recognition of the HIV Glycan Shield". Annual Review of Biophysics. 47 (1): 499–523. doi:10.1146/annurev-biophys-060414-034156. ISSN   1936-122X. PMC   6163090 . PMID   29595997.
  16. Guha, Debjani; Ayyavoo, Velpandi (2013). "Innate Immune Evasion Strategies by Human Immunodeficiency Virus Type 1". ISRN AIDS. 2013: 954806. doi: 10.1155/2013/954806 . ISSN   2090-939X. PMC   3767209 . PMID   24052891.
  17. 1 2 3 Salazar, Georgina; Zhang, Ningyan; Fu, Tong-Ming; An, Zhiqiang (10 July 2017). "Antibody therapies for the prevention and treatment of viral infections". npj Vaccines. 2 (1): 19. doi: 10.1038/s41541-017-0019-3 . PMC   5627241 . PMID   29263875.
  18. 1 2 Casadevall, A; Dadachova, E; Pirofski, LA (September 2004). "Passive antibody therapy for infectious diseases". Nature Reviews. Microbiology. 2 (9): 695–703. doi: 10.1038/nrmicro974 . PMID   15372080.
  19. Kreil, Thomas R. (March 2015). "Treatment of Ebola Virus Infection with Antibodies from Reconvalescent Donors". Emerging Infectious Diseases. 21 (3): 521–523. doi: 10.3201/eid2103.141838 . PMC   4344290 . PMID   25695274.
  20. Schmidt, Rebecca; Beltzig, Lea C.; Sawatsky, Bevan; Dolnik, Olga; Dietzel, Erik; Krähling, Verena; Volz, Asisa; Sutter, Gerd; Becker, Stephan; von Messling, Veronika (5 October 2018). "Generation of therapeutic antisera for emerging viral infections". npj Vaccines. 3 (1): 42. doi: 10.1038/s41541-018-0082-4 . PMC   6173733 . PMID   30323953.
  21. Hung, I. F.; To, K. K.; Lee, C.-K.; Lee, K.-L.; Chan, K.; Yan, W.-W.; Liu, R.; Watt, C.-L.; Chan, W.-M.; Lai, K.-Y.; Koo, C.-K.; Buckley, T.; Chow, F.-L.; Wong, K.-K.; Chan, H.-S.; Ching, C.-K.; Tang, B. S.; Lau, C. C.; Li, I. W.; Liu, S.-H.; Chan, K.-H.; Lin, C.-K.; Yuen, K.-Y. (19 January 2011). "Convalescent Plasma Treatment Reduced Mortality in Patients With Severe Pandemic Influenza A (H1N1) 2009 Virus Infection". Clinical Infectious Diseases. 52 (4): 447–456. doi: 10.1093/cid/ciq106 . PMC   7531589 . PMID   21248066.
  22. World Health Organization. "WHO | Use of convalescent whole blood or plasma collected from patients recovered from Ebola virus disease". WHO. Retrieved 5 April 2020.
  23. Shen, Chenguang; Wang, Zhaoqin; Zhao, Fang; Yang, Yang; Li, Jinxiu; Yuan, Jing; Wang, Fuxiang; Li, Delin; Yang, Minghui; Xing, Li; Wei, Jinli; Xiao, Haixia; Yang, Yan; Qu, Jiuxin; Qing, Ling; Chen, Li; Xu, Zhixiang; Peng, Ling; Li, Yanjie; Zheng, Haixia; Chen, Feng; Huang, Kun; Jiang, Yujing; Liu, Dongjing; Zhang, Zheng; Liu, Yingxia; Liu, Lei (27 March 2020). "Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma". JAMA. 323 (16): 1582–1589. doi: 10.1001/jama.2020.4783 . PMC   7101507 . PMID   32219428.
  24. Casadevall, Arturo; Pirofski, Liise-anne (13 March 2020). "The convalescent sera option for containing COVID-19". Journal of Clinical Investigation. 130 (4): 1545–1548. doi: 10.1172/JCI138003 . PMC   7108922 . PMID   32167489.
  25. 1 2 Bregenholt, S; Jensen, A; Lantto, J; Hyldig, S; Haurum, JS (2006). "Recombinant human polyclonal antibodies: A new class of therapeutic antibodies against viral infections". Current Pharmaceutical Design. 12 (16): 2007–15. doi:10.2174/138161206777442173. PMID   16787244.
  26. "Our Formulary". Infectious Diseases Laboratories. Centers for Disease Control and Prevention. Archived from the original on 16 December 2016. Retrieved 9 December 2016.
  27. Multi-National PREVAIL II Study Team, Davey RT Jr, Dodd L, Proschan MA, Neaton J, Neuhaus Nordwall J, Koopmeiners JS, Beigel J, Tierney J, Lane HC, Fauci AS, Massaquoi MBF, Sahr F, Malvy D, et al. (PREVAIL II Writing Group) (13 October 2016). "A Randomized, Controlled Trial of ZMapp for Ebola Virus Infection". New England Journal of Medicine. 375 (15): 1448–1456. doi: 10.1056/NEJMoa1604330 . PMC   5086427 . PMID   27732819.
  28. "Label - Palivizumab (Synagis), Medimmune, Incorporated" (PDF). Retrieved 4 February 2020.
