Antibody-dependent cellular cytotoxicity

Last updated
Antibody-dependent cellular cytotoxicity Antibody-dependent Cellular Cytotoxicity.svg
Antibody-dependent cellular cytotoxicity

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. [1] It is one of the mechanisms through which antibodies, as part of the humoral immune response, can act to limit and contain infection. [2]

Contents

ADCC is independent of the immune complement system that also lyses targets but does not require any other cell. ADCC requires an effector cell which classically is known to be natural killer (NK) cells that typically interact with immunoglobulin G (IgG) antibodies. [3] However, macrophages, neutrophils and eosinophils can also mediate ADCC, such as eosinophils killing certain parasitic worms known as helminths via IgE antibodies. [4]

In general, ADCC has typically been described as the immune response to antibody-coated cells leading ultimately to the lysing of the infected or non-host cell. In recent literature, its importance in regards to treatment of cancerous cells and deeper insight into its deceptively complex pathways have been topics of increasing interest to medical researchers.

NK cells

The typical ADCC involves activation of NK cells by antibodies in a multi-tiered progression of immune control. [5] A NK cell expresses Fcγ receptors. These receptors recognize and bind to the reciprocal portion of an antibody, such as IgG, which binds to the surface of a pathogen-infected target cell. The most common of these Fc receptors on the surface of an NK cell is CD16 or FcγRIII. Once the Fc receptor binds to the Fc region of the antibody, the NK cell releases cytotoxic factors that cause the death of the target cell.

During replication of a virus, some of the viral proteins are expressed on the cell surface membrane of the infected cell. Antibodies can then bind to these viral proteins. Next, the NK cells which have reciprocal Fcγ receptors will bind to that antibody, inducing the NK cell to release proteins such as perforin and proteases known as granzymes, which causes the lysis of the infected cell to hinder the spread of the virus.

Eosinophils

Large parasites like helminths are too big to be engulfed and killed by phagocytosis. They also have an external structure or integument that is resistant to attack by substances released by neutrophils and macrophages. After IgE coat these parasites, the Fc receptor (FcɛRI) of an eosinophil will recognize IgE. Subsequently, interaction between FcεRI and the Fc portion of helminth-bound IgE signals the eosinophil to degranulate.

In vitro assays

Several laboratory methods exist for determining the efficacy of antibodies or effector cells in eliciting ADCC. Usually, a target cell line expressing a certain surface-exposed antigen is incubated with antibody specific for that antigen. After washing, effector cells expressing Fc receptor CD16 are co-incubated with the antibody-labelled target cells. Effector cells are typically PBMCs (peripheral blood mononuclear cell), of which a small percentage are NK cells (natural killer cell); less often they are purified NK cells themselves. Over the course of a few hours a complex forms between the antibody, target cell, and effector cell which leads to lysis of the cell membrane of the target. If the target cell was pre-loaded with a label of some sort, that label is released in proportion to the amount of cell lysis. Cytotoxicity can be quantified by measuring the amount of label in solution compared to the amount of label that remains within healthy, intact cells.

The classical method of detecting this is the chromium-51 [51Cr] release assay; the sulfur-35 [35S] release assay is a little used radioisotope-based alternative. Target cell lysis is determined by measuring the amount of radiolabel released into the cell culture medium by means of a gamma counter or scintillation counter. A variety of non-radioactive methods are now in widespread use. Fluorescence-based methods include such things as direct labelling with a fluorescent dye like calcein or labelling with europium that becomes fluorescent when released Eu3+ binds to a chelator. Fluorescence can be measured by means of multi-well fluorometers or by flow cytometry methods. There are also enzymatic-based assays in which the contents of the lysed cells includes cellular enzymes like GAPDH that remain active; supplying a substrate for that enzyme can catalyze a reaction whose product can be detected by luminescence or by absorbance.

Medical applications

NK cells are involved in killing tumor cells and other cells that may lack MHC I on their surface, indicating a non-self cell. NK cells have been shown to behave similarly to memory cells due to their ability to react to destroy non-host cells only after interacting with a host cell. As NK cells are not themselves specific to certain pathways of immune control, they are utilized a majority of the time in ADCC as a less discriminate cell destroyer than antibody-specific apoptosis mechanisms. The ability of activated ex vivo NK cells has been a topic of interest for the treatment of tumors. After early clinical trials involving activation through cytokines produced poor results and severe toxicological side effects, more recent studies have produced success in regulating metastatic tumors using interleukin proteins to activate the NK cell. [6]

The effects against solid tumors of trastuzumab and rituximab monoclonal antibodies have been shown in experiments with mice to involve ADCC as an important mechanism of therapeutic action. [7] In the clinic, the FcgRIII 158V/F polymorphism interfere with the ability to generate ADCC responses in vitro during trastuzumab treatment.

