throbber
627
`
`Review
`
`Received: 24 April 2014,
`
`Revised: 14 May 2014,
`
`Accepted: 15 May 2014,
`
`Published online in Wiley Online Library
`
`(wileyonlinelibrary.com) DOI: 10.1002/jmr.2394
`
`Specificity, polyspecificity, and heterospecificity
`of antibody-antigen recognition
`Marc H. V. Van Regenmortela*
`
`The concept of antibody specificity is analyzed and shown to reside in the ability of an antibody to discriminate
`between two antigens. Initially, antibody specificity was attributed to sequence differences in complementarity
`determining regions (CDRs), but as increasing numbers of crystallographic antibody-antigen complexes were
`elucidated, specificity was analyzed in terms of six antigen-binding regions (ABRs) that only roughly correspond
`to CDRs. It was found that each ABR differs significantly in its amino acid composition and tends to bind different
`types of amino acids at the surface of proteins. In spite of these differences, the combined preference of the six ABRs
`does not allow epitopes to be distinguished from the rest of the protein surface. These findings explain the poor
`success of past and newly proposed methods for predicting protein epitopes. Antibody polyspecificity refers to the
`ability of one antibody to bind a large variety of epitopes in different antigens, and this property explains how the
`immune system develops an antibody repertoire that is able to recognize every antigen the system is likely to
`encounter. Antibody heterospecificity arises when an antibody reacts better with another antigen than with the one
`used to raise the antibody. As a result, an antibody may sometimes appear to have been elicited by an antigen with
`which it is unable to react. The implications of antibody polyspecificity and heterospecificity in vaccine development
`are pointed out. Copyright © 2014 John Wiley & Sons, Ltd.
`
`Keywords: antibody specificity; polyspecificity; heterospecificity; peptide hydropathic complementarity; epitope prediction;
`polyreactive antibodies; monogamous bivalent binding; HIV-1 vaccines
`
`INTRODUCTION
`
`Adaptive immune responses involve B cells that recognize native
`protein antigens and differentiate into antibody-secreting plasma
`cells and T cells that recognize unfolded peptide fragments of the
`antigen that have been processed in antigen-presenting cells.
`These peptide fragments known as T cell epitopes are presented
`in the groove of surface-exposed major histocompatibility
`complex class I and II molecules recognized by CD8+ and CD4+
`T cells, respectively. The pathways of antigen processing in
`antigen-presenting cells are fairly well understood[1] but fall
`outside the scope of this review.
`IgM
`The primary antibody repertoire consists mainly of
`molecules, generated independently of exogenous antigen, which
`arise in the bone marrow following immunoglobulin (Ig) variable
`(V) gene rearrangements and imprecise joining at the borders of
`integrated gene segments.
`Following encounter with antigen, B cells are activated and
`migrate into germinal center follicles where they proliferate.[2]
`Under the influence of antigen selection, a small number of
`these B cells undergo an Ig heavy chain class switch to IgG,
`IgA, or IgE molecules and produce the secondary antibody
`repertoire following a process of somatic hypermutation that
`introduces point mutations in the V regions at a rate of about
`4–10
`3 per base pair per generation.[3] Most B cells that fail
`10
`to be selected by antigen in the germinal centers undergo
`apoptosis.[4]
`Somatic hypermutation produces many amino acid substitu-
`tions in the complementarity determining regions (CDRs) of
`the V genes but fewer ones in the conserved Ig framework (FR)
`region. This leads to a considerable increase in the antibody
`
`(Ab) affinity for the antigen. The mechanisms by which B cells
`with higher affinity receptors (BCRs) are selected are not entirely
`clear. Progeny cells with higher affinity BCRs may be selected
`because they bind preferentially to the immunogen as its
`concentration decreases during antigen clearance. Another
`possibility is that unprocessed antigen molecules presented by
`follicular dendritic cells, perhaps via antibodies and Fc receptors
`picked up from the serum, are able to stimulate memory B cells
`for long periods.[5]
`Following further proliferation and antigen selection, B cells with
`increased receptor affinity differentiate into either Ab-producing
`plasma cells or memory B cells, a fate that is controlled by T cell
`help and the IL-21 cytokine.[6]
`The binding capacity of an Ab molecule resides in an antigen-
`binding cleft of 50–70 residues that is located in the N-terminal
`regions of the Ig heavy (H) and the light (L) chains. Each cleft
`harbors several overlapping binding subsites of 15–20 amino
`acid residues that are called paratopes. These subsites possess
`a particular structural and chemical complementarity to certain
`patches of residues present at the surface of protein antigens,
`known as B cell epitopes. If the context makes it clear that one
`refers to epitopes recognized by antibodies rather than by T
`cells, one simply calls them epitopes.
