7
Antibody-antigen interactions David M Webster, Andrew H Henry and Anthony R Rees University of Bath, Bath, UK The structural origins of antibody diversity are well understood as a result of X-ray crystallographic and molecular modelling studies of Fab fragments. A similar understanding of antibody specificity is beginning to emerge from an analysis of structures of hapten, peptide and protein antibody complexes. While the nature of the antibody-antigen interface, and any conformational changes that occur on complex formation, can be described in structural terms, a full explanation of the thermodynamic and mechanistic basis of affinity is less accessible from structure alone. A number of physiochemical studies carried out on wild type and mutant antibodies have raised questions about the natureof theenergetics of the interaction, and the possibility of a key role for water molecules has been discussed. The possibility of 'induced fit' as a common mechanism for antigen-antibody interactions has been raised, and a molecular basis for hapten and protein cross-reactivity proposed. These recent contributions to the field, as well as providing partial solutions to old problems, have provided exciting new insights. Current Opinion in Structural Biology 1994, 4:123-129 Introduction The interaction between an antibody and its antigen has become a paradigm for the study of molecular recognition. Ehrlich and Landsteiner in the early part of the century recognized the importance of antibod- ies [1]. The manner in which antibodies have been shown to be organized at the genetic level has re- vealed a potential for molecular diversity unmatched anywhere else in the genome. Translation of this pri- mary code into three-dimensional antibody structures is beginning to unravel the molecular basis of this di- versity, and in doing so, reveal unexpected subtleties and new mechanisms for debate. For example, a relationship between antigen type and the topography of antibody combining sites (ACS) has begun to emerge from X-ray crystallographic studies of antibody complexes (reviewed in [2]). In attempting to uncover the thermodynamics of such interactions by protein engineering and calorimetric studies, pro- posals have emerged to the effect that the affinity of binding is driven by entropic (hydrophobic) effects [3"], by enthalpic (van der Waals, hydrogen-bond, salt bridge) effects [4"], or by entropy-enthalpy compen- sation [5"]. There have also been attempts to explain the long observed phenomenon of 'cross reactivity' whereby an antibody reacts with a number of epi- topes [6", 7",8"]. In one instance, a crystallographic study of Fab-steroid complexes led to the assertion that functionally inert haptens such as steroids may not display sufficient 'molecular character' to induce high specificity antibodies [6"]. It has also been pro- posed that the primary mechanism by which antigens bind to antibodies is 'induced fit' [9"]. In reviewing recent work in this field, we will focus on the nature of the antigen interaction and how structural methods, physical chemistry, and molecular biology are together contributing to the debate. In passing, we will address the question: is extrapolation from a few observations to general theorems on the mecha- nism by which antigens and antibodies interact now possible? The antibody combining site: variation with antigen type The current Brookhaven database contains 31 antibody structures, of which 14 are complexes with antigen (see Table 1). Three classes of combining site can be described: cavity (hapten), groove (peptide, DNA, car- bohydrate) and planar (protein), as illustrated in Fig. 1. The amount of Fab surface area buried on complex for- mation increases in the order cavity < groove < planar (see Table 2), while the percentage of antigen surface area buried in the interface decreases as the size of the antigen increases. When the antibody-antigen interfaces are inspected, a high degree of shape complementarity is of- ten seen, particularly for protein complexes. In an Abbreviations ACS~antibody combining sites; CDR--complementarity determining regions. © Current Biology Ltd ISSN 0959-440X I 23

Antibody-antigen interactions

Embed Size (px)

Citation preview

Page 1: Antibody-antigen interactions

Antibody-antigen interactions David M Webster, Andrew H Henry and Anthony R Rees

Univers i t y of Bath, Bath, UK

The structural origins of antibody diversity are well understood as a result of X-ray crystallographic and molecular modelling studies of Fab fragments. A similar understanding of antibody specificity is beginning to emerge from an analysis of structures of hapten, peptide and protein antibody complexes. While the nature of the antibody-antigen interface, and any conformational changes that occur on complex formation, can be described in structural terms, a full explanation of the thermodynamic and mechanistic basis of affinity is less accessible from structure alone. A number of physiochemical studies carried out on wild type and mutant antibodies have raised questions about the natureof theenergetics of the interaction, and the possibility of a key role for water molecules has been discussed. The possibility of 'induced fit' as a common mechanism for antigen-antibody interactions has been raised, and a molecular basis for hapten and protein cross-reactivity proposed. These recent contributions to the field, as well as providing partial solutions to old

problems, have provided exciting new insights.

