throbber
Intern. Rev. lmmunol. 2, 1987. pp. 339-356
`Photocopying permitted by license only
`1:>1987 Harwood Academic Publishers GmbH
`Printed in the United States of America
`
`Perspectives on Antigenicity
`and Idiotypy
`
`THOMAS KIEBER-EMMONS,t ELIZABETH GETZOFF,*
`and HEINZ KOHLER..,
`Roswell Park Memorial Institute, 666
`
`tDepartment of Molecular Immunology.
`Elm St., Buffalo. NY 14263
`*Departments of Molecular Biology and Immunology,
`Research Institute of
`Scripps Clinic, La Jolla, CA 92037
`
`INTRODUCTION
`
`Over ten years have passed since the concept of using synthetic peptides to
`
`
`
`
`
`probe antigenicity was first developed [l, 2]. Since then, prominent among
`
`
`
`the applications of synthetic immunogen technology in biology and medi­
`cine (3, 4] is the utilization
`
`
`
`of synthetic peptides derived from the antigen for
`[5]. The network hypothesis
`vaccine development
`of Jerne [6] offers still
`
`another elegant concept for vaccine development. The anti-idiotype concept
`
`[7] provides an approach whereby an antigen can be substituted by an
`
`
`
`antibody possessing characteristics of that antigen. This can be demon­
`
`
`
`
`strated by using an anti-idiotypic antibody (Ab2) as a surrogate antigen that
`
`
`
`can stimulate an antigen-specific immune response [7]. This avenue
`
`
`
`provides an alternative in cases where the production of antigen based upon
`
`molecular biological approaches may not be feasible.
`
`
`
`
`The development of idiotope (Id) derived vaccines rests on the principle of
`
`
`
`molecular mimicry. An understanding of the structural basis of molecular
`
`
`
`mimicry could improve the production of idiotype vaccines, moving it from
`
`339
`
`1 of 18
`
`BI Exhibit 1054
`
`