`
`* Correspondence to: Marc H. V. Van Regenmortel, Institut de Recherche de l’Ecole
`de Biotechnologie de Strasbourg, CNRS/UDS, UMR 7242, Illkirch 67400, France.
`E-mail: vanregen@unistra.fr
`
`a M. H. V. Van Regenmortel
`Wallenberg Research Center, Stellenbosch Institute for Advanced Study,
`Stellenbosch University, Stellenbosch, South Africa
`
`J. Mol. Recognit. 2014; 27: 627–639
`
`Copyright © 2014 John Wiley & Sons, Ltd.
`
`Lassen - Exhibit 1056, p. 1
`
`

`

`Because an Ab always harbors several individual paratopes
`able to bind different epitopes present in one or other antigen,
`an Ab is never monospecific for a single binding partner
`because this would require that the remaining 50 or so residues
`in the antigen-binding cleft that are not involved in the one in-
`teraction are unable to bind any other antigenic structure,
`which is unlikely. Steric hindrance may prevent two antigen
`molecules from binding simultaneously to the same Ab-
`binding cleft, but if the two paratope subsites do not overlap,
`it is possible for two small antigens to bind simultaneously to
`the same Ab molecule.[7]
`
`ANTIBODY SPECIFICITY
`
`The concept of antibody specificity is widely used in immunology,
`although it has rarely been defined satisfactorily. The term
`specificity is derived from the word species, and “specific”
`properties were initially considered to be properties that allow
`the members of one species to be distinguished from those of
`another. This terminology goes back to the days when individual
`species were believed to be separated by permanent, sharp
`boundaries, and Darwinian evolution and the transformation of
`species by selection had not yet become universally accepted.
`As recounted by Mayr,[8] many biologists in the 19th century
`continued to view species as fixed entities separated by clear-
`cut discontinuities that gave rise to a belief in absolute immuno-
`logical specificity. Paul Ehrlich, for instance, believed that antisera
`raised against members of different species of pathogenic
`bacteria were completely specific, allowing the members of
`different species to be distinguished serologically with absolute
`certainty.[9] When it was later discovered that antisera raised
`against cells from different animal species cross-reacted serologi-
`cally with cells from many other species, it became clear that an
`antigen was able to elicit not only a single antibody of absolute
`specificity but also a whole spectrum of antibodies that could
`cross-react with many related antigens.[10] The specificity of anti-
`gen-antibody recognition was then no longer perceived as an all
`or none phenomenon but was interpreted as a matter of more
`or less good fit between molecules that possessed different
`degrees of stereochemical complementarity.[11]
`The most
`reliable method for
`identifying epitopes and
`paratopes is by solving the 3D structure of antigen-antibody
`complexes and determining which amino acids in the two
`
`M. H. V. VAN REGENMORTEL
`
`partners make contact with each other.[12] The surfaces of
`proteins always harbor many different epitopes, and each of
`them is able to recognize complementary antibodies. When
`two antigens are compared with a panel of monoclonal antibod-
`ies (Mabs) raised against them, they will appear to be identical if
`a Mab is used that recognizes an identical epitope present in
`both antigens (Figure 1). Such a cross-reaction between two
`antigens is usually referred to as shared reactivity.[14] On the
`other hand, if a Mab is used that recognizes an epitope present
`in only one of the antigens, both antigen molecules will appear
`to be unrelated. If an investigator wants to differentiate between
`the two antigens, the first type of Mab would be called nonspe-
`cific, whereas the second Mab would be called specific because
`it discriminates between the two antigens. Instead of speaking
`of specificity, it may thus be preferable to speak of the discrimi-
`nation potential of antibodies because it is the wish of the
`investigator to distinguish between two antigens that determine
`whether an Ab is considered to be specific.
`A third possibility referred to as true cross-reactivity[14] occurs
`when an Ab recognizes an epitope that is only structurally
`related by not identical in both antigens (Figure 1). In many cases
`but not always, the Ab will react with higher affinity with the
`homologous epitope used for raising the Ab than with the
`cross-reactive heterologous epitope. It is unfortunate that Abs
`are often given names derived from the antigen they are able
`to react with, because they cannot be specific for an entire
`protein antigen that harbors many different epitopes but only
`for one of its epitopes.
