You are on page 1of 29

Antibody

(Redirected from Immunoglobulins)

An antibody (Ab), also known as an immunoglobulin


(Ig),[1] is a large, Y-shaped protein used by the immune
system to identify and neutralize foreign objects such as
pathogenic bacteria and viruses. The antibody recognizes a
unique molecule of the pathogen, called an antigen.[2][3]
Each tip of the "Y" of an antibody contains a paratope
(analogous to a lock) that is specific for one particular
epitope (analogous to a key) on an antigen, allowing these
two structures to bind together with precision. Using this
binding mechanism, an antibody can tag a microbe or an
infected cell for attack by other parts of the immune system,
or can neutralize it directly (for example, by blocking a part
of a virus that is essential for its invasion).

To allow the immune system to recognize millions of


different antigens, the antigen-binding sites at both tips of
the antibody come in an equally wide variety.
In contrast, the
remainder of the antibody is relatively constant. It only
occurs in a few variants, which define the antibody's class or
isotype: IgA, IgD, IgE, IgG, and IgM.
The constant region at
the trunk of the antibody includes sites involved in Each antibody binds to a specific antigen;
interactions with other components of the immune system. an interaction similar to a lock and key.
The class hence determines the function triggered by an
antibody after binding to an antigen, in addition to some
structural features.
Antibodies from different classes also differ in where they are released in the body
and at what stage of an immune response.

Together with B and T cells, antibodies comprise the most important part of the adaptive immune
system.
They occur in two forms: one that is attached to a B cell, and the other, a soluble form, that is
unattached and found in extracellular fluids such as blood plasma.
Initially, all antibodies are of the
first form, attached to the surface of a B cell – these are then referred to as B-cell receptors (BCR).
After an antigen binds to a BCR, the B cell activates to proliferate and differentiate into either plasma
cells, which secrete soluble antibodies with the same paratope, or memory B cells, which survive in the
body to enable long-lasting immunity to the antigen.[4]
Soluble antibodies are released into the blood
and tissue fluids, as well as many secretions.
Because these fluids were traditionally known as humors,
antibody-mediated immunity is sometimes known as, or considered a part of, humoral immunity.[5]
The soluble Y-shaped units can occur individually as monomers, or in complexes of two to five units.

Antibodies are glycoproteins belonging to the immunoglobulin superfamily.


The terms antibody and
immunoglobulin are often used interchangeably,[1] though the term 'antibody' is sometimes reserved
for the secreted, soluble form, i.e. excluding B-cell receptors.[6]

Structure
Antibodies are heavy (~150 kDa) proteins of about 10 nm in
size,[7]
arranged in three globular regions that roughly form
a Y shape.

In humans and most mammals, an antibody unit consists of


four polypeptide chains; two identical heavy chains and two
identical light chains connected by disulfide bonds.[8]
Each
chain is a series of domains: somewhat similar sequences of
about 110 amino acids each.
These domains are usually
represented in simplified schematics as rectangles.
Light
chains consist of one variable domain VL and one constant
domain CL, while heavy chains contain one variable domain
VH and three to four constant domains CH1, CH2, ...[9]
Schematic structure of an antibody: two
Structurally an antibody is also partitioned into two antigen- heavy chains (blue, yellow) and the two
binding fragments (Fab), containing one VL, VH, CL, and CH1 light chains (green, pink). The antigen
domain each, as well as the crystallisable fragment (Fc), binding site is circled.
forming the trunk of the Y shape.[10]
In between them is a

A more accurate depiction of an antibody (3D structure at RCSB PDB (https://www.rcsb.org/3d-view/1IGY/1)).


Glycans in the Fc region are shown in black.

hinge region of the heavy chains, whose flexibility allows antibodies to bind to pairs of epitopes at
various distances, to form complexes (dimers, trimers, etc.), and to bind effector molecules more
easily.[11]

In an electrophoresis test of blood proteins, antibodies mostly migrate to the last, gamma globulin
fraction.
Conversely, most gamma-globulins are antibodies, which is why the two terms were
historically used as synonyms, as were the symbols Ig and γ.
This variant terminology fell out of use
due to the correspondence being inexact and due to confusion with γ (gamma) heavy chains which
characterize the IgG class of antibodies.[12][13]
Antigen-binding site

The variable domains can also be referred to as the FV region. It is the subregion of Fab that binds to
an antigen.
More specifically, each variable domain contains three hypervariable regions – the amino
acids seen there vary the most from antibody to antibody.
When the protein folds, these regions give
rise to three loops of β-strands, localized near one another on the surface of the antibody.
These loops
are referred to as the complementarity-determining regions (CDRs), since their shape complements
that of an antigen.
Three CDRs from each of the heavy and light chains together form an antibody-
binding site whose shape can be anything from a pocket to which a smaller antigen binds, to a larger
surface, to a protrusion that sticks out into a groove in an antigen.
Typically however only a few
residues contribute to most of the binding energy.[2]

The existence of two identical antibody-binding sites allows antibody molecules to bind strongly to
multivalent antigen (repeating sites such as polysaccharides in bacterial cell walls, or other sites at
some distance apart), as well as to form antibody complexes and larger antigen-antibody
complexes.[2] The resulting cross-linking plays a role in activating other parts of the immune system.

The structures of CDRs have been clustered and classified by Chothia et al.[14]
and more recently by
North et al.[15]
and Nikoloudis et al.[16] However, describing an antibody's binding site using only one
single static structure limits the understanding and characterization of the antibody's function and
properties. To improve antibody structure prediction and to take the strongly correlated CDR loop and
interface movements into account, antibody paratopes should be described as interconverting states
in solution with varying probabilities.[17]

In the framework of the immune network theory, CDRs are also called idiotypes. According to
immune network theory, the adaptive immune system is regulated by interactions between idiotypes.

Fc region

The Fc region (the trunk of the Y shape) is composed of constant domains from the heavy chains. Its
role is in modulating immune cell activity: it is where effector molecules bind to, triggering various
effects after the antibody Fab region binds to an antigen.[2][11]
Effector cells (such as macrophages or
natural killer cells) bind via their Fc receptors (FcR) to the Fc region of an antibody, while the
complement system is activated by binding the C1q protein complex. IgG or IgM can bind to C1q, but
IgA cannot, therefore IgA does not activate the classical complement pathway.[18]

Another role of the Fc region is to selectively distribute different antibody classes across the body. In
particular, the neonatal Fc receptor (FcRn) binds to the Fc region of IgG antibodies to transport it
across the placenta, from the mother to the fetus.

Antibodies are glycoproteins,[19] that is, they have carbohydrates (glycans) added to conserved amino
acid residues.[19][20]
These conserved glycosylation sites occur in the Fc region and influence
interactions with effector molecules.[19][21]

Protein structure

The N-terminus of each chain is situated at the tip.


Each immunoglobulin domain has a similar
structure, characteristic of all the members of the immunoglobulin superfamily:
it is composed of
between 7 (for constant domains) and 9 (for variable domains) β-strands, forming two beta sheets in a
Greek key motif.
The sheets create a "sandwich" shape, the immunoglobulin fold, held together by a
disulfide bond.

Antibody complexes

Secreted antibodies can occur as a single Y-shaped unit, a monomer.


However, some antibody classes also form dimers with two Ig units (as
with IgA), tetramers with four Ig units (like teleost fish IgM), or
pentamers with five Ig units (like shark IgW or mammalian IgM, which
occasionally forms hexamers as well, with six units).[22]

Antibodies also form complexes by binding to antigen: this is called an


antigen-antibody complex or immune complex.
Small antigens can cross-
link two antibodies, also leading to the formation of antibody dimers,
trimers, tetramers, etc.
Multivalent antigens (e.g., cells with multiple
epitopes) can form larger complexes with antibodies.
An extreme
example is the clumping, or agglutination, of red blood cells with Some antibodies form
antibodies in the Coombs test to determine blood groups: the large complexes that bind to
clumps become insoluble, leading to visually apparent precipitation. multiple antigen molecules.

B cell receptors

The membrane-bound form of an antibody may be called a surface immunoglobulin (sIg) or a


membrane immunoglobulin (mIg). It is part of the B cell receptor (BCR), which allows a B cell to
detect when a specific antigen is present in the body and triggers B cell activation.[23] The BCR is
composed of surface-bound IgD or IgM antibodies and associated Ig-α and Ig-β heterodimers, which
are capable of signal transduction.[24] A typical human B cell will have 50,000 to 100,000 antibodies
bound to its surface.[24] Upon antigen binding, they cluster in large patches, which can exceed 1
micrometer in diameter, on lipid rafts that isolate the BCRs from most other cell signaling
receptors.[24]
These patches may improve the efficiency of the cellular immune response.[25] In
humans, the cell surface is bare around the B cell receptors for several hundred nanometers,[24] which
further isolates the BCRs from competing influences.

Classes
Antibodies can come in different varieties known as isotypes or classes. In placental mammals there
are five antibody classes known as IgA, IgD, IgE, IgG, and IgM, which are further subdivided into
subclasses such as IgA1, IgA2.
The prefix "Ig" stands for immunoglobulin, while the suffix denotes the
type of heavy chain the antibody contains: the heavy chain types α (alpha), γ (gamma), δ (delta), ε
(epsilon), μ (mu) give rise to IgA, IgG, IgD, IgE, IgM, respectively.
The distinctive features of each
class are determined by the part of the heavy chain within the hinge and Fc region.[2]

The classes differ in their biological properties, functional locations and ability to deal with different
antigens, as depicted in the table.[8]
For example, IgE antibodies are responsible for an allergic
response consisting of histamine release from mast cells, often a sole contributor to asthma (though
other pathways exist as do exist symptoms very similar to yet not technically asthma). The antibody's
variable region binds to allergic antigen, for example house dust mite particles, while its Fc region (in
the ε heavy chains) binds to Fc receptor ε on a mast cell, triggering its degranulation: the release of
molecules stored in its granules.[26]

Antibody isotypes of mammals


Class Subclasses Description
Found in mucosal areas, such as the gut, respiratory tract and urogenital tract, and prevents
IgA 2
colonization by pathogens.[27] Also found in saliva, tears, and breast milk.

Functions mainly as an antigen receptor on B cells that have not been exposed to antigens.[28]
IgD 1
It has been shown to activate basophils and mast cells to produce antimicrobial factors.[29]
Binds to allergens and triggers histamine release from mast cells and basophils, and is
IgE 1 involved in allergy. Humans and other animals evolved IgE to protect against parasitic worms,
though in the present, IgE is primarily related to allergies and asthma.[5]
In its four forms, provides the majority of antibody-based immunity against invading
IgG 4 pathogens.[5] The only antibody capable of crossing the placenta to give passive immunity to
the fetus.
Expressed on the surface of B cells (monomer) and in a secreted form (pentamer) with very
IgM 1 high avidity. Eliminates pathogens in the early stages of B cell-mediated (humoral) immunity
before there is sufficient IgG.[5][28]

The antibody isotype of a B cell changes during cell development and activation. Immature B cells,
which have never been exposed to an antigen, express only the IgM isotype in a cell surface bound
form. The B lymphocyte, in this ready-to-respond form, is known as a "naive B lymphocyte." The naive
B lymphocyte expresses both surface IgM and IgD. The co-expression of both of these
immunoglobulin isotypes renders the B cell ready to respond to antigen.[30] B cell activation follows
engagement of the cell-bound antibody molecule with an antigen, causing the cell to divide and
differentiate into an antibody-producing cell called a plasma cell. In this activated form, the B cell
starts to produce antibody in a secreted form rather than a membrane-bound form. Some daughter
cells of the activated B cells undergo isotype switching, a mechanism that causes the production of
antibodies to change from IgM or IgD to the other antibody isotypes, IgE, IgA, or IgG, that have
defined roles in the immune system.

Light chain types

In mammals there are two types of immunoglobulin light chain, which are called lambda (λ) and
kappa (κ). However, there is no known functional difference between them, and both can occur with
any of the five major types of heavy chains.[2] Each antibody contains two identical light chains: both
κ or both λ. Proportions of κ and λ types vary by species and can be used to detect abnormal
proliferation of B cell clones. Other types of light chains, such as the iota (ι) chain, are found in other
vertebrates like sharks (Chondrichthyes) and bony fishes (Teleostei).

In non-mammalian animals

In most placental mammals, the structure of antibodies is generally the same.