`

`340
`
`T. KlEBER-EMMONS, E GETZOFF, and H. KOHLER
`
`an experimental state to a rational approach [8]. The importance of molecu­
`lar mimicry by monoclonal anti-idiotypes (anti-Id) is the ability to make T
`ceU-independent antigens T cell-dependent. For example, anti-idiotypes
`would allow the presentation of carbohydrate antigens as mimicked by the
`structure and conformation of the protein surrogate [9]. The mimicking
`abilities of such "internal image" antibodies also sets the stage for the
`possibility of producing fully synthetic idiotope vaccines using essential
`sequence information obtained from idiotope hybridoma antigens. The use
`of such designed idiotope-derived synthetic peptides would thwart problems
`associated with the administering of mouse hybridomas to humans (10).
`Of fundamental importance in designing new peptide antigens is the
`faithfulness or fidelity of the molecular mimicry. Antibody-antibody interac­
`tions are modulated by their large surface areas, so the complete description
`for an Id may entail contact points which are close in space but remote in
`sequence. Depending on the idiotope, there may be two components which
`contribute to the degree of mimicking fidelity: essential mimicking residues,
`and contact residues whose complementary interactions lend to the overall
`association constant for a particular complex formation. This latter compo­
`nent may also play an important secondary role in helping to stabilize a
`particular structural environment required for full antigenic mimicry. To
`disentangle these possible effects and ultimately achieve the successful
`development of a functional antibody or peptide vaccine, it is imperative to
`fully understand the structure of the antibody molecule, the basis of idiotypic
`expression in three dimensions and the mechanisms by which large surface
`areas on proteins modulate protein-protein interactions. These points are
`addressed in this volume. Here, we present an overview of the salient
`features of these topics.
`
`THE ANTIGENIC NATURE OF IMMUNOGWBULINS
`
`ldiotopes represent a particular category of antigenic determinants which
`can activate clones bearing complementary paratopes through a self-recogni­
`tion process. This behavior implies that idiotopes are auto-antigens: self­
`antigens recognized by the immune system. One model proposes that the
`response to such self-proteins is directed against sequence regions that
`exhibit the highest evolutionary variability [II]. Therefore, according to this
`model, sequence-variable regions are antigenic and evolutionarily conserved
`regions induce tolerance. From a structural perspective, variable, and there-
`
`2 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTIVES ON ANTIGENICITY AND ID!OTYPY
`
`341
`
`fore antigenic, regions can tolerate local changes in conformation and should
`correlate with sequence regions which are Oexible and surface exposed (12).
`Thus, the combination of intrinsic factors such as mobility and accessibility,
`and extrinsic host factors such as tolerance, immune response genes, idi­
`otype networking and structural gene repertoire appear to describe protein
`antigenic structure [ l l , 13).
`Historically, complementary relationships in the recognition properties of
`immunoglobulins have been attributed to the hypervariable nature of immu­
`noglobulin sequences; complementary determining regions (CDRs) [14).
`These regions have been typically associated with the antigen binding site
`[15] and have shown some correlation with the self-association of light and
`heavy chains [16, 17). Conversely, residues which are not classically hyper­
`variable can be complementary in the context of idiotope recognition [18).
`
`SURFACE VARIABILITY ANALYSIS
`
`Considering that the classical views of immunoglobulin (lg) hypervariability
`and binding site complementarity may not necessarily be equivalent con­
`cepts, it may be appropriate to re-evaluate variability from other viewpoints.
`A theoretical approach, referred to as surface variability analysis, couples
`both intrinsic and extrinsic factors of antigenicity by considering the evolu­
`tionary variability of protein surface regions [ 19). This method characterizes
`autoantigenic loci in protein families based upon examination of the vari­
`ability in the hydrophilic properties of evolutionarily variant protein se­
`quences. Surface variability is measured as a function of hydrophilicity and
`evolutionary sequence variation [19). For each sequence, hydration poten­
`tials defining the affinity of each amino acid side chain for solvent water (20,
`21) are averaged over six residues and inverted to make hydrophilic values
`positive. At each sequence position, the resulting hydrophilicity profiles are
`averaged to form a consensus value and assayed for variability according to
`the formula of Wu and Kabat [14): number of different (hydrophilicity)
`values divided by the frequency of the most common (hydrophilicity) value.
`The product of the consensus and variability of hydrophilicity values is used
`to define a surface variabilty index giving maximal values for surface­
`exposed sequences which varied significantly during evolution, i.e. those
`likely to form antigenic determinants. Advantages of surface variability
`analysis over consideration of surface-accessibility to antibodies are 1) a
`canonical ensemble of structures is evaluated, 2) parameters associated with
`
`3 of 18
`
`BI Exhibit 1054
`
`