`It is often assumed that Abs that possess a high binding
`affinity are likely to be also more specific because it is believed
`that they should possess a better stereochemical complementarity
`with their antigens than Abs of lower affinity. However, there is
`no necessary link between affinity and specificity because Abs
`of low affinity may be able to discriminate better between two
`antigens than Abs of high affinity. This is because these Abs
`may detect fewer cross-reactions between antigens than Abs
`of high affinity because weaker cross-reactions are more likely
`to occur below the level where they can still be detected with
`low affinity Abs.[13]
`Whereas Ab affinity is defined by a binary relationship
`between one epitope and one paratope, Ab specificity is a
`ternary relational property that has meaning only with respect
`to a minimum of three partners, for instance, one paratope and
`two epitopes. Specificity is only meaningful with respect to the
`
`Figure 1. Potential cross-reactivity between two related antigens 1 and 2. Epitope a is present on both antigens, and Mab anti-a will react in an
`identical fashion with antigens 1 and 2. Mabs anti-b and anti-d recognize unrelated epitopes on the two antigens and do not cross-react. They are
`specific for either Ag 1 or Ag 2. Mab anti-c reacts strongly with the homologous epitope c and cross-reacts weakly with the heterologous epitope e
`(from Van Regenmortel[13] reproduced with permission).
`
`628
`
`wileyonlinelibrary.com/journal/jmr
`
`Copyright © 2014 John Wiley & Sons, Ltd.
`
`J. Mol. Recognit. 2014; 27: 627–639
`
`Lassen - Exhibit 1056, p. 2
`
`

`

`Because most protein epitopes are discontinuous and usually
`involve numerous residues situated on different accessible loops
`of the protein, their structure is somewhat analogous to the
`discontinuous structure of paratopes built up from residues
`located on the different Ab hypervariable loops. Furthermore,
`because peptides of 6–12 residues corresponding to the CDRs
`of an Ab are sometimes able on their own to bind to the
`cognate intact antigen, they could be viewed as continuous
`paratopes, reminiscent of the binding capacity of continuous
`epitopes.[28] Such continuous paratopes that are able to
`recognize the antigen in a specific manner, albeit with a lower
`affinity, may contain residues found by crystallographic analysis
`of
`the antigen-antibody complex to be located at
`the
`paratope–epitope interface.[29]
`The functional binding capacity of a continuous paratope has
`been illustrated by the construction of a chimeric peptide
`consisting of one CDR of an antiviral antibody conjugated to a
`continuous epitope of another virus.[30] This construct made it
`possible to redirect the specificity of an antiviral Ab to allow it
`to recognize another virus and illustrates the functional equiva-
`lence of continuous paratopes and epitopes because they were
`able to mimic intact Abs and antigens, respectively.[28,30]
`The ability of linear peptides to bind to each other is usually
`attributed to hydropathic complementarity. This arises when
`peptides of opposite hydropathy bind to each other because
`hydrophilic residues in one peptide are oriented toward the
`aqueous solvent and liberate a space that can accommodate
`an hydrophobic residue from the other peptide.[31] An example
`illustrating the hydropathic complementarity between a contin-
`uous epitope of angiotensin II and a continuous paratope from
`the CDR-H1 of an angiotensin Ab is shown in Figure 2.
`Hydropathic complementarity is regularly observed between
`the sense and antisense peptides encoded by complementary
`sense and antisense messenger RNA.[32] There is in fact no
`exception to the rule that DNA or RNA codons (sense) and antico-
`dons (antisense) always code for amino acids of opposite
`hydropathicity (i.e., either hydrophilic or hydrophobic residues).[33]
`This remarkable pattern suggests that the genetic code may have
`evolved initially to favor the simultaneous emergence from two
`
`629
`
`Figure 2. Complementary hydropathic profiles of two interacting pep-
`tides: (●) continuous epitope (DRVYIHPF) of angiotensin II; (○) continuous
`paratope (TFNTDAMN) in the CDR-H1 of an angiotensin Mab. Hydro-
`pathic profiles were obtained using the Kyte and Doolittle hydropathic
`coefficients (from Boquet et al. 1995, Mol. Immunol. 32: 303, reproduced
`with permission).