Jawed fish appear to be
the most primitive animals that are able to make antibodies similar to those of mammals, although
many features of their adaptive immunity appeared somewhat earlier.[31]
Cartilaginous fish (such as sharks) produce heavy-chain-only antibodies (i.e., lacking light chains)
which moreover feature longer chain pentamers (with five constant units per molecule). Camelids
(such as camels, llamas, alpacas) are also notable for producing heavy-chain-only antibodies.[2][32]

Antibody classes not found in mammals


Class Types Description

IgY Found in birds and reptiles; related to mammalian IgG.[33]

IgW Found in sharks and skates; related to mammalian IgD.[34]

IgT/Z Found in teleost fish[35]

Antibody–antigen interactions
The antibody's paratope interacts with the antigen's epitope. An antigen usually contains different
epitopes along its surface arranged discontinuously, and dominant epitopes on a given antigen are
called determinants.

Antibody and antigen interact by spatial complementarity (lock and key). The molecular forces
involved in the Fab-epitope interaction are weak and non-specific – for example electrostatic forces,
hydrogen bonds, hydrophobic interactions, and van der Waals forces. This means binding between
antibody and antigen is reversible, and the antibody's affinity towards an antigen is relative rather
than absolute. Relatively weak binding also means it is possible for an antibody to cross-react with
different antigens of different relative affinities.

Function
The main categories of antibody action include the following:

Neutralisation, in which neutralizing antibodies block parts of the surface of a bacterial cell or
virion to render its attack ineffective
Agglutination, in which antibodies "glue together" foreign cells into clumps that are attractive
targets for phagocytosis
Precipitation, in which antibodies "glue together" serum-soluble antigens, forcing them to
precipitate out of solution in clumps that are attractive targets for phagocytosis
Complement activation (fixation), in which antibodies that are latched onto a foreign cell
encourage complement to attack it with a membrane attack complex, which leads to the following:
Lysis of the foreign cell
Encouragement of inflammation by chemotactically attracting inflammatory cells

More indirectly, an antibody can signal immune cells to present antibody fragments to T cells, or
downregulate other immune cells to avoid autoimmunity.

Activated B cells differentiate into either antibody-producing cells called plasma cells that secrete
soluble antibody or memory cells that survive in the body for years afterward in order to allow the
immune system to remember an antigen and respond faster upon future exposures.[4]
At the prenatal and neonatal stages of life, the presence of
antibodies is provided by passive immunization from the mother.
Early endogenous antibody production varies for different kinds of
antibodies, and usually appear within the first years of life. Since
antibodies exist freely in the bloodstream, they are said to be part
of the humoral immune system. Circulating antibodies are
produced by clonal B cells that specifically respond to only one
antigen (an example is a virus capsid protein fragment).
Antibodies contribute to immunity in three ways: They prevent
pathogens from entering or damaging cells by binding to them; 1. Antibodies (A) and
they stimulate removal of pathogens by macrophages and other pathogens (B) free roam in
cells by coating the pathogen; and they trigger destruction of the blood.
pathogens by stimulating other immune responses such as the 2. The antibodies bind to
complement pathway.[36] Antibodies will also trigger vasoactive pathogens, and can do so in
amine degranulation to contribute to immunity against certain different formations such as:
types of antigens (helminths, allergens).
a. opsonization,
b. neutralisation, and
Activation of c. agglutination.
complement 3. A phagocyte (C)
approaches the pathogen,
Antibodies that bind to surface and the Fc region (D) of the
antigens (for example, on antibody binds to one of the
bacteria) will attract the first Fc receptors (E) of the
component of the complement phagocyte.
cascade with their Fc region 4. Phagocytosis occurs as the
and initiate activation of the pathogen is ingested.
"classical" complement
The secreted mammalian IgM has system. [36] This results in the
five Ig units. Each Ig unit (labeled 1) killing of bacteria in two ways.[5] First, the binding of the antibody
has two epitope binding Fab and complement molecules marks the microbe for ingestion by
regions, so IgM is capable of phagocytes in a process called opsonization; these phagocytes are
binding up to 10 epitopes. attracted by certain complement molecules generated in the
complement cascade. Second, some complement system
components form a membrane attack complex to assist antibodies
to kill the bacterium directly (bacteriolysis).[37]

Activation of effector cells

To combat pathogens that replicate outside cells, antibodies bind to pathogens to link them together,
causing them to agglutinate. Since an antibody has at least two paratopes, it can bind more than one
antigen by binding identical epitopes carried on the surfaces of these antigens. By coating the
pathogen, antibodies stimulate effector functions against the pathogen in cells that recognize their Fc
region.[5]

Those cells that recognize coated pathogens have Fc receptors, which, as the name suggests, interact
with the Fc region of IgA, IgG, and IgE antibodies. The engagement of a particular antibody with the
Fc receptor on a particular cell triggers an effector function of that cell; phagocytes will phagocytose,
mast cells and neutrophils will degranulate, natural killer cells will release cytokines and cytotoxic
molecules; that will ultimately result in destruction of the invading microbe. The activation of natural
killer cells by antibodies initiates a cytotoxic mechanism known as antibody-dependent cell-mediated
cytotoxicity (ADCC) – this process may explain the efficacy of monoclonal antibodies used in
biological therapies against cancer. The Fc receptors are isotype-specific, which gives greater
flexibility to the immune system, invoking only the appropriate immune mechanisms for distinct
pathogens.[2]

Natural antibodies

Humans and higher primates also produce "natural antibodies" that are present in serum before viral
infection. Natural antibodies have been defined as antibodies that are produced without any previous
infection, vaccination, other foreign antigen exposure or passive immunization. These antibodies can
activate the classical complement pathway leading to lysis of enveloped virus particles long before the
adaptive immune response is activated. Many natural antibodies are directed against the disaccharide
galactose α(1,3)-galactose (α-Gal), which is found as a terminal sugar on glycosylated cell surface
proteins, and generated in response to production of this sugar by bacteria contained in the human
gut.[38] Rejection of xenotransplantated organs is thought to be, in part, the result of natural
antibodies circulating in the serum of the recipient binding to α-Gal antigens expressed on the donor
tissue.[39]

Immunoglobulin diversity
Virtually all microbes can trigger an antibody response. Successful recognition and eradication of
many different types of microbes requires diversity among antibodies; their amino acid composition
varies allowing them to interact with many different antigens.[40] It has been estimated that humans
generate about 10  billion different antibodies, each capable of binding a distinct epitope of an
antigen.[41] Although a huge repertoire of different antibodies is generated in a single individual, the
number of genes available to make these proteins is limited by the size of the human genome. Several
complex genetic mechanisms have evolved that allow vertebrate B cells to generate a diverse pool of
antibodies from a relatively small number of antibody genes.[42]

Domain variability

The chromosomal region that encodes an antibody is


large and contains several distinct gene loci for each
domain of the antibody—the chromosome region
containing heavy chain genes (IGH@) is found on
chromosome 14, and the loci containing lambda and
kappa light chain genes (IGL@ and IGK@) are found on
chromosomes 22 and 2 in humans. One of these domains
is called the variable domain, which is present in each
heavy and light chain of every antibody, but can differ in The complementarity determining regions of
different antibodies generated from distinct B cells. the heavy chain are shown in red (PDB: 1IGT
Differences between the variable domains are located on (https://www.rcsb.org/structure/1IGT)​)
three loops known as hypervariable regions (HV-1, HV-2
and HV-3) or complementarity-determining regions
(CDR1, CDR2 and CDR3). CDRs are supported within the variable domains by conserved framework
regions. The heavy chain locus contains about 65 different variable domain genes that all differ in
their CDRs. Combining these genes with an array of genes for other domains of the antibody generates
a large cavalry of antibodies with a high degree of variability. This combination is called V(D)J
recombination discussed below.[43]

V(D)J recombination

Somatic recombination of immunoglobulins, also known


as V(D)J recombination, involves the generation of a
unique immunoglobulin variable region. The variable
region of each immunoglobulin heavy or light chain is
encoded in several pieces—known as gene segments
(subgenes). These segments are called variable (V),
diversity (D) and joining (J) segments.[42] V, D and J
segments are found in Ig heavy chains, but only V and J
segments are found in Ig light chains. Multiple copies of
the V, D and J gene segments exist, and are tandemly
arranged in the genomes of mammals. In the bone
marrow, each developing B cell will assemble an
immunoglobulin variable region by randomly selecting
and combining one V, one D and one J gene segment (or
one V and one J segment in the light chain). As there are Simplified overview of V(D)J recombination of
multiple copies of each type of gene segment, and immunoglobulin heavy chains
different combinations of gene segments can be used to
generate each immunoglobulin variable region, this
process generates a huge number of antibodies, each with different paratopes, and thus different
antigen specificities.[44] The rearrangement of several subgenes (i.e. V2 family) for lambda light chain
immunoglobulin is coupled with the activation of microRNA miR-650, which further influences
biology of B-cells.

RAG proteins play an important role with V(D)J recombination in cutting DNA at a particular
region.[44] Without the presence of these proteins, V(D)J recombination would not occur.[44]

After a B cell produces a functional immunoglobulin gene during V(D)J recombination, it cannot
express any other variable region (a process known as allelic exclusion) thus each B cell can produce
antibodies containing only one kind of variable chain.[2][45]

Somatic hypermutation and affinity maturation

Following activation with antigen, B cells begin to proliferate rapidly. In these rapidly dividing cells,
the genes encoding the variable domains of the heavy and light chains undergo a high rate of point
mutation, by a process called somatic hypermutation (SHM). SHM results in approximately one
nucleotide change per variable gene, per cell division.[46] As a consequence, any daughter B cells will
acquire slight amino acid differences in the variable domains of their antibody chains.

This serves to increase the diversity of the antibody pool and impacts the antibody's antigen-binding
affinity.[47] Some point mutations will result in the production of antibodies that have a weaker
interaction (low affinity) with their antigen than the original antibody, and some mutations will
generate antibodies with a stronger interaction (high affinity).[48] B cells that express high affinity
antibodies on their surface will receive a strong survival signal during interactions with other cells,
whereas those with low affinity antibodies will not, and will die by apoptosis.[48] Thus, B cells
expressing antibodies with a higher affinity for the antigen will outcompete those with weaker
affinities for function and survival allowing the average affinity of antibodies to increase over time.
The process of generating antibodies with increased binding affinities is called affinity maturation.
Affinity maturation occurs in mature B cells after V(D)J recombination, and is dependent on help
from helper T cells.[49]

Class switching

Isotype or class switching is a biological process occurring


after activation of the B cell, which allows the cell to
produce different classes of antibody (IgA, IgE, or
IgG).[44] The different classes of antibody, and thus
effector functions, are defined by the constant (C) regions
of the immunoglobulin heavy chain. Initially, naive B cells
express only cell-surface IgM and IgD with identical
antigen binding regions. Each isotype is adapted for a
distinct function; therefore, after activation, an antibody
with an IgG, IgA, or IgE effector function might be
required to effectively eliminate an antigen. Class
switching allows different daughter cells from the same
activated B cell to produce antibodies of different
isotypes. Only the constant region of the antibody heavy
chain changes during class switching; the variable Mechanism of class switch recombination that
regions, and therefore antigen specificity, remain allows isotype switching in activated B cells
unchanged. Thus the progeny of a single B cell can
produce antibodies, all specific for the same antigen, but
with the ability to produce the effector function appropriate for each antigenic challenge. Class
switching is triggered by cytokines; the isotype generated depends on which cytokines are present in
the B cell environment.[50]

Class switching occurs in the heavy chain gene locus by a mechanism called class switch
recombination (CSR). This mechanism relies on conserved nucleotide motifs, called switch (S)
regions, found in DNA upstream of each constant region gene (except in the δ-chain). The DNA
strand is broken by the activity of a series of enzymes at two selected S-regions.[51][52] The variable
domain exon is rejoined through a process called non-homologous end joining (NHEJ) to the desired
constant region (γ, α or ε). This process results in an immunoglobulin gene that encodes an antibody
of a different isotype.[53]

Specificity designations

An antibody can be called monospecific if it has specificity for the same antigen or epitope,[54] or
bispecific if they have affinity for two different antigens or two different epitopes on the same
antigen.[55] A group of antibodies can be called polyvalent (or unspecific) if they have affinity for
various antigens[56] or microorganisms.[56] Intravenous immunoglobulin, if not otherwise noted,
consists of a variety of different IgG (polyclonal IgG). In contrast, monoclonal antibodies are identical
antibodies produced by a single B cell.
Asymmetrical antibodies

Heterodimeric antibodies, which are also asymmetrical antibodies, allow for greater flexibility and
new formats for attaching a variety of drugs to the antibody arms. One of the general formats for a
heterodimeric antibody is the "knobs-into-holes" format. This format is specific to the heavy chain
part of the constant region in antibodies. The "knobs" part is engineered by replacing a small amino
acid with a larger one. It fits into the "hole", which is engineered by replacing a large amino acid with a
smaller one. What connects the "knobs" to the "holes" are the disulfide bonds between each chain. The
"knobs-into-holes" shape facilitates antibody dependent cell mediated cytotoxicity. Single chain
variable fragments (scFv) are connected to the variable domain of the heavy and light chain via a short
linker peptide. The linker is rich in glycine, which gives it more flexibility, and serine/threonine,
which gives it specificity. Two different scFv fragments can be connected together, via a hinge region,
to the constant domain of the heavy chain or the constant domain of the light chain.[57] This gives the
antibody bispecificity, allowing for the binding specificities of two different antigens.[58] The "knobs-
into-holes" format enhances heterodimer formation but doesn't suppress homodimer formation.