`

`342
`
`T. KlEBER-EMMONS. E. GETZOFF, and H. KOHLER
`
`the intrinsic factors of protein structure are related to the extrinsic biological
`factors of evolution that play a role in defining antigenicity, and 3) the
`distinction shown between strong and weak immunodominant regions corre­
`lates with serological trends.
`Surface variability analysis of variable domain lg sequences highlights
`potential autogenic surface regions, referred to as idiotope determining
`regions (IDRs). Surface variability profiles (Fig. l) for a family of2S mouse
`and human lg variable region sequences [22] show that, although the major­
`ity of IDRs correspond to hypervariable regions, IDRs also occur in frame­
`work regions. It is clear from the profiles that the majority of surface variabil­
`ity in this lg family resides in the heavy chain. In Figure 2, the antigenic
`topography of the Fv region is illustrated by mapping surface variability
`values (classified into 4 categories from Figure I: most variable, more
`variable, Jess variable, and least variable) onto the surface of the 3-dimen­
`sional structure of MPC603 (23, 24]. IDRs are depicted by the most brightly
`colored surface regions (Fig. 2B) contributed by residues with high surface
`variability (shown with labels in Fig. 2A). Clearly, IDRs cover a continuum
`of binding sites in the variable region. The large repertoire of IDRs should
`allow many combinatorial possibilities for idiotope expression in three
`dimensions, including those formed solely by light chain residues, those
`formed solely by heavy chain residues, and those formed by residues of both
`chains. Topographic mapping of one idiotypic system has shown a linear
`idiotope map spanning from the antigen binding site to the vicinity of the
`constant region (see Greenspan and Monafo, this issue). Surface variable
`regions including framework residues may be recognized by more cross­
`reactive anti-idiotypic antibodies, since fewer CDR residues are involved.
`Of the framework residues in the light chain, only 49 and 85 (numbered
`sequentially according to MPC603) exhibit high surface variability (Fig. 2).
`Both are isolated sequentially and spatially from the CDRs. In the heavy
`chain, framework residues with high sequence variability include those
`adjacent in sequence to CDRs (30, 49, and 99-100), those conformationally
`adjacent to CDRs on the surface (76-79), and those distant from the CDRs
`(84, 86, 88). Two clusters of heavy chain framework residues (76-79 and 84,
`86, 88) form likely IDRs; residues 76-79 form a protruding beta bend made
`up of most variable residues, while residues 84, 86, and 88 (outwardly facing
`residues along a beta strand) form a relatively small flat surface patch of more
`variable residues. These surface topography and surface variability charac­
`teristics suggest that the region emcompassing residues 76-79 forms the
`most likely IDR outside of the antibody-combining site.
`
`4 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTIVES ON ANTIGENICITY AND IDlaTYPY
`
`343
`
`Llghl Choin
`
`0
`
`20
`
`40
`
`60
`
`80
`
`100
`
`120
`
`A
`
`Heovy Chein
`
`3000
`
`2500
`
`� 2000
`:E 1500
`Cl
`·c c
`> 1000
`
`500
`
`0
`
`:}()()()
`
`2500
`
`.... 2000
`::
`:E 1500
`Cl
`;: c > 1000
`
`500
`
`0
`
`0
`
`40
`
`80
`60
`Sequence DUJnber
`
`100
`
`1 20
`
`B
`
`FIGURE I Plots of the linear relarionship of surface variability to sequence position in the
`variable regions of the light (A) and heavy (B) chains from 25 mouse and human immu­
`noglobulins [22]. Residues are numbered sequentially to match the sequence of MPC603.
`Variability is averaged over six residues and plotted a1 1he third position to allow appropriate
`mapping onto the three-dimensional structure (see Fig. 2A). Complementarity-determining
`regions are shown by horizomal bars: light chain CORI (residues 24-40). CDR2 (56-62). and
`CDR3 (95-103) and heavy chain CORI (31-35), CDR2 (50-68), and CDR3 (IOI-Ill). Long
`dashed horizontal lines separate the four categories of surface variability used to color code
`Figure 2B. The residues included in each category for the light chain: most variable (residues
`25-26, 28, 29, 36, 38. 96-100). more variable (27, 37, 39, 49, 58-60, 85. 95, IOI), less
`variable (22-24, 40, 42, 45-48, 50, 56-57, 61-63. 84, 86-89, 102-103, 108, 112), and least
`variable (1-21. 30-35. 41, 43-44, 51-55. 64-83, 90-94, 104-107, 109-lll, 113-115); aod for
`the heavy chain: most variable (32-33, 51-55. 58-64, 66. 77-79, 99-105), more variable (30.
`35, 49-50, 65, 76, 84, 86, 88. 106-!09), less variable (4-5. 7, 18, 28-29, 31, 34, 36-37, 45,
`47-48, 68-69, 71-75, 80-83, 85, 87. 89-91. 98. 110-111), and least variable (1-3, 6. 8-17,
`19-27, 38-44, 46, 56-57, 67, 70, 92-97, 112-122).
`
`5 of 18
`
`BI Exhibit 1054
`
`