  29. VanBlargan, Laura A.; Goo, Leslie; Pierson, Theodore C. (26 October 2016). "Deconstructing the Antiviral Neutralizing-Antibody Response: Implications for Vaccine Development and Immunity". Microbiology and Molecular Biology Reviews. 80 (4): 989–1010. doi: 10.1128/MMBR.00024-15 . PMC   5116878 . PMID   27784796.
  30. Kwong, P. D.; Mascola, J. R.; Nabel, G. J. (1 September 2011). "Rational Design of Vaccines to Elicit Broadly Neutralizing Antibodies to HIV-1". Cold Spring Harbor Perspectives in Medicine. 1 (1): a007278. doi: 10.1101/cshperspect.a007278 . PMC   3234457 . PMID   22229123.
  31. 1 2 Burton, Dennis R. (2002). "Antibodies, viruses and vaccines". Nature Reviews Immunology. 2 (9): 706–713. doi:10.1038/nri891. ISSN   1474-1733. PMID   12209139. S2CID   9376285.
  32. Kaslow, R. A.; Stanberry, L.R.; Le Duc, J. W., eds. (2014). Viral Infections of Humans: Epidemiology and Control (5th ed.). Springer. p. 56. ISBN   9781489974488 . Retrieved 4 April 2020.
  33. Santoro, MM; Perno, CF (2013). "HIV-1 Genetic Variability and Clinical Implications". ISRN Microbiology. 2013: 481314. doi: 10.1155/2013/481314 . PMC   3703378 . PMID   23844315.
  34. 1 2 Kumar, R; Qureshi, H; Deshpande, S; Bhattacharya, J (August 2018). "Broadly neutralizing antibodies in HIV-1 treatment and prevention". Therapeutic Advances in Vaccines and Immunotherapy. 6 (4): 61–68. doi: 10.1177/2515135518800689 . PMC   6187420 . PMID   30345419.
  35. Cohen, J. (2013). "Bound for Glory". Science. 341 (6151): 1168–1171. Bibcode:2013Sci...341.1168C. doi:10.1126/science.341.6151.1168. PMID   24030996.
  36. Simek, MD; Rida, W; Priddy, FH; Pung, P; Carrow, E; Laufer, DS; Lehrman, JK; Boaz, M; Tarragona-Fiol, T; Miiro, G; Birungi, J; Pozniak, A; McPhee, DA; Manigart, O; Karita, E; Inwoley, A; Jaoko, W; Dehovitz, J; Bekker, LG; Pitisuttithum, P; Paris, R; Walker, LM; Poignard, P; Wrin, T; Fast, PE; Burton, DR; Koff, WC (July 2009). "Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm". Journal of Virology. 83 (14): 7337–48. doi: 10.1128/JVI.00110-09 . PMC   2704778 . PMID   19439467.
  37. Haynes, Barton F.; Burton, Dennis R.; Mascola, John R. (30 October 2019). "Multiple roles for HIV broadly neutralizing antibodies". Science Translational Medicine. 11 (516): eaaz2686. doi: 10.1126/scitranslmed.aaz2686 . PMC   7171597 . PMID   31666399.
  38. "HIV Databases". Los Alamos National Laboratory.
  39. Corti, D; Cameroni, E; Guarino, B; Kallewaard, NL; Zhu, Q; Lanzavecchia, A (June 2017). "Tackling influenza with broadly neutralizing antibodies". Current Opinion in Virology. 24: 60–69. doi:10.1016/j.coviro.2017.03.002. PMC   7102826 . PMID   28527859.
  40. Colbert, MD; Flyak, AI; Ogega, CO; Kinchen, VJ; Massaccesi, G; Hernandez, M; Davidson, E; Doranz, BJ; Cox, AL; Crowe JE, Jr; Bailey, JR (15 July 2019). "Broadly Neutralizing Antibodies Targeting New Sites of Vulnerability in Hepatitis C Virus E1E2". Journal of Virology. 93 (14). doi: 10.1128/JVI.02070-18 . PMC   6600205 . PMID   31068427.
  41. Durham, ND; Agrawal, A; Waltari, E; Croote, D; Zanini, F; Fouch, M; Davidson, E; Smith, O; Carabajal, E; Pak, JE; Doranz, BJ; Robinson, M; Sanz, AM; Albornoz, LL; Rosso, F; Einav, S; Quake, SR; McCutcheon, KM; Goo, L (10 December 2019). "Broadly neutralizing human antibodies against dengue virus identified by single B cell transcriptomics". eLife. 8. doi: 10.7554/eLife.52384 . PMC   6927745 . PMID   31820734.
  42. Goo, L; Debbink, K; Kose, N; Sapparapu, G; Doyle, MP; Wessel, AW; Richner, JM; Burgomaster, KE; Larman, BC; Dowd, KA; Diamond, MS; Crowe JE, Jr; Pierson, TC (January 2019). "A protective human monoclonal antibody targeting the West Nile virus E protein preferentially recognizes mature virions". Nature Microbiology. 4 (1): 71–77. doi:10.1038/s41564-018-0283-7. PMC   6435290 . PMID   30455471.
  43. Bhiman, Jinal N.; Lynch, Rebecca M. (27 March 2017). "Broadly neutralizing antibodies as treatment: effects on virus and immune system". Current HIV/AIDS Reports. 14 (2): 54–62. doi:10.1007/s11904-017-0352-1. ISSN   1548-3568. PMC   5401706 . PMID   28349376.