Current Opinion in Structural Biology 1994, 4:123-129

Introduction

The interaction between an antibody and its antigen has become a paradigm for the study of molecular recognition. Ehrlich and Landsteiner in the early part of the century recognized the importance of antibod- ies [1]. The manner in which antibodies have been shown to be organized at the genetic level has re- vealed a potential for molecular diversity unmatched anywhere else in the genome. Translation of this pri- mary code into three-dimensional antibody structures is beginning to unravel the molecular basis of this di- versity, and in doing so, reveal unexpected subtleties and new mechanisms for debate.

For example, a relationship between antigen type and the topography of antibody combining sites (ACS) has begun to emerge from X-ray crystallographic studies of antibody complexes (reviewed in [2]). In attempting to uncover the thermodynamics of such interactions by protein engineering and calorimetric studies, pro- posals have emerged to the effect that the affinity of binding is driven by entropic (hydrophobic) effects [3"], by enthalpic (van der Waals, hydrogen-bond, salt bridge) effects [4"], or by entropy-enthalpy compen- sation [5"]. There have also been attempts to explain the long observed phenomenon of 'cross reactivity' whereby an antibody reacts with a number of epi- topes [6", 7",8"]. In one instance, a crystallographic study of Fab-steroid complexes led to the assertion that functionally inert haptens such as steroids may not display sufficient 'molecular character' to induce

high specificity antibodies [6"]. It has also been pro- posed that the primary mechanism by which antigens bind to antibodies is ' induced fit' [9"].

In reviewing recent work in this field, we will focus on the nature of the antigen interaction and how structural methods, physical chemistry, and molecular biology are together contributing to the debate. In passing, we will address the question: is extrapolation from a few observations to general theorems on the mecha- nism by which antigens and antibodies interact now possible?

The antibody combining site: variation with antigen type

The current Brookhaven database contains 31 antibody structures, of which 14 are complexes with antigen (see Table 1). Three classes of combining site can be described: cavity (hapten), groove (peptide, DNA, car- bohydrate) and planar (protein), as illustrated in Fig. 1. The amount of Fab surface area buried on complex for- mation increases in the order cavity < groove < planar (see Table 2), while the percentage of antigen surface area buried in the interface decreases as the size of the antigen increases.

When the antibody-antigen interfaces are inspected, a high degree of shape complementarity is of- ten seen, particularly for protein complexes. In an

Abbreviations ACS~antibody combining sites; CDR--complementarity determining regions.

© Current Biology Ltd ISSN 0959-440X I 23

Page 2: Antibody-antigen interactions

124 Folding and binding

Table 1. List of antibodies and their fragments whose X-ray coordinates have appeared in the Brookhaven database of protein structures during

the past year,

Brookhaven Resolution Antigen entry Name (I~) type Reference

1 dba, 1 dbb DB3 2.7-3.0 hapten 6 1 ncd NC41 2.9 protein 10 1 igf/2igf B1312 2.8 peptide 20 1 hil/1 hin/1 him 17/9 2.0 peptide 22 lggi, lggb, l g g c 50.1 2.8 peptide 23 ldfb 3D6 2.7 peptide 37 2fbj J539 1.95 CHO 39 ligi 26-10 2.7 hapten 40 7lab NEW 2.0 Myeloma 41 Imam YS*T9.1 2.45 Cl iO 42 I baf ANO2 2.9 hapten 43 8lab HIL 1.8 myeloma a 1 bbd 8F5 2.8 hapten 44 ligm POT 2.3 peptide 45

aFA Saul and RJ Poljak, unpublished results

analysis of the interface of the influenza virus N9 neuraminidase-NC41 Fab complex it was found that the average packing density of atoms at the inter- face was lower than for a typical protein core [10]. This result is in contrast with the analysis of Walls and Sternberg [11] who found that the packing densi- ties of the three anti-hen egg-white lysozyme antibody interfaces were as high as the protein cores. In con- trast, electrostatic complementarity is not always high [12]. For example, in the N9 neuraminidase-NC41 com- plex, a number of polar residues in the interface are not formally neutralized, though many are in close proxim- ity to shielding residues.

It has also been suggested that water molecules, present at the interface, may mediate antibody-antigen interactions by increasing the packing density and contributing to charge complementarity [10,13]. Wa- ter molecules have been found in the interface of the anti-lysozyme antibody D1.3 [13-15], around the periphery of some anti-protein interfaces [10,15,16], and in the combining site of some anti-peptide anti- bodies [17]. The proposal that water molecules play a general role in protein recognition may be a little premature in the presence of only a handful of struc- tures [13]. However, it is a reasonable speculation that, where complementarity is imperfect, filling-in by water molecules might occur.