`

`344
`
`T. KIEBER-EMMONS, E. GETZOFF. and H. KOHLER
`
`A
`
`FIGURE 2 Computer graphics images of immunoglobulin surface variability mapped onto
`the light and heavy chain variable domains of MPC603. Labeled alpha carbon backbones (A)
`and solid external molecular surfaces (B) are shown in the same orien1a1ion, looking down into
`the combining site from the solvent. The alpha carbon back.bone (A) of the light chain (top) is
`shown in blue with yellow CDRs and the alpha carbon backbone of the heavy chain (bottom) is
`shown in purple with green CDRs. In both the light and heavy chains, residues with high average
`surface variability are labeled (A) by number at the alpha carbon position in yellow (more
`variable) and red (most variable). The solid external molecular surface (B) is color coded by the
`surface variability (defined in texl and shown in Fig. I) using a radiating body color scale with
`increasing variability corresponding to increasingly brighter color. The highly (most and more)
`variable regions (lightest colors in B) are defined 10 be potential lDRs; the majority are also
`CDRs. The most prominent highly variable surface region outside the CDRs is formed by heavy
`chain residues 76-79 (bottom right), which are conformationally adjacent to CDR2. Other
`framework residues with high surface variability are light chain 49 (behind) and 85 (upper
`righti, heavy chain 30, 49 and 99-100 (adjacent in sequence to CORI, CDR2, and CDR3.
`
`6 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTIVES ON ANTIGENICITY AND IDIOTYPY
`
`345
`
`respectively), and heavy chain 84, 86, and 88 (lower left). The backbone is displayed using the
`graphics programs GRAMPS [25) and GRANNY (26). The molecular surface, defined by
`mathern<ttically rolling a probe sphere (1.4 A radius) representing a solvent water molecule over
`the van der Waals surface of the protein, is calculared and displayed using the programs AMS
`and RAMS (27, 28). Crystallographic coordinates [23, 24) are taken from the Brookhaven
`Protein .Data Base [29). (See Color Plates I and II in the color secrion of this issue.)
`
`B
`
`7 of 18
`
`BI Exhibit 1054
`
`

`

`346
`
`T. KIEBER-EMMONS. E. GETZOFF. and H. KOHLER
`
`IDIOTOPE/PARATOPE OVERLAP
`
`IDRs agree quite well with idiotope locations deduced from sequence anal­
`
`
`
`ysis (Table I; see also Chen, et al., this issue). IDR areas have been shown 10
`
`
`
`be surface-exposed (see Novotny in this issue). Surface variability analysis
`has implied that 1) there are more IDR than CDR loci and 2) all CDR loci are
`
`
`
`also IDR loci. Structural and functional implications of this relationship
`
`between CDRs and IDRs have been discussed [22) (see also Stevens and
`
`Schiffer, this issue). The overlap between idiotopes and paratopes [22]
`
`
`
`as binding sites; the dual roles of implies that idiotopes can participate
`binding to a determinant or being bound by another antibody may be
`
`
`
`
`
`expressed simultaneously by the same topographic site. Consequently, sites
`
`
`on Igs may not be easily characterized functionally [22). In support of this
`
`Correspondence of Experimentally Deduced ldiotype Locations and Predicred IDRs
`
`TABLE l
`
`System
`
`Anti-Dextran
`
`Anti-Phosphocholine
`Anti-Galactan
`
`Ars·A
`
`Anti·lnulin
`
`B lymphocyte
`Anti-4-hydroxy-3-nitro-S-idiopbenyl
`IgM-RF
`
`Experimental
`Position
`
`H96-97
`H53-54
`HFR3
`HV3
`H96-97