To further improve the function of heterodimeric antibodies, many scientists are looking towards
artificial constructs. Artificial antibodies are largely diverse protein motifs that use the functional
strategy of the antibody molecule, but aren't limited by the loop and framework structural constraints
of the natural antibody.[59] Being able to control the combinational design of the sequence and three-
dimensional space could transcend the natural design and allow for the attachment of different
combinations of drugs to the arms.

Heterodimeric antibodies have a greater range in shapes they can take and the drugs that are attached
to the arms don't have to be the same on each arm, allowing for different combinations of drugs to be
used in cancer treatment. Pharmaceuticals are able to produce highly functional bispecific, and even
multispecific, antibodies. The degree to which they can function is impressive given that such a change
of shape from the natural form should lead to decreased functionality.

History
The first use of the term "antibody" occurred in a text by Paul Ehrlich. The term Antikörper (the
German word for antibody) appears in the conclusion of his article "Experimental Studies on
Immunity", published in October 1891, which states that, "if two substances give rise to two different
Antikörper, then they themselves must be different".[60] However, the term was not accepted
immediately and several other terms for antibody were proposed; these included Immunkörper,
Amboceptor, Zwischenkörper, substance sensibilisatrice, copula, Desmon, philocytase, fixateur, and
Immunisin.[60] The word antibody has formal analogy to the word antitoxin and a similar concept to
Immunkörper (immune body in English).[60] As such, the original construction of the word contains a
logical flaw; the antitoxin is something directed against a toxin, while the antibody is a body directed
against something.[60]

The study of antibodies began in 1890 when Emil von Behring and Kitasato Shibasaburō described
antibody activity against diphtheria and tetanus toxins. Von Behring and Kitasato put forward the
theory of humoral immunity, proposing that a mediator in serum could react with a foreign
antigen.[64][65] His idea prompted Paul Ehrlich to propose the side-chain theory for antibody and
antigen interaction in 1897, when he hypothesized that receptors (described as "side-chains") on the
surface of cells could bind specifically to toxins  – in a "lock-and-key" interaction  – and that this
binding reaction is the trigger for the production of antibodies.[66] Other researchers believed that
antibodies existed freely in the blood and, in 1904, Almroth
Wright suggested that soluble antibodies coated bacteria to label
them for phagocytosis and killing; a process that he named
opsoninization.[67]

In the 1920s, Michael


Heidelberger and Oswald
Avery observed that antigens
Angel of the West (2008) by Julian
could be precipitated by
Voss-Andreae is a sculpture based
antibodies and went on to
on the antibody structure published show that antibodies are made
by E. Padlan.[61] Created for the of protein.[68] The biochemical
Florida campus of the Scripps properties of antigen-
Research Institute,[62] the antibody antibody-binding interactions
is placed into a ring referencing were examined in more detail
Leonardo da Vinci's Vitruvian Man in the late 1930s by John Michael Heidelberger
Marrack. [69] The next major
thus highlighting the similarity of the
antibody and the human body. [63] advance was in the 1940s,
when Linus Pauling confirmed the lock-and-key theory proposed
by Ehrlich by showing that the interactions between antibodies
and antigens depend more on their shape than their chemical composition.[70] In 1948, Astrid
Fagraeus discovered that B cells, in the form of plasma cells, were responsible for generating
antibodies.[71]

Further work concentrated on characterizing the structures of the antibody proteins. A major advance
in these structural studies was the discovery in the early 1960s by Gerald Edelman and Joseph Gally of
the antibody light chain,[72] and their realization that this protein is the same as the Bence-Jones
protein described in 1845 by Henry Bence Jones.[73] Edelman went on to discover that antibodies are
composed of disulfide bond-linked heavy and light chains. Around the same time, antibody-binding
(Fab) and antibody tail (Fc) regions of IgG were characterized by Rodney Porter.[74] Together, these
scientists deduced the structure and complete amino acid sequence of IgG, a feat for which they were
jointly awarded the 1972 Nobel Prize in Physiology or Medicine.[74] The Fv fragment was prepared
and characterized by David Givol.[75] While most of these early studies focused on IgM and IgG, other
immunoglobulin isotypes were identified in the 1960s: Thomas Tomasi discovered secretory antibody
(IgA);[76] David S. Rowe and John L. Fahey discovered IgD;[77] and Kimishige Ishizaka and Teruko
Ishizaka discovered IgE and showed it was a class of antibodies involved in allergic reactions.[78] In a
landmark series of experiments beginning in 1976, Susumu Tonegawa showed that genetic material
can rearrange itself to form the vast array of available antibodies.[79]

Medical applications

Disease diagnosis

Detection of particular antibodies is a very common form of medical diagnostics, and applications
such as serology depend on these methods.[80] For example, in biochemical assays for disease
diagnosis,[81] a titer of antibodies directed against Epstein-Barr virus or Lyme disease is estimated
from the blood. If those antibodies are not present, either the person is not infected or the infection
occurred a very long time ago, and the B cells generating these specific antibodies have naturally
decayed.
In clinical immunology, levels of individual classes of immunoglobulins are measured by
nephelometry (or turbidimetry) to characterize the antibody profile of patient.[82] Elevations in
different classes of immunoglobulins are sometimes useful in determining the cause of liver damage
in patients for whom the diagnosis is unclear.[1] For example, elevated IgA indicates alcoholic
cirrhosis, elevated IgM indicates viral hepatitis and primary biliary cirrhosis, while IgG is elevated in
viral hepatitis, autoimmune hepatitis and cirrhosis.

Autoimmune disorders can often be traced to antibodies that bind the body's own epitopes; many can
be detected through blood tests. Antibodies directed against red blood cell surface antigens in immune
mediated hemolytic anemia are detected with the Coombs test.[83] The Coombs test is also used for
antibody screening in blood transfusion preparation and also for antibody screening in antenatal
women.[83]

Practically, several immunodiagnostic methods based on detection of complex antigen-antibody are


used to diagnose infectious diseases, for example ELISA, immunofluorescence, Western blot,
immunodiffusion, immunoelectrophoresis, and magnetic immunoassay. Antibodies raised against
human chorionic gonadotropin are used in over the counter pregnancy tests.

New dioxaborolane chemistry enables radioactive fluoride (18F) labeling of antibodies, which allows
for positron emission tomography (PET) imaging of cancer.[84]

Disease therapy

Targeted monoclonal antibody therapy is employed to treat diseases such as rheumatoid arthritis,[85]
multiple sclerosis,[86] psoriasis,[87] and many forms of cancer including non-Hodgkin's
lymphoma,[88] colorectal cancer, head and neck cancer and breast cancer.[89]

Some immune deficiencies, such as X-linked agammaglobulinemia and hypogammaglobulinemia,


result in partial or complete lack of antibodies.[90] These diseases are often treated by inducing a
short-term form of immunity called passive immunity. Passive immunity is achieved through the
transfer of ready-made antibodies in the form of human or animal serum, pooled immunoglobulin or
monoclonal antibodies, into the affected individual.[91]

Prenatal therapy

Rh factor, also known as Rh D antigen, is an antigen found on red blood cells; individuals that are Rh-
positive (Rh+) have this antigen on their red blood cells and individuals that are Rh-negative (Rh–) do
not. During normal childbirth, delivery trauma or complications during pregnancy, blood from a fetus
can enter the mother's system. In the case of an Rh-incompatible mother and child, consequential
blood mixing may sensitize an Rh- mother to the Rh antigen on the blood cells of the Rh+ child,
putting the remainder of the pregnancy, and any subsequent pregnancies, at risk for hemolytic disease
of the newborn.[92]

Rho(D) immune globulin antibodies are specific for human RhD antigen.[93] Anti-RhD antibodies are
administered as part of a prenatal treatment regimen to prevent sensitization that may occur when a
Rh-negative mother has a Rh-positive fetus. Treatment of a mother with Anti-RhD antibodies prior to
and immediately after trauma and delivery destroys Rh antigen in the mother's system from the fetus.
It is important to note that this occurs before the antigen can stimulate maternal B cells to
"remember" Rh antigen by generating memory B cells. Therefore, her humoral immune system will
not make anti-Rh antibodies, and will not attack the Rh antigens of the current or subsequent babies.
Rho(D) Immune Globulin treatment prevents sensitization that can lead to Rh disease, but does not
prevent or treat the underlying disease itself.[93]

Research applications
Specific antibodies are produced by injecting an antigen into a
mammal, such as a mouse, rat, rabbit, goat, sheep, or horse for
large quantities of antibody. Blood isolated from these animals
contains polyclonal antibodies—multiple antibodies that bind to
the same antigen—in the serum, which can now be called
antiserum. Antigens are also injected into chickens for generation
of polyclonal antibodies in egg yolk.[94] To obtain antibody that is
specific for a single epitope of an antigen, antibody-secreting
lymphocytes are isolated from the animal and immortalized by
fusing them with a cancer cell line. The fused cells are called
hybridomas, and will continually grow and secrete antibody in
culture. Single hybridoma cells are isolated by dilution cloning to
Immunofluorescence image of the
generate cell clones that all produce the same antibody; these
eukaryotic cytoskeleton.
antibodies are called monoclonal antibodies.[95] Polyclonal and Microtubules as shown in green, are
monoclonal antibodies are often purified using Protein A/G or marked by an antibody conjugated
antigen-affinity chromatography.[96] to a green fluorescing molecule,
FITC.
In research, purified antibodies are used in many applications.
Antibodies for research applications can be found directly from
antibody suppliers, or through use of a specialist search engine. Research antibodies are most
commonly used to identify and locate intracellular and extracellular proteins. Antibodies are used in
flow cytometry to differentiate cell types by the proteins they express; different types of cells express
different combinations of cluster of differentiation molecules on their surface, and produce different
intracellular and secretable proteins.[97] They are also used in immunoprecipitation to separate
proteins and anything bound to them (co-immunoprecipitation) from other molecules in a cell
lysate,[98] in Western blot analyses to identify proteins separated by electrophoresis,[99] and in
immunohistochemistry or immunofluorescence to examine protein expression in tissue sections or to
locate proteins within cells with the assistance of a microscope.[97][100] Proteins can also be detected
and quantified with antibodies, using ELISA and ELISpot techniques.[101][102]

Antibodies used in research are some of the most powerful, yet most problematic reagents with a
tremendous number of factors that must be controlled in any experiment including cross reactivity, or
the antibody recognizing multiple epitopes and affinity, which can vary widely depending on
experimental conditions such as pH, solvent, state of tissue etc. Multiple attempts have been made to
improve both the way that researchers validate antibodies[103][104] and ways in which they report on
antibodies. Researchers using antibodies in their work need to record them correctly in order to allow
their research to be reproducible (and therefore tested, and qualified by other researchers). Less than
half of research antibodies referenced in academic papers can be easily identified.[105] Papers
published in F1000 in 2014 and 2015 provide researchers with a guide for reporting research antibody
use.[106][107] The RRID paper, is co-published in 4 journals that implemented the RRIDs Standard for
research resource citation, which draws data from the antibodyregistry.org as the source of antibody
identifiers[108] (see also group at Force11[109]).
Antibody regions can be used to further biomedical research by acting as a guide for drugs to reach
their target. Several application involve using bacterial plasmids to tag plasmids with the Fc region of
the antibody such as pFUSE-Fc plasmid.

