selecting and screening of ant antibody libraries

Upload: soujig

Post on 07-Apr-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    1/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1105

    Selecting and screening recombinantantibody librariesHennie R Hoogenboom

    During the past decade several display methods and other library screening techniques have been developed for isolating

    monoclonal antibodies (mAbs) from large collections of recombinant antibody fragments. These technologies are now widely

    exploited to build human antibodies with high affinity and specificity. Clever antibody library designs and selection concepts are

    now able to identify mAb leads with virtually any specificity. Innovative strategies enable directed evolution of binding sites with

    ultra-high affinity, high stability and increased potency, sometimes to a level that cannot be achieved by immunization. Automationof the technology is making it possible to identify hundreds of different antibody leads to a single therapeutic target. With the first

    antibody of this new generation, adalimumab (Humira, a human IgG1 specific for human tumor necrosis factor (TNF)), already

    approved for therapy and with many more in clinical trials, these recombinant antibody technologies will provide a solid basis for

    the discovery of antibody-based biopharmaceuticals, diagnostics and research reagents for decades to come.

    In humans, the immune system is capable of creating thousands of

    millions of different antibodies from which suitable antigen-binding

    antibodies are rapidly selected. Envious of this unsurpassed powerful

    system for making binding sites, scientists have been investigating for

    decades methods to recreate systems to build immunoglobulin-based

    binding sites using recombinant approaches (reviewed by Winter and

    Milstein1). One of the first breakthroughs came in 1989 with an inno-

    vative technology that enabled the cloning of antibody genes2, thereby

    bypassing hybridomasa hybrid cell produced by the fusion of an

    antibody-producing lymphocyte with a tumor cell, which was the tra-

    ditional means of manufacturing mAbs. In the new method, antibody

    genes were cloned directly from lymphocytes of immunized animals and

    expressed as a single-domain library3 of antibody heavy- or light-chain

    variable regions or as a combinatorial library of antigen-binding frag-

    ment (Fab) fragments in bacteria4. To screen combinatorial librar ies, a

    slow and cumbersome colony-lifting and filter-based screening method

    with radio-labeled antigen was then used to identify the few antigen-

    reactive antibodies in libraries from millions of clones.

    Within a year, a method based on the expression of functional anti-

    body fragments on the surface of filamentous phage was described,

    which provided a way to quickly select antibodies from libraries on thebasis of the ant igen-binding behavior of individual clones5. A few years

    later this technique, called phage display, in combination with PCR-

    based cloning of antibody repertoires2,4, was successfully used to isolate

    murine6 and human7,8 antibodies from recombinant antibody libraries

    built from natural sources, such as from animal or human B lympho-

    cytes, and eventually libraries were created entirely by in vitro cloning

    techniques (reviewed in ref. 9).

    Fifteen years later, phage, and more recently, ribosome- and yeast-

    display technologies (described below) have turned into mainstream

    antibody and protein engineering platforms. Display technology has

    also become one of the three major technologies for creating mAbs for

    human therapy, in addition to the use of immunized t ransgenic mice

    and the humanization of mAbs. This review covers the most important,

    currently used selection platforms for recombinant antibody libraries,

    the methods for selecting and screening different types of libraries, sev-

    eral antibody affinity and stability optimization strategies and finally, the

    impact of library-based approaches on antibody humanization, with a

    focus on the developments (and citations) of the past few years.

    Selection platforms for antibody libraries

    The antigen-binding site of an antibody is composed of six comple-

    mentarity determining regions (CDRs) or hypervariable regionsthree

    within the light-chain variable domain (VL) and three within the heavy-

    chain variable domain (VH). In the immune system, a large collection of

    different antibody binding sites is created by the combinatorial assembly

    of germline-encoded segments (Fig. 1). This produces a repertoire of

    naive B-cell lymphocytes, each expressing a unique antibody binding site

    on their surface. Exposure to antigen selects from this repertoire thoselymphocytes that produce antigen-reactive antibodies, and tr iggers the

    incorporation of somatic mutations in the V genes, allowing subsequent

    selection of mutations that improve the affinity of the antibody for the

    antigen.

    Antibodies can also be isolated from recombinant antibody libraries

    in the laboratory, using one of the platforms for selection that in essence

    mimics this in vivo process. Many of these selection platforms share

    four key steps with the procedure for antibody generation in the in vivo

    immune system: first, the generation (or cloning) of genotypic diversity;

    second, the coupling of genotype to phenotype; third, the application of

    selective pressure; and four th, amplification (Fig. 2a). This process first

    leads to a diverse collection of recombinant antibody genes, such as those

    Ablynx NV, Technologiepark 4, 9052 Ghent, Belgium. Correspondence should be

    addressed to H.R.H. ([email protected]).

    Published online 7 September 2005; doi:10.1038/nbt1126

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    2/12

    1106 VOLUME 23 NUMBER 9 SEPTEMBER 2005 NATURE BIOTECHNO LO GY

    from the B lymphocytes of immunized animals. This collection of genes

    is then cloned to provide a physical link between each antibodys phe-

    notype (antigen-binding behavior) and the encoding genotype.Rather

    than screening the clones directly for antigen binding, antibody libraries

    are enriched by rounds of selection with target antigens and amplifica-

    tion and after a few rounds, individual clones are screened for antigen

    reactivity (Fig. 2a). Box 1 and Figure 2b describe some of the latest

    and most frequently used selection platforms for antibody isolation and

    engineering; in Table 1 the three main platforms are compared.

    Phage display. Antibody display on the surface of two types of

    bacteriophage, fd and M13, is currently the most widespread method for

    the display and selection of large collections of antibodies and for the

    engineering of selected antibodies (reviewed in ref. 10). It owes its favor-

    able status to being robust, simple to use and highly versatile; the selec-

    tion process can be adapted to many specific conditions that foil most

    other display platforms, including selections on whole cells and t issues

    and even in animals. The most successful applications of phage antibody

    display include the following: first, the de novo isolation of high-affinity

    human antibodies from nonimmune and synthetic libraries1115, includ-

    ing antibodies against self antigens; second, the generation of picomolar

    affinity antibodies by in vitro affinity maturation1618; and third, the

    discovery of antibodies with unique properties from nonimmune19,20

    and immune libraries from animal or human donors21,22.

    Ribosome and mRNA display. The most developed forms ofin vitrodis-

    play rely on the stable formation of a complex of antibody fragment and

    its encoding mRNA (for a review, see ref. 23). The mRNAs from selected

    complexes are then amplified. The most successful applications of ribo-

    some display are in the field of affinity maturation of antibodies2426.

    The built-in affinity maturation feature of this display system, caused

    by the error-prone process of reverse transcriptase and amplification,contributes to the efficient maturation of picomolar concentrations of

    antibodies27,28.

    Microbial cell display. Surface display on the yeast Saccharomyces cerevi-

    siae29provides the possibility to select repertoires of cells by flow cytom-

    etry. In combination with random mutagenesis methods to diversify the

    VH and VL genes (see below), the yeast-display method has yielded the

    highest affinity (48 fM) for any antibody30. The use of many other micro-

    bial display formats that have been used successfully with pept ides or

    enzymes, such as surface display on the bacterial cell walls ofEscherichia

    coli, Staphylococcus aureusand Zymomonas mobilisor on spores of cer-

    tainBacillusstrains, has been limited by problems with the ant ibodys

    * *

    *

    Antibody variable regiongermline segments

    DNArearrangement

    Selection forantigen binding

    Somatichypermutation

    and affinityselection

    Library of naivelymphocytes

    Ag-selectedlymphocyte(s)

    Maturedlymphocyte(s)

    Naive/nonimmunebinding site library

    Immunebinding site library

    Unique Ag-bindingsite surface

    Improved Ag-bindingsite surface

    L2 H3

    L3

    H1

    L1 H2

    Clonal level

    Library levelL2 H3

    L3

    H1

    L1 H2

    L2 H3

    L3

    H1

    L1 H2

    Low affinity

    L2 H3

    L3

    H1

    L1 H2

    High affinity

    +

    VH

    VL

    D J

    V J

    H1 H2 H3

    L1 L2 L3

    VH

    VL

    H1 H2 H3

    L1 L2 L3

    VH

    VL

    H1 H2 H3

    L1 L2 L3

    + +

    V

    Figure 1 Generating binding site diversity in the immune system. In the immune system, a large collection of different antibody-binding sites is created by

    the combinatorial assembly of germline-encoded segments (V, D and J for the heavy-chain variable region VH, V and J for the light-chain variable region VL).

    This DNA reshuffling process targets most diversity in the primary repertoire to the heart of the binding site, the two CDR3 regions of heavy and light chains,

    respectively, creating a large chemical diversity that is of primary importance for the potential recognition of many different types of antigenic structures.

    This is depicted in a schematic representation of a collection of antibody-binding-site surfaces (in green), for simplicity indicated with H3 and L3. The other

    CDR/hypervariable regions (indicated with H1, H2, L1 and L2) are encoded in the human germ line by ~100 different functional V-gene segments; from

    a structural perspective, they are located at the periphery of the binding site surface, surrounding the H3 and L3 regions (lower level of diversity is shown

    in light green). Exposure to antigen (red) selects from this naive repertoire those lymphocytes that produce antigen-reactive antibodies and triggers the

    incorporation of somatic mutations in the V genes (indicated by the green lines at the clonal level) and subsequent selection of mutations that improve the

    affinity of the antibody for the antigen (green stars and circles at clonal and library level, respectively). Recombinant antibody libraries are made by rescue of

    genes from these various V-gene pools; note that nonimmune libraries, depending on their exact construction, may contain a mix of V genes from both naive

    and other B-cell sources (indicated with the dotted arrows). Ag, antigen.

    KatieRis

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    3/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1107

    heterologous expression, secretion and fold-

    ing, with proteolysis and antigen-antibody

    accessibility. Therefore, many of these display

    and screening systems, although elegant in

    nature31,32, are not widely used today for anti-

    bodies. However, a recently described approachbypasses most of these problems: it is based

    on anchoring the antibody fragment on the

    periplasmic face of the inner membrane ofE.

    coli followed by disruption of the outer mem-

    brane, incubation with fluorescently labeled

    antigen and sorting of the protoplasts. This

    very promising and versatile display method

    is directly compat ible with (filamentous) phage display, combines the

    ease ofE. coli-based library constructions with the power of cell sorting,

    and therefore, is likely to become widely used.