`H5 3
`L30
`H59
`HV3
`LV3
`LS3
`L56
`L53
`H94-99
`HV3
`H94-99
`L28-31
`L53
`HV2
`LFR2
`LFR3
`
`Predicted
`Position
`
`IDRE-H
`IDRC-H
`IDRD-H
`IDRE-H
`IDRE-H
`IDRC-H
`IDRB-L
`IDRD-H
`IDRE-H
`IDRG-L
`IDRD-L
`IDRD-L
`IDRD-L
`IDRE-H
`IDRE-H
`IDRE-H
`TDRB-L
`IDRD·L
`IDRC-H
`IDRC-L
`IDRE-L
`
`References
`
`30, 31
`32, 33
`18
`34
`35
`
`36
`
`32
`
`37
`38
`39-44
`
`45
`
`Experimental positions correspond to H (heavy), L (light) chain designation with standard
`numbering scheme (93]. HV and LV designation for heavy chain variable and light chain
`variable regions I to 3, respectively (93]. The nomenclature for corresponding IDR predicted
`positions are from [22].
`
`8 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTrVES ON ANTIGENICITY AND IDrOTYPY
`
`347
`
`notion is the recent description of two antibody structures, one with a unique
`V-V packing arrangement (see Stevens and Schiffer in this issue) and the
`other emphasizing the expanse of contact area across the surface of the
`antibody [46).
`Jdiotypic and anti-idiotypic interactions have been classified according to
`the location of the idiotope on the lg surface relative to their paratope [ 6, 47-
`50). Definitions derived from such topographic relationships are arbitrary in
`that an idiotypic antibody (Ab!) that is defined for one system can be an anti­
`idiotypic antibody (Ab2) in another system. The fact that the same antibody
`can bind epitopes while presenting itself as an antigen further emphasizes
`the correlation between paratopes and idiotopes.
`
`STRUCTURAL VERSUS TOPOGRAPHIC DETERMINANTS
`
`The phenomenon of molecular mimicry would appear to suggest a chemicaV
`structural equivalence between the antigen and anti-Id antibody. However,
`similar three-dimensional surface environments do not imply that the sec­
`ondary and tertiary folding pattern of the antibody and anti-idiotype are the
`same. For protein antigens, the identification of homologous sequence
`regions between the nominal antigen and anti-Id does not ensure the identi­
`fication of the "internal image," since different three-dimensional environ­
`ments influence the folding patterns of the related sequences (51, 52). On the
`other hand, the framework (FR) region of an antibody represents a simple
`beta sheet scaffold onto which binding sites may be built, implying that the
`structure of CDRs is relatively independent of the FR context. The confor­
`mation of CDR loops between beta strands depends on loop sizes and
`specific interactions between the loop and the beta sheet. Studies on the
`conformational attributes of the antibody molecule [53-57] have empha­
`sized antigen binding. Here, we are interested in how the folding patterns if
`Igs can be related to the folding pattern of nominal antigens, in particular,
`whether certain CDRs and FR regions influence the expression of idiotopes
`in terms of sequence or backbone conformation. Historically, determinants
`that seem to be affected by single residue changes have been described as
`topographic, because the immune system must be responding to local
`changes in the surface topography of the molecule [58]. Alternatively,
`determinant that involve changes in the backbone conformation were called
`conformational determinants or structural determinants [59). Neither of
`these definitions preclude that the determinants may be discontinuous. An
`
`9 of 18
`
`BI Exhibit 1054
`
`