`
`SPECIFICITY, POLYSPECIFICITY, AND HETEROSPECIFICITY
`
`capacity of an Ab to react differently with two or more antigens
`and thereby to discriminate between them.[13]
`Which selection pressure gave rise during evolution to the huge
`primary repertoire of specific paratopes[15] has been debated for
`many years, the most common explanation being that the
`immune system needed to remove either exogenous pathogens
`or antibodies to self-antigens.[16–20] However, this somewhat
`theoretical debate falls outside the scope of this review.
`
`THE STRUCTURAL BASIS OF PARATOPE–EPITOPE
`RECOGNITION
`
`Epitopes and paratopes are relational entities defined by their
`mutual complementarity, and they depend on each other to
`acquire a recognizable identity. Epitopes and paratopes are thus
`not intrinsic structural features of an antigen and Ab molecule,
`respectively, because they cannot exist in the absence of a
`relational nexus between the two partners. This means that the
`number of epitopes in a protein is equal to the number of
`different Mabs that can be raised against it.
`In this way, the
`insulin molecule can be said to have 115 epitopes[21] and the
`BLyS molecule more than a thousand.[22]
`This relational dependence means that as soon as an epitope
`is slightly altered and binding to the paratope is affected, the
`paratope is also no longer the same. This analysis differs from
`the classification of binding sites introduced by Cohn,[23] which
`defines a paratopic clan as a family of paratopes, distinguishable
`from each other, that are all functionally capable of binding a
`given single epitope. Cohn also defined a mimotopic array as a
`set of distinguishable epitopes that are all able to bind to a given
`single paratope.
`Protein epitopes are usually classified as continuous or
`discontinuous, depending on whether the amino acids that form
`the epitope are contiguous in the peptide chain or not. The
`majority of protein epitopes is discontinuous and consists of
`two to five short stretches of residues that are distant in the
`protein sequence and are brought together at the surface of
`the protein by the folding of the peptide chain.[24]
`Discontinuous epitopes are defined structurally by the amino
`acids that are found to be in contact with paratope residues in a
`crystallographic
`complex. However, discontinuous epitopes
`cannot be extracted from the protein antigen to demonstrate that
`they possess binding activity outside the context of the native
`protein, and it is also extremely difficult to reconstitute them in ac-
`tive form with sufficient 3D precision by peptide synthesis.[25,26]
`Continuous epitopes, on the other hand, always have fuzzy
`boundaries because they are identified functionally by the ability
`of short peptide fragments of the protein to bind to an antibody
`and not by establishing that all the residues in the epitope
`interact with a paratope.
`In most cases, all the residues in a
`continuous epitope do not interact with a paratope, and only
`some of its residues will be located at the surface of a native
`protein where they are usually part of a more complex discontin-
`uous epitope.[26,27]
`that
`through six CDRs
`Antibodies
`recognize antigens
`because of their enormous sequence variability are able to form
`millions of different
`antigen-binding paratopes.
`These
`hypervariable loops are denoted L1, L2, and L3 in the Ig light
`chain and H1, H2, and H3 in the Ig heavy chain, and they
`associate noncovalently at the tip of Ig Fab fragments to form
`the antigen-binding site.
`
`J. Mol. Recognit. 2014; 27: 627–639
`
`Copyright © 2014 John Wiley & Sons, Ltd.
`
`wileyonlinelibrary.com/journal/jmr
`
`Lassen - Exhibit 1056, p. 3
`
`

`

`M. H. V. VAN REGENMORTEL
`
`ABRs included as much as 96% of all the residues that actually
`bind the antigen.[51] It was also found that several residues in
`the FR region and constant regions of IgGs contributed signifi-
`cantly to antigen binding.[52]
`Using the Paratome web server, Kunik and Ofran[53] recently
`examined the amino acid composition of the 6 ABRs in 200 anti-
`body-antigen complexes, which is the largest number of com-
`plexes ever analyzed. The average lengths of the six ABRs and
`the number of SDRs in each ABR are presented in Figure 3.