Structural basis of specificity, affinity and cross-reactivity

Analysis of cavity-type antibodies suggests a correla- tion between buried surface area in combination with

van der Waal contacts, and affinity (Table 2, upper panel). However, it is likely that to generate high affinity and specificity, hydrogen-bonds and/or salt bridges are also required. This is evident when the characteristics of the anti-steroid antibody, DB3, are compared with those of the anti-fluorescein antibody, 4-4-20 (Table 2, upper panel). In the former case, cross- reactivity of the antibody is seen with several steroids which, in the X-ray structures, are seen to bind to alter- native subsites in the combining site [6%18"°], depend- ing on whether the steroid A-ring adopts a syn or anti conformation. The shape complementarity of this ACS to the progesterone immunogen is not totally specific. In comparison, the sheep anti-progesterone antibody, D7B2 has an affinity 200-times higher than DB3, and appears to lack the syn/anti dual accommodation al- lowed by DB3 [191. By contrast, the fluorescein cavity exhibits close complementarity to the hapten and the additionaJ~specificity and affinity observed are likely to result/from extra hydrogen-bonds and a buried salt bridge.

These comparisons draw attention to the fact that our notions of absolute specificity and maximum affinity arise from the systems we have available for inspection. Experience with other systems where small molecule-protein interactions occur, such as biotin-avidin, suggests that the practical upper limit of affinity may be about 1015M -1. This is at least 104 times greater than that of the best murine anti- body. While affinities of naturally occurring human antibodies are unknown, sheep antibodies routinely reach affinities of 1011-1014M -1, which suggests that the study of monoclonal antibodies from inbred mouse strains may have placed limits on antigen affinity that are atypical of outbred mammalian species ([19]; P Har- rison, personal communication).

Groove-type binding sites have been seen with anti- peptide antibodies. The first antibody-peptide com- plex studied by X-ray crystallography involved a 19 residue peptide derived from myohemerythrin [20], of which 10 residues were not visible in the electron den- sity, presumably due to their high mobility. Peptide mobility in the bound state has been studied directly using NMR [21]. While some residues may become very tightly bound on complex formation, others may be as mobile as in the free peptide. Recently, further anti-peptide antibody complexes have been reported [17,22",23"]. The central panel of Table 2 shows that for three complexes there is no obvious trend be- tween surface area, number of contacts and affinity. This may be due to the fact that the flexibility of peptide antigens, and its effect on the entropic com- ponent of the free energy of binding cannot easily be quantified.

Cross-reactivity was also seen [7"] in the binding of a series of peptides of increasing size to the light chain dimer Mcg. In a manner sin~ilar in many respects to the DB3-steroid binding, Mcg was able to bind equiv- alent parts of different peptides to the same region of the combining site, while other parts of the pep-

Page 3: Antibody-antigen interactions

Antibody-antigen interactions Webster et al. 125

(a)

P

(b) (c)

Fig. 1. Topographic classes of antibody combining site: (a) cavity type, exemplified by anti-fluomscein antibody 4-4-20 [30]; (b) groove type, exemplified by anti-peptide (myohemerythrin) antibody B13 12 [21 ]; (c) planar type, exemplified by influenza virus N9 neuraminidase-NC41 complex [13]. The antigen is shown in dark grey at the top of the figure. The antibody Fv has been pulled apart from the antigen and is the centre image. Residues which lie within 4,~ of any atom in the antigen are coloured dark grey. The bottom image shows the antibody Fv rotated through 90" to highlight the centre of the ACS.

tides adopted different binding modes. These studies draw attention to the so-called 'order-disorder para- dox', observed where anti-peptide antibodies are able to cross-react with the native protein, while anti-pro- tein antibodies do not recognize the peptide [24]. It has been suggested that such cross-reactivity is caused by antibodies that only recognize the denatured pro- tein [25]. However, this position is at odds with recent work in which such cross-reactivity has been clearly observed [26,27"]. This phenomenon still seems to be unresolved.

The planar-type binding site is characteristic of anti- protein antibodies (Fig. 1). Although the great ma- jority of antibodies have been raised against protein antigens, only five high resolution X-ray structures are known for antibody-protein complexes. These are summarized in the lower panel of Table 2. The affinities of the NC41-neuraminidase and D1.3-anti- D1.3 complexes have not been published, but they are likely to be at least 109M -1. What do these data tell us about protein antibody-antigen interactions? First, there is no obvious correlation between the

number or type of interaction seen (van der Waals, hydrogen-bond, salt bridge) and affinity. Second, the buried surface area is not a good indicator of affin- ity, as might be the case if burial of hydropho- bic residues and release of water molecules were the major contributor to t h e free energy of bind- ing. In fact, recent work [4"] provides a contrasting view. During measurements of the enthalpic and en- tropic contributions to the association of hen egg- white lysozyme with D1.3, using microcalorimetry, it was found that the binding was enthalpically driven, while the entropy decreased (AGo-47.9 kJ mol-1; AH -90.0kJmol-1; AS -42.1 kJmol-1). While this may not always be the case, it is clear that the affinities ob- served in antibody-antigen complexes have their ori- gins in rather different profiles of interaction.

In a recent study of protein cross-reactivity [8"], the structures of antibody FvD13.11 complexed with pheasant egg lysozyme and a number of uncomplexed cross-reactive avian lysozymes were compared. Of par- ticular interest was the observation of a heteroclitic re- sponse (an immune response to an antigen which pro-

Page 4: Antibody-antigen interactions

126 Folding and binding

Table 2. Some Physico-chemical characteristics of (top panel) antibody-hapten, (centre panel) antibody-peptide and (bottom panel) ant ibody-protein complexes.

Buried surface vdW Salt Antigen area, t~2 contacts H-bonds bridges Ka, M -1 Antibody name

phosphocholine 137 30 2 3 1.7 x 105 McPC603 a aetiocholanolone 227 27 2 0 4.8 x 107 DB3 phenyloxazalone 170 >15* >1 0 1.0 x 108 NQ10/12.5 5,[~-androstane 223 20 2 0 1.3 x 108 DB3 progesterone 11 ,et-hemisuccinate 291 68 2 0 2.8 x 109 DB3 ftuorescein 266 65 5 1 3.4 x 1010 4-4-20

Cholera toxin 503 21 10 1 1.2 x 106 TE33 HIV peptide 475 27 4 0 4.5 x 106 50.1 Influenza haemagglutinin peptide 400 74-81 13-15 1 5.0 x 107 17/9

Hen egg lysozyme 680 75 15 0 1 x 109 D1.3 " " 774 111 14 1 5x109 HyHEL10 " " 750 74 10 3 2 x 1 0 lo HyHEL5

Influenza neuraminidase 879 108 23 1 NA NC41 D1.3 800 99t 9 1 NA anti-D1.3

aData for McPC603, 4-4-20, D1.3, HyHEL10, HyHEL5 and NC41 are taken from [46]; the D1.3-anti-D1.3 data are taken from [47]. Peptide data are as follows: 50.1 [23]; 17/9 [22]; TE33 [17]. The hDB3 structures are from [6]. (t author's estimate, * not yet fully refined).

duces antibodies having a higher affinity to a similar, but not identical antigen) for two of the lysozymes. Residues that differed between the lysozymes were located around the edge of the epitope. Changes in conformational flexibility, coupled with the presence of water molecules around the periphery of the inter- acting surfaces, may have reduced the stereochemical constraints on complex formation.

How far could protein antigen cross-reactivity go? Given the radius of curvature of most proteins and the size of the antibody combining site, multiple paratopes would be forced to overlap. If such redundancy does exist, then proteins rather more different from one another than avian lysozymes may bind to the same antibody, thus increasing the available repertoire far beyond its present size.

Conformational change

The generality of induced fit as a mechanism governing antibody-antigen interactions has frequently been dis- cussed [2,9",13]. In some instances, small changes have been seen in sidechain position o r b a c k b o n e confor- mation [8",10,18"]; in others, no significant changes to either antigen or antibody are seen on complex for- mation [6%15]. For situations where the changes are restricted to sidechain reorientation, such that a pre- existing rotamer is preferentially selected in the bound state, the term 'induced fit' may be inappropriate. Per- haps the term 'mutual fit' better describes these types of interactions.

When Fab D1.3 binds to hen egg-white lysozyme [15] no significant changes are seen in the free and bound antigen. No structure for the free D1.3 Fab has yet been determined, but a small difference was seen in the VL-VH pairing of the free and bound forms of FvD1.3 [14]. The shift improves the contacts between the VH domain and the antigen, but is vanishingly small. Without the free Fab, the possibility that the observed change arises from loss of the anchoring effect of CH1 cannot be ruled out. Larger shifts in VL-VH pairing have been reported for the HIV anti- peptide antibody, Fab 50.1 (RL Stanfield et al., unpub- lished results).

Large movements of complementarity determining regis (CDRs) on complex formation were first seen in an anti-peptide antibody (17/9; central panel Table 2) which showed significant changes between the free and bound forms [22"]. This change was defined as a twisting of the two strands about the long axis of the H3 loop, giving rise to residue movements of around 2-5A. Similar CDR changes have recently been ob- served in the anti-peptide antibody Fab 50.1 ([23], RL Stanfield et al., unpublished results) and an anti-DNA antibody [28]. For the anti-peptide antibodies, the free conformation of the peptide is unknown but the as- sumption must be that it is disordered. In this situation, the notion of induced fit is entirely appropriate, since large changes in both antigen and antibody appear to be required to attain the complex state. The flexibility exhibited by peptide antigens, however, may impose thermodynamic (particularly entropic) constraints on the maximum affinity attainable.

Page 5: Antibody-antigen interactions

It has been suggested [29 °] that such flexible ligands may have an upper limit for their association constant of about 108M -1. Thus, for generating antibodies that are required to be cross-reactive with the native pro- tein, peptides may not be the ideal starting point.

Dissection of the antibody interaction and prospects for design

The first attempts to modify antibody affinity in a ra- tional and successful manner were carried out in 1987 using site-directed mutagenesis, in combination with molecular modelling [30]. Despite the arrival of phage display technology [31,32], most systematic studies of the combining site have continued to employ site-spe- cific methods.

'Alanine scanning' mutagenesis was used to identify residues important for antigen binding in the. anti- epidermal growth factor receptor antibody, hu-4D5-5 [3"']. The alanine mutations were chosen to line a shallow depression predicted by a homology model. Only four of the twenty-two mutations carried out re- sulted in a decrease of more than 10-fold in affinity, as would have been predicted by the 'energetic epi- tope ' model of Novotny [331, whereby the residues of a sub-region of the physical epi tope contribute the ma- jority of the binding energy. Titration calorimetry of the mutants led to the conclusion that antigen bind- ing was driven by the hydrophobic effect. Of partic- ular interest was the observation that the four critical residues did not exhibit enthalpy-entropy compen- sation during mutagenesis. This is in contrast to the observations of Ito et al. [5"] who, in a study of the D1.3 combining site using a pseudo-rational library (where each residue position is varied using ratio- nally designed subsets of all available amino acids) in which nine putative antigen contact residues were targeted, observed entropy-enthalpy compensat ion in all instances. On the basis of this study these au- thors conclude, 'It should be very difficult to design mutant antibodies with enhanced affinity based on modelling of antibody-antigen interactions'. Clearly, entropy--enthalpy compensat ion is not a universal phe- nomenon in antibody recognition and generalisations of this type are therefore not always supportable. Mu- tation of the high affinity (>109 M -1) anti-digoxin anti- body, 26-10, has also been carried out using a homol- ogy model [34]. The mutants were screened with a panel of digoxin analogues which showed few large differences in specificity or selectivity. This suggests that the affinity was due to a combination of many weak interactions, though it is also possible that the residues with strong interactions were not mutated. Thus, the concept of the energetic epitope, in which only a small number of the interfacial residues con- tribute to the binding constant, may not be universal [33]. Where good structures or models are known, rational mutations may be more easily designed [29,36"]. A

Antibody-antigen interactions Webster et al. 127

recent study in which nuclear Overhauser effect data were used to identify residues close to the antigen in an anti-2-phenyloxazol-5-one antibody [36"] supports this notion. Candidate residues were randomly mutated and screened by phage display, allowing identification of a mutant exhibiting a ten-fold increase in binding affinity.

Conclusions

During the past year, evidence has emerged to suggest that general combining site shape may be dictated by particular CDR length combinations. In addition to the three shape classes discussed previously, we have sug- gested the existence of a fourth, the 'H3 finger' class, characterized by long (>17 residue) H3 loops [29"]. Such loops are frequently seen in anti-viral antibodies, which may bind by a novel mechanism involving an H3 'finger-canyon' (the H3 finger-viral canyon) interaction [38",39].

In attempting to design an antibody combining site a b

initio, t w o premises are implicit. First, that the rules governing the overall coarse topography are known, and second, that fine specificity can be predicted by selection of the most appropriate antigen contact residues. The problem of predicting which antibody residues should form the partners in antibody-antigen pairwise interactions is, at present, a more distant prospect. Furthermore, where mutual, or induced fit processes are at work, the problem will become even more severe.

It is to be hoped that the rapidity with which X-ray structures can now be produced, and the power of phage technology in concert with random mutagene- sis, will allow the rules for design to emerge slowly and broaden yet more the available antibody repertoire.

Acknowledgements

We would like to thank SERC, British Biotechnology Ltd, Wellcome Research Ltd and Oxford Molecular Ltd for financial support. All Henry acknowledges SERC for a studentship.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as: • of special interest • . of outstanding interest

1. MILSTE1N C: The Croonian Lecture: Antibodies- a Paradigm for the Biology of Molecular Recognition. Proc Roy Soc Lond B 1990, 239:1-16.

2. DAVIES DR, CHACKO S: Antibody Structure. Acc Chem Res 1993, 26:421-427.

Page 6: Antibody-antigen interactions

128 Folding and binding

3. KELLY RE, O'CONNELL MP: Thermodynamic Analysis of an An- • • tibody Functional Epitope. Biochemistry 1993, 32:6828-6835. The first comprehensive thermodynamic analysis of antibody bind- ing. In this study, four out of twenty-two antibody residues mutated accounted for the majority of the binding energy, which was shown to bepredominantly entropy driven. This led to the suggestion that the binding was dominated by the hydrophobic effect.

4. TELLO D, GOLDBAUM FA, MARIUZZA RA, YSERN X, SCHWARZ • . FP, POLJAK RJ: Three-Dimensional Structure and Thermo-

dynamics of Antigen Binding by Anti-Lysozyme Antibodies. Biochem Soc Trans, 1994, in press.

A study of the thermodynamics of antibody-lysozyme binding with various antibodies. In most antilxxlies the binding was enthalpicaUy driven. The surprising result was that, for most antibodies, the AS of binding was negative, thus calling into quest ion the generality of the hypothesis that protein-protein interactions are driven by the hydrophobic effect.

5. ITO W, IBA Y, KUROSAWA Y: Effects of Substitutions of • Closely Related Amino Acids at the Contact Surface in

an Antibody-Antigen Complex on Thermodynamic Param- eters. J Btol Chem 1993, 268:16639-16647.

A thermodynamic analysis of antigen binding to a series of antibody mutants. In all instances the mutations showed entropy-enthalpy compensat ion leading to the suggest ion that improvement of affinity by design may be difficult.

6. AREVALO JH, TAUSSIG MJ, WILSON IA: Molecular Basis of • • Cross-Reactivity and the Limits of Antibody-Antigen Com-

plementarity. Nature 1993, 365:859-863. Thought-provoking article describing antibody cross-reactivity with different steroid haptens. The paper gives a structural insight into how cross-reactive steroids use different regions of the same anti- body combining site, thus exploiting to the full the available topog- raphy.

7. EDMUNDSON AB, HARRIS DL, FAN Z-C, GUDDAT LW, SCHLEY • BT, HANSON BL, TRIBBICK G, GEUSEN HM: Priciples and Pit-

falls in Designing Site-Directed Peptide Ligands. P r o t e i n s - Struct Funct Genet 1993, 16:246-267.

Crystallographic study of the interaction of peptide ligands of differ- ent sequence and length with the antibody combining site.

8. CHITARRA V, ALZARI PM, BENTLEY GA, BHKI" TN, EISEI.E J- • L, HOUDUSSE A, LESCAR J, SOUCHON H, POLJAK RJ: Three-

Dimensional Structure of a Heteroclitic Antibody-Antigen Cross-Reaction Complex. Proc Natl A cad Sci USA 1993, 90:7711-7715.

An interesting study of the cross-reactivity of an antibody with a num- ber of avian lysozymes. For one antibody, certain lysozymes exhib- ited binding affinities greater than that of the original immunogen. The possible role of water molecules in cross reactivity is discussed.

9. WILSON IA, STANFIELD RE: Antibody-Antigen Interactions. • Curr Opin Struct Biol 1993, 3:113-118. Recent review on antilx~dy-antigen interactions, focusing mainly on crystallographic results. A proposal was made, and discussed, that conformational change on binding may be a general mechanism.

10. TULIP WR, VARGHESE JN, LAVER WG, WEBSTER RG, COLMAN PM: Refined Crystal Structure of the Influenza Virus N9 Neuraminidase-NC41 Fab Complex. J Mol Biol 1992, 227:122-148.

11. WALLS PH, STERNBERG MJE: New Algorithm to Model Protein-Protein Recognition Based on Surface Complemen- tarity. J Mol Biol 1992, 228:277-297.

12. NOVOTNY J, SHARP K: Electrostatic Fields in Antibodies and Antibody/Antigen Complexes. Prog Biophys Molec Biol 1992, 58:203-224.

13. MAmUZZA RA, POLJAK RJ: The Basics and Mechanisms of Antigen Recognition and Mimicry by Antibodies. Curr Opin Immuno l 1993, 5:50-55.

14. BHAT TN, BENTLEY GA, FISCHMANN TO, BOULOT G, POLJAK RJ: Small Rearrangements in Structures of Fv and Fab Frag- ments of Antibody D1.3 on Antigen Binding. Nature 1990, 347:483-485.

15. FISCHMANN TO, BENTLEY GA, BHAT TN, BOULOT G, MARIUZZA RA, PHILLIPS SEV, TELLO D, POLJAK RJ: Crystallographic Re- f inement of the Three-dimensional Structure of the FabD1.3- Lysozyme Complex at 2.5A Resolution. J Btol Chern 1991, 266:12915-12920.

17. SHOHAM M: Crystal Structure of an Anticholera Toxin Pep- tide Complex at 2.3A. J Mol Biol 1993, 232:1169-1175.

18. AREVALO JH, STURA EA, TAUSSIG i J , WILSON IA: Three-Di- ** mensional Structure of an Anti-Steroid Fab'and Progester-

one-Fab ' Complex. J Mol Biol 1993, 231:103-118. The detailed structural version of [6"*], where the manne r in which different steroids bind to an antibody revealed a novel mechanism for cross-reactivity.

19. GROVES DJ, SAUER MJ, PAYMENT P, FOULKES JA, MORRIS BA: The Preparation of an Ovine Monoclonal Antibody to Pro- gesterone. J Endocrinol 1990, 126:217-222.

20. STANFIELD RL, FIESER TM, LERNER RA, WILSON IA: Crystal Structures of an Antibody to a Peptide and its Complex with Peptide Antigen at 2.8A Resolution. Science 1990, 248:712-719.

21. CHEETHAM JC, RALEIGH DP, GRIEST RE, RE~DF1ELD C, DOBSON CM, REES AR: Antigen Mobility in the Combining Site of an Anti-Peptide Antibody. Proc Natl Acad Sci USA 1991, 88:7968-7972.

22. R1NI JM, SCHULZE-GAHMEN U, WILSON IA: Structural Evidence for Induced Fit as a Mechanism for Antibody-Antigen Recog- nition. Science 1992, 255:959-965.

Description of large structural change in the conformation of CDR H3 on binding a pept ide from influenza virus hemagglutinin. This study draws attention to the possibility of induced fit as a general mechan i sm in antigen- antibody interactions.

23. RINJ JM, STANFIELD RE, S'IZIRA EA, SALINAS PA, PROFY AT, • WILSON IA: Crystal Structure of a Human Immunodefic iency

Virus Type 1 Neutralizing Antibody, 50.1, in Complex with Its V3 Loop Peptide Antigen. Proc Natl Acad Set USA 1993, 90:6325-6329.

This paper describes a crystal structure of an Fab-pept ide com- plex at 2.8A resolution. The peptide, which derived from HIV1, adopted an extended conformation not seen in other anti-peptide complexes. Movements at the VH-VL interface on antigen binding were proposed al though the structure of the free Fab has not been determined.

24. DYSON HJ, RANCE M, HOUGHTEN RA, WRIGHT PE, LERNER RA: Folding of Immunogenic Peptide Fragments of Proteins in Water Solution II. The Nascent Helix. J Mol Biol 1988, 201:201-217.

25. LAVER WG, MR GM, WEBSTER RG, SMITH-GILL SJ: Epi- topes on Protein Antigens: Misconceptions and Realities. Cell, 61:553-556.

26. VAN I{EGENMORTEL MHV: Molecular Dissection of Protein Antigens. In Structure o f Antigens Vol 1. Ed. Van Regen- mortel MHV. CRC Press; Boca Patton: 1992: 1-27.

27. ZEDER-LUTZ G, ALTSCHtJH D, GEYSEN HM, TRIFILIEFF E, • SOMMERMEYER G, VAN REGENMORTEL MHV: Monoclonal

Antipeptide Antibodies: Affinity and Kinetic Rate Constants Measured for the Peptide and the Cognate Protein using a Biosensor Technology. Mol Immuno l 1993, 30:145-155.

Detailed study of the kinetics and affinities of antibodies binding to peptides and cross-reacting proteins using the BIAcore method (Pharmacia). The authors conclude that anti-peptide antibodies bind to the native protein with 50-100 times lower affinities than to the peptide, and that this reduction is primarily due to a change in the association constant.

28. HERRON JN, HE XM, BALLARD DW, BLtER PR, PACE PE, BO'II-IWELL ALM, VOSS EW, EDMUNDSON AB: An Auto- Antibody to Single Stranded DNA: Comparison of the Three-Dimensional Structures of the Unliganded Fab and a Deoxynucleotide-Fab Complex. Proteins Struct Func Genet 1989, 5:271-280.

Page 7: Antibody-antigen interactions

29. SEARLE JS, PEDERSEN JT, HENRY AH, WEBSTER DM, REES AR: • Antibody Structure and Function. In Antibody Engineering

Manual. Edited by Borrebaeck K. Oxford University Press 1993, in press.

A comprehensive description of antibody variable region .structure, antibody modelling, in which preferred conformations of CDR H3 are described for the first time, and antibody engineering principles.

30. ROBERTS S, CHEETHAM JC, REES AR: Generation of an Anti- body with Enhanced Affinity and Specificity for its Antigen by Protein Engineering. Nature 1987, 328:731-734.

31. MCCAFFERTY J, GRIFFITHS AD, WINTER G, CHISWELL DJ: Phage Antibodies: Filamentous Phage Displaying Antibody Variable Domains. Nature 1990, 348:552-554.

32. KANG AS, BARBAS CF, JANDA KD, BENKOVIC SJ, LERNER RA: Linkage of Recognition and Replication Functions by As- sembling Combinatorial Antibody Fab Libraries Along Phage Surfaces. Proc Natl Acad Sci USA 1991, 88:4363-4366.

33. NOVOTNY J, BRUCCOLERI RE, SAUL FA: On the Attribution of Binding Energy in Antibody-Antigen Complexes McPC603, D1.3 and HyHEL-5. Biochemistry 1989, 28:4735--4749.

34. NEAR RI, MUDGETF-HUNTER M, NOVOTNY J, BRUCCOLERI RE, NG SC: Characterization of an Anti-Digoxin Antibody Binding Site by Site-Directed in Vitro Mutagenesis. Molec Immunol 1993, 30:369-377.

35. REES AR, MARTIN ACR, PEDERSEN JT, SEARLE SMJ: AbM TM, a • • Computer Program for Modelling Variable Regions of Anti-

bodies. 1992, Oxford Molecular Ltd, Oxford, UK. The first automated program for generating models of the complete variable regions of antibodies from sequence data alone. Framework accuracy is about 0.4A RMSD while CDRs have accuracies within the range 1.0-2.5,~, RMSD (after global fitting).

36. REICHMANN L, WEILL M: Phage Display and Selection of a Site- . Directed Randomized Singie-Chain Antibody Fv Fragment for

Its Affinity Improvement. Biochemistry 1993, 32:884843855. A study of antibody affinity in which an X-ray structure of the anti- body and NOE data on the hapten binding were used to guide the construction of a phage library. This resulted in selection of a mutant with a 10-fold higher affinity than the wild type.

37. HE XM, RUKER F, CASALE E, CARTER DC: Structure of a Hu- • man Monoclonal Antibody Fab Fragment Against gp41 of

Human Immunodeficiency Virus Type 1. Proc Natl Acad Sci USA 1992, 89:7154-7158.

The structure of an anti-peptide Fab containing the longest CDR H3 (17 residues) for which an X-ray structure is available. The H3 is con-

Antibody-antigen interactions Webster et al. 129

formationaUy flexible and may undergo large changes on binding the peptide antigen.

38. KABAT EA, WU "Ir, PERRY HM, GOTrESMAN KS, FOELLER C: Sequences of Proteins of Immunological Interest, 1991, US Department of Health and Human Services, NIH, Bethesda, MD USA.

39. MAINHART CR, POTFER M, FELDMAN RM: A Refined Model for the Variable Domains (Fv) of the J539 ~(1,6) D-galactan Binding Immunoglobulin. Mol Immunol 1984, 21:469-478.

40. JEFFREY PD, STRONG RK, SIEKER LC, CHANG CY, CAMPBELL RL, PETSKO GA, HABER E, MARGOLIES MN, SHERIFF S: 26-10 Fab: Digoxin Complex - - Affinity and Specificity Due to Surface Complementarity. Proc Natl Acad Set USA 1993, 90: 10310°10314.

41. SAUL FA, POLJAK RJ: Crystal Structure of Human Im- munoglobulin Fragment Fab New Refined at 2.02k Resolu- tion. Proteins: Stmtct Func Genet 1992, 14:363-371.

42. ROSE DR, PRZYBYLSKA M, TO RJ, KAYDEN CS, OOMEN RP, VORBERG E, YOUNG NM, BUNDLE DR: Crystal Structure to 2.45,~ Resolution of a Monoclonal Fab Specific for Bru- celia A Cell Wall Polysaccharide Moiety. Protein Sci 1993, 2:1106-1113.

43. BRUNGER AT, LEAHY DJ, HYNES TR, FOX RO: 2.9,~ Reso- lution Structure of an Anti-Dinitrophenyl-Spin-Label Mono- clonal Antibody Fab Fragment with Bound Hapten. J Mol Biol 1991, 221:257-270.

44. TROMO J, STADLER E, SKERN T, AUER H, KANZLER O, BE'I'ZEL C, BLASS D, FITA I: 3-dimensional Structure of the Fab Fragment of a Neutralizing Antibody to Human Rhinovirus Serotype-2. Protein Set 1992, 1:1154-1161.

45. FAN Z-C, SHAN L, GUDDAT LW, HE X-M, GRAY WR, RAISON l{L, EDMUNDSON AB: 3-Dimensional Structure of an Fv from a Human-IgM Immunoglobulin. J Mol Biol 1992, 228:188-207.

46. DAVIES DR, PADLAN EA: Antibody-Antigen Complexes. Ann Rev Biochem 1990, 59:439--473.

47. BENTLEY GA, BOULOT G, RIOTFOT MM, POLJAK RJ: 3-dimen- sional Structure of an Idiotope-Anti-Idiotope Complex. Na- ture 1990, 348:254-257.

DM Webster, AH Henry and AR Rees, School of Biology and Bio- chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.