`

`348
`
`T. KIEBER-EMMONS, E. GETZOFF. and H. KOHLER
`
`important difference in the two determinant features is that a synthetic
`peptide may not be able to adopt a particular conformational determinant.
`The subtle difference between these determinant types may also influence
`the expression of cross-reactive idiotopes which certainly play a role as
`regulatory idiotopes [18).
`The nature of antibody-antibody interactions implies that extended areas
`of the protein surface contribute to binding, leading to a multisite interaction
`model for idiotope recognition. The required thermodynamic conditions for
`such interactions can be achieved via several sources (13] and can be quite
`precise in that the interactions can be affected by a single amino acid change
`(60-63]. Experimental observations idicate that both sequence and confor­
`mation dictate an immune response (64, 65]. In either case, the degree of
`spatial adjustments in the molecular partners results in different free energies
`of association for any given antibody with any other given antibody or
`antigen. The overall energetics of the interaction is determined by the free
`energy cost (G) of any conformational changes experienced by either the
`antigen (anti-Id) or the antibody upon complex formation. At the molecular
`level, the relative energy cost of the spatial displacement of interacting
`groups is intrinsically associated with the relative mobility of a region. The
`concept of macromolecules as flexible entities and range of the dynamical
`nature of proteins has been extensively discussed [66-72].
`Determinants are often comprised of discontinuous parts of an antigen. In
`the crystal structure of Amit and coworkers [46], the lysozyme determinant
`is made up of two stretches of polypeptide chain comprising residues 18 to 27
`and 116 to 129. Surface variability analysis of lysozyme [19] is in relatively
`good agreement, identifying the majority of residues in this epitope (resi­
`dues 14-21 and residues 115-126) as autogenic loci. While CDR3 of the
`heavy chain of the antilysozyme forms the principle contacts, all six CDR
`regions are involved in contacting lysozyme. Molecular modelling studies of
`Jysozyme-antilysozyme complexes [73] also suggest that the loop region
`epitope (residues 57-84) of lysozyme is described by autogenic loci (19]
`centered on positions 67-79. In these complexes all six CDR regions are also
`involved in defining contacts.
`ldiotopes (and antigenic determfoants in general) have been localized by
`evaluating the reactivity of antibodies to the protein with synthetic peptides
`derived from different parts of a protein [33) (see Chen, et al., this issue).
`Small peptides are thought to exist in a multiplicity of transient conforma­
`tional states in dynamic equilibrium, in contrast to the relatively stable
`structure of a protein in solution (74]. Nevertheless, small peptides can
`
`10 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTIVES ON ANTIGENICITY AND IDIOTYPY
`
`349
`
`induce antibodies with a sequence and structural requirement for binding
`antigen comparable to antibodies raised against the native protein [3].
`Whether this is because certain peptides adopt a conformation similar to that
`found in the native protein [75] or this results from induced fit remains to be
`seen. The energetic considerations which mediate antipeptide-peptide com­
`plexes and the relationship to antipeptide-antibody interactions with the
`parent molecule have been discussed [13, 64, 65].
`The degree of specificity of a particular complex, as well as the possible
`description of epitope regions, relies on the consorted, dynamical nature of
`an antigen-antibody interaction [12, 76, 77]. However, in some circum­
`stances a restriction in the conformational freedom of immunizing peptides
`will result in antibodies with the same specificity as those induced by the
`proteins themselves [78, 79]. This result focuses on the supporting structural
`role of regions remote in sequence to a primary epitope center. Short
`synthetic linear peptides may not account for such environmental contribu­
`tions [80]. The influence of a tertiary environment on the natural conforma­
`tions of short peptide segments is clearly evident, in that identical sequence
`segments in native proteins can have different conformations [51, 52]. The
`possible fine specificity in antigenic recognition by B cells apparently
`extends to T cells, where different determinants can be formed by the same
`peptide and la molecule [81].
`
`MIMICRY OF CONTACT RESIDUES
`
`The possible ways in which anti-idiotypic antibodies can mimic antigens has
`been summarized (82). Sequence homology with the protein antigen has
`been suggested for the mimicking capabilities of monoclonal antibodies in
`the reovirus system [83). Sequence analysis of a hybridoma that mimics the
`reovirus antigen indicates sequence homology in an expected IDR of the
`kappa light chain [83]. A linear synthetic decapeptide derived from the anti­
`Id (LV2;hypervariable region 2 of the light chain;CDR2), which is homolo­
`gous to reovirus in five positions and has three conservative substitutions is
`capable of inducing biological responses similar to the reovirus antigen as
`well as the anti-Id [83 and M. Greene, personal communication]. Essential
`contact residues which define this reovirus epitope appear to be retained in
`the Ab2 in a homologous, linear fashion. In a random fashion, the likelihood
`of homologous matching of five residues in this hypervariable region, as
`evidence in the reovirus system, is 7 :20s. Although such homology would be
`
`11 of 18
`
`BI Exhibit 1054
`
`