Regulations

Production and testing

Traditionally, most antibodies are produced by hybridoma cell lines through immortalization of
antibody-producing cells by chemically-induced fusion with myeloma cells. In some cases, additional
fusions with other lines have created "triomas" and "quadromas". The manufacturing process should
be appropriately described and validated. Validation studies should at least include:

The demonstration that the process is able to produce in good quality (the process should be
validated)
The efficiency of the antibody purification (all impurities and virus must be eliminated)
The characterization of purified antibody (physicochemical characterization, immunological
properties, biological activities, contaminants, ...)
Determination of the virus clearance studies

Before clinical trials


Product safety testing: Sterility (bacteria and fungi), in vitro and in vivo testing for adventitious
viruses, murine retrovirus testing..., product safety data needed before the initiation of feasibility
trials in serious or immediately life-threatening conditions, it serves to evaluate dangerous
potential of the product.
Feasibility testing: These are pilot studies whose objectives include, among others, early
characterization of safety and initial proof of concept in a small specific patient population (in vitro
or in vivo testing).

Preclinical studies
Testing cross-reactivity of antibody: to highlight unwanted interactions (toxicity) of antibodies with
previously characterized tissues. This study can be performed in vitro (reactivity of the antibody or
immunoconjugate should be determined with a quick-frozen adult tissues) or in vivo (with
appropriates animal models).
Preclinical pharmacology and toxicity testing: preclinical safety testing of antibody is designed to
identify possible toxicity in humans, to estimate the likelihood and severity of potential adverse
events in humans, and to identify a safe starting dose and dose escalation, when possible.
Animal toxicity studies: Acute toxicity testing, repeat-dose toxicity testing, long-term toxicity testing
Pharmacokinetics and pharmacodynamics testing: Use for determinate clinical dosages, antibody
activities, evaluation of the potential clinical effects

Structure prediction and computational antibody design


The importance of antibodies in health care and the biotechnology industry demands knowledge of
their structures at high resolution. This information is used for protein engineering, modifying the
antigen binding affinity, and identifying an epitope, of a given antibody. X-ray crystallography is one
commonly used method for determining antibody structures. However, crystallizing an antibody is
often laborious and time-consuming. Computational approaches provide a cheaper and faster
alternative to crystallography, but their results are more equivocal, since they do not produce
empirical structures. Online web servers such as Web Antibody Modeling (WAM)[110] and Prediction
of Immunoglobulin Structure (PIGS)[111] enables computational modeling of antibody variable
regions. Rosetta Antibody is a novel antibody FV region structure prediction server, which
incorporates sophisticated techniques to minimize CDR loops and optimize the relative orientation of
the light and heavy chains, as well as homology models that predict successful docking of antibodies
with their unique antigen.[112] However, describing an antibody's binding site using only one single
static structure limits the understanding and characterization of the antibody's function and
properties. To improve antibody structure prediction and to take the strongly correlated CDR loop and
interface movements into account, antibody paratopes should be described as interconverting states
in solution with varying probabilities.[17]

The ability to describe the antibody through binding affinity to the antigen is supplemented by
information on antibody structure and amino acid sequences for the purpose of patent claims.[113]
Several methods have been presented for computational design of antibodies based on the structural
bioinformatics studies of antibody CDRs.[114][115][116]

There are a variety of methods used to sequence an antibody including Edman degradation, cDNA,
etc.; albeit one of the most common modern uses for peptide/protein identification is liquid
chromatography coupled with tandem mass spectrometry (LC-MS/MS).[117] High volume antibody
sequencing methods require computational approaches for the data analysis, including de novo
sequencing directly from tandem mass spectra[118] and database search methods that use existing
protein sequence databases.[119][120] Many versions of shotgun protein sequencing are able to increase
the coverage by utilizing CID/HCD/ETD[121] fragmentation methods and other techniques, and they
have achieved substantial progress in attempt to fully sequence proteins, especially antibodies. Other
methods have assumed the existence of similar proteins,[122] a known genome sequence,[123] or
combined top-down and bottom up approaches.[124] Current technologies have the ability to assemble
protein sequences with high accuracy by integrating de novo sequencing peptides, intensity, and
positional confidence scores from database and homology searches.[125]

Antibody mimetic
Antibody mimetics are organic compounds, like antibodies, that can specifically bind antigens. They
consist of artificial peptides or proteins, or aptamer-based nucleic acid molecules with a molar mass of
about 3 to 20 kDa. Antibody fragments, such as Fab and nanobodies are not considered as antibody
mimetics. Common advantages over antibodies are better solubility, tissue penetration, stability
towards heat and enzymes, and comparatively low production costs. Antibody mimetics have being
developed and commercialized as research, diagnostic and therapeutic agents.[126]

Binding antibody unit


BAU (binding antibody unit, often as BAU/mL) is a measurement unit defined by the WHO for the
comparison of assays detecting the same class of immunoglobulins with the same
specificity.[127][128][129]
See also
Affimer Magnetic immunoassay
Anti-mitochondrial antibodies Microantibody
Anti-nuclear antibodies Monoclonal antibody
Antibody mimetic Neutralizing antibody
Aptamer Optimer Ligand
Colostrum Secondary antibodies
ELISA Single-domain antibody
Humoral immunity Slope spectroscopy
Immunology Synthetic antibody
Immunosuppressive drug Western blot normalization
Intravenous immunoglobulin (IVIg)

References
1. Rhoades RA, Pflanzer RG (2002). Human Physiology (https://archive.org/details/humanphysiology
w00rodn/page/584) (5th ed.). Thomson Learning. p. 584 (https://archive.org/details/humanphysiol
ogyw00rodn/page/584). ISBN 978-0-534-42174-8.
2. Janeway C (2001). Immunobiology (https://archive.org/details/immunobiology00char) (5th ed.).
Garland Publishing. ISBN 978-0-8153-3642-6.
3. Litman GW, Rast JP, Shamblott MJ, Haire RN, Hulst M, Roess W, Litman RT, Hinds-Frey KR,
Zilch A, Amemiya CT (January 1993). "Phylogenetic diversification of immunoglobulin genes and
the antibody repertoire" (https://doi.org/10.1093%2Foxfordjournals.molbev.a040000). Molecular
Biology and Evolution. 10 (1): 60–72. doi:10.1093/oxfordjournals.molbev.a040000 (https://doi.org/
10.1093%2Foxfordjournals.molbev.a040000). PMID 8450761 (https://pubmed.ncbi.nlm.nih.gov/84
50761).
4. Borghesi L, Milcarek C (2006). "From B cell to plasma cell: regulation of V(D)J recombination and
antibody secretion". Immunologic Research. 36 (1–3): 27–32. doi:10.1385/IR:36:1:27 (https://doi.o
rg/10.1385%2FIR%3A36%3A1%3A27). PMID 17337763 (https://pubmed.ncbi.nlm.nih.gov/173377
63). S2CID 27041937 (https://api.semanticscholar.org/CorpusID:27041937).
5. Pier GB, Lyczak JB, Wetzler LM (2004). Immunology, Infection, and Immunity. ASM Press.
ISBN 978-1-55581-246-1.
6. "MeSH Browser – Immunoglobulins" (https://meshb.nlm.nih.gov/record/ui?ui=D007136).
meshb.nlm.nih.gov. Retrieved 25 October 2020.
7. Reth M (August 2013). "Matching cellular dimensions with molecular sizes" (http://www.slas.ac.cn/
upload/20130815-4.pdf) (PDF). Nature Immunology. 14 (8): 765–7. doi:10.1038/ni.2621 (https://do
i.org/10.1038%2Fni.2621). PMID 23867923 (https://pubmed.ncbi.nlm.nih.gov/23867923).
S2CID 24333875 (https://api.semanticscholar.org/CorpusID:24333875).
8. Woof JM, Burton DR (February 2004). "Human antibody-Fc receptor interactions illuminated by
crystal structures". Nature Reviews. Immunology. 4 (2): 89–99. doi:10.1038/nri1266 (https://doi.or
g/10.1038%2Fnri1266). PMID 15040582 (https://pubmed.ncbi.nlm.nih.gov/15040582).
S2CID 30584218 (https://api.semanticscholar.org/CorpusID:30584218).
9. Barclay AN (August 2003). "Membrane proteins with immunoglobulin-like domains—a master
superfamily of interaction molecules". Seminars in Immunology. 15 (4): 215–23.
doi:10.1016/S1044-5323(03)00047-2 (https://doi.org/10.1016%2FS1044-5323%2803%2900047-
2). PMID 14690046 (https://pubmed.ncbi.nlm.nih.gov/14690046).
10. Putnam FW, Liu YS, Low TL (April 1979). "Primary structure of a human IgA1 immunoglobulin. IV.
Streptococcal IgA1 protease, digestion, Fab and Fc fragments, and the complete amino acid
sequence of the alpha 1 heavy chain" (https://doi.org/10.1016%2FS0021-9258%2817%2930153-
9). The Journal of Biological Chemistry. 254 (8): 2865–74. doi:10.1016/S0021-9258(17)30153-9 (h
ttps://doi.org/10.1016%2FS0021-9258%2817%2930153-9). PMID 107164 (https://pubmed.ncbi.nl
m.nih.gov/107164).
11. Delves PJ, Martin SJ, Burton DR, Roitt IM (2017). Roitt's essential immunology (https://www.world
cat.org/title/roitts-essential-immunology/oclc/949912256) (13th ed.). Chichester, West Sussex.
ISBN 978-1-118-41577-1. OCLC 949912256 (https://www.worldcat.org/oclc/949912256).
12. "MeSH Browser – gamma-Globulins" (https://meshb.nlm.nih.gov/record/ui?ui=D005719).
meshb.nlm.nih.gov. Retrieved 18 October 2020.
13. "Recommendations for the nomenclature of human immunoglobulins" (https://pubmed.ncbi.nlm.ni
h.gov/5031329). Journal of Immunology. 108 (6): 1733–4. June 1972.
doi:10.4049/jimmunol.108.6.1733 (https://doi.org/10.4049%2Fjimmunol.108.6.1733).
PMID 5031329 (https://pubmed.ncbi.nlm.nih.gov/5031329).
14. Al-Lazikani B, Lesk AM, Chothia C (November 1997). "Standard conformations for the canonical
structures of immunoglobulins". Journal of Molecular Biology. 273 (4): 927–48.
doi:10.1006/jmbi.1997.1354 (https://doi.org/10.1006%2Fjmbi.1997.1354). PMID 9367782 (https://
pubmed.ncbi.nlm.nih.gov/9367782).
15. North B, Lehmann A, Dunbrack RL (February 2011). "A new clustering of antibody CDR loop
conformations" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3065967). Journal of Molecular
Biology. 406 (2): 228–56. doi:10.1016/j.jmb.2010.10.030 (https://doi.org/10.1016%2Fj.jmb.2010.1
0.030). PMC 3065967 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3065967). PMID 21035459
(https://pubmed.ncbi.nlm.nih.gov/21035459).
16. Nikoloudis D, Pitts JE, Saldanha JW (2014). "A complete, multi-level conformational clustering of
antibody complementarity-determining regions" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC41
03072). PeerJ. 2 (e456): e456. doi:10.7717/peerj.456 (https://doi.org/10.7717%2Fpeerj.456).
PMC 4103072 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4103072). PMID 25071986 (https://
pubmed.ncbi.nlm.nih.gov/25071986).
17. Fernández-Quintero ML, Georges G, Varga JM, Liedl KR (2021). "Ensembles in solution as a new
paradigm for antibody structure prediction and design" (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC8158028). mAbs. 13 (1): 1923122. doi:10.1080/19420862.2021.1923122 (https://doi.org/10.1
080%2F19420862.2021.1923122). PMC 8158028 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
8158028). PMID 34030577 (https://pubmed.ncbi.nlm.nih.gov/34030577).
18. Woof JM, Russell RW (2011). "Structure and function relationships in IgA" (https://doi.org/10.103
8%2Fmi.2011.39). Mucosal Immunology. 4 (6): 590–597. doi:10.1038/mi.2011.39 (https://doi.org/1
0.1038%2Fmi.2011.39). PMID 21937984 (https://pubmed.ncbi.nlm.nih.gov/21937984).
19. Maverakis E, Kim K, Shimoda M, Gershwin ME, Patel F, Wilken R, Raychaudhuri S, Ruhaak LR,
Lebrilla CB (February 2015). "Glycans in the immune system and The Altered Glycan Theory of
Autoimmunity: a critical review" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4340844). Journal
of Autoimmunity. 57 (6): 1–13. doi:10.1016/j.jaut.2014.12.002 (https://doi.org/10.1016%2Fj.jaut.20
14.12.002). PMC 4340844 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4340844).
PMID 25578468 (https://pubmed.ncbi.nlm.nih.gov/25578468).
20. Mattu TS, Pleass RJ, Willis AC, Kilian M, Wormald MR, Lellouch AC, Rudd PM, Woof JM, Dwek
RA (January 1998). "The glycosylation and structure of human serum IgA1, Fab, and Fc regions
and the role of N-glycosylation on Fcα receptor interactions" (https://doi.org/10.1074%2Fjbc.273.4.
2260). The Journal of Biological Chemistry. 273 (4): 2260–72. doi:10.1074/jbc.273.4.2260 (https://
doi.org/10.1074%2Fjbc.273.4.2260). PMID 9442070 (https://pubmed.ncbi.nlm.nih.gov/9442070).
21. Cobb BA (March 2020). "The history of IgG glycosylation and where we are now" (https://www.ncb
i.nlm.nih.gov/pmc/articles/PMC7109348). Glycobiology. 30 (4): 202–213.
doi:10.1093/glycob/cwz065 (https://doi.org/10.1093%2Fglycob%2Fcwz065). PMC 7109348 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC7109348). PMID 31504525 (https://pubmed.ncbi.nlm.ni
h.gov/31504525).
22. Roux KH (October 1999). "Immunoglobulin structure and function as revealed by electron
microscopy". International Archives of Allergy and Immunology. 120 (2): 85–99.
doi:10.1159/000024226 (https://doi.org/10.1159%2F000024226). PMID 10545762 (https://pubme
d.ncbi.nlm.nih.gov/10545762). S2CID 12187510 (https://api.semanticscholar.org/CorpusID:12187
510).
23. Parker DC (1993). "T cell-dependent B cell activation". Annual Review of Immunology. 11 (1):
331–60. doi:10.1146/annurev.iy.11.040193.001555 (https://doi.org/10.1146%2Fannurev.iy.11.0401
93.001555). PMID 8476565 (https://pubmed.ncbi.nlm.nih.gov/8476565).
24. Wintrobe MM (2004). Greer JG, Foerster F, Lukens JN, Rodgers GM, Paraskevas F (eds.).
Wintrobe's clinical hematology (11 ed.). Hagerstown, MD: Lippincott Williams & Wilkins. pp. 453–
456. ISBN 978-0-7817-3650-3.
25. Tolar P, Sohn HW, Pierce SK (February 2008). "Viewing the antigen-induced initiation of B-cell
activation in living cells" (https://zenodo.org/record/1230708). Immunological Reviews. 221 (1):
64–76. doi:10.1111/j.1600-065X.2008.00583.x (https://doi.org/10.1111%2Fj.1600-065X.2008.0058
3.x). PMID 18275475 (https://pubmed.ncbi.nlm.nih.gov/18275475). S2CID 38464264 (https://api.s
emanticscholar.org/CorpusID:38464264).
26. Williams CM, Galli SJ (May 2000). "The diverse potential effector and immunoregulatory roles of
mast cells in allergic disease" (https://doi.org/10.1067%2Fmai.2000.106485). The Journal of
Allergy and Clinical Immunology. 105 (5): 847–59. doi:10.1067/mai.2000.106485 (https://doi.org/1
0.1067%2Fmai.2000.106485). PMID 10808163 (https://pubmed.ncbi.nlm.nih.gov/10808163).
27. Underdown BJ, Schiff JM (1986). "Immunoglobulin A: strategic defense initiative at the mucosal
surface". Annual Review of Immunology. 4 (1): 389–417.
doi:10.1146/annurev.iy.04.040186.002133 (https://doi.org/10.1146%2Fannurev.iy.04.040186.0021
33). PMID 3518747 (https://pubmed.ncbi.nlm.nih.gov/3518747).
28. Geisberger R, Lamers M, Achatz G (August 2006). "The riddle of the dual expression of IgM and
IgD" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1782314). Immunology. 118 (4): 429–37.
doi:10.1111/j.1365-2567.2006.02386.x (https://doi.org/10.1111%2Fj.1365-2567.2006.02386.x).
PMC 1782314 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1782314). PMID 16895553 (https://
pubmed.ncbi.nlm.nih.gov/16895553).
29. Chen K, Xu W, Wilson M, He B, Miller NW, Bengtén E, Edholm ES, Santini PA, Rath P, Chiu A,
Cattalini M, Litzman J, B Bussel J, Huang B, Meini A, Riesbeck K, Cunningham-Rundles C,
Plebani A, Cerutti A (August 2009). "Immunoglobulin D enhances immune surveillance by
activating antimicrobial, proinflammatory and B cell-stimulating programs in basophils" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC2785232). Nature Immunology. 10 (8): 889–98.
doi:10.1038/ni.1748 (https://doi.org/10.1038%2Fni.1748). PMC 2785232 (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC2785232). PMID 19561614 (https://pubmed.ncbi.nlm.nih.gov/19561614).
30. Goding JW (1978). "Allotypes of IgM and IgD receptors in the mouse: a probe for lymphocyte
differentiation". Contemporary Topics in Immunobiology. 8: 203–43. doi:10.1007/978-1-4684-
0922-2_7 (https://doi.org/10.1007%2F978-1-4684-0922-2_7). ISBN 978-1-4684-0924-6.
PMID 357078 (https://pubmed.ncbi.nlm.nih.gov/357078).
31. Litman GW, Rast JP, Fugmann SD (August 2010). "The origins of vertebrate adaptive immunity" (h
ttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC2919748). Nature Reviews. Immunology. 10 (8):
543–53. doi:10.1038/nri2807 (https://doi.org/10.1038%2Fnri2807). PMC 2919748 (https://www.nc
bi.nlm.nih.gov/pmc/articles/PMC2919748). PMID 20651744 (https://pubmed.ncbi.nlm.nih.gov/2065
1744).
32. Litman GW, Rast JP, Fugmann SD (August 2010). "The origins of vertebrate adaptive immunity" (h
ttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC2919748). Nature Reviews. Immunology. John Wiley
& Sons, Ltd. 10 (8): 543–53. doi:10.1002/9783527699124.ch4 (https://doi.org/10.1002%2F978352
7699124.ch4). ISBN 978-3-527-69912-4. PMC 2919748 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC2919748). PMID 20651744 (https://pubmed.ncbi.nlm.nih.gov/20651744).
33. Lundqvist ML, Middleton DL, Radford C, Warr GW, Magor KE (2006). "Immunoglobulins of the
non-galliform birds: antibody expression and repertoire in the duck" (https://www.ncbi.nlm.nih.gov/
pmc/articles/PMC1317265). Developmental and Comparative Immunology. 30 (1–2): 93–100.
doi:10.1016/j.dci.2005.06.019 (https://doi.org/10.1016%2Fj.dci.2005.06.019). PMC 1317265 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC1317265). PMID 16150486 (https://pubmed.ncbi.nlm.ni
h.gov/16150486).
34. Berstein RM, Schluter SF, Shen S, Marchalonis JJ (April 1996). "A new high molecular weight
immunoglobulin class from the carcharhine shark: implications for the properties of the primordial
immunoglobulin" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC39599). Proceedings of the
National Academy of Sciences of the United States of America. 93 (8): 3289–93.
Bibcode:1996PNAS...93.3289B (https://ui.adsabs.harvard.edu/abs/1996PNAS...93.3289B).
doi:10.1073/pnas.93.8.3289 (https://doi.org/10.1073%2Fpnas.93.8.3289). PMC 39599 (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC39599). PMID 8622930 (https://pubmed.ncbi.nlm.nih.gov/862
2930).
35. Salinas, I., & Parra, D. (2015). Fish mucosal immunity: Intestine. In Mucosal Health in
Aquaculture. Elsevier Inc. https://doi.org/10.1016/B978-0-12-417186-2.00006-6
36. Ravetch JV, Bolland S (2001). "IgG Fc receptors". Annual Review of Immunology. 19 (1): 275–90.
doi:10.1146/annurev.immunol.19.1.275 (https://doi.org/10.1146%2Fannurev.immunol.19.1.275).
PMID 11244038 (https://pubmed.ncbi.nlm.nih.gov/11244038).
37. Rus H, Cudrici C, Niculescu F (2005). "The role of the complement system in innate immunity".
Immunologic Research. 33 (2): 103–12. doi:10.1385/IR:33:2:103 (https://doi.org/10.1385%2FIR%
3A33%3A2%3A103). PMID 16234578 (https://pubmed.ncbi.nlm.nih.gov/16234578).
S2CID 46096567 (https://api.semanticscholar.org/CorpusID:46096567).
38. Racaniello, Vincent (6 October 2009). "Natural antibody protects against viral infection" (http://ww
w.virology.ws/2009/10/06/natural-antibody-protects-against-viral-infection/). Virology Blog.
Archived (https://web.archive.org/web/20100220015318/http://www.virology.ws/2009/10/06/natural
-antibody-protects-against-viral-infection/) from the original on 20 February 2010. Retrieved
22 January 2010.
39. Milland J, Sandrin MS (December 2006). "ABO blood group and related antigens, natural
antibodies and transplantation". Tissue Antigens. 68 (6): 459–66. doi:10.1111/j.1399-
0039.2006.00721.x (https://doi.org/10.1111%2Fj.1399-0039.2006.00721.x). PMID 17176435 (http
s://pubmed.ncbi.nlm.nih.gov/17176435).
40. Mian IS, Bradwell AR, Olson AJ (January 1991). "Structure, function and properties of antibody
binding sites". Journal of Molecular Biology. 217 (1): 133–51. doi:10.1016/0022-2836(91)90617-F
(https://doi.org/10.1016%2F0022-2836%2891%2990617-F). PMID 1988675 (https://pubmed.ncbi.
nlm.nih.gov/1988675).
41. Fanning LJ, Connor AM, Wu GE (April 1996). "Development of the immunoglobulin repertoire".
Clinical Immunology and Immunopathology. 79 (1): 1–14. doi:10.1006/clin.1996.0044 (https://doi.o
rg/10.1006%2Fclin.1996.0044). PMID 8612345 (https://pubmed.ncbi.nlm.nih.gov/8612345).
42. Nemazee D (October 2006). "Receptor editing in lymphocyte development and central tolerance".
Nature Reviews. Immunology. 6 (10): 728–40. doi:10.1038/nri1939 (https://doi.org/10.1038%2Fnri
1939). PMID 16998507 (https://pubmed.ncbi.nlm.nih.gov/16998507). S2CID 2234228 (https://api.
semanticscholar.org/CorpusID:2234228).
43. Peter Parham. The Immune System. 2nd ed. Garland Science: New York, 2005. pg.47–62
44. Market E, Papavasiliou FN (October 2003). "V(D)J recombination and the evolution of the
adaptive immune system" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC212695). PLOS Biology.
1 (1): E16. doi:10.1371/journal.pbio.0000016 (https://doi.org/10.1371%2Fjournal.pbio.0000016).
PMC 212695 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC212695). PMID 14551913 (https://pu
bmed.ncbi.nlm.nih.gov/14551913).
45. Bergman Y, Cedar H (October 2004). "A stepwise epigenetic process controls immunoglobulin
allelic exclusion". Nature Reviews. Immunology. 4 (10): 753–61. doi:10.1038/nri1458 (https://doi.or
g/10.1038%2Fnri1458). PMID 15459667 (https://pubmed.ncbi.nlm.nih.gov/15459667).
S2CID 8579156 (https://api.semanticscholar.org/CorpusID:8579156).
46. Diaz M, Casali P (April 2002). "Somatic immunoglobulin hypermutation" (https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC4621002). Current Opinion in Immunology. 14 (2): 235–40.
doi:10.1016/S0952-7915(02)00327-8 (https://doi.org/10.1016%2FS0952-7915%2802%2900327-
8). PMC 4621002 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4621002). PMID 11869898 (http
s://pubmed.ncbi.nlm.nih.gov/11869898).
47. Honjo T, Habu S (1985). "Origin of immune diversity: genetic variation and selection". Annual
Review of Biochemistry. 54 (1): 803–30. doi:10.1146/annurev.bi.54.070185.004103 (https://doi.or
g/10.1146%2Fannurev.bi.54.070185.004103). PMID 3927822 (https://pubmed.ncbi.nlm.nih.gov/39
27822).
48. Or-Guil M, Wittenbrink N, Weiser AA, Schuchhardt J (April 2007). "Recirculation of germinal center
B cells: a multilevel selection strategy for antibody maturation". Immunological Reviews. 216: 130–
41. doi:10.1111/j.1600-065X.2007.00507.x (https://doi.org/10.1111%2Fj.1600-065X.2007.00507.x).
PMID 17367339 (https://pubmed.ncbi.nlm.nih.gov/17367339). S2CID 37636392 (https://api.seman
ticscholar.org/CorpusID:37636392).
49. Neuberger MS, Ehrenstein MR, Rada C, Sale J, Batista FD, Williams G, Milstein C (March 2000).
"Memory in the B-cell compartment: antibody affinity maturation" (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC1692737). Philosophical Transactions of the Royal Society of London. Series B,
Biological Sciences. 355 (1395): 357–60. doi:10.1098/rstb.2000.0573 (https://doi.org/10.1098%2F
rstb.2000.0573). PMC 1692737 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1692737).
PMID 10794054 (https://pubmed.ncbi.nlm.nih.gov/10794054).
50. Stavnezer J, Amemiya CT (August 2004). "Evolution of isotype switching". Seminars in
Immunology. 16 (4): 257–75. doi:10.1016/j.smim.2004.08.005 (https://doi.org/10.1016%2Fj.smim.
2004.08.005). PMID 15522624 (https://pubmed.ncbi.nlm.nih.gov/15522624).
51. Durandy A (August 2003). "Activation-induced cytidine deaminase: a dual role in class-switch
recombination and somatic hypermutation". European Journal of Immunology. 33 (8): 2069–73.
doi:10.1002/eji.200324133 (https://doi.org/10.1002%2Feji.200324133). PMID 12884279 (https://p
ubmed.ncbi.nlm.nih.gov/12884279). S2CID 32059768 (https://api.semanticscholar.org/CorpusID:3
2059768).
52. Casali P, Zan H (November 2004). "Class switching and Myc translocation: how does DNA
break?" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4625794). Nature Immunology. 5 (11):
1101–3. doi:10.1038/ni1104-1101 (https://doi.org/10.1038%2Fni1104-1101). PMC 4625794 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC4625794). PMID 15496946 (https://pubmed.ncbi.nlm.ni
h.gov/15496946).
53. Lieber MR, Yu K, Raghavan SC (September 2006). "Roles of nonhomologous DNA end joining,
V(D)J recombination, and class switch recombination in chromosomal translocations". DNA
Repair. 5 (9–10): 1234–45. doi:10.1016/j.dnarep.2006.05.013 (https://doi.org/10.1016%2Fj.dnare
p.2006.05.013). PMID 16793349 (https://pubmed.ncbi.nlm.nih.gov/16793349).
54. p. 22 (https://books.google.com/books?id=TfW5sUfeM5gC&pg=PA22) in: Shoenfeld Y, Meroni PL,
Gershwin ME (2007). Autoantibodie. Amsterdam; Boston: Elsevier. ISBN 978-0-444-52763-9.
55. Spiess C, Zhai Q, Carter PJ (October 2015). "Alternative molecular formats and therapeutic
applications for bispecific antibodies" (https://doi.org/10.1016%2Fj.molimm.2015.01.003).
Molecular Immunology. 67 (2 Pt A): 95–106. doi:10.1016/j.molimm.2015.01.003 (https://doi.org/10.
1016%2Fj.molimm.2015.01.003). PMID 25637431 (https://pubmed.ncbi.nlm.nih.gov/25637431).
56. Farlex dictionary > polyvalent (http://medical-dictionary.thefreedictionary.com/polyvalent) Citing:
The American Heritage Medical Dictionary. 2004
57. Gunasekaran K, Pentony M, Shen M, Garrett L, Forte C, Woodward A, Ng SB, Born T, Retter M,
Manchulenko K, Sweet H, Foltz IN, Wittekind M, Yan W (June 2010). "Enhancing antibody Fc
heterodimer formation through electrostatic steering effects: applications to bispecific molecules
and monovalent IgG" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2885242). The Journal of
Biological Chemistry. 285 (25): 19637–46. doi:10.1074/jbc.M110.117382 (https://doi.org/10.1074%
2Fjbc.M110.117382). PMC 2885242 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2885242).
PMID 20400508 (https://pubmed.ncbi.nlm.nih.gov/20400508).
58. Muller KM (1998). "The first constant domain (CH1 and CL) of an antibody used as
heterodimerization domain for bispecific miniantibodies" (https://doi.org/10.1016%2Fs0014-5793%
2898%2900021-0). FEBS Letters. 422 (2): 259–264. doi:10.1016/s0014-5793(98)00021-0 (https://
doi.org/10.1016%2Fs0014-5793%2898%2900021-0). PMID 9490020 (https://pubmed.ncbi.nlm.ni
h.gov/9490020). S2CID 35243494 (https://api.semanticscholar.org/CorpusID:35243494).
59. Gao C, Mao S, Lo CH, Wirsching P, Lerner RA, Janda KD (May 1999). "Making artificial
antibodies: a format for phage display of combinatorial heterodimeric arrays" (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC26829). Proceedings of the National Academy of Sciences of the
United States of America. 96 (11): 6025–30. Bibcode:1999PNAS...96.6025G (https://ui.adsabs.har
vard.edu/abs/1999PNAS...96.6025G). doi:10.1073/pnas.96.11.6025 (https://doi.org/10.1073%2Fp
nas.96.11.6025). PMC 26829 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC26829).
PMID 10339535 (https://pubmed.ncbi.nlm.nih.gov/10339535).
60. Lindenmann J (April 1984). "Origin of the terms 'antibody' and 'antigen' ". Scandinavian Journal of
Immunology. 19 (4): 281–5. doi:10.1111/j.1365-3083.1984.tb00931.x (https://doi.org/10.1111%2Fj.
1365-3083.1984.tb00931.x). PMID 6374880 (https://pubmed.ncbi.nlm.nih.gov/6374880).
S2CID 222200504 (https://api.semanticscholar.org/CorpusID:222200504).
61. Padlan EA (February 1994). "Anatomy of the antibody molecule" (https://zenodo.org/record/12583
37). Molecular Immunology. 31 (3): 169–217. doi:10.1016/0161-5890(94)90001-9 (https://doi.org/1
0.1016%2F0161-5890%2894%2990001-9). PMID 8114766 (https://pubmed.ncbi.nlm.nih.gov/8114
766).
62. Sauter E (10 November 2018). "New Sculpture Portraying Human Antibody as Protective Angel
Installed on Scripps Florida Campus" (http://www.scripps.edu/newsandviews/e_20081110/sculptur
e.html). News & Views. Vol. 8, no. 34. The Scripps Research Institute. Archived (https://web.archiv
e.org/web/20110110070639/http://www.scripps.edu/newsandviews/e_20081110/sculpture.html)
from the original on 10 January 2011. Retrieved 12 December 2008.
63. Pescovitz D (22 October 2008). "Protein sculpture inspired by Vitruvian Man" (https://www.boingbo
ing.net/2008/10/22/protein-sculpture-in.html). boingboing (Blog). Archived (https://web.archive.org/
web/20101104033646/http://boingboing.net/2008/10/22/protein-sculpture-in.html) from the original
on 4 November 2010. Retrieved 12 December 2008.
64. Emil von Behring – Biographical. NobelPrize.org. Nobel Media AB 2020. Mon. 20 January 2020.
<https://www.nobelprize.org/prizes/medicine/1901/behring/biographical/>
65. AGN (August 1931). "The Late Baron Shibasaburo Kitasato" (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC382621). Canadian Medical Association Journal. 25 (2): 206. PMC 382621 (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC382621). PMID 20318414 (https://pubmed.ncbi.nlm.nih.gov/203
18414).
66. Winau F, Westphal O, Winau R (July 2004). "Paul Ehrlich—in search of the magic bullet".
Microbes and Infection. 6 (8): 786–9. doi:10.1016/j.micinf.2004.04.003 (https://doi.org/10.1016%2
Fj.micinf.2004.04.003). PMID 15207826 (https://pubmed.ncbi.nlm.nih.gov/15207826).
67. Silverstein AM (May 2003). "Cellular versus humoral immunology: a century-long dispute" (https://
doi.org/10.1038%2Fni0503-425). Nature Immunology. 4 (5): 425–8. doi:10.1038/ni0503-425 (http
s://doi.org/10.1038%2Fni0503-425). PMID 12719732
(https://pubmed.ncbi.nlm.nih.gov/12719732). S2CID 31571243 (https://api.semanticscholar.org/Co
rpusID:31571243).
68. Van Epps HL (January 2006). "Michael Heidelberger and the demystification of antibodies" (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC2118068). The Journal of Experimental Medicine. 203
(1): 5. doi:10.1084/jem.2031fta (https://doi.org/10.1084%2Fjem.2031fta). PMC 2118068 (https://w
ww.ncbi.nlm.nih.gov/pmc/articles/PMC2118068). PMID 16523537 (https://pubmed.ncbi.nlm.nih.go
v/16523537).
69. Marrack JR (1938). Chemistry of antigens and antibodies (2nd ed.). London: His Majesty's
Stationery Office. OCLC 3220539 (https://www.worldcat.org/oclc/3220539).
70. "The Linus Pauling Papers: How Antibodies and Enzymes Work" (http://profiles.nlm.nih.gov/MM/Vi
ews/Exhibit/narrative/specificity.html). Archived (https://web.archive.org/web/20101205061247/htt
p://profiles.nlm.nih.gov/MM/Views/Exhibit/narrative/specificity.html) from the original on 5
December 2010. Retrieved 5 June 2007.
71. Silverstein AM (December 2004). "Labeled antigens and antibodies: the evolution of magic
markers and magic bullets" (https://web.archive.org/web/20090325001032/http://users.path.ox.ac.
uk/~seminars/halelibrary/Paper%2018.pdf) (PDF). Nature Immunology. 5 (12): 1211–7.
doi:10.1038/ni1140 (https://doi.org/10.1038%2Fni1140). PMID 15549122 (https://pubmed.ncbi.nl
m.nih.gov/15549122). S2CID 40595920 (https://api.semanticscholar.org/CorpusID:40595920).
Archived from the original (http://users.path.ox.ac.uk/~seminars/halelibrary/Paper%2018.pdf)
(PDF) on 25 March 2009.
72. Edelman GM, Gally JA (August 1962). "The nature of Bence-Jones proteins. Chemical similarities
to polypetide chains of myeloma globulins and normal gamma-globulins" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC2137388). The Journal of Experimental Medicine. 116 (2): 207–27.
doi:10.1084/jem.116.2.207 (https://doi.org/10.1084%2Fjem.116.2.207). PMC 2137388 (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC2137388). PMID 13889153 (https://pubmed.ncbi.nlm.nih.gov/
13889153).
73. Stevens FJ, Solomon A, Schiffer M (July 1991). "Bence Jones proteins: a powerful tool for the
fundamental study of protein chemistry and pathophysiology" (https://digital.library.unt.edu/ark:/67
531/metadc1400136/). Biochemistry. 30 (28): 6803–5. doi:10.1021/bi00242a001 (https://doi.org/1
0.1021%2Fbi00242a001). PMID 2069946 (https://pubmed.ncbi.nlm.nih.gov/2069946).
74. Raju TN (September 1999). "The Nobel chronicles. 1972: Gerald M Edelman (b 1929) and
Rodney R Porter (1917–85)". Lancet. 354 (9183): 1040. doi:10.1016/S0140-6736(05)76658-7 (htt
ps://doi.org/10.1016%2FS0140-6736%2805%2976658-7). PMID 10501404 (https://pubmed.ncbi.n
lm.nih.gov/10501404). S2CID 54380536 (https://api.semanticscholar.org/CorpusID:54380536).
75. Hochman J, Inbar D, Givol D (March 1973). "An active antibody fragment (Fv) composed of the
variable portions of heavy and light chains". Biochemistry. 12 (6): 1130–5.
doi:10.1021/bi00730a018 (https://doi.org/10.1021%2Fbi00730a018). PMID 4569769 (https://pubm
ed.ncbi.nlm.nih.gov/4569769).
76. Tomasi TB (October 1992). "The discovery of secretory IgA and the mucosal immune system".
Immunology Today. 13 (10): 416–8. doi:10.1016/0167-5699(92)90093-M (https://doi.org/10.1016%
2F0167-5699%2892%2990093-M). PMID 1343085 (https://pubmed.ncbi.nlm.nih.gov/1343085).
77. Preud'homme JL, Petit I, Barra A, Morel F, Lecron JC, Lelièvre E (October 2000). "Structural and
functional properties of membrane and secreted IgD". Molecular Immunology. 37 (15): 871–87.
doi:10.1016/S0161-5890(01)00006-2 (https://doi.org/10.1016%2FS0161-5890%2801%2900006-
2). PMID 11282392 (https://pubmed.ncbi.nlm.nih.gov/11282392).
78. Johansson SG (2006). "The discovery of immunoglobulin E". Allergy and Asthma Proceedings. 27
(2 Suppl 1): S3–6. PMID 16722325 (https://pubmed.ncbi.nlm.nih.gov/16722325).
79. Hozumi N, Tonegawa S (October 1976). "Evidence for somatic rearrangement of immunoglobulin
genes coding for variable and constant regions" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4
31171). Proceedings of the National Academy of Sciences of the United States of America. 73
(10): 3628–32. Bibcode:1976PNAS...73.3628H (https://ui.adsabs.harvard.edu/abs/1976PNAS...7
3.3628H). doi:10.1073/pnas.73.10.3628 (https://doi.org/10.1073%2Fpnas.73.10.3628).
PMC 431171 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC431171). PMID 824647 (https://pubm
ed.ncbi.nlm.nih.gov/824647).
80. "Animated depictions of how antibodies are used in ELISA assays" (https://web.archive.org/web/2
0110614091640/http://www.elispot-analyzers.de/english/elisa-animation.html). Cellular Technology
Ltd.—Europe. Archived from the original (http://www.immunospot.eu/elisa-animation.html) on 14
June 2011. Retrieved 8 May 2007.
81. "Animated depictions of how antibodies are used in ELISPOT assays" (https://web.archive.org/we
b/20110516142529/http://www.elispot-analyzers.de/english/elispot-animation.html). Cellular
Technology Ltd.—Europe. Archived from the original (http://www.immunospot.eu/elispot-animatio
n.html) on 16 May 2011. Retrieved 8 May 2007.
82. Stern P (2006). "Current possibilities of turbidimetry and nephelometry" (https://web.archive.org/w
eb/20080410032918/http://www.clsjep.cz/odkazy/kbm0603-146.pdf) (PDF). Klin Biochem Metab.
14 (3): 146–151. Archived from the original (http://www.clsjep.cz/odkazy/kbm0603-146.pdf) (PDF)
on 10 April 2008.
83. Dean L (2005). "Chapter 4: Hemolytic disease of the newborn" (https://www.ncbi.nlm.nih.gov/book
s/bv.fcgi?rid=rbcantigen.chapter.ch4). Blood Groups and Red Cell Antigens. NCBI Bethesda (MD):
National Library of Medicine (US).
84. Rodriguez EA, Wang Y, Crisp JL, Vera DR, Tsien RY, Ting R (May 2016). "New Dioxaborolane
Chemistry Enables [(18)F]-Positron-Emitting, Fluorescent [(18)F]-Multimodality Biomolecule
Generation from the Solid Phase" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4916912).
Bioconjugate Chemistry. 27 (5): 1390–1399. doi:10.1021/acs.bioconjchem.6b00164 (https://doi.or
g/10.1021%2Facs.bioconjchem.6b00164). PMC 4916912 (https://www.ncbi.nlm.nih.gov/pmc/articl
es/PMC4916912). PMID 27064381 (https://pubmed.ncbi.nlm.nih.gov/27064381).
85. Feldmann M, Maini RN (2001). "Anti-TNF alpha therapy of rheumatoid arthritis: what have we
learned?". Annual Review of Immunology. 19 (1): 163–96. doi:10.1146/annurev.immunol.19.1.163
(https://doi.org/10.1146%2Fannurev.immunol.19.1.163). PMID 11244034 (https://pubmed.ncbi.nl
m.nih.gov/11244034).
86. Doggrell SA (June 2003). "Is natalizumab a breakthrough in the treatment of multiple sclerosis?".
Expert Opinion on Pharmacotherapy. 4 (6): 999–1001. doi:10.1517/14656566.4.6.999 (https://doi.
org/10.1517%2F14656566.4.6.999). PMID 12783595
(https://pubmed.ncbi.nlm.nih.gov/12783595). S2CID 16104816 (https://api.semanticscholar.org/Co
rpusID:16104816).
87. Krueger GG, Langley RG, Leonardi C, Yeilding N, Guzzo C, Wang Y, Dooley LT, Lebwohl M
(February 2007). "A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis".
The New England Journal of Medicine. 356 (6): 580–92. doi:10.1056/NEJMoa062382 (https://doi.
org/10.1056%2FNEJMoa062382). PMID 17287478 (https://pubmed.ncbi.nlm.nih.gov/17287478).
88. Plosker GL, Figgitt DP (2003). "Rituximab: a review of its use in non-Hodgkin's lymphoma and
chronic lymphocytic leukaemia". Drugs. 63 (8): 803–43. doi:10.2165/00003495-200363080-00005
(https://doi.org/10.2165%2F00003495-200363080-00005). PMID 12662126 (https://pubmed.ncbi.
nlm.nih.gov/12662126).
89. Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, Slamon DJ, Murphy
M, Novotny WF, Burchmore M, Shak S, Stewart SJ (2001). "First-line Herceptin monotherapy in
metastatic breast cancer". Oncology. 61. 61 (Suppl. 2): 37–42. doi:10.1159/000055400 (https://do
i.org/10.1159%2F000055400). PMID 11694786 (https://pubmed.ncbi.nlm.nih.gov/11694786).
S2CID 24924864 (https://api.semanticscholar.org/CorpusID:24924864).
90. LeBien TW (July 2000). "Fates of human B-cell precursors" (https://web.archive.org/web/2010042
9220000/http://bloodjournal.hematologylibrary.org/cgi/content/full/96/1/9). Blood. 96 (1): 9–23.
doi:10.1182/blood.V96.1.9 (https://doi.org/10.1182%2Fblood.V96.1.9). PMID 10891425 (https://pu
bmed.ncbi.nlm.nih.gov/10891425). Archived from the original (http://bloodjournal.hematologylibrar
y.org/cgi/content/full/96/1/9) on 29 April 2010. Retrieved 31 March 2007.
91. Ghaffer A (26 March 2006). "Immunization" (http://pathmicro.med.sc.edu/ghaffar/immunization.ht
m). Immunology — Chapter 14. University of South Carolina School of Medicine. Archived (https://
web.archive.org/web/20101018004057/http://pathmicro.med.sc.edu/ghaffar/immunization.htm)
from the original on 18 October 2010. Retrieved 6 June 2007.
92. Urbaniak SJ, Greiss MA (March 2000). "RhD haemolytic disease of the fetus and the newborn".
Blood Reviews. 14 (1): 44–61. doi:10.1054/blre.1999.0123 (https://doi.org/10.1054%2Fblre.1999.0
123). PMID 10805260 (https://pubmed.ncbi.nlm.nih.gov/10805260).
93. Fung Kee Fung K, Eason E, Crane J, Armson A, De La Ronde S, Farine D, Keenan-Lindsay L,
Leduc L, Reid GJ, Aerde JV, Wilson RD, Davies G, Désilets VA, Summers A, Wyatt P, Young DC
(September 2003). "Prevention of Rh alloimmunization". Journal of Obstetrics and Gynaecology
Canada. 25 (9): 765–73. doi:10.1016/S1701-2163(16)31006-4 (https://doi.org/10.1016%2FS1701-
2163%2816%2931006-4). PMID 12970812 (https://pubmed.ncbi.nlm.nih.gov/12970812).
94. Tini M, Jewell UR, Camenisch G, Chilov D, Gassmann M (March 2002). "Generation and
application of chicken egg-yolk antibodies". Comparative Biochemistry and Physiology. Part A,
Molecular & Integrative Physiology. 131 (3): 569–74. doi:10.1016/S1095-6433(01)00508-6 (http
s://doi.org/10.1016%2FS1095-6433%2801%2900508-6). PMID 11867282 (https://pubmed.ncbi.nl
m.nih.gov/11867282).
95. Cole SP, Campling BG, Atlaw T, Kozbor D, Roder JC (June 1984). "Human monoclonal
antibodies". Molecular and Cellular Biochemistry. 62 (2): 109–20. doi:10.1007/BF00223301 (http
s://doi.org/10.1007%2FBF00223301). PMID 6087121 (https://pubmed.ncbi.nlm.nih.gov/6087121).
S2CID 12616168 (https://api.semanticscholar.org/CorpusID:12616168).
96. Kabir S (2002). "Immunoglobulin purification by affinity chromatography using protein A mimetic
ligands prepared by combinatorial chemical synthesis". Immunological Investigations. 31 (3–4):
263–78. doi:10.1081/IMM-120016245 (https://doi.org/10.1081%2FIMM-120016245).
PMID 12472184 (https://pubmed.ncbi.nlm.nih.gov/12472184). S2CID 12785078 (https://api.seman
ticscholar.org/CorpusID:12785078).
97. Brehm-Stecher BF, Johnson EA (September 2004). "Single-cell microbiology: tools, technologies,
and applications" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC515252). Microbiology and
Molecular Biology Reviews. 68 (3): 538–59, table of contents. doi:10.1128/MMBR.68.3.538-
559.2004 (https://doi.org/10.1128%2FMMBR.68.3.538-559.2004). PMC 515252 (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC515252). PMID 15353569 (https://pubmed.ncbi.nlm.nih.gov/153535
69).
98. Williams NE (2000). Immunoprecipitation procedures (https://archive.org/details/tetrahymenatherm
62acad/page/449). Methods in Cell Biology. Vol. 62. San Diego, CA : Academic Press. pp. 449–53
(https://archive.org/details/tetrahymenatherm62acad/page/449). doi:10.1016/S0091-
679X(08)61549-6 (https://doi.org/10.1016%2FS0091-679X%2808%2961549-6). ISBN 978-0-12-
544164-3. PMID 10503210 (https://pubmed.ncbi.nlm.nih.gov/10503210).
99. Kurien BT, Scofield RH (April 2006). "Western blotting". Methods. 38 (4): 283–93.
doi:10.1016/j.ymeth.2005.11.007 (https://doi.org/10.1016%2Fj.ymeth.2005.11.007).
PMID 16483794 (https://pubmed.ncbi.nlm.nih.gov/16483794).
100. Scanziani E (1998). "Immunohistochemical staining of fixed tissues" (https://archive.org/details/my
coplasmaprotoc00roge/page/133). Mycoplasma Protocols. Methods in Molecular Biology.
Vol. 104. Totowa, N.J. : Humana Press. pp. 133–40 (https://archive.org/details/mycoplasmaprotoc
00roge/page/133). doi:10.1385/0-89603-525-5:133 (https://doi.org/10.1385%2F0-89603-525-5%3
A133). ISBN 978-0-89603-525-6. PMID 9711649 (https://pubmed.ncbi.nlm.nih.gov/9711649).
101. Reen DJ (1994). "Enzyme-linked immunosorbent assay (ELISA)". Basic Protein and Peptide
Protocols. Methods in Molecular Biology. Vol. 32. pp. 461–6. doi:10.1385/0-89603-268-X:461 (http
s://doi.org/10.1385%2F0-89603-268-X%3A461). ISBN 978-0-89603-268-2. PMC 2366430 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC2366430). PMID 7951745 (https://pubmed.ncbi.nlm.nih.go
v/7951745).
102. Kalyuzhny AE (2005). "Chemistry and biology of the ELISPOT assay". Handbook of ELISPOT.
Methods in Molecular Biology. Vol. 302. pp. 15–31. doi:10.1385/1-59259-903-6:015 (https://doi.or
g/10.1385%2F1-59259-903-6%3A015). ISBN 978-1-59259-903-5. PMID 15937343 (https://pubme
d.ncbi.nlm.nih.gov/15937343).
103. Saper CB (December 2005). "An open letter to our readers on the use of antibodies" (https://doi.or
g/10.1002%2Fcne.20839). The Journal of Comparative Neurology. 493 (4): 477–8.
doi:10.1002/cne.20839 (https://doi.org/10.1002%2Fcne.20839). PMID 16304632 (https://pubmed.
ncbi.nlm.nih.gov/16304632). S2CID 14082678 (https://api.semanticscholar.org/CorpusID:1408267
8).
104. "NOT-OD-16-011: Implementing Rigor and Transparency in NIH & AHRQ Research Grant
Applications" (https://grants.nih.gov/grants/guide/notice-files/NOT-OD-16-011.html).
grants.nih.gov.
105. Vasilevsky NA, Brush MH, Paddock H, Ponting L, Tripathy SJ, Larocca GM, Haendel MA (2
September 2013). "On the reproducibility of science: unique identification of research resources in
the biomedical literature" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3771067). PeerJ. 1:
e148. doi:10.7717/peerj.148 (https://doi.org/10.7717%2Fpeerj.148). PMC 3771067 (https://www.n
cbi.nlm.nih.gov/pmc/articles/PMC3771067). PMID 24032093 (https://pubmed.ncbi.nlm.nih.gov/240
32093).
106. Bandrowski A, Brush M, Grethe JS, Haendel MA, Kennedy DN, Hill S, et al. (2015). "The
Resource Identification Initiative: A cultural shift in publishing" (https://www.ncbi.nlm.nih.gov/pmc/a
rticles/PMC4648211). F1000Research. 4: 134. doi:10.12688/f1000research.6555.2 (https://doi.or
g/10.12688%2Ff1000research.6555.2). PMC 4648211 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC4648211). PMID 26594330 (https://pubmed.ncbi.nlm.nih.gov/26594330).
107. Helsby MA, Fenn JR, Chalmers AD (23 August 2013). "Reporting research antibody use: how to
increase experimental reproducibility" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3829129).
F1000Research. 2: 153. doi:10.12688/f1000research.2-153.v2 (https://doi.org/10.12688%2Ff1000
research.2-153.v2). PMC 3829129 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3829129).
PMID 24358895 (https://pubmed.ncbi.nlm.nih.gov/24358895).
108. "The Antibody Registry" (https://antibodyregistry.org/). antibodyregistry.org.
109. "Resource Identification Initiative" (https://www.force11.org/group/resource-identification-initiative).
FORCE11. 14 August 2013. Retrieved 18 April 2016.
110. Archived (https://web.archive.org/web/20110717212251/http://antibody.bath.ac.uk/abmod.html) 17
July 2011 at the Wayback Machine