Multiple myeloma can be treated with daratumumab (Darzalex) monoclonal antibody. [8] Studies with in vitro materials and patient materials indicate that ADCC is an important mechanism, along with CDC (complement-dependent cytotoxicity). [9]

ADCC as used in immune control is typically more useful for viral infections than bacterial infections due to IgG antibodies binding to virus-related antigens over prokaryotic cells. [10] Instead of ADCC removing outside toxins, immunoglobulins neutralize products of infecting bacteria and encase infected host cells that have had bacterial toxins directly inserted through the cell membrane.

ADCC is also important in the use of vaccines, as creation of antibodies and the destruction of antigens introduced to the host body are crucial to building immunity through small exposure to viral and bacterial proteins. Examples of this include vaccines targeting repeats in toxins (RTX) that are structurally crucial to a wide variety of erythrocyte-lysing bacteria, described as hemolysins. [11] These bacteria target the CD18 portion of leukocytes, which has historically been shown to impact ADCC in adhesion-deficient cells. [12]

See also

Related Research Articles

<span class="mw-page-title-main">Natural killer cell</span> Type of cytotoxic lymphocyte

Natural killer cells, also known as NK cells or large granular lymphocytes (LGL), are a type of cytotoxic lymphocyte critical to the innate immune system. They belong to the rapidly expanding family of known innate lymphoid cells (ILC) and represent 5–20% of all circulating lymphocytes in humans. The role of NK cells is analogous to that of cytotoxic T cells in the vertebrate adaptive immune response. NK cells provide rapid responses to virus-infected cell and other intracellular pathogens acting at around 3 days after infection, and respond to tumor formation. Most immune cells detect the antigen presented on major histocompatibility complex (MHC) on infected cell surfaces, but NK cells can recognize and kill stressed cells in the absence of antibodies and MHC, allowing for a much faster immune reaction. They were named "natural killers" because of the notion that they do not require activation to kill cells that are missing "self" markers of MHC class I. This role is especially important because harmful cells that are missing MHC I markers cannot be detected and destroyed by other immune cells, such as T lymphocyte cells.

<span class="mw-page-title-main">CD32</span> Surface receptor glycoprotein

CD32, also known as FcγRII or FCGR2, is a surface receptor glycoprotein belonging to the Ig gene superfamily. CD32 can be found on the surface of a variety of immune cells. CD32 has a low-affinity for the Fc region of IgG antibodies in monomeric form, but high affinity for IgG immune complexes. CD32 has two major functions: cellular response regulation, and the uptake of immune complexes. Cellular responses regulated by CD32 include phagocytosis, cytokine stimulation, and endocytic transport. Dysregulated CD32 is associated with different forms of autoimmunity, including systemic lupus erythematosus. In humans, there are three major CD32 subtypes: CD32A, CD32B, and CD32C. While CD32A and CD32C are involved in activating cellular responses, CD32B is inhibitory.

Opsonins are extracellular proteins that, when bound to substances or cells, induce phagocytes to phagocytose the substances or cells with the opsonins bound. Thus, opsonins act as tags to label things in the body that should be phagocytosed by phagocytes. Different types of things ("targets") can be tagged by opsonins for phagocytosis, including: pathogens, cancer cells, aged cells, dead or dying cells, excess synapses, or protein aggregates. Opsonins help clear pathogens, as well as dead, dying and diseased cells.

<span class="mw-page-title-main">Cancer immunotherapy</span> Artificial stimulation of the immune system to treat cancer

Cancer immunotherapy (immuno-oncotherapy) is the stimulation of the immune system to treat cancer, improving on the immune system's natural ability to fight the disease. It is an application of the fundamental research of cancer immunology and a growing subspecialty of oncology.

<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">Antibody opsonization</span> Immune system process

Antibody opsonization is a process by which a pathogen is marked for phagocytosis.

<span class="mw-page-title-main">Fc receptor</span> Surface protein important to the immune system

In immunology, an Fc receptor is a protein found on the surface of certain cells – including, among others, B lymphocytes, follicular dendritic cells, natural killer cells, macrophages, neutrophils, eosinophils, basophils, human platelets, and mast cells – that contribute to the protective functions of the immune system. Its name is derived from its binding specificity for a part of an antibody known as the Fc region. Fc receptors bind to antibodies that are attached to infected cells or invading pathogens. Their activity stimulates phagocytic or cytotoxic cells to destroy microbes, or infected cells by antibody-mediated phagocytosis or antibody-dependent cell-mediated cytotoxicity. Some viruses such as flaviviruses use Fc receptors to help them infect cells, by a mechanism known as antibody-dependent enhancement of infection.