`ABR H2 was found to have the longest median length of 14 res-
`idues, followed by H3 and L2 with a median length of 11 resi-
`dues. H3 showed the highest length diversity, whereas H1, L2,
`and L3 showed limited length diversity. These lengths vary
`somewhat from earlier results,[45,54,55] which may be due to the
`different methods that were used to define CDRs. The number
`of SDRs in each ABR is shown in Figure 3B. H2 and H3 have the
`largest median number of SDRs (six residues), but H3 is more di-
`verse because it may contain as many as 14 SDRs. L2 sometimes
`
`Figure 3. Lengths of antigen-binding regions (ABRs) and number of an-
`tigen-binding residues. (A) ABRs length. The black bold lines represent
`the median length. The second and third quartiles are depicted by light
`and dark gray boxes, respectively. The range of lengths is indicated by
`vertical lines. (B) Number of antigen-binding residues in each ABR. The
`black bold lines and the light and dark gray zones are as in (A). The max-
`imal and minimal number of antigen-binding residues within each ABR
`are indicated by vertical lines (from Kunik & Ofran,[53] reproduced with
`a permission).
`
`complementary nucleic acid strands, of sense and antisense pep-
`tides that are able to interact with each other as do receptors and
`ligands.[34,35]
`The specificity of interactions between two peptides is en-
`hanced by the ability of amino acids to form complementary
`protrusions and cavities and by the presence of amino acids of
`opposite charge.[36] In a recent application of the phenomenon
`of hydropathic complementarity to the study of human immu-
`nodeficiency virus type 1 (HIV-1) immune responses, a comple-
`mentary HIV gp120 antisense peptide of inverted hydropathy
`corresponding to the cA1 T cell epitope peptide was used to in-
`duce cellular immunity against HIV-1.[37]
`Additional types of protein epitopes have been recognized
`such as neotopes that arise from the quaternary structure of pro-
`tein aggregates and mimotopes that are peptides that bind to
`antiprotein Abs but show little or no sequence similarity with
`the protein used to raise these Abs.[26] Because numerous re-
`views of the antigenic structure of proteins are available,[38–41]
`the various types of epitopes will not be further discussed here.
`In recent years,
`increasing numbers of antibody structures
`have been elucidated by crystallographic analysis of antibody-
`antigen complexes, and this has given us a much better under-
`standing of the structural basis of antibody specificity. The CDRs
`of Abs vary considerably in length, whereas the individual
`paratopes within the antigen-binding site usually consist of
`10–20 residues. Residues found to be in contact with the antigen
`are often referred to as specificity-determining residues (SDRs),
`and they are more variable than residues that are not in contact
`with it.[42,43] Recently, SDRs were compared with residues that
`had undergone somatic hypermutation during affinity matura-
`tion. The results based on an analysis of 140 antibody-antigen
`complexes showed that somatic replacements occurred mainly
`in residues that were not involved in contacts with the anti-
`gen.[44] This observation is consistent with the fact that the
`hypermutation process is stochastic and occurs in a nonselective
`manner irrespective of whether residues are in contact with the
`antigen or not. However, non-SDRs may contribute to binding
`activity by helping to maintain the conformation of the binding
`site.[43]
`Initially, CDRs were identified by aligning a limited number of
`antibody sequences and determining the positions of the most
`variable residues.[45] As increasing numbers of 3D Ab structures
`became available, the hypervariable loop and constant FR re-
`gions could be located in the Ab structure, and the CDRs were
`found to adopt a restricted set of conformations termed canon-
`ical structures.[46] Different combinations of canonical structures
`alter the topography of paratopes and determine the size of the
`antigen surface with which the Ab is able to interact.[43,47]
`A third approach to define CDRs was developed using the ex-
`tended database of variable Ig genome sequences.[48,49] All
`these approaches produce slightly different residue numbering
`systems, mainly because nucleotide insertions are accommo-
`dated differently.
`More recently, a fourth approach using the Paratome web
`server was developed on the basis of a multiple structural align-
`ment of all antibody-antigen complexes available in the Protein
`Data Base.[50,51] This method identified regions of structural con-
`sensus called antigen-binding regions (ABRs), roughly corre-
`sponding to CDRs, in which the pattern of structural positions
`that bind the antigen was found to be very similar among all an-
`tibodies. The superiority of ABRs compared with previously used
`CDR identification tools was demonstrated by the fact that the
`
`630
`
`wileyonlinelibrary.com/journal/jmr
`
`Copyright © 2014 John Wiley & Sons, Ltd.