    Other selection platforms. Directed evolution platforms recently devel-

    oped for antibody fragments include retroviral display34, display based

    on protein-DNA linkage35,36, microbead display by in vitro compart-

    mentalization37, in vivo-based growth selection based on the protein

    fragment complementation assay (PCA)38 or other systems39 and even

    single-molecule sorting40. Although each of these methods will have

    specific theoretical advantages, to date, their validation with antibody

    fragment libraries has been limited, and their advantages over more

    established systems (e.g. regarding the truly monovalent nature of the

    method, eukaryotic expression advantages, increase in library size or

    selection efficiency) remain to be demonstrated. (For a more in-depth

    discussion of library-display technologies, including PCA and two-

    hybrid systems, that are available but have not yet been used in combi-

    nation with ant ibody fragments, see ref. 41.)

    To establish a platform to select recombinant antibody libraries in

    the IgG format, the preferred format for many applications, researchersrecently displayed small libraries of IgGs on the surface of mammalian

    cells. After homologous integration of a single-gene copy in each cell,

    the population was sorted by flow cytometry to obtain a clone with

    sevenfold affinity improvement (W.D. Shen, Amgen, personal commu-

    nication). In the future, bigger combinatorial IgG formatbased libraries

    may be built using vaccinia virusbased vectors42, or diversity may be

    introduced in vivo by using B-cell lines that hypermutate a carr ier anti-

    body gene constitutively43 or upon induction44 or that harbor induc-

    ible hypermutable enzymes involved in this process in nature45. Some

    of these newer selection and diversification methods may open novel

    applications for the directed evolution of antibodies and other proteins

    (see also accompanying review on p. 11261136).

    Strategies to select and screen antibody libraries

    Individual clones of a recombinant single-chain Fv (scFv) or Fab library

    theoretically can be directly screened for antigen binding, for example,

    using binding assays based on ELISA or filter-based screening. Screening

    is limited by the number of clones that can be examined, hence in many

    applications the frequency of antigen-reactive clones is too low, and

    the libraries too large (with tens of millions to billions of clones) to

    do this efficiently. The connection between genotype and phenotype

    in phage- or ribosome-display libraries provides a means to select for

    clones binding to a desirable antigen, thereby increasing the frequency of

    antigen-reactive clones, enriching the clones with best binding affinity,

    or the clones with certain predefined binding characteristics. Typically

    many more clones can therefore be sampled compared with screen-

    ing procedures. Many different selection methods and experimental

    approaches have been developed that separate clones that bind from

    those that do not (Fig. 3).

    Selection procedures. For phage-display libraries, selection involves

    exposure to ant igen to allow antigen-specific phage antibodies to bind

    their targets during biopanning. This is followed by recovery of antigen-bound phage and subsequent infection in bacteria. Although ideally,

    only one round of selection would be required, nonspecific binding

    limits the enrichment that can be achieved per selection round and

    therefore, in most cases, recursive rounds of selection and amplification

    are needed to select the best binders from the library (Fig. 2a).

    Phage displaybased selections are now a relatively standard procedure

    in many molecular biology laboratories (a more detailed description of

    these procedures is provided elsewhere10,46 and references therein). For

    more complex selections such as those using cells or tissues, it can be

    instructive to use enrichment studies with control phage antibodies to

    optimize the efficiency of the selection method and to compare different

    selection approaches, and then tune the selection strategy accordingly to

    Phage display

    Protein-mRNAlink via:

    Protein-DNAdisplay

    Growthselection via:

    Display on:

    Microbeadvia in vitro

    compartmentalization

    Coupling of genoto phenotype

    Selective pressureon phenotype

    Screening Amplification

    +

    Antibody gene pool

    Displayed library

    Selected antibody lead

    Synthetic DNA

    Cloning ofgenetic diversity

    B-cells

    Selectioncycle

    Mutagenesisand

    selectioncycle

    -ribosome display

    -mRNA display

    -Yeast-Bacteria-Mammalian cells-Retroviruses-.....

    -Yeast 2-hybrid-Protein fragmentcomplementation

    a bSteps in antibody selection Selection platformsFigure 2 Creating and selecting recombinantantibody libraries. (a) First, antibody diversity

    is generated from synthetic V genes or cloned

    from B cells. Next, antibody phenotype (boxes

    in green, blue and orange) is coupled to its

    genotype (wavy line) via a phenotype-genotype

    link (green) packaged in a host (purple) (shown

    here schematically for phage display). As a

    result, each host particle expresses (or displays)

    a unique antibody on i ts surface. The repertoire

    of antibodies displayed on these host particles

    is subjected to The process is repeated and

    eventually antibodies binding to antigen are

    confirmed by screening. (b) Different selection

    platforms for conventional antibodies. Color code

    as for a (see text for details and citations).

    KatieRis

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    4/12

    1108 VOLUME 23 NUMBER 9 SEPTEMBER 2005 NATURE BIOTECHNO LO GY

    maximally sample the repertoire47. Fewer selection methods have been

    tested on nonphage-display systems, but both ribosome and mRNA-dis-

    play work part icularly well with purified antigen and yeast celldisplay

    libraries are preferentially selected by cell sorting.

    Selection antibody libraries for target binding. A variety of protocols

    have been described for selecting antibodies from phage- and r ibosome-

    display libraries with improved binding affinity or kinetics toward the

    target antigen. For example, by using limited and decreasing amounts of

    antigen, the selection favors clones with lower Kd; by using long washing

    steps after the incubation of target antigen, library clones with improved

    off-rate are selected; and by using very short incubation times prefer-

    entially, clones with improved on-rate are selected. In yeast display, the

    optimization of ant igen concentration and time for dissociation of anti-

    body-antigen complexes enables cell sorting with flow cytometry on the

    basis of small differences in the kinetic parameters of antigen-an tibody

    interactions, at least for monomeric antigens48. Antibodies may also be

    selected with or for a particular functional activity, such as for recep-

    tor cross-linking49, signaling or gene transfer (reviewed in refs. 10 and

    50). Special selection procedures have been developed to identify phage

    antibodies that catalyze certain chemical reactions51 or that bind to and

    are internalized by cells52; the latter property is useful for targeting ofimmunoliposomes and other payloads.

    Screening antibody libraries for target binding. The outcome of any selec-

    tion procedure is a mixture of antibody clones with different target-binding

    properties that then need to be individually screened. Antigen-binding

    of poly- or monoclonal phage antibodies is tested using typical anti-

    body-binding assays, ranging from ELISAs to immunoprecipitation.

    The best screening assays are fast, robust, amenable to automation (e.g.,

    in 96- or 384-well format) and use the display host (phage or yeast cells)

    or the soluble antibody fragment equipped with tags for detection and

    purification. The diversity of the clones present in the selected antibody

    library (which is tracked by restriction enzymebased fingerprinting or

    by high-throughput DNA sequencing of selected clones) can be used as

    a guideline to define at what stage to screen the library. When finding

    drug candidates among the selected antibody leads, it may be advanta-

    geous to screen for biological function using biochemical or cell-based

    bioassays because binding affinity and potency are not always corre-

    latedfor example, this applies when identifying antibodies that neu-

    tralize an interaction, agonize or antagonize receptor binding. Screening

    (or selection) procedures can also be tailored to identify antibody leads

    that cross-react with the murine antigen or bind to different isoforms

    of the antigen. In ribosome/mRNA display, selected populations are

    first cloned and individual antibodies are expressed either by the host

    cell in vivo or by translation in vitro. The preferred screening method in

    yeast display is flow cytometry, which under the right conditions yields

    informat ion on affinity, expression level and epitope binding53.

    Automating the process. Although automation seems particularly

    straightforward for in vitro display-based approaches (e.g., ribosome

    display and microbead display by in vitro compartmentalization37,

    Several different molecular selection strategies for isolating and

    engineering human antibodies are currently in use. Described here

    are the three best established platforms.

    Phage display. To express an antibody fragment on the surface

    of the phage particle, its encoding gene is fused in-frame to oneof the phage coat proteins and cloned in a vector that can be

    packaged as a phage particle. Different display systems can lead to

    monovalent (single copy) or to multivalent (multiple copy) display

    of an antibody, depending on the type of anchor protein and display

    vector used. The most popular system is to use monovalent display,

    which is convenient for selecting antibodies of higher affinity,

    achieved by using a direct fusion or a disulfide-bridged link to

    a minor coat protein, pIII, and by using phagemids into which

    antibody libraries are easier to clone than phage vectors. Libraries

    with over 1010 clones can be made using recombination-based

    protocols11,159, but more frequently are made by conventional

    transformations with genetically stable phagemid vectors. Selection

    efficiency is improved by using mult ivalent di splay in t he first

    step(s), for example, using helper phage variants (reviewed inref. 46), inducible promoters160,161 or bivalent display162,163.

    Multivalent display is also used for selecting antibodies that

    mediate receptor-mediated endocytosis164 , panels of antibodies for

    target discovery165 and to rapidly select antibody-antigen pairs on

    the tip of the phage166 .

    Ribosome and mRNA display. In ribosome display167 , the link

    between antibody and encoding mRNA is made by the ri bosome,

    which at the end of translating this mRNA is made to stop

    without releasing the polypeptide168,169. The ternary complex

    as a whole is used for the selection. In mRNA display, there is

    a covalent bond between antibody and mRNA established via

    puromycin as an adaptor molecule170 . These display methods

    are carried out enti rely in vitro, t hereby eliminating the need

    for cell transformation. The other advantage is that it is very

    amenable to mutagenesis to provide additional diversity be-

    tween generati ons (e.g., by nonproofreading polymerases),

    without t he need to t ransform the cloned li brary into E. coli.

    Although there are many examples of anti body-ribosome display,

    the mRNA-display format has been used more exclusively for

    single-domain protein171 and has only recently been used for

    conventional antibodies23 .