WAM (http://antibody.bath.ac.uk/abmod.html)
111. Marcatili P, Rosi A, Tramontano A (September 2008). "PIGS: automatic prediction of antibody
structures" (http://biocomputing.it/pigs/). Bioinformatics. 24 (17): 1953–4.
doi:10.1093/bioinformatics/btn341 (https://doi.org/10.1093%2Fbioinformatics%2Fbtn341).
PMID 18641403 (https://pubmed.ncbi.nlm.nih.gov/18641403). Archived (https://web.archive.org/w
eb/20101126235204/http://arianna.bio.uniroma1.it/pigs/) from the original on 26 November 2010.

Prediction of Immunoglobulin Structure (PIGS) (http://biocomputing.it/pigs/) Archived (https://web.


archive.org/web/20101126235204/http://arianna.bio.uniroma1.it/pigs/) 26 November 2010 at the
Wayback Machine
112. Archived (https://web.archive.org/web/20110719215959/http://antibody.graylab.jhu.edu/) 19 July
2011 at the Wayback Machine

RosettaAntibody (http://antibody.graylab.jhu.edu)
113. Park H. "Written Description Problems of the Monoclonal Antibody Patents after Centocor v.
Abbott" (https://web.archive.org/web/20141213031525/http://jolt.law.harvard.edu/digest/patent/writ
ten-description-problems-of-the-monoclonal-antibody-patents-after-centocor-v-abbott).
jolt.law.harvard.edu. Archived from the original (http://jolt.law.harvard.edu/digest/patent/written-des
cription-problems-of-the-monoclonal-antibody-patents-after-centocor-v-abbott) on 13 December
2014. Retrieved 12 December 2014.
114. Adolf-Bryfogle J, Kalyuzhniy O, Kubitz M, Weitzner BD, Hu X, Adachi Y, et al. (April 2018).
"RosettaAntibodyDesign (RAbD): A general framework for computational antibody design" (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC5942852). PLOS Computational Biology. 14 (4):
e1006112. Bibcode:2018PLSCB..14E6112A (https://ui.adsabs.harvard.edu/abs/2018PLSCB..14E
6112A). doi:10.1371/journal.pcbi.1006112 (https://doi.org/10.1371%2Fjournal.pcbi.1006112).
PMC 5942852 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5942852). PMID 29702641 (https://
pubmed.ncbi.nlm.nih.gov/29702641).
115. Lapidoth GD, Baran D, Pszolla GM, Norn C, Alon A, Tyka MD, Fleishman SJ (August 2015).
"AbDesign: An algorithm for combinatorial backbone design guided by natural conformations and
sequences" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4881815). Proteins. 83 (8): 1385–406.
doi:10.1002/prot.24779 (https://doi.org/10.1002%2Fprot.24779). PMC 4881815 (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC4881815). PMID 25670500 (https://pubmed.ncbi.nlm.nih.gov/25670
500).
116. Li T, Pantazes RJ, Maranas CD (2014). "OptMAVEn--a new framework for the de novo design of
antibody variable region models targeting specific antigen epitopes" (https://www.ncbi.nlm.nih.gov/
pmc/articles/PMC4143332). PLOS ONE. 9 (8): e105954. Bibcode:2014PLoSO...9j5954L (https://u
i.adsabs.harvard.edu/abs/2014PLoSO...9j5954L). doi:10.1371/journal.pone.0105954 (https://doi.o
rg/10.1371%2Fjournal.pone.0105954). PMC 4143332 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC4143332). PMID 25153121 (https://pubmed.ncbi.nlm.nih.gov/25153121).
117. Pham V, Henzel WJ, Arnott D, Hymowitz S, Sandoval WN, Truong BT, et al. (May 2006). "De novo
proteomic sequencing of a monoclonal antibody raised against OX40 ligand". Analytical
Biochemistry. 352 (1): 77–86. doi:10.1016/j.ab.2006.02.001 (https://doi.org/10.1016%2Fj.ab.2006.
02.001). PMID 16545334 (https://pubmed.ncbi.nlm.nih.gov/16545334).
118. Ma B, Zhang K, Hendrie C, Liang C, Li M, Doherty-Kirby A, Lajoie G (2003). "PEAKS: powerful
software for peptide de novo sequencing by tandem mass spectrometry". Rapid Communications
in Mass Spectrometry. 17 (20): 2337–42. Bibcode:2003RCMS...17.2337M (https://ui.adsabs.harva
rd.edu/abs/2003RCMS...17.2337M). doi:10.1002/rcm.1196 (https://doi.org/10.1002%2Frcm.1196).
PMID 14558135 (https://pubmed.ncbi.nlm.nih.gov/14558135).
119. Zhang J, Xin L, Shan B, Chen W, Xie M, Yuen D, et al. (April 2012). "PEAKS DB: de novo
sequencing assisted database search for sensitive and accurate peptide identification" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC3322562). Molecular & Cellular Proteomics. 11 (4):
M111.010587. doi:10.1074/mcp.M111.010587 (https://doi.org/10.1074%2Fmcp.M111.010587).
PMC 3322562 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3322562). PMID 22186715 (https://
pubmed.ncbi.nlm.nih.gov/22186715).
120. Perkins DN, Pappin DJ, Creasy DM, Cottrell JS (December 1999). "Probability-based protein
identification by searching sequence databases using mass spectrometry data". Electrophoresis.
20 (18): 3551–67. doi:10.1002/(SICI)1522-2683(19991201)20:18<3551::AID-
ELPS3551>3.0.CO;2-2 (https://doi.org/10.1002%2F%28SICI%291522-2683%2819991201%292
0%3A18%3C3551%3A%3AAID-ELPS3551%3E3.0.CO%3B2-2). PMID 10612281 (https://pubme
d.ncbi.nlm.nih.gov/10612281). S2CID 42423655 (https://api.semanticscholar.org/CorpusID:42423
655).
121. Bandeira N, Tang H, Bafna V, Pevzner P (December 2004). "Shotgun protein sequencing by
tandem mass spectra assembly". Analytical Chemistry. 76 (24): 7221–33. doi:10.1021/ac0489162
(https://doi.org/10.1021%2Fac0489162). PMID 15595863 (https://pubmed.ncbi.nlm.nih.gov/15595
863).
122. Liu X, Han Y, Yuen D, Ma B (September 2009). "Automated protein (re)sequencing with MS/MS
and a homologous database yields almost full coverage and accuracy" (https://doi.org/10.1093%2
Fbioinformatics%2Fbtp366). Bioinformatics. 25 (17): 2174–80. doi:10.1093/bioinformatics/btp366
(https://doi.org/10.1093%2Fbioinformatics%2Fbtp366). PMID 19535534 (https://pubmed.ncbi.nlm.
nih.gov/19535534).
123. Castellana NE, Pham V, Arnott D, Lill JR, Bafna V (June 2010). "Template proteogenomics:
sequencing whole proteins using an imperfect database" (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC2877985). Molecular & Cellular Proteomics. 9 (6): 1260–70. doi:10.1074/mcp.M900504-
MCP200 (https://doi.org/10.1074%2Fmcp.M900504-MCP200). PMC 2877985 (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC2877985). PMID 20164058 (https://pubmed.ncbi.nlm.nih.gov/2016405
8).
124. Liu X, Dekker LJ, Wu S, Vanduijn MM, Luider TM, Tolić N, et al. (July 2014). "De novo protein
sequencing by combining top-down and bottom-up tandem mass spectra". Journal of Proteome
Research. 13 (7): 3241–8. doi:10.1021/pr401300m (https://doi.org/10.1021%2Fpr401300m).
PMID 24874765 (https://pubmed.ncbi.nlm.nih.gov/24874765).
125. Tran NH, Rahman MZ, He L, Xin L, Shan B, Li M (August 2016). "Complete De Novo Assembly of
Monoclonal Antibody Sequences" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4999880).
Scientific Reports. 6: 31730. Bibcode:2016NatSR...631730T (https://ui.adsabs.harvard.edu/abs/20
16NatSR...631730T). doi:10.1038/srep31730 (https://doi.org/10.1038%2Fsrep31730).
PMC 4999880 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4999880). PMID 27562653 (https://
pubmed.ncbi.nlm.nih.gov/27562653).
126. Gebauer M, Skerra A (June 2009). "Engineered protein scaffolds as next-generation antibody
therapeutics". Current Opinion in Chemical Biology. 13 (3): 245–55.
doi:10.1016/j.cbpa.2009.04.627 (https://doi.org/10.1016%2Fj.cbpa.2009.04.627). PMID 19501012
(https://pubmed.ncbi.nlm.nih.gov/19501012).
127. Kristiansen, Paul A.; Page, Mark; Bernasconi, Valentina; Mattiuzzo, Giada; Dull, Peter; Makar,
Karen; Plotkin, Stanley; Knezevic, Ivana (10 April 2021) [2021-02-23]. "WHO International
Standard for anti-SARS-CoV-2 immunoglobulin" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7
987302). The Lancet. World Health Organization / Elsevier. 397 (10282): 1347–1348.
doi:10.1016/S0140-6736(21)00527-4 (https://doi.org/10.1016%2FS0140-6736%2821%2900527-
4). PMC 7987302 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7987302). PMID 33770519 (http
s://pubmed.ncbi.nlm.nih.gov/33770519).
128. Knezevic, Ivana; Mattiuzzo, Giada; Page, Mark; Minor, Philip; Griffiths, Elwyn; Nuebling, Micha;
Moorthy, Vasee (26 October 2021). "WHO International Standard for evaluation of the antibody
response to COVID-19 vaccines: call for urgent action by the scientific community" (https://www.nc
bi.nlm.nih.gov/pmc/articles/PMC8547804). The Lancet Microbe. World Health Organization /
Elsevier. 3 (3): e235–e240. doi:10.1016/S2666-5247(21)00266-4 (https://doi.org/10.1016%2FS26
66-5247%2821%2900266-4). PMC 8547804 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8547
804). PMID 34723229 (https://pubmed.ncbi.nlm.nih.gov/34723229).
129. Knezevic, Ivana (10 November 2021). "Training webinar for the calibration of quantitative serology
assays using the WHO International Standard for anti-SARS-CoV-2 immunoglobulin" (https://cdn.
who.int/media/docs/default-source/biologicals/covid-19/agenda-ppt-q-a-ik.pdf?sfvrsn=89c3ddfa_5)
(PDF). Archived (https://web.archive.org/web/20220218031435/https://cdn.who.int/media/docs/def
ault-source/biologicals/covid-19/agenda-ppt-q-a-ik.pdf?sfvrsn=89c3ddfa_5) (PDF) from the
original on 18 February 2022. Retrieved 5 March 2022. (68 pages)

External links
Mike's Immunoglobulin Structure/Function Page (https://web.archive.org/web/20070403013202/htt
p://www.path.cam.ac.uk/~mrc7/mikeimages.html) at University of Cambridge
Antibodies as the PDB molecule of the month (https://pdb101.rcsb.org/motm/21) Discussion of the
structure of antibodies at RCSB Protein Data Bank
A hundred years of antibody therapy (https://web.archive.org/web/20070405170923/http://users.p
ath.ox.ac.uk/~scobbold/tig/new1/mabth.html) History and applications of antibodies in the
treatment of disease at University of Oxford
How Lymphocytes Produce Antibody (http://www.cellsalive.com/antibody.htm) from Cells Alive!

Retrieved from "https://en.wikipedia.org/w/index.php?title=Antibody&oldid=1136401805"

You might also like