`

`350
`
`T. KIEBER-EMMONS, E. GETZOFF, and H KOHLER
`
`rare in randomly generated sequences. analysis of viral protein sequences
`suggests that rudimentary viral epitope sequences are established within the
`lg germ lines (Kieber-Emmons and Kohler, unpublished).
`In Table II, light chain sequences of the crystallographically known lg
`structures are aligned with those of the proposed reovirus epitope and its
`mimicking anti-Id. The resulting homology points out the possible structural
`similarities between the mimicking monoclonal 87. 92.6 and MPC603 in this
`region. In Table III, heavy chain-sequence comparisons with reovirus indi
`cate additional homology including the identical reovirus sequence (NSYSGS).
`Comparison of the crystallographic structures of HV2 (hypervariable region
`2 of the heavy chain;CDR2) regions from NEWM and MPC603 also indi­
`cates that the conformational properties are highly constrained in spite of
`sequence differences.
`The alignments in HV2 and LV2 provide an opportunity to compare the
`conformational properties of the sequences of the proposed reovirus epitope
`and the mimicking monoclonal. The sequences in the two tables can be
`considered as structural variations of beta bends. The structural similarities
`provide a data base [64] to help elucidate residues essential for antibody
`87.92.6 recognition and binding and residues integral to the idiotope be­
`cause they stabilize secondary structure essential to the presentation of the
`antigenic site.
`The identification of sequence similarities between an antibody and an
`antigen is nongermane in the analysis of anti-idiotypic antibodies mimicking
`carbohydrates or haptenic antigens. For these cases, conceptual physical/
`chemical models (e.g. the proper alignment of functionally reactive groups,
`or of residues with van der Waals interaction tendencies, shape or other
`physical attributes resembling those of the antigen) must be invoked as an
`initial basis for mimicry of nonprotein antigens. Such models must also be
`examined in cases where the mimicking antibody and protein do not exhibit
`sequence homology. Thus, the degree of chemical or sequence similarities
`will certainly affect the degree of mimicry fidelity or faithfulness.
`An anti-TIS hybridoma, 4CJI, is capable of inducing biological effects
`similar to those induced by phosphorylcholine (PC) (85, 86]. Competitive
`bindings assays have shown that PC is a successful inhibitor of 4Cll-Tl5
`binding. Crystallographic analysis of the PC-binding antibody MPC603 and
`PC-binding studies provide a basis for examining potential contact residues
`in the variable domains of 4Cll that could provide chemical mimicry of PC.
`Conformational attributes of model peptides interacting in a manner similar
`to PC with MPC603 suggests that the tetrapeptide Lys-Gly-Gly-Asp is
`
`12 of 18
`
`BI Exhibit 1054
`
`

`

`Alignment of Struc1urally Known LV2 Regions with Reovirus and Anti-Id
`
`TABLED
`
`MPC603
`Reovirus
`87.91.6
`NEWM
`KOL
`
`Leu
`Ue
`Leu
`Leu
`Leu
`
`Leu
`Val
`Leu
`Leu
`Leu
`
`Tyr
`Gly
`Ile
`Ser
`Ser
`Tyr
`Tyr Ser
`Ile
`Arg
`lie 'fyr
`Ile
`Tyr
`Arg
`
`Ser Thr
`Ala
`Gly Ser
`Gly
`Gly
`Ser
`Thr
`Lys
`Asp
`Asp
`Ala Met
`Asp
`
`Gin
`Arg
`Ser
`Asn
`Trp
`Leu
`Leu
`Gin
`Ser
`Arg Pro
`Ser
`Pro Ser
`Arg
`
`Gly
`Arg
`Gly
`Gly
`Gly
`
`The sequences for MPC603, NEWM, KOL, and reovirus were obtained from Protein Identification Resource [84]; the sequence for 87. 91.6 is from
`[83].
`
`Alignmenl of Selected HV2 Regions wilh Reovirus and Mimicking Anti-Id
`
`TABLE 01
`
`Reovirus
`36-60
`NEWM
`MPC603
`87.91.6
`
`Gln Ser Met - Trp
`Ile Gly
`Ile
`Asn Lys
`Leu Glu His Met Gly Tyr
`Trp Ile Gly 'fyr
`Lys Gly Leu Glu
`Lys Arg Leu Glu
`Ala Ala
`Trp
`Ile
`Gtn Gly Leu Glu
`Trp
`Ile Gly Arg
`Sequences for reovirus. 36-60, NEWM, and MPC603 are from lhe Protein Identification Resource [84]; lhe sequence for 87.91.6 is from (83].
`
`Tyr Ser Gly
`Val
`Ser
`Ser Gly Leu Asn
`Ser Tyr Ser Gly
`Ile
`Ser Thr
`Tyr Tyr
`Val Phe
`Tyr His Gly Thr
`Ser Asp Asp
`Ser Arg Asn
`Lys
`Tyr Thr
`Lys Gly Asn
`Asp Pro Ala Asn Gly Asn Thr Tyr
`lie
`
`;:g
`;;o
`"'
`�
`rn
`�
`�
`Ci
`�
`fi
`::i
`-<
`�
`8 s -<
`
`�
`
`"" .,,,
`
`.:D
`...
`- ct
`z;
`...
`• m
`r-
`
`�-;z
`,.
`..... v
`
`13 of 18
`
`BI Exhibit 1054
`
`