<span class="mw-page-title-main">Fragment crystallizable region</span> Tail region of an antibody

The fragment crystallizable region is the tail region of an antibody that interacts with cell surface receptors called Fc receptors and some proteins of the complement system. This region allows antibodies to activate the immune system, for example, through binding to Fc receptors. In IgG, IgA and IgD antibody isotypes, the Fc region is composed of two identical protein fragments, derived from the second and third constant domains of the antibody's two heavy chains; IgM and IgE Fc regions contain three heavy chain constant domains in each polypeptide chain. The Fc regions of IgGs bear a highly conserved N-glycosylation site. Glycosylation of the Fc fragment is essential for Fc receptor-mediated activity. The N-glycans attached to this site are predominantly core-fucosylated diantennary structures of the complex type. In addition, small amounts of these N-glycans also bear bisecting GlcNAc and α-2,6 linked sialic acid residues.

Zalutumumab is a fully human IgG1 monoclonal antibody (mAb) directed towards the epidermal growth factor receptor (EGFR). It is a product developed by Genmab in Utrecht, the Netherlands. Specifically, zalutumumab is designed for the treatment of squamous cell carcinoma of the head and neck (SCCHN), a type of cancer.

CD16, also known as FcγRIII, is a cluster of differentiation molecule found on the surface of natural killer cells, neutrophils, monocytes, macrophages, and certain T cells. CD16 has been identified as Fc receptors FcγRIIIa (CD16a) and FcγRIIIb (CD16b), which participate in signal transduction. The most well-researched membrane receptor implicated in triggering lysis by NK cells, CD16 is a molecule of the immunoglobulin superfamily (IgSF) involved in antibody-dependent cellular cytotoxicity (ADCC). It can be used to isolate populations of specific immune cells through fluorescent-activated cell sorting (FACS) or magnetic-activated cell sorting, using antibodies directed towards CD16.

A bispecific monoclonal antibody is an artificial protein that can simultaneously bind to two different types of antigen or two different epitopes on the same antigen. Naturally occurring antibodies typically only target one antigen. BsAbs can be manufactured in several structural formats. BsAbs can be designed to recruit and activate immune cells, to interfere with receptor signaling and inactivate signaling ligands, and to force association of protein complexes. BsAbs have been explored for cancer immunotherapy, drug delivery, and Alzeimer's disease.

<span class="mw-page-title-main">FCAR</span> Mammalian protein found in Homo sapiens

Fc fragment of IgA receptor (FCAR) is a human gene that codes for the transmembrane receptor FcαRI, also known as CD89. FcαRI binds the heavy-chain constant region of Immunoglobulin A (IgA) antibodies. FcαRI is present on the cell surface of myeloid lineage cells, including neutrophils, monocytes, macrophages, and eosinophils, though it is notably absent from intestinal macrophages and does not appear on mast cells. FcαRI plays a role in both pro- and anti-inflammatory responses depending on the state of IgA bound. Inside-out signaling primes FcαRI in order for it to bind its ligand, while outside-in signaling caused by ligand binding depends on FcαRI association with the Fc receptor gamma chain.

Signaling lymphocytic activation molecule (SLAM) is a family of genes. Homophilic binding between SLAMs is involved in cell-to-cell adhesion during antigen presentation.

The following outline is provided as an overview of and topical guide to immunology:

Urelumab is a fully human, non‐ligand binding, CD137 agonist immunoglobulin‐γ 4 (IgG4) monoclonal antibody. It was developed utilizing Medarex's UltiMAb(R) technology by Bristol-Myers Squibb for the treatment of cancer and solid tumors. Urelumab promotes anti-tumor immunity, or an immune response against tumor cells, via CD137 activation. The application of Urelumab has been limited due to the fact that it can cause severe liver toxicity.

<span class="mw-page-title-main">Daratumumab</span> Monoclonal antibody

Daratumumab, sold under the brand name Darzalex, is an anti-cancer monoclonal antibody medication. It binds to CD38, which is overexpressed in multiple myeloma cells. Daratumumab was originally developed by Genmab, but it is now being jointly developed by Genmab along with the Johnson & Johnson subsidiary Janssen Biotech, which acquired worldwide commercialization rights to the drug from Genmab.

Cytokine-induced killer cells (CIK) cells are a group of immune effector cells featuring a mixed T- and natural killer (NK) cell-like phenotype. They are generated by ex vivo incubation of human peripheral blood mononuclear cells (PBMC) or cord blood mononuclear cells with interferon-gamma (IFN-γ), anti-CD3 antibody, recombinant human interleukin (IL)-1 and recombinant human interleukin (IL)-2.