    Yeast cell display. Antibodies are displayed on the yeast S.

    cerevisiaecell surface via fusion to the -agglutinin yeast

    adhesion receptor, which is located on the yeast cell wall. The

    display level on the cell is variable (on average about 3 10 4

    fusions per cell for a scFv), but the intrinsic avidity of this display

    system is counteracted by the power of cell sorting. By staining

    the cells with both f luorescently labeled antigen and anti-epitope

    tag reagent, the yeast cells can be sorted according to the level

    of antigen binding and antibody expression on the cell surface.Limi ting factors such as the transformation eff iciency of yeast

    and the cell sorting speed are currently being tackled. This

    approach has been used to build a nonimmune human scFv

    library172 with over 109 clones, which is efficiently selected

    with magnetic beads172174 and then sorted by flow cytometry

    to yield single-nanomolar affini ty antibodies172 . Combinations

    of phage with yeast-display platforms have been described174 ,

    and recent approaches include the use of the yeast cells mating

    system to create combinatorial diversity estimated to be 1014 in

    size175,176 and in another study the use of the yeast homologous

    recombination system for in vivogene diversification177 .

    Box 1 Selection platforms

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    5/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1109

    which in theory avoids complicated host cell and phage manipulations,

    in practice these methods rely on PCR which is highly sensitive to con-

    tamination and nonspecific amplification. Therefore to date most prog-ress has been made by adapting the more robust phage displaybased

    selection and screens to robotics54.

    Automation of the selection process is required when handling many

    antigens in parallel, for example, when generating thousands of anti-

    bodies for use in proteomic screens or for antibody arrays55. In these

    approaches, antigens are immobilized to a surface (Fig. 3ac) and all

    selection and amplification steps are carried ou t in microtiter plate for-

    mat5658. The materials used for such selections are most often produced

    by antigen-derived, surface-exposed linear peptides59,60 or recombinant

    approaches54,55,61. Devices have also been built to carry out semi-auto-

    mated cell selections using capillary flow chambers62. Automation of

    the screening process has been used to test thousands of different anti-

    body leads15,63.These high-throughput screening platforms consist of

    a combination of robotic colony-pickers and workstations, incubators

    for high-throughput expression, fluid-handling robots for perform-

    ing ELISAs, high-throughput cloning and high-throughput purifica-

    tion, detection devices, PCR-machines, sequencing apparatus and data

    handling systems and software to integrate the data from all steps64.

    Further efforts to streamline the screening procedures65,66, including

    miniaturization, in vitroexpression67, multiplexing and signal detection

    and data processing, will increase the throughput of these screening

    systems. Finally, high-density gridding of bacteria followed by protein

    array screening68 and filter-based colony screening69,70 have been used

    to bypass the selection step and directly screen antibody libraries.

    In the future, protein microarrays may also become particularly use-

    ful for high-throughput analysis of antibody specificity71 and affinity72.

    Arguably, the greatest bottleneck in screening today is analysis furtherdownstream, including kinetic analysis and in vitro/in vivo functional

    and bioactivity analysis. Functional and potency assays often require the

    recloning of antibody genes for expression in the IgG format, which is

    also amenable to high-throughput methods7375.

    Recombinant antibody library types

    MAb libraries can be based on immune fragments (that is, biased

    towards certain specificities present in immunized animals6 or natu-

    rally immunized, or infected, humans8) or naive fragments (not biased

    toward specificities found in the immune system). The latter type of

    fragment can be derived from n onimmune natural or semi-synthetic

    sources.

    Antibodies from immune antibody libraries. These libraries are con-

    structed with VH and VL gene pools that are cloned from source B-

    cells (from diverse lymphoid sources including peripheral blood, bonemarrow, spleen or tonsils) by PCR-based2,4 or similar76 cloning tech-

    nologies, cloned into an appropriate vector for expression as a random

    combinatorial antibody library, and subsequently selected for and/or

    screened. Compared with the yield using hybridoma technology, many

    more antibodies can be derived from a recombinant immune library

    made with the material of a single immunized donor, and in vitro selec-

    tion can enrich for rare antibody specificities. Further, human immune

    or disease-associated antibody libraries have identified antibodies with

    very interesting proper ties21,22,77 unlikely to be present in nonimmune

    or synthetic libraries. These libraries also facilitate the investigation of

    the humoral immune system at a molecular level78. If required, natural

    pairings of heavy and light chain can be maintained by in-cell PCR-link-

    ing of the V genes, or by parallel amplifying in high-throughput the VH

    and VLgenes of single antibodyproducing cells79. Librar ies made with

    this procedure may form a better reflection of the composition of the

    natural immune response compared with random combinatorial librar-

    ies with artificially paired chains in immune or nonimmune libraries,

    but it remains to be seen whether this translates into antibodies with

    higher affinity and more potency or with a lower immunogenicity when

    used in humans.

    Antibodies from nonimmune and semisynthetic libraries. This type

    of library is comprised of antibody fragments from a source of genes

    that is not explicitly biased to contain clones binding to antigen; as such

    they are useful for selecting antibodies against a wide variety of antigens

    (see below). Nonimmune (or naive) libraries are derived from natural,

    unimmunized, rearranged V genes (e.g., from the IgM B-cell pool) toreduce antigen-induced biases in the repertoire, and were the first librar-

    ies used to isolate anti-self antibodiesotherwise difficult to obtain by

    immunization.

    Synthetic antibody libraries are constructed entirely in vitro using

    oligonucleotides that introduce areas of complete or tailored degeneracy

    into the CDRs of one or more V genes. Synthetic diversity bypasses

    the natural biases and redundancies1 of antibody repertoires created

    in vivo and allows control over the genetic makeup of V genes and the

    introduction of diversity.The first reports of synthetic antibodies in

    1992 (refs. 80,81) were followed by many different design strategies,

    including mimicking the natural pattern of diversity in the immune

    system by randomizing CDR positions at the center of the binding site

    Table 1 Comparing the main selection platforms for antibodies

    Name

    Valency of

    display

    Typical max.

    library size

    Selection

    scope Main application Main strength Main weakness

    Suitable

    formats

    Phage Monovalent

    Multivalent

    10 10 to 1011 Versatile mAbs from natural

    libraries

    mAbs from synthetic

    libraries

    Affinit y maturation

    Stability increase

    Large mAb panels

    Technically robust

    Easy to use

    Automated

    Introduction of diversity

    by cloning is slow

    Large li braries diff icult

    to make

    Not t ruly monovalent

    scFv

    Fab

    Fab 2dAb

    Diabody

    Ribosome Monovalent 1012 to 1013 Limited Affinity maturation

    Stability increase

    Intrinsic mutagenesis

    Fastest of all systems

    Amenable to

    automation

    Small mAb panels

    Limited selection scope

    Technicall y sensiti ve

    scFv

    dAb

    Yeast cell Multivalent 107 Sorting Affinity maturation

    Expression increase

    Stability increase

    Fast in combination

    with random

    mutagenesis

    Direct screening for

    kinetics with cells

    Sorting expertise and

    equipment needed

    Transformation

    efficiency

    scFv

    Fab

    dAb

    dAb, domain antibody

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    6/12

    1110 VOLUME 23 NUMBER 9 SEPTEMBER 2005 NATURE BIOTECHNO LO GY

    and using a set of V-gene germline segments to provide a low level of

    diversity in peripheral regions11. Alternatively, CDR positions known to

    be involved in antigen binding can be identified on structural grounds

    and randomized in the context of an antibody with known structure82,

    or CDR positions chosen for randomization based on the relatively high

    mutation frequency of these positions in natural antibody repertoires,

    and then randomized in a set of frequently used V germline segments83.

    In another design consensus, V-gene segments optimized for expression

    inE. coli and setup for downstream engineering are used in combination

    with tr inucleotide-mediated diversity in the CDR3 regions14. Libraries

    with improved quality and/or downstream characteristics of antibodies

    may also be built by selecting expressed V genes for functionality beforelibrary construction84,85.

    Semi-synthetic libraries have combinations of natural and synthetic

    diversity; they are often created to increase natural diversity while main-

    taining a certain level of functional diversity. For example, such libraries

    have been created by shuffling natural CDR regions86 or by introduc-

    ing naturally rearranged and highly functional CDR3 sequences from

    human B-cells with synthetic CDR1 and CDR2 diversity15. Examples of

    library designs are schematically depicted in Figure 4.

    If sufficiently large and diverse, all these types of librar ies are a source

    of antibodies against a large number of different antigens, including self,

    nonimmunogenic and toxic antigens, and for this reason these libraries

    are now extensively used in the industry and in academia (reviewed in

    refs. 87,88). Medium-affinity an tibodies are readily isolated from rela-

    tively small nonimmune libraries or from synthetic antibody libraries

    diversified in just one or two CDRs or in just one of the two chains,

    which reflects the situation in transgenic mice with restricted antibody

    repertoires89,90. The most current and successful antibody libraries,

    however, display (natural or synthetic) diversity in multiple CDRs and

    routinely yield single-digit nanomolar and sometimes subnanomolar

    affinity ant ibodiesthe latter having affinities equal to the affinities of

    antibodies regularly isolated from immunized mice or from recombi-

    nant immune libraries. In general, antibody affinities from these libraries

    are proportional to the size of the libraryup to 10 nM for libraries with

    107 to 108 clones, and up to 0.1 nM for the best libraries with over 1010members. More importantly, these libraries, in association with high-

    throughput screening, deliver panels with thousands of antibodies that

    bind distinct epitopes on the same target antigen15,63. Antibody leads

    with the highest potencies can then be identified. A subnanomolar affin-

    ity is also readily obtainable when selecting even relatively small librar-

    ies with ribosome display, in which the V genes are mutated between

    selections, although the complexity of selected antibody panels appears

    limited24,25,91,92.