`

`352
`
`T. KJEBER-EMMONS, E. GETZOFF, and H. KOHLER
`
`representative of the major antigenic determinant of 4C! l which interacts
`with Tl 5 (87]. Competitive inhibition assays have shown that the synthesized
`model tetrapeptide is capable of inhibiting 4Cll-T15 binding, though not as
`effectively as PC (87). Sequence analysis (88) and molecular modelling of
`4CI I indicates that a loose chemical mimicry of the dipolar character of PC
`localized in CD2 oftbe heavy chain may have a dominant role in determining
`the antigenic topography of the hybridoma. For a small haptenic antigen like
`PC, which is being mimicked by a comparatively large molecule like an
`antibody, mimicry may be mediated by appropriately placed contact residues
`that reside on different parts of the hybridoma. The mode of interaction of an
`antibody with a small hapten and with a large protein are different. So there
`may be multiple ways in which mimicry can be realized. Residues of anti-PC
`antibodies that interact with PC may be utilized in 4CI l binding, but other
`residues of the anti-PC antibodies that do not contact PC may now contact
`4Cl 1. Anti-PC antibodies with different sequences may not bind 4Cl I or may
`bind with very low affinity, due to the proximity of noncornplementary
`residues over a large surface area in the complexes.
`
`SUMMARY
`
`The recent crystal determination of a lysozyme-antilysozyme complex
`provides a three-dimensional prototype of the manner in which contacts in
`idiotype-anti-idiotype interactions may be realized [46]. Such interactions
`can be approximated by two complementary "flat" surfaces. Each IDR
`(autoantigenic locus) location might provide a particular recognition feature
`between two interacting partners. The combinatorial manner in which IDR
`domains are recognized by anti-idiotypic antibodies describe the repertoire
`of private and public (crossreactive) idiotopes of an antibody.
`Several interesting features emerge from consideration of the Ab contact
`residues in the crystal structure. First, framework residues are implicated in
`contacting tbe antigen: Thr 30 (FRI) of the heavy chain and Tyr 49 (FR2) of
`the kappa light chain. Both of these residues lie within predicted lDRs (22].
`Framework regions have recently been suggested to be involved in several
`anti-idiotypic systems (18, 45), although such regions have, in the past, been
`disregarded based solely upon sequence analysis. The surface variabilty
`analysis, which identifies the repertoire of complementary interacting sur­
`faces, depicts the immunoglobulin as having more variability than generally
`
`14 of 18
`
`BI Exhibit 1054
`
`

`

`PERSPECTIVES ON ANTIGENICITY AND IDIOTYPY
`
`353
`
`thought. This variability may also extend to T cell receptors since T cell
`chains express an extensive surface variable repertoire similar to that of the
`irnmunoglobulin bght chains (Kieber-Emmons and Kohler, unpublished).
`Second, the D region plays a critical role in the generation of the anti­
`lysozyme combining sites. Similarly, the D segment makes up the largest
`component of an IDR (22]. Third, while the CDR3 of the heavy chain
`contributes most to the antibody-lysozyme complex it is not the most
`surface-exposed (see Novotny, this issue). Nevertheless, surface variability
`analysis indicates that this region is generally immunodominant (22] which
`is also observed experimentally [33). Together, these results indicate that
`perhaps certain JDR regions are intrinsically more antigenic.
`ldiotypic structures must be accessible for antibody recognition and
`binding. From a structural viewpoint, a single antibody molecule has a
`continuum or several different combining sites (89]. Subsequently, a single
`residue can be contained in several overlapping idiotypic determinants.
`Surface variability analysis suggests that the hypervariable regions of lgs
`provide a diverse idiotope repertoire that can be utilized for binding. Mono­
`clonal antibodies have been shown to have multiple specificities (90-92) and
`this capacity for multiple binding is also intrinsic to the definitions that have
`emerged for anti-idiotypic antibodies.
`From an application perspective, although vaccination against bacterial
`and viral diseases has been a major achievement of immunology, there sti

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