Afucosylated monoclonal antibodies are monoclonal antibodies engineered so that the oligosaccharides in the Fc region of the antibody do not have any fucose sugar units. When antibodies are afucosylated, antibody-dependent cellular cytotoxicity (ADCC) is increased.

Complement-dependent cytotoxicity (CDC) is an effector function of IgG and IgM antibodies. When they are bound to surface antigen on target cell, the classical complement pathway is triggered by bonding protein C1q to these antibodies, resulting in formation of a membrane attack complex (MAC) and target cell lysis.

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. Hashimoto, G.; Wright, P. F.; Karzon, D. T. (1983-11-01). "Antibody-dependent cell-mediated cytotoxicity against influenza virus-infected cells". The Journal of Infectious Diseases. 148 (5): 785–794. doi:10.1093/infdis/148.5.785. ISSN   0022-1899. PMID   6605395.
  2. Pollara, Justin; Hart, Lydia; Brewer, Faraha; Pickeral, Joy; Packard, Beverly Z.; Hoxie, James A.; Komoriya, Akira; Ochsenbauer, Christina; Kappes, John C. (2011-08-01). "High-throughput quantitative analysis of HIV-1 and SIV-specific ADCC-mediating antibody responses". Cytometry Part A. 79 (8): 603–612. doi:10.1002/cyto.a.21084. ISSN   1552-4930. PMC   3692008 . PMID   21735545.
  3. Wang, W; Erbe, AK; Hank, JA; Morris, ZS; Sondel, PM (2015). "NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy". Front Immunol. 6: 368. doi: 10.3389/fimmu.2015.00368 . PMC   4515552 . PMID   26284063.
  4. Capron, M; Kazatchkine, MD; Fischer, E; Joseph, M; Butterworth, AE; et al. (1987). "Functional role of the alpha-chain of complement receptor type 3 in human eosinophil-dependent antibody-mediated cytotoxicity against schistosomes". J Immunol. 139 (6): 2059–65. doi: 10.4049/jimmunol.139.6.2059 . PMID   2957447. S2CID   44940057.
  5. Lo Nigro, Cristiana; Macagno, Marco; Sangiolo, Dario; Bertolaccini, Luca; Aglietta, Massimo; Merlano, Marco Carlo (March 2019). "NK-mediated antibody-dependent cell-mediated cytotoxicity in solid tumors: biological evidence and clinical perspectives". Annals of Translational Medicine. 7 (5): 105. doi: 10.21037/atm.2019.01.42 . ISSN   2305-5839. PMC   6462666 . PMID   31019955.
  6. Cheng, Min; Chen, Yongyan; Xiao, Weihua; Sun, Rui; Tian, Zhigang (May 2013). "NK cell-based immunotherapy for malignant diseases". Cellular & Molecular Immunology. 10 (3): 230–252. doi: 10.1038/cmi.2013.10 . ISSN   2042-0226. PMC   4076738 . PMID   23604045.
  7. Clynes, RA; Towers, TL; Presta, LG; Ravetch, JV (2000). "Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets". Nat Med. 6 (4): 443–6. doi:10.1038/74704. PMID   10742152. S2CID   20629632.
  8. Sanchez, L; Wang, Y; Siegel, DS (2016). "Daratumumab: a first-in-class CD38 monoclonal antibody for the treatment of multiple myeloma". J Hematol Oncol. 9 (1): 51. doi: 10.1186/s13045-016-0283-0 . PMC   4929758 . PMID   27363983.
  9. de Weers, M; Tai, YT; Bakker, JM; Vink, T; Jacobs, DC; et al. (2011). "Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors". J Immunol. 186 (3): 1840–8. doi: 10.4049/jimmunol.1003032 . PMID   21187443 . Retrieved 28 April 2017.
  10. Sawa, T.; Kinoshita, M.; Inoue, K.; Ohara, J.; Moriyama, K. (2019). "Immunoglobulin for Treating Bacterial Infections: One More Mechanism of Action". Antibodies. 8 (4): 52. doi: 10.3390/antib8040052 . PMC   6963986 . PMID   31684203.
  11. Frey, J. (2019). "RTX Toxins of Animal Pathogens and Their Role as Antigens in Vaccines and Diagnostics". Toxins. 11 (12): 719. doi: 10.3390/toxins11120719 . PMC   6950323 . PMID   31835534.
  12. Majima, T.; Ohashi, Y.; Nagatomi, R.; Iizuka, A.; Konno, T. (1993). "Defective mononuclear cell antibody-dependent cellular cytotoxicity (ADCC) in patients with leukocyte adhesion deficiency emphasizing on different CD11/CD18 requirement of Fc gamma RI versus Fc gamma RII in ADCC". Cellular Immunology. 148 (2): 385–396. doi:10.1006/cimm.1993.1120. PMID   8098672.

Further reading