`
`J. Mol. Recognit. 2014; 27: 627–639
`
`Lassen - Exhibit 1056, p. 4
`
`

`

`SPECIFICITY, POLYSPECIFICITY, AND HETEROSPECIFICITY
`
`contains as many as 10 SDRs, although its median number of
`SDRs is only 1. For all ABRs, there are instances where they do
`not contain a single SDR.
`Some authors have concluded that the amino acid composition
`of protein epitopes does not differ significantly from that of
`protein surfaces,[56] whereas others suggested that epitopes may
`be enriched with certain types of amino acids.[57,58] In their study,
`Kunik and Ofran[53] determined the frequency of the five most
`abundant SDRs in each ABR. This is a finer analysis than that
`carried out in earlier studies that always determined the frequency
`of SDRs averaged over an entire paratope rather than in individual
`ABRs.[54,56,58]
`As shown in Figure 4, Tyr was the most frequent SDR in the
`ABRs followed by Asp, Asn, Arg, and Trp. The five most common
`SDRs in each ABR covered 63% of L1, 60% of L2, 59% of H3, 58%
`of H1, 54% of H2, and 48% of L3. Although Tyr, Ser, Asn, and Trp
`have previously been reported to be the most abundant residues
`in paratopes,[54,56,59] exactly the same distribution of residues
`was not found when the frequencies of each SDR in individual
`ABRs were considered. Tyr makes the greatest energetic contri-
`bution to antigen binding,
`in line with its ability to mediate
`different types of contacts (i.e., van der Waals, aromatic interac-
`tions, and hydrogen bonds). It should be noted that there is no
`link between the abundance of an amino acid in SDRs and its
`energetic contribution to antigen binding. The most energeti-
`cally important residues in the ABRs were found to be Tyr, Asp,
`Asn, and Arg. H3 has the highest percentage (29%) of SDRs that
`are energetically important for binding followed by L1 (24%) and
`H2 (22%).
`The analysis of Kunik and Ofran[53] for the first time revealed
`that the six ABRs differ significantly in their amino acid composi-
`tions and that each ABR tends to bind different types of amino
`acids at the surface of proteins. Because the six ABRs, because of
`their significantly different amino acids compositions, have differ-
`ent contact preferences for certain epitope residues,
`it seems
`plausible that epitopes might also possess distinguishable amino
`acid compositions. However, when the amino acid composition
`of epitopes was compared with that of entire protein surfaces,
`
`In other
`no noticeable differences were observed (Figure 5).
`words, although each ABR has a unique set of contact preferences
`favoring certain epitope residues over others, the combination of
`all these individual ABR preferences yields a collective amino acid
`composition of epitopes that is very similar to the composition of
`protein surfaces in general.[53,56]
`Because the entire accessible surface of a protein is a continuum
`of potential epitopes,[39] it could be argued that it would be advan-
`tageous for Abs to bind any protein surface patch without requiring
`specialized sites of increased stickiness. It seems that antibodies are
`in fact able to achieve this because they have evolved a set of ABRs
`where each ABR binds different types of amino acids, while the
`combined preference of the entire set is for epitopes that are
`indistinguishable from the rest of the protein surface.[53] The situa-
`tion is different in most protein–protein complexes, for instance,
`virus–host receptor partners, which have optimized their mutual
`complementarity over long periods of biological evolution. This
`coevolution has favored the selection of small “sticky” areas at the
`surface of proteins that are enriched in hydrophobic, aromatic,
`and charged residues.[60] This is not the case with epitope–paratope
`partners because paratopes need to optimize their complementarity
`to epitopes fairly rapidly (i.e., weeks or months) within the context
`of an individual immune system.
`
`THE EPITOPE PREDICTION CONUNDRUM
`
`The recent results of Kunik and Ofran[53] and Kringelum et al.[56]
`have revealed a major difficulty when attempts are made to
`predict epitopes at the surface of proteins. Because no amino
`acid was found to be significantly over represented in epitope
`regions compared with the rest of the surface and because the
`surface is an antigenic continuum, most of the protein surface
`can be expected to be part of some epitope potentially recogniz-
`able by one or other antibody. Success in predicting that some
`residues are part of an epitope will thus improve as the number
`of examined antibodies raised against the antigen increases[61]
`and not necessarily because some residues are inherently more
`
`631
`
`Figure 4. The five most frequent antigen-binding amino acids in each antigen-binding region (ABR). For each ABR, the list of residues that contact the
`antigen is listed, and the frequency of each amino acid is indicated (from Kunik & Ofran,[53] reproduced with permission).