    There seems to be no major differences in performance when compar-

    ing the best nonimmune and synthetic antibody phage-display libraries

    in use today, with regard to the frequency of binders and top affinity of

    selected clones. However, the success of the drug discovery process is

    Table 2 Examples of antibodies subjected to, or obtained by,in vitroselection and/or optimization

    Antibody/developer Target Mutagenesis Strategy

    Increase in affinity/

    resulta Reference

    Vitaxin (MEDI-522 )/Medarex,

    Gaithersburg, MD, USA

    v3 Focused on CDR Screening 80 96

    Synagis (pali vizumab)/MedImmune,

    Gaithersburg, MD, USA

    Respiratory

    syncytial virus

    Focused on CDR Screening 100 off-rate;

    5 on-rate

    94

    RFB4/National Cancer Institute,

    Bethesda, MD, USA

    CD22 Hot-spot mutagenesis Screening 15 175

    b4/12/Scripps Research Institute/

    La Jolla, CA, USA

    gp120 CDR walking Selection on phage 420 to 15 pM 16

    C6.5/University of California,

    San Francisco

    c-erbB2 CDR3 mutagenesis Selection on phage 1,230 to 13 pM 17

    L19/University of Siena, Italy Fibronectin CDR3 mutagenesis Selection on phage 1,317 to 54 pM 99

    G8/University of Maastricht,

    The Netherlands

    MHC peptide Chain shuff ling and CDR3

    mutagenesis

    Selection on phage 18 to 14 nM 174

    Fab-12/Genentech,

    S. San Francisco, CA, USA

    VEGF CDR3 mutagenesis Selection on phage 100 potency 176

    1121/ImClone System/New York VEGF receptor Chain shuffling Selection on phage >30 to 0.1 nM 18

    Humira (adalimumab)/

    Abbott Laboratories,

    Deerfield, IL, USA

    TNF Guided selection Selection on phage 0.3 nM 106

    A4.6.1b/Genentech/S. San Francisco, CA, USA

    VEGF Framework-region library of mAb Selection on phage 125 102

    H6/University of Zurich/Switzerland GCN4 Error-prone PCR and DNA shuffling Selection on ribosomes 500 to 5 pM 28

    C1 1L3 4/Universi ty of Zuri ch/Swi tzerland GCN4 I mmune l ibrary and error-prone PCR Sel ecti on on ri bosomes 6 5 t o 4 0 pM 2 7

    4M5.3 /University of Il linois,

    Urbana, IL, USA

    FITC Error-prone PCR and DNA shuffling Sorting using yeast 1,800 to 48 fM 30

    smE3/Massachusetts Institute of

    Technology (MIT), Cambridge

    CEA Error-prone PCR and DNA shuffling Sorting using yeast 2 85 to 3 0 pM/expres-

    sion increases

    116

    VL-12.3/MIT Huntington

    (htt) protein

    Error-prone PCR/homologous

    recombination

    Sorting using yeast 10 177

    1 4B7 /Uni versi ty of Texas, Aust in, Texas Ant hrax t oxi n Error-prone PCR Sort ing usi ng bact eri a 2 00 -f ol d improvement

    to 21 pM

    33

    a indicates increase in affinity unless otherwise specified. bAvastin (bevacizumab) is a humanized version of this murine mAb. MHC, major hi stocompatibi lit y complex; VEGF, vascularendothelial growth f actor; GCN4, general control protein 4 ; FITC, fluoroisothiocyanate; CEA, carcinoembryonic antigen.

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    7/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1111

    influenced by several impor tant differences among libraries, including

    antibody format and display levels, sequence diversity in selected anti-

    body populations, average expression levels, tendency to multimerize,

    compatibility with expression screening (e.g., automation) and with

    affinity maturation and finally the ease of conversion to other antibody

    formats, display or selection systems.

    Affinity maturation of antibodies.Although initial antibody leads from

    display libraries or from hybridomas often have a number of desirable

    characteristics, their potency is sometimes insufficient for therapeutic

    applications or for use in sensitive diagnoses. Frequently, an antibodys

    potency is governed by its affinity for antigen, and an affinity increase

    may help to increase pharmacokinetic and safety profiles and reduce

    dosing, toxicity and cost of therapy. Indeed, for many well-studied

    cases, increased affinities have translated into improved biological effi-

    cacy (Table 2 and citations therein). For some applications, there is nostrict correlation between affinity and efficacy above a certain affinity

    threshold, such as in some virus neutralizations93,94 and in tumor tar-

    geting, where engineered high-affinity antibodies have been shown to

    not necessarily have superior tumor targeting efficacy compared with

    low-affinity variants, but on the contrary, may display diminished pen-

    etration into solid tumors95

    In the immune system, antibodies are affinity matured in a stepwise

    fashion by incorporating mutations and selecting variants under increas-

    ing selective pressures. In a first and simplest form ofin vitro affinity

    maturation, small libraries with focused diversity at a small number of

    residues that are most likely to interact with ant igen, the CDRs, are built

    using oligonucleotides and PCR. These libraries are then screened to

    identify variants with improved affinity, and mutations conferring the

    highest affinities are combined in a single clone96. Randomization may

    also be introduced at positions frequently mutated in vivo, which are

    most likely to generate improved affinity (hot-spot mutagenesis97) or

    influence affinity based on structural analysis. Focused mutagenesis has

    the advantage that only very small libraries need to be screened initially,

    but the disadvantage is that extensive screening of variant combinations

    is required to find clones with the desired affinity or kinetic charac-

    teristics. Further, the screening assay is the bot tleneck: its throughput

    will determine how many different residues can be efficiently sampled

    in each library, and the whole approach is crucially dependent on its

    capability to discriminate relatively small differences in affinity between

    clones.

    The second approach involves the display of millions of antibody

    variants and selection under conditions that favor clones with improved

    affinity or binding kinetics, in a procedure that mimics the in vivo matu-ration process. This procedure is much faster and allows sampling of a

    much larger sequence space with up to billions of variants, limited only

    by transformation efficiency or scale of the production of the in vitro

    (ribosome) display library. This approach has yielded impressive affin-

    ity gains for selected an tibody fragments: 1,000-fold improvements in

    potency are not uncommon and affinities as low as 48 fM have been cited

    (Table 2), indicating that this in vitroprocedure does not suffer from the

    kinetic and affinity limits inherent in the immune system98.

    Many mutagenesis and selection strategies have been used to provide

    subnanomolar affinities (Fig. 4). Broadly applicable mutagenesis strat-

    egies target the CDRs or the whole V gene with high or low levels of

    randomization, respectively. When clones from (semi)synthetic libraries

    a Immobilized Ag

    b Biotinylated Ag

    c Recombinant Ag

    d Bacteria displaying Ag

    e Subcellular fractions

    f Cells displayingantigen/internalizing Ag

    g Alternating selectionson Ag+/ cells

    h Subtractive cell sorting

    i Tissues with Ag

    j Proximity to other Ag

    k In vivo selection

    l Trypsin digestion

    m Mild reduction(release phage only)

    n Mild reduction(release phage +Ag)

    o Competitive elution

    SS

    SS

    Figure 3 Methods for in vitroselection for

    binding. Selections from display libraries have

    been carried out using several methods (or

    any combination of them). (a) Antigen (Ag)

    immobili zed onto solid supports, columns, pi ns

    or cellulose/poly(vinylidene fluoride) membranes/

    other filters, deposited on BIAcore sensorchips or

    immobili zed indi rectly via capture;

    (b) bioti nylated antigen (bioti n (red) is captured

    via streptavidin-coated beads (gray)); (c) diverserecombinant antigens, includi ng antigens

    incorporated into paramagnetic liposomes

    (left ) and immunoadhesins (right); (d) fixed

    prokaryotic cells displaying the (recombinant)

    antigen; (e) enriched subcellular f ractions or

    membrane fractions; (f) transfected or tumor

    cells to select f or binding or internalization;

    (g) alternating antigen-displaying cells and

    depleting antigen-negative cells; (h) subtractive

    selection (e.g., using sorting procedures

    shown by a flow cytometric analysis of a cell

    population with sorting window (in red)); (i)

    enrichment on tissues (e.g., on appropriately

    prepared tissue slides); (j) proximity to another

    bound ligand (green); and finally, (k) injection

    into living animals and recovery of the relevantcells/tissues. Elution conditions can also be

    used to drive the selection towards the desired

    population, for example, (l) via trypsin-digestion

    of a proteolytically sensitive phage, via mild

    disulfi de-bridge reduction to release (m) phage

    (CysDisplay) or (n) antigen and phage, or (o)

    via competitive elution with a ligand binding to

    the antigen and displacing the relevant phage

    antibody. (For citations, see text and refs.

    10,46.) KatieRis

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    8/12

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    9/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1113

    tissue-specific antigens123. Combining this approach with expression

    cloning, immunoprecipitation and mass spectrometry has already led to

    the identification of novel target molecules on malignant cells124126.

    Intracellular selection. Intracellular antibodies (or intrabodies), when

    folded properly, are valuable tools for studying biological processes and

    for blocking proteins inside cells127. Individual scFv antibodies can be

    evolved directly for stable cytoplasmic expression by growth selection

    in bacteria128, although it may be faster to functionally identify pools

    of phage-selected antibodies that have been recloned and expressed

    intracellularly in mammalian cells129,130. A modified yeast two-hybrid

    selection strategy was previously described that can directly select and

    isolate several functional antigen-binding intrabodies127,131,. More

    recently, libraries have been engineered to contain a high percentage

    of functional intrabodies using scFv frameworks132 or antibody heavy-

    chain variable domains133selected directly in the intracellular environ-

    ment , which were then employed to build single-framework intrabody

    libraries. Although not yet applied to the screening of large libraries oraffinity optimization, combinations with fully automated two-hybrid

    systems may eventually yield a platform suitable for generat ing antibod-

    ies of medium affinity to panels of antigens for large-scale functional

    proteomics projects, as recently suggested by work done with other bind-

    ing proteins134.

    Future developments

    Library technology has led to one human antibody so far approved

    for therapy and many more antibodies in clinical and preclinical trials

    (Table 3). Although a full discussion on the immunogenicity of these

    and other engineered antibodies135 is beyond the scope of this review, it

    is accepted in the field that the risk of immunogenicity may be reduced

    by using antibodies that are as human as possible. With t ime, library

    designs and affinity-maturation strategies may be even more tuned

    towards the ideal antibody composition: an as-close-to-human germ-

    line sequence with optimal affinity yet with a minimal number of T-cell

    epitopes and a human-like heavy chain CDR3 (ref. 136). Library designs

    may go even further by reducing the difficulties in downstream develop-

    ment by avoiding potentially problematic amino acids137 in the variable

    regions (e.g., methionine oxidation, asparagine deamidation or aspartate

    isomerization). Fortunately, as has been demonstrated recently138,139, a

    reduction in diversity to just four or even two well-chosen amino acids,

    does not necessarily limit library performance. Also, future selection

    procedures against instability and aggregation behavior may help to

    reduce potential immunogenicity and increase solubility.

    There are now many different molecular selection strategies for iso-

    lating and engineering human antibodies. The three main selection

    platforms (phage, ribosome/mRNA and microbial cell display) are

    somewhat complementary in their use, but they all fall short of the

    ideal: a selection system that provides within a few days a large panelof antibodies to a large number of epitopes on the target antigen of

    choice, a range of selected affinities, a certain level of stability and

    expression and a precisely targeted sequence diversity. To build a better

    antibody molecular evolution machine, we need to make further refine-

    ments in several areas: first, improve methods and predictive designs

    for introducing diversification into antibody genes to build libraries

    with a higher quality in functionality and biophysical properties; sec-

    ond, build normalized selection and amplification strategies to reduce

    biases towards nondesirable variables (e.g., reduce advantages due to

    PCR, infection, growth, multimerization); third, combine affinity and

    expression maturation for populations rather than individual clones to

    increase throughput and simultaneously maintain or improve multiple

    Table 3 Examples of therapeutic antibodies derived from recombinant antibody libraries

    Name Target Indication Company Clinical phase

    Humira

    (adalimumab)

    TNF Autoimmune diseases Abbott/CambridgeAntibody Technology (CaT)

    Approved for arthritis

    (in phase 2/3 for others)

    Numax

    (MEDI-524)

    Respiratory syncitial virus RSV prophylaxis MedImmune Phase 3

    ABT-874 Interleukin 12 Multiple sclerosis Abbott/CaT Phase 2

    CAT-192B

    (belimumab)

    Transforming growth factor 1 Systemic sclerosis Genzyme/CaT Phase 2

    LymphoStat-B B-cell activating factor Lupus/rheumatoid arthritis Human Genome Sciences/CaT Phase 2

    MT201 Epithelial cell adhesion molecule Breast and prostate cancer Micromet Phase 2

    HGS-ETR1 TRAIL-R1 Non-Hodgkin lymphoma Human Genome Sciences/CaT Phase 2

    CAT-213 Eotaxin1 Allergic rhinitis CaT Phase 2

    MYO-029 Growth differentiation factor-8 Muscular dystrophy Wyeth/CaT Phase 1

    ABthrax Protective antigen Anthrax Human Genome Sciences/CaT Phase 1 finished

    HGS-ETR2 TRAIL-R2 Solid tumors Human Genome Sciences/CaT Phase 1

    CAT-354 Interleukin 13 Asthma CaT Phase 1

    1D09C3 MHC class II Non-Hodgkin lymphoma GPC Biotech/MorphoSys Phase 1

    IMC-11F8 EGFR Solid tumors ImClone Phase 1

    IMC-1121b VEGFR-2 Solid tumors ImClone Phase 1

    GC-1008 Transforming growth factor Idiopathic pulmonary fibrosis Genzyme/CaT Preclinical

    IMC-A12 Insulin-like growth factor receptor Solid tumors ImClone Preclinical

    MOR102 Intracellular adhesion molecule 1 Autoimmune diseases MorphoSys Preclinical

    DX-2240 Tie-1 Cancer Dyax Preclinical

    AZD3102 Undisclosed Alzheimer disease AstraZeneca/Dyax Preclinical

    GMCSFR Granulocyte-macrophagecolony-stimulating factor

    receptor

    Autoimmune diseases CaT/AMRAD Preclinical

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    10/12

    1114 VOLUME 23 NUMBER 9 SEPTEMBER 2005 NATURE BIOTECHNO LO GY

    parameters; and four th, integrate all these in a high-throughput screen

    that allows multiple biophysical parameters to be tracked for thousands

    of antibody leads in parallel. Computational kinetic models would

    help to set quantitative biophysical goals140 and protein modeling and

    design141 would help build improved libraries.

    Although synthetic diversity yields a structural diversity significantly

    greater than that observed in nature142, this approach has not yet been

    exploited to target epitopes not readily recognized by naturally derived

    antibodies, such as narrow cavities and carbohydrate ant igens. Although

    such features were already successfully achieved using engineered pro-

    teins based on single-binding protein domains (see review by p. 1126

    1136; ref. 41), future modeling-based and experimentally assisted de

    novobinding-site design may also lead to libraries of conventional mAbs

    with a propensity either to bind such types of epitopes143or to bind to a

    chosen antigen144 with an extraordinary configuration145, structure146

    or as has been recently shown, a biosensor incorporated into the bind-

    ing site147.

    By not only learning from nature but also liberating antibodies from

    the many restrictions imposed by nature, we have amassed a large col-

    lection of selection platforms that now make it possible to engineer

    antibodies with exquisite and unusual binding affinities, binding kinetics

    and sequence/biophysical characteristics. These platforms have maturedto the point where we can glimpse the promised land that Paul Ehrlich

    wrote about over 100 years ago148: the land which [] will yield rich

    treasures for biology and therapeutics.

    ACKNOWLEDGMENTS

    I thank many colleagues including Jane Osbourn and Lutz Jermutus, Clive Wood,

    Zhenping Zhu, Patr ick Bauerle, Herren Wu, David Chen and Lex Bakker for sharing

    unpublished results and am grateful to Mark Alfenito for reviewing the manuscript.

    COMPETING INTERESTS STATEMENT

    The author declares that he has no competing financial interests.

    Published online at http://www.nature.com/naturebiotechnology/

    1. Winter, G. & Milstein, C. Man-made antibodies. Nature349 , 293299 (1991).

    2. Orlandi, R., Gussow, D.H., Jones, P.T. & Winter, G. Cloning immunoglobul in variabl e

    domains for expression by the polymerase chain reaction. Proc. Natl. Acad. Sci. USA

    86 , 38333837 (1989).

    3. Ward, E.S., Gussow, D., Grif fi ths, A.D., Jones, P.T. & Winter, G. Binding activit ies of a

    repertoire of single immunoglobulin variable domains secreted from Escherichia coli.

    Nature341 , 544546 (1989).

    4. Huse, W.D. et al. Generation of a large combinatorial library of the immunoglobulin

    repertoire in phage lambda. Science246 , 12751281 (1989).

    5. McCafferty, J., Griffi ths, A.D., Winter, G. & Chiswell, D.J. Phage antibodies: filamen-

    tous phage displaying antibody variable domains. Nature348 , 552554 (1990).

    6. Clackson, T., Hoogenboom, H.R., Grif fi ths, A.D. & Winter, G. Making antibody frag-

    ments using phage display libraries. Nature352 , 624628 (1991).

    7. Marks, J.D. et al. By-passing immuni zation: human antibodies from V-gene libraries

    displayed on phage. J. Mol. Biol.222 , 581597 (1991).

    8. Burton, D.R. et al. A large array of human monoclonal antibodies to type 1 human

    immunodeficiency virus from combinatorial libraries of asymptomatic individuals.

    Proc. Natl. Acad. Sci. USA88, 1013410137 (1991).

    9. Winter, G., Grif fi ths, A.D., Hawkins, R.E. & Hoogenboom, H.R. Making antibody by

    phage display technology. Annu. Rev. Immunol.12, 433455 (1994).

    10. Hoogenboom, H.R. Overview of antibody phage-display technology and its applica-tions. Methods Mol. Biol.178 , 137 (2002).

    11. Griff iths, A.D. et al. Isolation of high affinity human antibodies directly from large

    synthetic repertoires. EMBO J.13 , 32453260 (1994).

    12 . Vaughan, T.J. et al. Human antibodies with sub-nanomolar affinities isolated from a

    large non-immunized phage display l ibrary. Nat. Biotechnol.14, 309314 (1996).

    13. de Haard, H.J. et al. A large non-immunized human Fab fragment phage library that

    permits rapid isolation and kinetic analysis of high affinity antibodies. J. Biol. Chem.

    274 , 1821818230 (1999).

    14. Knappik, A. et al. Fully synthetic human combinatorial antibody libraries (HuCAL)

    based on modular consensus frameworks and CDRs randomized with trinucleotides.

    J. Mol. Biol.296 , 5786 (2000).

    15. Hoet, R.M. et al. Generation of high-affinity human antibodies by combining donor-

    derived and synthetic complementarity-determining-region diversity. Nat. Biotechnol.

    23 , 344348 (2005).

    16 . Yang, W.P. et al. CDR walking mutagenesis for the affinity maturation of a potent

    human anti-HIV-1 antibody into the picomolar range. J. Mol. Biol.254 , 392403

    (1995).

    17. Schier, R. et al. Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by

    molecular evolution of the complementarity determining regions in the center of the

    antibody binding site. J. Mol. Biol.263 , 551567 (1996).

    18. Lu, D. et al. Tailoring in vitroselection for a picomolar affinity human antibody

    directed against vascular endothelial growth f actor receptor 2 for enhanced neutral-

    izing activit y. J. Biol. Chem.278 , 4349643507 (2003).

    19. Chames, P., Huf ton, S.E., Coulie, P.G., Uchanska-Ziegler, B. & Hoogenboom, H.R.

    Direct selection of a human antibody fragment directed against the tumor T-cell

    epitope HLA-A1-MAGE-A1 from a nonimmunized phage-Fab library. Proc. Natl. Acad.

    Sci. USA97 , 79697974 (2000).

    20. Huie, M.A. et al. Antibodies to human fetal erythroid cell s from a nonimmune phage

    antibody library. Proc. Natl. Acad. Sci. USA98 , 26822687 (2001).21. Moulard, M. et al. Broadly cross-reactive HI V-1-neutral izing human monoclonal Fab

    selected for binding to gp120CD4-CCR5 complexes. Proc. Natl. Acad. Sci. USA

    99 , 69136918 (2002).

    22. Kramer, R.A. et al. The human antibody repertoire specif ic f or rabies virus glycopro-

    tein as selected from immune libraries. Eur. J. Immunol.35 , 21312145 (2005).

    23. Lipovsek, D. & Pluckthun, A. In-vitroprotein evolution by ribosome display and mRNA

    display. J. Immunol. Methods290 , 5167 (2004).

    24. Hanes, J., Schaff it zel, C., Knappik, A. & Pluckt hun, A. Picomolar affinit y antibodies

    from a fully synthetic naive library selected and evolved by ribosome display. Nat.

    Biotechnol.18 , 12871292 (2000).

    25. Schaffi tzel, C. et al.In vitrogenerated antibodies specific for telomeric guanine-

    quadruplex DNA react with Stylonychia lemnae macronuclei. Proc. Natl. Acad. Sci.

    USA98 , 85728577 (2001).

    26. Jermutus, L., Honegger, A., Schwesinger, F., Hanes, J. & Pluckthun, A. Tailoring in

    vitroevolution for protein affinity or stabili ty. Proc. Natl. Acad. Sci. USA98 , 7580

    (2001).

    27 . Hanes, J., Jermutus, L., Weber-Bornhauser, S., Bosshard, H.R. & Pluckthun, A.

    Ribosome display efficiently selects and evolves high-affini ty antibodies in vitrofrom

    immune libraries. Proc. Natl. Acad. Sci. USA95 , 1413014135 (1998).

    28. Zahnd, C. et al. Directed in vitroevolut ion and crystallographic analysis of a peptide-

    binding single chain antibody fragment (scFv) with low picomolar affinity. J. Biol.

    Chem.279 , 1887018877 (2004).

    29. Boder, E.T. & Wit trup, K.D. Yeast surface display for screening combinatorial poly-

    peptide l ibraries. Nat. Biotechnol.15 , 553557 (1997).

    30. Boder, E.T., Midelfort, K.S. & Wit trup, K.D. Directed evoluti on of antibody fragments

    with monovalent femtomolar antigen-binding aff ini ty. Proc. Natl . Acad. Sci. USA97 ,

    1070110705 (2000).

    31. Daugherty, P.S., Olsen, M.J., Iverson, B.L. & Georgiou, G. Development of an opti-

    mized expression system for the screening of antibody libraries displayed on the

    Escherichia colisurface. Protein Eng.12 , 613621 (1999).

    32 . Chen, G. et al. Isolation of high-affi nity l igand-binding proteins by periplasmic expres-

    sion with cytometric screening (PECS). Nat. Biotechnol.19 , 537542 (2001).

    33. Harvey, B.R. et al. Anchored periplasmic expression, a versatile technology for the

    isolation of high-affinit y antibodies from Escherichia coli-expressed libraries. Proc.

    Natl. Acad. Sci. USA101 , 91939198 (2004).

    34. Urban, J.H. et al. Selection of functional human antibodies from retroviral display

    libraries. Nucleic Acids Res.33 , e35 (2005).35. Odegrip, R. et al. CIS display: In vitroselection of peptides from libraries of protein-

    DNA complexes. Proc. Natl. Acad. Sci. USA101 , 28062810 (2004).

    36. Reiersen, H. et al. Covalent antibody displayan in vitroantibody-DNA library selec-

    tion system. Nucleic Acids Res.33, e10 (2005).

    37. Sepp, A., Tawfik, D.S. & Grif fi ths, A.D. Microbead display by in vitrocompartmen-

    talisation: selection for binding using flow cytometry. FEBS Lett.532 , 455458

    (2002).

    38. Mossner, E., Koch, H. & Pluckt hun, A. Fast selecti on of antibodies without antigen

    purification: adaptation of the protein fragment complementation assay to select

    antigen-antibody pairs. J. Mol. Biol.308 , 115122 (2001).

    39 . Urech, D.M., Licht len, P. & Barberis, A. Cell growth selecti on system to detect

    extracellular and transmembrane protein int eractions. Biochim. Biophys. Acta1622,

    117127 (2003).

    40. Jermutus, L. et al. Ligand binding of a ribosome-displayed protein detected in

    solution at the single molecule level by fluorescence correlation spectroscopy. Eur.

    Biophys. J.31 , 179184 (2002).

    41. Binz, H.K., Amstutz, P. & Pluckthun, A. The matrix reloaded: specifi c binding proteins

    based on nonimmunoglobuli n domains. Nat. Biotechnol.23, in the press (2005).

    42. Zauderer, M. & Smith, E.S. In vitromethods of producing and identifying immuno-

    globulin molecules in eukaryotic cells. US patent application 20,020,123,057A1

    (2002).

    43. Cumbers, S.J. et al. Generation and iterative affinity maturation of antibodies in vitro

    using hypermutating B-cell lines. Nat. Biotechnol.20 , 11291134 (2002).

    44. Weller, S. et al. Hypermutation in human B cells in vivoand in vitro. Ann. NY Acad.

    Sci.987 , 158165 (2003).

    45. Nicolaides, N.C., Grasso, L. & Sass, P.M. Methods for generating geneticall y altered

    antibody producing cell lines with improved antibody characteristics. US patent

    6,808,894 (2004).

    46. Bradbury, A.R. & Marks, J.D. Antibodies from phage antibody libraries. J. Immunol.

    Methods290 , 2949 (2004).

    47. Mutuberria, R., Hoogenboom, H.R., van der Linden, E., de Brune, A.P. & Roovers,

    R.C. Model systems to study the parameters determining the success of phage anti-

    body selections on complex anti gens. J. Immunol. Methods231 , 6581 (1999).

    48. VanAntwerp, J.J. & Wit trup, K.D. Fine affinity discrimination by yeast surface display

    and fl ow cytometry. Biotechnol. Prog.16 , 3137 (2000).

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    11/12

    NATURE BIOTECHN O LO GY VOLUME 23 NUMBER 9 SEPTEMBER 2005 1115

    49. Zaccolo, M., Griff iths, A.P., Prospero, T.D., Winter, G. & Gherardi, E. Dimerization of

    Fab fragments enables ready screening of phage antibodies that affect hepatocyte

    growth factor/scatter factor activity on target cells. Eur. J. Immunol.27 , 618623

    (1997).

    50. Larocca, D. et al. Evolving phage vectors for cell targeted gene delivery. Curr. Pharm.

    Biotechnol.3, 4557 (2002).

    51. Janda, K.D. et al. Direct selection for a catalytic mechanism from combinatorial

    antibody libraries. Proc. Natl. Acad. Sci. USA91 , 25322536 (1994).

    52 . Becerril, B., Poul, M.A. & Marks, J.D. Toward selection of internalizing antibodies

    from phage libraries. Biochem. Biophys. Res. Commun.255 , 386393 (1999).

    53. Feldhaus, M.J. & Siegel, R.W. Yeast display of antibody fragments: a discovery and

    characterization platform. J. Immunol. Methods290 , 6980 (2004).54. Bradbury, A. et al. Antibodies in proteomics II: screening, high-throughput charac-

    terization and downstream applications. Trends Biotechnol.21, 312317 (2003).

    55 . Chambers, R.S. High-throughput antibody production. Curr. Opin. Chem. Biol.9,

    4650 (2005).

    56. Lou, J. et al. Antibodies in haystacks: how selection strategy infl uences the outcome

    of selection from molecular diversity libraries. J. Immunol. Methods253 , 233242

    (2001).

    57. Liu, B., Huang, L., Sihlbom, C., Burli ngame, A. & Marks, J.D. Towards proteome-wide

    production of monoclonal antibody by phage display. J. Mol. Biol.315 , 10631073

    (2002).

    58. Walter, G., Konthur, Z. & Lehrach, H. High-throughput screening of surface displayed

    gene products. Comb. Chem. High Throughput Screen.4, 193205 (2001).

    59 . Pavlik, P. et al. Predicting antigenic peptides suitable for the selection of phage

    antibodies. Hum. Antibodies12 , 99112 (2003).

    60. Van Beijnum, J.R. et al. Target validation for genomics using pept ide-specifi c phage

    antibodies: a study of fi ve gene products overexpressed in colorectal cancer. Int. J.

    Cancer101 , 118127 (2002).

    61. Frisch, C. et al. From EST to IHC: human antibody pipeline for target research.

    J. Immunol. Methods275 , 203212 (2003).

    62. Hoet, R. et al. Method and apparatus for washing magneticall y responsive particl es.

    US patent application 20,030,170,686A1 (2003).

    63 . Edwards, B.M. et al. The remarkable flexibility of the human antibody repertoire;

    isolation of over one thousand different antibodies to a single protein, BLyS. J. Mol.

    Biol.334 , 103118 (2003).

    64 . Hallborn, J. & Carlsson, R. Automated screening procedure for high-throughput

    generation of antibody f ragments. BiotechniquesSuppl., 3037 (2002).

    65. Angenendt, P. et al. Seeing bett er through a MIST: evaluati on of monoclonal recom-

    binant antibody fragments on microarrays. Anal. Chem.76 , 29162921 (2004).

    66 . Vanhercke, T., Ampe, C., Tirry, L. & Denolf, P. Rescue and in situselection and

    evaluati on (RISE): a method for high-throughput panning of phage display libraries.

    J. Biomol. Screen.10 , 108117 (2005).

    67 . Rungpragayphan, S. et al. High-throughput, cloning-independent protein library con-

    struction by combining single-molecule DNA amplification with in vitroexpression.

    J. Mol. Biol.318 , 395405 (2002).

    68 . Holt, L.J., Bussow, K., Walter, G. & Tomlinson, I.M. By-passing selection: direct

    screening for antibody-antigen int eractions using protein arrays. Nucleic Acids Res.

    28 , e72 (2000).69. Watkins, J.D. et al. Discovery of human antibodies to cell surface antigens by capture

    li ft screening of phage-expressed antibody libraries. Anal. Biochem.256 , 169177

    (1998).

    70 . Pini, A., Ricci , C. & Bracci, L. Phage display and colony fi lter screening for high-

    throughput selection of antibody libraries. Comb. Chem. High Throughput Screen.

    5, 503510 (2002).

    71. Michaud, G.A. et al. Analyzing anti body specif ici ty wit h whole proteome microarrays.

    Nat. Bi otechnol.21 , 15091512 (2003).

    72 . Poetz, O. et al. Protein microarrays for antibody profiling: specificity and affinity

    determination on a chip. Proteomics5, 24022411 (2005).

    73. Jostock, T. et al. Rapid generation of functional human IgG antibodies derived from

    Fab-on-phage display l ibraries. J. Immunol. Methods289 , 6580 and corrigendum

    in 294 , 209 (2004).

    74. Sarantopoulos, S., Kao, C.Y., Den, W. & Sharon, J. A method for linki ng VL and VH

    region genes that allows bulk transfer between vectors for use in generating polyclonal

    IgG libraries. J. Immunol.152 , 53445351 (1994).

    75. Persic, L. et al. An int egrated vector system for the eukaryotic expression of antibod-

    ies or their fragments after selection from phage display libraries. Gene187 , 918

    (1997).

    76. Schoonbroodt, S. et al. Oligonucleotide-assisted cleavage and ligation: a novel direc-

    tional DNA cloning technology to capture cDNAs. Application in the construction of a

    human immune antibody phage-display library. Nucleic Acids Res.33, e81 (2005).

    77. Marzari, R. et al. Molecular dissection of the tissue transglutaminase autoantibody

    response in celiac disease. J. Immunol.166 , 41704176 (2001).

    78 . Roovers, R.C. et al. Evidence for a bias toward intracellular antigens in the local

    humoral anti-tumor immune response of a colorectal cancer patient revealed by

    phage display. Int. J. Cancer93 , 832840 (2001).

    79. Haurum, J. & Bregenholt, S. Recombinant polyclonal antibodies: therapeutic anti-

    body technologies come full circle. IDrugs8, 404409 (2005).

    80. Hoogenboom, H.R. & Winter, G. By-passing immunisation. Human antibodies from

    synthetic repertoires of germline VH gene segments rearranged in vitro.J. Mol. Biol.

    227 , 381388 (1992).

    81 . Barbas, C.F., Bain, J.D., Hoekstra, D.M. & Lerner, R. Semisynthetic combinatorial

    libraries: a chemical solution to the diversity problem. Proc. Natl. Acad. Sci. USA

    89 , 44574461 (1992).

    82. Sidhu, S.S. et al. Phage-displayed antibody libraries of synthetic heavy chain comple-

    mentarity determining regions. J. Mol. Biol.338 , 299310 (2004).

    83. Silacci, M. et al. Design, construction, and characterization of a large synthetic human

    antibody phage display li brary. Proteomics5, 23402350 (2005).

    84. de Wildt, R.M., Mundy, C.R., Gorick, B.D. & Tomlinson, I.M. Antibody arrays for high-

    throughput screening of antibody-antigen interactions. Nat. Biotechnol.18, 989994

    (2000).

    85. Loset, G.A. et al. Construction, evaluation and refinement of a large human antibody

    phage library based on the IgD and IgM variable gene repertoire. J. Immunol. Methods

    299 , 4762 (2005).

    86. Soderlind, E. et al. Recombining germline-derived CDR sequences for creating diverse

    single-framework antibody l ibraries. Nat. Biotechnol.18, 852856 (2000).87. Hust, M. & Dubel, S. Mating antibody phage display with proteomics. Trends

    Biotechnol.22 , 814 (2004).

    88 . Hoogenboom, H.R. & Chames, P. Natural and designer bindi ng sit es made by phage

    display technology. Immunol. Today21, 371378 (2000).

    89. Xu, J.L. & Davis, M.M. Diversity in the CDR3 region of V(H) is sufficient for most

    antibody specificities. Immunity13, 3745 (2000).

    90. Senn, B.M. et al. Combinatorial immunoglobulin light chain variability creates suf-

    ficient B cell diversity to mount protective antibody responses against pathogen infec-

    tions. Eur. J. Immunol.33, 950961 (2003).

    91. Lee, M.S. et al. Selection of scFvs specific for HBV DNA polymerase using ribosome

    display. J. Immunol. Methods284 , 147157 (2004).

    92. He, M. et al. Selection of a human anti-progesterone antibody f ragment f rom a trans-

    genic mouse library by ARM ribosome display. J. Immunol. Methods231 , 105117

    (1999).

    93. Bakker, A. et al. Novel human antibody combination effectively neutralizing natural

    rabies virus variants and individual in vitroescape mutants. J. Virol.79, 90629068

    (2005).

    94. Wu, H. et al. Ultra-potent antibodies against Respiratory Syncytial Virus: effects

    of binding kinetics and binding valency on viral neutralization. J. Mol. Biol.350 ,

    126144 (2005).

    95. Weiner, L.M. & Carter, P. Tunable antibodies. Nat. Biotechnol. 23 , 556557

    (2005).

    96. Wu, H. et al. Stepwise in vitroaffinity maturation of Vitaxin, an alphav beta3-specific

    humanized mAb. Proc. Natl. Acad. Sci. USA95 , 60376042 (1998).

    97. Ho, M., Kreitman, R.J., Onda, M. & Pastan, I. In vitroantibody evolution targeting

    germline hot spots to increase activity of an anti-CD22 immunotoxin. J. Biol. Chem.

    280 , 607617 (2005).

    98. Foote, J. & Eisen, H.N. Kineti c and aff ini ty limits on antibodies produced during

    immune responses. Proc. Natl. Acad. Sci. USA92, 12541256 (1995).

    99. Pini, A. et al. Design and use of a phage display library. Human antibodies with sub-

    nanomolar aff ini ty against a marker of angiogenesis eluted from a two-dimensional

    gel. J. Biol. Chem.273 , 2176921776 (1998).

    100. Rauchenberger, R. et al. Human combinatorial Fab library yielding specific and func-

    tional antibodies against the human f ibroblast growth factor receptor 3. J. Biol. Chem.

    278 , 3819438205 (2003).

    101 . Midelfort, K.S. et al. Substantial energetic improvement with minimal structural per-

    turbation in a high affinity mutant antibody. J. Mol. Biol.343 , 685701 (2004).102. Baca, M. & Presta, L.G. SJ, OConnor, S.J. & Wells, J.A. Anti body humanization using

    monovalent phage display. J. Biol. Chem.272 , 1067810684 (1997).

    103. Wu, H., Nie, Y., Huse, W.D. & Watkins, J.D. Humanization of a murine monoclonal

    antibody by simultaneous optimization of framework and CDR residues. J. Mol. Biol.

    294 , 151162 (1999).

    104. Dallacqua, W.F. et al. Antibody humanization by framework shuffling. Methods36,

    4360 (2005).

    105. Beiboer, S.H. et al. Guided selection of a pan carcinoma specific antibody reveals

    similar binding characteristics yet structural divergence between the original murine

    antibody and its human equivalent. J. Mol. Biol.296 , 833849 (2000).

    106. Salfeld, J. et al. Human antibodies that bind human TNF. US patent 6,090,382

    (2000).

    107 . Naundorf, S. et al.In vitroand in vivoactivity of MT201, a fully human monoclonal

    antibody for pancarcinoma treatment. Int . J. Cancer100 , 101110 (2002).

    108. Jespers, L.S., Roberts, A., Mahler, S.M., Winter, G. & Hoogenboom, H.R. Guiding the

    selection of human antibodies f rom phage display repertoires to a single epitope of

    an antigen. Bio/Technology12 , 899903 (1994).

    109. Rader, C., Cheresh, D.A. & Barbas, C.F., I II . A phage display approach for rapid anti-

    body humanization: designed combinatorial V gene libraries. Proc. Natl. Acad. Sci.

    USA95 , 89108915 (1998).

    110. Jung, S., Honegger, A. & Pluckthun, A. Selection for improved protein stabili ty by

    phage display. J. Mol. Biol.294 , 163180 (1999).

    111. Kristensen, P. & Winter, G. Proteolytic selection for protein folding using fil amentous

    bacteriophages. Fold. Des.3, 321328 (1998).

    112. Brockmann, E.C., Cooper, M., Stromsten, N., Vehniainen, M. & Saviranta, P. Selecting

    for antibody scFv fragments with improved stabili ty using phage display with denatur-

    ation under reducing conditions. J. Immunol. Methods296 , 159170 (2005).

    113. Jespers, L., Schon, O., Famm, K. & Winter, G. Aggregation-resistant domain antibodies

    selected on phage by heat denaturation. Nat. Biotechnol.22, 11611165 (2004).

    114. Shusta, E.V., Raines, R.T., Pluckthun, A. & Wit trup, K.D. Increasing the secretory

    capacity of Saccharomyces cerevisiaefor production of single-chain antibody frag-

    ments. Nat. Biotechnol.16 , 773777 (1998).

    115. Shusta, E.V., Holler, P.D., Kieke, M.C., Kranz, D.M. & Witt rup, K.D. Directed evolution

    of a stable scaffold for T-cell receptor engineering. Nat. Bi otechnol.18 , 754759

    (2000).

    REVIEW

    200

    5NaturePublishingGroup

    http://w

    ww.nature.com/naturebiotechnology

  • 8/6/2019 Selecting and Screening of ant Antibody Libraries

    12/12

    1116 VOLUME 23 NUMBER 9 SEPTEMBER 2005 NATUREBIOTECHNO LO GY

    116. Graff , C.P., Chester, K., Begent, R. & Wit trup, K.D. Directed evolut ion of an anti -

    carcinoembryonic antigen scFv with a 4-day monovalent dissociation half-time at

    37 degrees C. Protein Eng. Des. Sel.17 , 293304 (2004).

    117. Roodveldt, C., Aharoni, A. & Tawfik, D.S. Directed evoluti on of proteins for heterolo-

    gous expression and stabil it y. Curr. Opin. Struct. Biol.15 , 5056 (2005).

    118. Topping, K.P., Hough, V.C., Monson, J.R. & Greenman, J. Isolation of human colorec-

    tal tumour reactive antibodies using phage display technology. Int. J. Oncol.16 ,

    187195 (2000).

    119. Roovers, R.C., van der Linden, E., de Brune, A.P., Arends, J.W. & Hoogenboom, H.R.

    Identification of colon tumour-associated antigens by phage antibody selections on

    primary colorectal carcinoma. Eur. J. Cancer37 , 542549 (2001).

    120. Tur, M.K. et al. A novel approach for immunization, screening and characterizationof selected scFv libraries using membrane fracti ons of tumor cells. Int. J. Mol. Med.

    11 , 523527 (2003).

    121. Mutuberria, R. et al. Isolation of human antibodies to tumor-associated endothelial

    cell markers by in vitrohuman endothelial cell selecti on with phage display libraries.

    J. Immunol. Methods287 , 3147 (2004).

    122. Liu, B., Conrad, F., Cooperberg, M.R., Ki rpotin, D.B. & Marks, J.D. Mapping tumor

    epitope space by direct selection of single-chain Fv antibody libraries on prostate

    cancer cells. Cancer Res.64 , 704710 (2004).

    123. Nizak, C. et al. Recombinant antibodies against subcellular fractions used to track

    endogenous Golgi protein dynamics in vivo. Traffic4, 739753 (2003).

    124. Ridgway, J.B. et al. Identification of a human anti-CD55 single-chain Fv by subtrac-

    ti ve panning of a phage library using tumor and nontumor cell l ines. Cancer Res.59 ,

    27182723 (1999).

    125. Geuijen, C.A. et al. A proteomic approach to tumour target identi fication using phage

    display, affinity purification and mass spectrometry. Eur. J. Cancer41 , 178187

    (2005).

    126. Bakker, A.B. et al. C-type lectin-li ke molecule-1: a novel myeloid cell surface marker

    associated with acute myeloid leukemia. Cancer Res.64 , 84438450 (2004).

    127 . Visinti n, M., Meli , G.A., Cannistraci, I . & Cattaneo, A. Intracellular antibodies for

    proteomics. J. Immunol. Methods290 , 135153 (2004).

    128. Martineau, P., Jones, P. & Winter, G. Expression of an anti body fragment at high

    levels in the bacterial cytoplasm. J. Mol. Biol.280 , 117127 (1998).

    129. Gargano, N. & Cattaneo, A. Rescue of a neutralizing anti-viral antibody fragment f rom

    an intracellular polyclonal repertoire expressed in mammalian cells. FEBS Lett.414 ,

    537540 (1997).

    130. Gennari, F. et al. Direct phage to intrabody screening (DPIS): demonstration by

    isolation of cytosolic intrabodies against the TES1 site of Epstein Barr virus latent

    membrane protein 1 (LMP1) that block NF-kappaB transactivation. J. Mol. Biol.

    335 , 193207 (2004).

    131. Visinti n, M., Tse, E., Axelson, H., Rabbitt s, T.H. & Cattaneo, A. Selection of antibodies

    for int racellular functi on using a two-hybrid in vivosystem. Proc. Natl. Acad. Sci.

    USA96 , 1172311728 (1999).

    132. auf der Maur, A. et al. Direct in vivoscreening of intrabody libraries constructed on a

    highly stable single-chain framework. J. Biol. Chem.277 , 4507545085 (2002).

    133. Tanaka, T., Lobato, M.N. & Rabbit ts, T.H. Single domain intracellular antibodies: a

    minimal fragment for direct in vivoselection of antigen-specific intrabodies. J. Mol.

    Biol.331 , 11091120 (2003).134. Amstutz, P. et al. Intracellular kinase inhibitors selected from combinatorial libraries

    of designed ankyrin repeat proteins. J. Biol. Chem.280 , 2471524722 (2005).

    135. Pendley, C., Schantz, A. & Wagner, C. Immunogenicit y of therapeutic monoclonal

    antibodies. Curr. Opin. Mol. Ther.5, 172179 (2003).

    136. Zemlin, M. et al. Expressed murine and human CDR-H3 intervals of equal length

    exhibit distinct repertoires that diff er in t heir amino acid compositi on and predicted

    range of structures. J. Mol. Biol.334 , 733749 (2003).

    137. Harris, R.J., Shire, S.J. & Winter, C. Commercial manufacturi ng scale formulation

    and analytical characterization of therapeutic recombinant antibodies. Drug Dev.

    Res.61 , 137154 (2004).

    138. Fellouse, F.A., Wiesmann, C. & Sidhu, S.S. Synthetic ant ibodies from a four-amino-

    acid code: a dominant role for tyrosine in antigen recognition. Proc. Natl. Acad. Sci.

    USA101 , 1246712472 (2004).

    139. Fellouse, F.A. et al. Molecular recognition by a binary code. J. Mol. Biol. 348 ,

    11531162 (2005).

    140. Rao, B.M., Lauffenburger, D.A. & Wit trup, K.D. Int egrating cell-level kinetic modeling

    into the design of engineered protein therapeutics. Nat. Bi otechnol.23 , 191194

    (2005).

    141. De Genst, E., Areskoug, D., Decanniere, K., Muyldermans, S. & Andersson, K. Kineticand affini ty predicti ons of a protein-protein int eraction using multivariate experimen-

    tal design. J. Biol. Chem.277 , 2989729907 (2002).

    142. Bond, C.J., Wiesmann, C., Marsters, J.C. Jr. & Sidhu, S.S. A structure-based database

    of antibody variable domain diversity. J. Mol. Biol.348 , 699709 (2005).

    143 . Almagro, J.C. I dentification of dif ferences in the specifici ty-determining residues

    of antibodies that recognize antigens of different size: implications for the rational

    design of anti body repertoires. J. Mol. Recognit.17 , 132143 (2004).

    144. Kirkham, P.M., Neri, D. & Winter, G. Towards the design of an antibody that recog-

    nises a given protein epitope. J. Mol. Biol.285 , 909915 (1999).

    145. Calarese, D.A. et al. Antibody domain exchange is an immunological solution to

    carbohydrate cluster recognition. Science300 , 20652071 (2003).

    146. Darbha, R. et al. Crystal structure of the broadly cross-reactive HIV-1-neutralizing

    Fab X5 and fine mapping of its epitope. Biochemistry43 , 14101417 (2004).

    147. Jespers, L., Bonnert, T.P. & Winter, G. Selecti on of optical biosensors from chemi-

    synthetic antibody libraries. Protein Eng. Des. Sel.17 , 709713 (2004).

    148 . Ehrlich, P. On autoimmunity with special references to cell lif e. Proc. R. Soc.66 ,

    424448 (1900).

    149. Salvatore, G., Beers, R., Margulies, I. , Kreitman, R.J. & Pastan, I. Improved cytotoxic

    activity toward cell lines and fresh leukemia cells of a mutant anti-CD22 immunotoxin

    obtained by ant ibody phage display. Clin. Cancer Res.8, 9951002 (2002).

    150. Chames, P. et al. TCR-like human antibodies expressed on human CTLs mediate anti-

    body affinity-dependent cytolytic activity. J. Immunol.169 , 11101118 (2002).

    151. Chen, Y. et al. Selection and analysis of an optimized anti-VEGF antibody: crystal

    structure of an affinity-matured Fab in complex with antigen. J. Mol. Biol.293 ,865881 (1999).

    152. Colby, D.W. et al. Potent inhibition of huntingtin aggregation and cytotoxicity by a

    disulfide bond-free single-domain intracellular antibody. Proc. Natl. Acad. Sci. USA

    101 , 1761617621 (2004).

    153. Desiderio, A. et al. A semi-synthetic repertoire of intrinsically stable antibody

    fragments derived from a single-framework scaffold. J. Mol. Biol.310 , 603615

    (2001).

    154. Azriel-Rosenfeld, R., Valensi, M. & Benhar, I. A human synthetic combinatorial

    library of arrayable single-chain antibodies based on shuffling in vivoformed CDRs

    into general framework regions. J. Mol. Biol.335 , 177192 (2004).

    155. Yau, K.Y. et al. Affinity maturation of a V(H)H by mutational hotspot randomization.

    J. Immunol. Methods297 , 213224 (2005).

    156. De Pascalis, R. et al. In vitro affinity maturation of a specificity-determining

    region-grafted humanized anticarcinoma antibody: isolation and characterization

    of minimally immunogenic high-affi nity variants. Clin. Cancer Res.9, 55215531

    (2003).

    157. Maynard, J.A. et al. Protection against anthrax toxin by recombinant antibody frag-

    ments correlates with antigen affinity. Nat. Bi otechnol.20 , 597601 (2002).

    158. Hoogenboom, H.R.J.M. & Somers, V. Hybridi zation control of sequence variation. US

    patent application 20,040,00 5,709A1 (2004).

    159. Sblattero, D. & Bradbury, A. Exploiting recombination in single bacteria to make large

    phage antibody libraries. Nat. Bi otechnol.18 , 7580 (2000).

    160 . Lutz, R. & Bujard, H. Independent and tight regulation of transcriptional units in

    Escherichia coli via the LacR/O, the TetR/O and AraC/I1I2 regulatory elements.

    Nucleic Acids Res.25 , 12031210 (1997).

    161. Hoet, R., Ladner, R.C. & Frans, N. Methods and compositi ons for controll ing valency

    of phage display. US patent application 20,040,180,422A1 (2004).

    162. McGuinness, B.T. et al. Phage diabody repertoires for selecti on of l arge numbers of

    bispecific antibody fragments. Nat. Biotechnol.14 , 11491154 (1996).

    163. Lee, C.V., Sidhu, S.S. & Fuh, G. Bivalent antibody phage display mimics natural

    immunoglobulin. J. Immunol. Methods284 , 119132 (2004).

    164. Poul, M.A. & Marks, J.D. Targeted gene delivery to mammalian cells by fil amentous

    bacteriophage. J. Mol. Biol.288 , 203211 (1999).

    165. OConnell, D., Becerril, B., Roy-Burman, A., Daws, M. & Marks, J.D. Phage versus

    phagemid libraries for generation of human monoclonal antibodies. J. Mol. Biol.321 ,

    4956 (2002).

    166. de Wildt, R.M., Tomlinson, I.M., On