`
`J. Mol. Recognit. 2014; 27: 627–639
`
`Copyright © 2014 John Wiley & Sons, Ltd.
`
`wileyonlinelibrary.com/journal/jmr
`
`Lassen - Exhibit 1056, p. 5
`
`

`

`M. H. V. VAN REGENMORTEL
`
`Figure 5. Amino acid composition of antigen surface residues and of epitopes. The frequency of each amino acid is calculated for exposed residues
`and for epitope residues, using a nonredundant set of antibody-antigen complexes (from Kunik & Ofran,[53] reproduced with permission).
`
`immunologically active than others. This may be one reason why
`attempts to predict protein epitopes using various amino acid
`propensities have been notoriously unsuccessful.[26,27,62–70] How-
`ever, there may also be other reasons for this lack of success. For
`many years, investigators concentrated mainly on the prediction
`of continuous epitopes because these correspond to short pep-
`tides that can easily be synthesized and could readily be used
`to replace pathogenic antigens in diagnostic immunoassays[71,72]
`or to act as immunogens for raising antipeptide antibodies that
`cross-react with the cognate protein.[73] Unfortunately, when
`one refers to continuous epitopes of a protein, the impression
`is created that these epitopes exist as such in the native protein.
`In reality, these so-called protein epitopes are mostly short linear
`peptide fragments of more complex discontinuous epitopes that
`cross-react only weakly with certain antiprotein Abs and possess
`only limited structural similarity with small regions of the protein
`surface that may be as short as dipeptide or tripeptide se-
`quences.[27,74,75,70] Predicting continuous epitopes is thus of lim-
`ited value for analyzing the antigenicity and immunogenicity of
`native proteins.
`It is astonishing that in spite of innumerable
`failed attempts to use continuous epitopes as potential synthetic
`vaccines,[26,76,77] many investigators continue to use short linear
`peptides, possibly because they are dubbed continuous epitopes
`of a pathogen, as promising candidates for developing synthetic
`vaccines.
`Because it is increasingly accepted that the vast majority of pro-
`tein epitopes are discontinuous, many investigators have
`attempted to develop computational prediction algorithms that
`they claim allow the prediction of discontinuous epitopes.[26,65–70]
`What these methods do is to predict that a small number of
`residues located at the protein surface are likely to be part of a
`discontinuous epitope. What these methods are unable to do is
`to identify which full set of residues from distant parts of the
`protein sequence need to be assembled in a precise conformation
`to form an active site endowed with the antigenic and immuno-
`genic properties of a discontinuous epitope present in the native
`protein.[27,65–71,78] To claim that such methods allow discontinuous
`epitopes to be “predicted” is rather ambiguous because they are
`only able to “map” or “identify” a limited number of residues pres-
`ent in complex discontinuous epitope. Discontinuous epitopes are
`only defined structurally by determining which residues of an anti-
`gen are in contact with paratope residues and not functionally by
`showing that this complete set of residues, when it is not embed-
`ded in the protein, achieves adequate immunological mimicry. Be-
`cause the main purpose of epitope prediction is to replace a
`complete protein antigen by a small fragment of the molecule that
`possesses the antigenic and/or immunogenic properties of one of
`its epitopes, the feasibility of predicting functional discontinuous
`epitopes by computation remains at present a rather elusive goal.
`
`Recently, a completely different approach to epitope prediction
`has been proposed that utilizes the paratope sequences of a
`series of Mabs raised against an antigen in order to predict which
`surface regions of the antigen are likely to be recognized by these
`Mabs.[61]
`Instead of predicting which residues of an antigen
`possess a superior capacity for binding to any potential Ab, this
`method attempts to predict patches of about five residues located
`at the surface of a protein that are likely to be recognized by one
`or a few available Mabs.
`The method is based on residue pairing preferences that have
`been shown to exist in ABRs and epitopes,[53] and it predicts
`potential matches between a given Ab and a given epitope. It
`also utilizes cross-blocking inhibition assays with different Abs
`to assess if the epitopes they recognize overlap or not.
`Because patches of five residues cannot represent a complete
`discontinuous epitope, their usefulness to serve as potential
`diagnostic reagents or immunogens is likely to be rather limited,
`especially because such applications always
`involve the
`participation of a large variety of different Abs. The method also
`de

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket