16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

Embed Size (px)

Citation preview

  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    1/7

    A revival of bispecific antibodiesPeter Kufer1,2, Ralf Lutterbuse1 and Patrick A. Baeuerle1

    1Micromet AG, Staffelseestr. 2, 81477 Munich, Germany2Institute of Immunology, Goethestr. 31, 80336 Munich, Germany

    Bispecific antibodies usually do not occur in nature but

    are constructed by recombinant DNA or cell-fusion

    technologies. Most are designed to recruit cytotoxic

    effector cells of the immune system effectively against

    pathogenic target cells. This complex task explains

    why, after more than 15 years of extensive research,

    many different formats of bispecific antibodies havebeen developed but only a few have advanced to clini-

    cal trials. Here, we give a brief history of bispecific anti-

    bodies and review very recent progress towards

    formats that are beginning to solve the major issues of

    earlier formats. These improved bispecific antibodies

    are expected to show clinical efficacy in patients with

    cancer and other diseases, in a way that monoclonal

    antibodies have shown in recent years.

    Until now, the hybridoma technology invented by Kohler

    and Milstein to generate monoclonal antibodies has

    nourished the hope for therapeutic breakthroughs in

    diseases with high medical needs not served sufficientlyby conventional therapies [1]. The hallmark of monoclonal

    antibodies is their specific binding to a particular antigen,

    which enables them to find their target precisely in vivo

    while ignoring antigen-negative sites. Bound to a target,

    therapeutic antibodies can deliver a toxic payload, act as

    agonists or antagonists of receptors, or as neutralizers of

    ligands. Antibodies might even bind many targets that are

    not recognizable by small-molecule drugs.

    Monoclonal antibodies of the IgG type contain two

    identical antigen-binding arms and a constant fragment,

    (Fc)g. The Fc part enables the antibody to function as an

    adaptor protein, linking antibody-bound cells to immune

    cells bearing Fcg receptors. Because there are different

    Fcg receptors and other proteins binding to Fc portions of

    antibodies, such as complement, monoclonal antibodies

    can mediate multiple effects ranging from the recruitment

    of immune effector functions to mere increase of serum

    half-life by retention of IgG on non-signaling Fc receptors.

    It was observed recently that human antibodies of the

    IgG4 type can exchange their halves with each other,

    potentially creating antibodies with dual specificity [2].

    However, the biological relevance of this observation

    remains obscure.

    For treatment of malignant diseases, monoclonal

    antibodies typically need to be modified to enhance efficacy

    and to use them in humans. One important modification is

    the reduction of immunogenicity of rodent monoclonalantibodies by chimerization, humanization through

    grafting of complementarity determining regions (CDRs)

    or using various technologies for recovery of fully human

    antibodies, such as phage display libraries or transgenic

    mice expressing human antibody repertoires. Reduced

    immunogenicity of antibodies can prolong their half life

    and, in the absence of a neutralizing immune response,

    enable prolonged treatment. Another important modifi-cation is arming the humanized antibody with additional

    cytotoxic mechanisms, be it radioisotopes, bacterial toxins,

    inflammatory cytokines, chemotherapeutics or prodrugs.

    There is a growing number of approved cancer thera-

    peutics that are efficacious either as chimerized antibody

    (Rituximab [3]) or humanized IgG1 (Herceptin [4] and

    Campath-1H [5]), or as conjugate with chemotherapeutics

    (Mylotarg [6]) or a radioisotope (Zevalin [7] and Bexxar

    [8]). In spite of this progress, the efficacy of monoclonal

    antibodies for cancer treatment is still limited [9], leaving

    great potential for further improvements. One class of

    antibody derivatives with the promise of enhanced potency

    for cancer treatment are bispecific antibodies.

    From monoclonal to bispecific antibodies

    Antibodies with a dual specificity in their binding arms

    usually do not occur in nature and, therefore, had to be

    crafted with the help of recombinant DNA or cell-fusion

    technology. Among the first bispecific antibodies were

    constructs designed to redirect T cells against cancer

    target cells [10]. Target cells were killed when cytotoxic

    T lymphocytes were tethered to tumor cells and simul-

    taneously triggered by one arm of the bispecific antibody

    that interacted with the T-cell receptor (TCR) CD3

    complex. The use of the monomorphic CD3 complex for

    triggering T cells circumvented the restrictions of clono-

    typic T-cell specificity and enabled a polyclonal cytotoxic

    T lymphocyte response against target cells bearing the

    antigen recognized by the second arm of the bispecific

    antibody [11]. It is important to note that cytotoxic

    T lymphocytes (CTL), which are considered to be the

    most potent killer cells of the immune system, cannot be

    engaged by monoclonal antibodies because they lack Fcg

    receptors. Another development is bispecific antibodies

    that simultaneously bind tumor cells and an activating

    Fcg receptor, for example, CD64/FcgRI on monocytes [12].

    Their binding to Fcg receptors can elicit effector cell

    activation, without being competed by simultaneously

    binding normal IgG.

    Why so many bispecific antibody formats?

    Production of bispecific antibodies in sufficient amounts

    and purity was an obvious challenge from the beginning.Corresponding author: Peter Kufer ([email protected]).

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004

    www.sciencedirect.com 0167-7799/$ - see front matter q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2004.03.006

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    2/7

    Although producing small amounts for in vitro studies and

    animal models was relatively straightforward, high and

    affordable production yields as needed for clinical grade

    material required major efforts. Thus, the intense parallel

    development of various molecular formats of bispecific

    antibodies was largely driven by an attempt to solve

    production problems (Figure 1).

    The first approach to construct and produce bispecific

    antibodies was the quadroma technology [1315]. It is

    based on the somatic fusion of two different hybridoma cell

    lines expressing murine monoclonal antibodies with the

    desired specificities of the bispecific antibody. Because of

    the random pairing of two different Ig heavy and light

    chains within the resulting hybridhybridoma (or quad-

    roma) cell line, up to ten different immunogloblin species

    are generated of which only one is the functional bispecific

    antibody. The presence of mispaired by-products reduced

    production yields significantly and required sophisticatedpurification procedures. A major improvement to the

    conventional quadroma approach was the somatic fusion

    of a murine and a rat hybridoma cell line expressing

    monoclonal antibodies with two IgGsubclasses selected for

    their preferential pairing [16]. Bispecific antibodies from

    quadroma cell lines still closely resemble conventional

    monoclonal antibodies. Besides the dual-specific antigen

    binding fragment (Fab) parts, they contain an Fcg part and

    can thus be considered trispecific. The additional inter-

    action of such trispecific antibodies with Fcg receptors

    might have desired as well as undesired biological effects.

    To avoid undesired effects, bispecific F(ab0)2 fragments

    were prepared by removing the Fcg part through

    enzymatic digestion [17]. Other approaches used chemical

    conjugation of two different monoclonal antibodies or

    smaller antibody fragments [18]. A straightforward

    method was the coupling of two parental antibodies with

    a hetero-bifunctional crosslinker, but the resulting prep-

    arations of bispecific antibodies suffered from a significant

    molecular heterogeneity because reaction of the cross-linker with the parental antibodies was not site-directed.

    To obtain more homogeneous preparations of bispecific

    antibodies two different Fab fragments have been chemi-

    cally crosslinked at their hinge cysteine residues in a site-

    directed manner [19].

    Recombinant DNA technology promised to overcome

    shortcomings of conventional bispecific antibody pro-

    duction. Bispecific antibodies resembling the quadroma-

    derived format but consisting of human instead of murine

    and/or rat sequences could be designed by the elegant

    knobs-into-holes strategy [20]. The mispairing of Ig heavy

    chains was reduced in this technology by mutating

    selected amino acids forming the interface of the CH3domains in human IgG. At positions within the CH3domain at which the two heavy chains interact directly, an

    amino acid with a small side chain (hole) was introduced

    into the sequence of one heavy chain and an amino acid

    with a large side chain (knob) into that of the other one. As

    a result, the more favorable protein interaction between

    knobs and holes led to the formation of up to 90% of the

    correct bispecific human IgG by transfected mammalian

    host cells.

    Alternative recombinant strategies focused on smaller

    bispecific antibody constructs. A simpler structure and

    smaller molecular size should enable bacterial expression

    of larger quantities in Escherichia coli. One strategy

    made use of the natural ability of certain protein

    domains to associate as heterodimers. The leucine

    zipper domains of transcription factors Fos and Jun

    were joined to the C terminus of two different single-

    chain Fv antibody fragments [21], or, to the corresponding

    heavy chain segments in a Fab-based approach [22].

    However, expression in E. coli revealed preferential

    formation of stable FosFos and JunJun homodimers.

    Therefore, bispecific heterodimers had to be generated

    finally in vitro in a second step by mixing the dissociated

    halves of homodimers. A similar approach relying on the

    heterodimerization of the constant Ig domains Ck

    and CH1led to the successful association of two different single-

    chain Fv antibody fragments during periplasmicexpression in E. coli [23]. In contrast to the Fos Jun

    strategy, no additional in vitro-engineering steps were

    required.

    Figure 1. Alternative bispecific antibody formats. (a) Quadroma-derived bispecific

    antibody, resembling monospecific monoclonal antibodies in structure and size.

    (bd) Heterodimeric bispecific antibodies of medium size, comprising constant

    immunoglobulin regions and/or heterodimerization domains. (ef) Bispecific anti-

    bodies of minimal size, consisting only of variable immunoglobulin regions.Abbreviations: CH13, constant regions of Ig-heavy chain; CL, constant regions of

    Ig-light chain; Fab, antigen binding fragment; Fc, constant fragment; VH, variable

    region of Ig-heavy chain; VL, variable region of Ig-light chain. The difference in col-

    ors (i.e. gray and purple) represents different binding specificities.

    TRENDS in Biotechnology

    S-S

    VL

    VH

    CH1

    CL

    CH2

    CH3

    CH1Ck

    (c)

    (a)

    (e)

    VL

    VH

    VH

    VL VL VH

    VL

    VH

    CH1

    CL

    VH

    VL

    Fab

    Fc

    Quadroma F(ab)2

    Heterodimeric scFv Heterodimer ic Fab

    Diabody Tandem scFv

    VH VL

    (b)

    (d)

    (f)

    FosJun

    VL

    VH

    CH1

    CL

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004 239

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    3/7

    Recombinant DNA technology was also used to

    generate particularly small bispecific antibodies consist-

    ing of only two VH and two VL domains from two different

    antibodies. In diabodies, each VL domain is connected by a

    short peptide linker with the VH domain of the other

    antigen specificity and vice versa [24]. The peptide linker

    is three to 12 amino acids in length, which is too short to

    enable single-chain Fv formation between non-cognate

    Vregions. Coexpression of respective VHVL fusions in the

    periplasm of E. coli resulted in stable bispecific diabodies.

    A further development of diabodies are so-called tandem

    diabodies [25]. Although diabody crystal structures

    revealed considerable flexibility [26], the connection of

    the antigen-binding arms by two amino acid linkers might

    nevertheless cause steric restrictions. This is in line with

    the observation that the antigen-binding arms in diabo-

    dies with VLVH configuration are more flexible than in

    diabodies with VH VL domain arrangement [27]. Thus,diabodies in general are capable of bridging surface

    antigens between different cells [28]; however, this

    might become problematic in situations in which the

    simultaneous accessibility of two antigens on two different

    cells itself is restricted.

    An alternative small format is the arrangement of two

    single-chain antibody fragments (scFv) connected by a

    flexible polypeptide linker on a single polypeptide chain.

    First attempts to produce such tandem scFvs were made in

    E. coli [29,30]. From these and more recent experiments

    [31] it became clear that, in contrast to diabodies,

    expression of functional bispecific single-chain antibodies

    in E. coli was not feasible. Renaturation of denaturedprotein fromE. coli inclusion bodies was no solution either

    as it resulted in poor yields of functional bispecific

    antibody. A breakthrough for bispecific single-chain

    antibodies came from their expression in mammalian

    cells [32]. It seemed that the secretory pathway of higher

    eukaryotic cells was capable of properly folding the four

    consecutively aligned Ig domains, and secrete the tandem

    scFv in decent yields as fully functional bispecific antibody

    into the cell culture medium. Affinity purification by a

    poly-histidine tag attached to the C terminus, and size-

    exclusion chromatography enabled preparation of highly

    homogeneous bispecific antibodies. The flexible GlySer

    linker connecting the two scFvs was chosen to be short to

    prevent mispairing of non-cognate V regions, although

    longer linkers turned out to work as well [32,33]. In

    particular, there was no need for peptide linkers forming

    secondary structures to keep the different antigen-binding

    sites apart [34]. In contrast to diabodies, the two binding

    sites in single-chain bispecific antibodies can rotate freely

    and their axes can be kinked, which might facilitate

    simultaneous binding of two antigen epitopes juxtaposed

    on two different cell surfaces.

    Biological effects of bispecific antibodies and clinical

    experience

    The vast majority of bispecific antibodies were designed to

    redirect cytotoxic effector cells against target cells thathave a key function in disease processes (Figure 2). With

    respect to target cells, most approaches selected anti-

    bodies for bispecific antibody development recognizing

    tumor-associated surface antigens to eliminate malignant

    cells causing cancer. With respect to cytotoxic effector cells,

    a variety of antibodies against triggering molecules of

    cytotoxic effector cells were tested for bispecific antibody

    development.

    The best characterized triggering molecules for recruit-

    ing myeloid effector cells are the high affinity IgG receptor

    FcgRI/CD64 and the IgA receptor FcaRI/CD89. CD64 is an

    activating receptor found on monocytes, macrophages anddendritic cells and can be upregulated on neutrophils

    by G-CSF, GM-CSF and IFN-g [35,36]. Monocyte-

    mediated tumor-cell lysis by CD64-directed bispecific

    antibodies required preactivation of the effector cells by

    IFN-g and showed a dose response relationship similar to

    that found with antibody-dependent cellular cytotoxicity

    (ADCC) induced by a humanized IgG1 antibody against

    the same target [37]. Likewise, CD64-mediated recruit-

    ment of neutrophils requires preactivation of the effector

    cells [38,39]. So far, no consistent antitumor activity was

    observed in clinical trials with the bispecific F(ab0)2antibodies MDX-210 (Her2/neu CD64), MDX-H210

    ( humanized MDX-210) and MDX-447 (EGFR CD64)

    [40,41], although high doses (20 mg m22) were welltolerated and biopsies of metastatic lesions showed

    tumor localization of bispecific antibody [42]. The only

    exception reported was a Phase II clinical trial in patients

    with advanced Her2/neu-expressing prostate cancer trea-

    ted with humanized MDX-H210 plus GM-CSF, in which

    35% of the evaluable patients showed a prostate-specific

    antigen (PSA) response [43]. Limited clinical efficacy

    might be explained by preclinical data on MDX-210

    showing that measurable tumor-cell lysis requires bispe-

    cific antibody concentrations of 0.11 mg ml21 and high

    effector-to-target (E:T) cell ratios of at least 40:1 even when

    human neutrophils that had been prestimulated with IFN-g

    and G-CSF were used [44].CD89 is expressed primarily on neutrophils, monocytes,

    macrophages and eosinophils [45]. In contrast to CD64,

    expression of this activating receptor on neutrophils is

    Figure 2. Redirected target-cell lysis by a cytotoxic effector cell, mediated by a

    bispecific antibody. The simultaneous engagement by a bispecific antibody of

    both a surface antigen on the target cell and a triggering receptor on the effector

    cell induces the effector cell to kill the target cell by delivering a cytotoxic payload.

    TRENDS in Biotechnology

    Triggering receptor

    Surfaceantigen

    Cytotoxicgranules

    Bispecificantibody

    Redirectedlysis

    Target cell

    Cytotoxic effector cell

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004240

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    4/7

    constitutive. Bispecific antibodies triggering CD89 could

    be demonstrated to mediate redirected lysis of tumor cells

    by freshly isolated neutrophils and monocytes without any

    need for prestimulation [46]. To obtain acceptable killing

    rates in vitro, however, high E:T ratios and concentrations

    of bispecific antibody were required (Table 1). CD89-

    directed bispecific antibodies have not yet been tested in

    clinical trials.

    Bispecific antibodies designed to redirect the cytotoxic

    activity of lymphocytic effector cells against tumor targets

    mainly engaged CD16/FcgRIII or CD3 as triggering

    molecules. CD16 is the major activating receptor on

    natural killer (NK) cells and mediates low affinity binding

    of IgG. The hybridhybridoma-derived bispecific antibody

    2B1 (Her2/neu CD16) showed few minor clinical

    responses in a Phase I trial in patients with advanced

    Her2/neu-overexpressing cancer [47]. Infusion of 2B1

    induced significant increases in the levels of circulatingcytokines, and maximally tolerated dose (MTD) was

    reached at 2.5 mg m22. The toxicity of the molecule has

    been explained by its Fcg part leading to systemic

    leukocyte activation by simultaneous engagement of

    CD16 and other Fcg receptors [48]. Compared with 2B1,

    recombinant bispecfic Her2/neu CD16 constructs in the

    tandem scFv format devoid of the Fcg part, required a

    100-fold higher concentration in vitro to achieve the same

    peak tumor-cell lysis by peripheral blood lymphocytes

    prestimulated with IL-2 [49]. Thus, the Fcg part is not only

    responsible for clinical side effects but also contributes to

    the biological activity of CD16-directed bispecific anti-

    bodies. HRS-3/A9 is another CD16-directed bispecificantibody already tested in clinical trials [50]. The

    compound is a bispecific F(ab0)2 antibody (without Fcg

    part) targeting the CD30 antigen on Hodgkin and Reed-

    Sternberg cells in patients with Hodgkins Disease (HD).

    HRS-3/A9 was significantly less toxic than 2B1, and

    maximum tolerated dose was not reached at 64 mg m22

    when the first Phase III trial ran out of clinical test

    material because of the limitations of the production

    process. The first study reported one complete remission

    (CR) and one partial remission (PR) in 15 treated patients

    [51], which could be confirmed by 25% objective antitumor

    responses (1 CR 3 PR) in a second clinical trial [52].

    Therefore, HRS-3/A9 can be regarded as the first clinicalproof of the bispecific antibody concept, although the

    problem of insufficient drug supply remains unresolved.

    CD3 is chosen as the triggering molecule in most

    bispecific antibody approaches aimed at redirecting

    cytotoxic T-cell activity against tumor cells. CD3 is a

    T-cell-specific complex of three different monomorphic

    chains (1, g and d) associated as signal transduction units

    with the polymorphic TCR, which is devoid of signalling

    properties. Thus, CD3 engagement by bispecific antibodies

    can mediate TCR signaling by circumventing the clono-

    typic antigen specificity of T cells. However, stimulation of

    T-cell activity is a complex process strictly regulated by

    several molecules, such as the costimulatory molecule

    CD28. Indeed, most CD3-directed bispecific antibodies

    require costimulation or prestimulation of T cells to elicit

    cytotoxic activity against target (tumor) cells. This

    dependency has been observed in vitro and in animal

    models and applies to the conventional bispecific F(ab 0)2format as well as to various recombinant diabodies [28,

    5358]. Most T-cell-redirecting bispecific antibodies

    require an excess of effector cells for significant tumor-

    cell lysis (E:T ratios of at least 2:1), and a broad range of

    bispecific antibody concentrations is reported in the

    literature for efficient induction of redirected T-cellcytotoxicity (Table 1). So far, indications for clinical efficacy

    of CD3-directed bispecific antibodies have come mainly

    from locoregional or intraperitoneal treatment of cancer

    patients [5962], or from adoptive transfer of Tcells coated

    ex vivo with bispecific antibody [63,64]. However, systemic

    application, which is the desired route for bispecific

    antibody therapy of, for instance, metastatic cancer, has

    not shown clinical efficacy up to now. In one study, patients

    with B-cell malignancies were treated by intravenous

    infusion of a hybrid hybridoma-derived bispecific anti-

    body against CD19 and CD3 with or without IL-2 [65].

    Doses of up to 5 mg were administered, which caused a

    massive systemic release of cytokines but no clinicalresponses. Another Phase I clinical trial in renal cell

    cancer patients receiving bispecific F(ab0)2 antibody

    against Ep-CAM, CD3 and IL-2 also revealed systemic

    cytokine release, which, in this case, was dose-limiting at

    5 mg kg21 [66]. No antitumor responses could be observed

    at the tested dose levels.

    In addition to Fc receptors and the TCRCD3 complex,

    several other, less potent triggering molecules such as

    CD2, which is present on T and NK cells [67] were

    evaluated for effector cell recruitment through bispecific

    antibodies [68].

    Except for the recruitment of immune effector cells,

    bispecific antibodies have been designed for clearingpathogens from the bloodstream through specific targeting

    to CR1 (complement receptor 1) on erythrocytes [69,70] and

    for specific interference with complement regulators on

    tumor cells [71]. Moreover, bispecific antibodies can be used

    for the specific targeting of toxins [72], chemotherapeutic

    drugs [73] and radioisotopes to tumor cells [74]. In yet

    Table 1. Characteristics of bispecific antibodies recruiting cytotoxic effector cells and requirements for redirected target-cell lysis

    Triggering molecule CD64 CD89 CD16 CD3

    Bispecific antibody format F(ab0)2 F(ab0)2

    Quadroma

    F(ab0)2Tandem scFv

    F(ab0)2Diabody

    BiTE

    Effector cells Monocytes and neutrophils NK cells T cells

    Requirement of pre- or costimulation Yes No Yes Yes NoED50 range [mg ml

    21] 0.1 1 0.1 1 0.03 1 0.001 1 10251024

    E:T ratio $40:1 100:1 200:1 50:1 $2:1 # 1:10

    Refs [44] [46] [49] [28,57] [80]

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004 241

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    5/7

    another approach, bispecific antibodies are used to target

    two different tumor-associated antigens simultaneously, to

    achieve enhanced and more specific tumor targeting [75].

    BiTEs: a promising new class of bispecific antibodies

    Several factors limit the biological activity of most tumor-

    directed bispecific antibodies. Even those bispecific anti-

    bodies triggering the most potent activating receptors on

    respective effector cells, i.e. CD64 or CD89 on monocytes

    and neutrophils, CD16 on NK cells, and CD3 on T cells,

    suffer from at least one of the following shortcomings:

    (i) the need for additional signals inducing expression of

    the triggering receptor and/or a pre- or costimulation of

    effector cells; (ii) high concentrations of bispecific antibody

    for decent target-cell lysis, which cannot be realized in

    patients because of limited drug supply; (iii) dose-limiting

    toxicity, and (iv) an excess of effector over target cells,

    which is rarely encountered in vivo (Table 1).It now seems possible to overcome most if not all

    previous limitations by a new class of bispecific antibodies,

    designated bispecific T-cell engager (BiTEs). BiTEs are

    recombinant bispecific single-chain antibodies consisting

    of two scFv antibody fragments directed against a surface

    antigen on target cells and CD3 on T cells. BiTEs can be

    efficiently produced as fully functional molecules by

    mammalian cells. In contrast to other CD3-directed

    bispecific antibodies, BiTEs are capable of efficiently

    redirecting T-cell cytotoxicity against various different

    target cells without any requirement for pre- or costimula-

    tion of effector T cells [32,7678]. This might be explained

    by their ability to induce immunological cytolytic synapsesbetween target cells and cytotoxic T cells that are

    indistinguishable in composition, subdomain arrange-

    ment and size from synapses induced by regular T-cell

    stimuli (S. Offner et al., unpublished). Typically, engage-

    ment of only a few TCR molecules per T cell is sufficient to

    trigger synapse formation and T-cell cytotoxicity efficiently

    [79]. Likewise, target cells obviously need to be decorated

    with only a few BiTE molecules to be killed specifically by

    T cells. This is in line with the observation that BiTE

    concentrations as low as 10 100 pg ml21 (,0.2 2 pM) are

    usually sufficient for half-maximal target-cell lysis [80].

    Sub-microgram amounts of a BiTE were also sufficient to

    prevent tumor outgrowth in a mouse model [81]. Data from

    video-assisted microscopy show that BiTEs mediate serial

    killing of many target cells by single cytotoxic T cells

    (P. Hoffmann et al., unpublished), explaining how BiTEs

    can lead to a complete lysis of target cells at E:T ratios as

    low as 1:10 [82]. Obviously, cytotoxic activity at particu-

    larly low E:T ratios is one of the most important

    prerequisites for clinical efficacy, which has not been met

    by any other bispecific antibody format so far. Moreover, it

    is an attractive feature of BiTEs that no coadministration

    of costimulatory agents is necessary that would otherwise

    inevitably result in demanding multistep or multicom-

    pound treatment regimens.

    Future perspectivesTherapeutics based on bispecific antibodies have not yet

    yielded the anticipated clinical success. However, this

    review shows that further developments are ongoing with

    considerable pace and ingenuity. Bispecific formats that

    are still actively pursued in preclinical and early clinical

    development include diabodies and tandem diabodies,

    crosslinked F(ab0)2, trispecific quadroma antibodies and

    single-chain bispecifics. Their success in the clinic will

    hinge on the quality of the target selected for recognizing

    cancer cells, whether large-scale production is feasible,

    and on the biological activities of recruited effector cell

    population.

    The critical selection of the antigen for tumor targeting

    is a challenge shared with all other antibody-based

    approaches exploiting the exquisite specificity of anti-

    bodies. Antibodies against a great variety of tumor-

    associated antigens have been characterized over the

    past 25 years. Those that have shown good tumor imaging

    and an acceptable safety profile in the clinic are good

    candidates for an enhancement of efficacy through

    conversion into a bispecific format that is expected tohave a higher potency and a particular benefit through

    using natural immune effector mechanisms.

    Complexity of production and cost of goods is a

    permanent issue with biologicals and, in particular, with

    antibody derivatives. Simplicity and robustness of a

    bispecific construct, reduction of the number of production

    steps, and use of powerful production systems are key

    parameters to solve the production issues that have

    dominated the field of bispecific antibodies for a long time.

    The race is still open as to which effector cell population

    of the immune system, after recruitment through bispe-

    cific antibodies, is best suited to eliminate tumor cells. The

    increasing list of immune evasion mechanisms discoveredin late-stage tumor cells [83] strongly suggests that T cells

    are the worst enemies of tumor cells. MHC-class-I-

    negative tumors, which are no longer recognized by

    T cells, are frequent in the metastatic situation [84]. In

    earlier stages in which tumors are smaller, genetically

    more homogenous and better penetrable by antibodies

    other immune effector cells bearing CD16 or CD64 as

    targets might have good therapeutic effects when

    recruited by bispecific antibodies. Much more has to be

    learned about the potential of the various immune effector

    cells recruited by bispecific antibodies for cancer treat-

    ment. Because the bispecific antibody essentially works as

    a catalyst, repeatedly tethering a complex toxic payload

    namely an entire immune effector cell to the cancer cell,

    it is vital to understand what immune cells can achieve at

    which stage of cancer after being recruited by a bispecific

    antibody. The number of immune cells in the periphery

    and target tissue, their state of activity, cytotoxic potential,

    ability to penetrate (tumor) tissue, ability to effect serial

    killing, and susceptibility to evasion mechanisms of

    tumors are just a few of the parameters that must be

    studied in this respect.

    References1 Kohler, G. and Milstein, C. (1975) Continuous cultures of fused cells

    secreting antibody of predefined specificity. Nature 256, 495497

    2 Schuurman, J. et al. (2001) The inter-heavy chain disulfide bonds ofIgG4 are in equilibrium with intra-chain disulfide bonds. Mol.

    Immunol. 38, 18

    3 McLaughlin, P.etal. (1998) Rituximab chimeric anti-CD20 monoclonal

    antibody therapy for relapsed indolent lymphoma: half of patients

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004242

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    6/7

    respond to a four-dose treatment program. J. Clin. Oncol. 16,

    28252833

    4 Pegram, M.D. et al. (1998) Phase II study of receptor-

    enhanced chemosensitivity using recombinant humanized anti-

    p185HER2/neu monoclonal antibody plus cisplatin in patientswith HER2/neu-overexpressing metastatic breast cancer refractory to

    chemotherapy treatment. J. Clin. Oncol. 16, 26592671

    5 Moreton, P. and Hillmen, P. (2003) Alemtuzumab therapy in B-cell

    lymphoproliferative disorders. Semin. Oncol. 30, 493501

    6 Sievers, E.L. et al. (2001) Efficacy and safety of gemtuzumab

    ozogamicin in patients with CD33-positive acute myeloid leukemia

    in first relapse. J. Clin. Oncol. 19, 32443254

    7 Wiseman, G.A. et al. (2002) Radiation dosimetry results for Zevalin

    radioimmunotherapy of rituximab-refractory non-Hodgkin lym-

    phoma. Cancer 94, 13491357

    8 Zelenetz, A.D. (2003) A clinical and scientific overview of tositumomab

    and iodine I 131 tositumomab. Semin. Oncol. 30, 2230

    9 Fishelson, Z. et al. (2003) Obstacles to cancer immunotherapy:

    expression of membrane complement regulatory proteins (mCRPs)

    in tumors. Mol. Immunol. 40, 109123

    10 Staerz, U.D. et al. (1985) Hybrid antibodies can target sites for attackby T cells. Nature 314, 628631

    11 Perez, P. et al. (1985) Specific targeting of cytotoxic T cells by anti-T3

    linked to anti-target cell antibody. Nature 316, 354356

    12 Karpovsky, B. et al. (1984) Production of target-specific effector cells

    using hetero-cross-linked aggregates containing anti-target cell and

    anti-Fc gamma receptor antibodies. J. Exp. Med. 160, 16861701

    13 Staerz, U.D. and Bevan, M.J. (1986) Hybrid hybridoma producing a

    bispecific monoclonal antibody that can focus effector T-cell activity.

    Proc. Natl. Acad. Sci. U. S. A. 83, 14531457

    14 Milstein, C. and Cuello, A.C. (1983) Hybrid hybridomas and their use

    in immunohistochemistry. Nature 305, 537540

    15 Suresh,M.R.etal. (1986) Bispecific monoclonal antibodies fromhybrid

    hybridomas. Methods Enzymol. 121, 210228

    16 Lindhofer, H. et al. (1995) Preferential species-restricted heavy/light

    chain pairing in rat/mouse quadromas. Implications for a single-step

    purification of bispecific antibodies. J. Immunol. 155, 21922517 Mezzanzanica, D. et al. (1988) Human ovarian carcinoma lysis by

    cytotoxic T cells targeted by bispecific monoclonal antibodies: analysis

    of the antibody components. Int. J. Cancer 41, 609615

    18 Brennan, M. et al. (1985) Preparation of bispecific antibodies by

    chemical recombination of monoclonal immunoglobulin G1 fragments.

    Science 229, 8183

    19 Glennie, M.J. et al. (1987) Preparation and performance of bispecific

    F(ab0 gamma)2 antibody containing thioether-linked Fab0 gamma

    fragments. J. Immunol. 139, 23672375

    20 Merchant,A.M.etal. (1998)An efficientrouteto human bispecific IgG.

    Nat. Biotechnol. 16, 677681

    21 de Kruif, J. and Logtenberg, T. (1996) Leucine zipper dimerized

    bivalent and bispecific scFv antibodies from a semi-synthetic antibody

    phage display library. J. Biol. Chem. 271, 76307634

    22 Kostelny, S.A.etal. (1992)Formation of a bispecificantibodyby theuse

    of leucine zippers. J. Immunol. 148, 15471553

    23 Muller, K.M.etal. (1998) Thefirst constant domain(C(H)1 andC(L)) of

    an antibody used as heterodimerization domain for bispecific

    miniantibodies. FEBS Lett. 422, 259264

    24 Holliger, P. et al. (1993) Diabodies: small bivalent and bispecific

    antibody fragments. Proc. Natl. Acad. Sci. U. S. A. 90, 64446448

    25 Kipriyanov, S.M. et al. (1999) Bispecific tandem diabody for tumor

    therapy with improved antigen binding and pharmacokinetics.J. Mol.

    Biol. 293, 4156

    26 Perisic, O. et al. (1994) Crystal structure of a diabody, a bivalent

    antibody fragment. Structure 2, 12171226

    27 Carmichael, J.A. et al. (2003) The crystal structure of an anti-CEA

    scFv diabody assembled from T84.66 scFvs in V(L)-to-V(H) orien-

    tation: implications for diabody flexibility. J. Mol. Biol. 326, 341351

    28 Zhu, Z. et al. (1996) High level secretion of a humanized bispecific

    diabody from Escherichia coli. Biotechnology 14, 19219629 Mallender, W.D. and Voss, E.W. Jr (1994) Construction, expression,

    and activity of a bivalent bispecific single-chain antibody. J. Biol.

    Chem. 269, 199206

    30 Gruber, M. et al. (1994) Efficient tumor cell lysis mediated by a

    bispecific single chain antibody expressed in Escherichia coli.

    J. Immunol. 152, 53685374

    31 Kipriyanov, S.M. et al. (2003) Effect of domain order on the activity of

    bacteriallyproducedbispecific single-chain Fv antibodies.J.Mol.Biol.

    330, 99111

    32 Mack, M. et al. (1995) A small bispecific antibody construct expressed

    as a functional single-chain molecule with high tumor cell cytotoxicity.

    Proc. Natl. Acad. Sci. U. S. A. 92, 70217025

    33 De Jonge,J.etal. (1998)In vivo retargeting of Tcell effector function by

    recombinant bispecific single chain Fv (anti-CD3 x anti-idiotype)

    induces long-term survival in the murine BCL1 lymphoma model.

    J. Immunol. 161, 14541461

    34 Jost, C.R.et al. (1996) A single-chain bispecific Fv2 molecule produced

    in mammalian cells redirects lysis by activated CTL. Mol. Immunol.

    33, 211219

    35 Shen, L. et al. (1987) Polymorphonuclear leukocyte function triggered

    through the high affinity Fc receptor for monomeric IgG. J. Immunol.

    139, 534538

    36 Valerius, T. et al. (1993) Involvement of the high-affinity receptor for

    IgG (Fc gamma RI; CD64) in enhanced tumor cell cytotoxicity of

    neutrophils during granulocyte colony-stimulating factor therapy.Blood 82, 931939

    37 Goldstein, J.etal. (1997) Cytolytic and cytostatic propertiesof an anti-

    human Fc gammaRI (CD64) x epidermal growth factor bispecific

    fusion protein. J. Immunol. 158, 872879

    38 Heijnen, I.A. et al. (1997) Generation of HER-2/neu-specific cytotoxic

    neutrophils in vivo: efficient arming of neutrophils by combined

    administration of granulocyte colony-stimulating factor and Fcgamma

    receptor I bispecific antibodies. J. Immunol. 159, 56295639

    39 Honeychurch, J. et al. (2000) Therapeutic efficacy of FcgammaRI/

    CD64-directed bispecific antibodies in B-cell lymphoma. Blood 96,

    35443552

    40 Curnow, R.T. (1997) Clinical experience with CD64-directed immu-

    notherapy. An overview. Cancer Immunol. Immunother. 45, 210215

    41 Posey, J.A. et al. (1999) A pilot trial of GM-CSF and MDX-H210 in

    patients with erbB-2-positive advanced malignancies. J. Immunother.

    22, 37137942 Valone, F.H. et al. (1995) Phase Ia/Ib trial of bispecific antibody MDX-

    210 in patients with advanced breast or ovarian cancer that

    overexpresses the proto-oncogene HER-2/neu. J. Clin. Oncol. 13,

    22812292

    43 James, N.D. et al. (2001) A phase II study of the bispecific antibody

    MDX-H210 (anti-HER2 x CD64) with GM-CSF in HER2 advanced

    prostate cancer. Br. J. Cancer 85, 152156

    44 Valone, F.H.etal. (1995) Clinical trials of bispecific antibody MDX-210

    in women with advanced breast or ovarian cancer that overexpresses

    HER-2/neu. J. Hematother. 4, 471475

    45 Monteiro,R.C.and Van De Winkel, J.G. (2003)IgA Fc receptors.Annu.

    Rev. Immunol. 21, 177204

    46 Deo, Y.M. et al. (1998) Bispecific molecules directed to the Fc receptor

    for IgA (Fc alpha RI, CD89) and tumor antigens efficiently promote

    cell-mediated cytotoxicity of tumor targets in wholeblood.J. Immunol.

    160, 1677168647 Weiner, L.M. et al. (1995) Phase I trial of 2B1, a bispecific monoclonal

    antibody targeting c-erbB-2 and Fc gamma RIII. Cancer Res. 55,

    45864593

    48 Weiner, L.M.etal. (1996) Humanneutrophil interactions of a bispecific

    monoclonal antibody targeting tumor and human Fc gamma RIII.

    Cancer Immunol. Immunother. 42, 141150

    49 McCall, A.M. et al. (2001) Increasing the affinity for tumor antigen

    enhances bispecific antibody cytotoxicity. J. Immunol. 166,

    61126117

    50 Hombach, A.etal. (1993) A CD16/CD30bispecific monoclonal antibody

    induces lysis of Hodgkins cells by unstimulated natural killer cells in

    vitro and in vivo. Int. J. Cancer 55, 830836

    51 Hartmann, F. et al. (1997) Treatment of refractory Hodgkins disease

    with an anti-CD16/CD30 bispecific antibody. Blood 89, 20422047

    52 Hartmann, F. et al. (2001) Anti-CD16/CD30 bispecific antibody

    treatment for Hodgkins disease: role of infusion schedule andcostimulation with cytokines. Clin. Cancer Res. 7, 18731881

    53 Titus, J.A. et al. (1987) Human T cells targeted with anti-T3 cross-

    linked to antitumor antibody prevent tumor growth in nude mice.

    J. Immunol. 138, 40184022

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004 243

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/
  • 8/8/2019 16504266 Review TRENDS in Biotechnology Vol22 No5 May 2004 A

    7/7

    54 Weiner, G.J. and Hillstrom, J.R. (1991) Bispecific anti-idiotype/anti-

    CD3 antibody therapy of murine B cell lymphoma. J. Immunol. 147,

    40354044

    55 Demanet, C. et al. (1996) Bispecific antibody-mediated immunother-

    apy of the BCL1 lymphoma: increased efficacy with multiple injections

    and CD28-induced costimulation. Blood 87, 43904398

    56 Renner, C.etal. (1994) Cureof xenografted humantumors by bispecific

    monoclonal antibodies and human T cells. Science 264, 833835

    57 Kipriyanov, S.M.etal. (1998) Bispecific CD3 x CD19 diabody for T cell-

    mediated lysis of malignant human B cells. Int. J. Cancer 77, 763772

    58 Cochlovius, B. et al. (2000) Cure of Burkitts lymphoma in severe

    combined immunodeficiency mice by T cells, tetravalent CD3 x CD19

    tandem diabody, and CD28 costimulation. Cancer Res. 60, 43364341

    59 Nitta, T. et al. (1990) Preliminary trial of specific targeting therapy

    against malignant glioma. Lancet 335, 368371

    60 Canevari, S.etal. (1995) Regression of advanced ovarian carcinoma by

    intraperitoneal treatment with autologous T lymphocytes retargeted

    by a bispecific monoclonal antibody. J. Natl. Cancer Inst. 87,

    14631469

    61 Manzke, O. et al. (2001) Locoregional treatment of low-grade B-cell

    lymphoma with CD3xCD19 bispecific antibodies and CD28 costimula-

    tion. I. Clinical phase I evaluation. Int. J. Cancer 91, 508515

    62 Marme, A. et al. (2002) Intraperitoneal bispecific antibody (HEA125x-

    OKT3) therapy inhibits malignant ascites production in advanced

    ovarian carcinoma. Int. J. Cancer 101, 183189

    63 Bolhuis, R.L. et al. (1992) Adoptive immunotherapy of ovarian

    carcinoma with bs-MAb-targeted lymphocytes: a multicenter study.

    Int. J. Cancer Suppl. 7, 7881

    64 Canevari, S. et al. (1997) Approaches to implement bispecific antibody

    treatment of ovarian carcinoma. Cancer Immunol. Immunother. 45,

    187189

    65 De Gast, G.C. et al. (1995) Clinical experience with CD3 x CD19

    bispecific antibodies in patients with B cell malignancies.

    J. Hematother. 4, 433437

    66 Kroesen, B.J. et al. (1994) Phase I study of intravenously applied

    bispecific antibody in renal cell cancer patients receivingsubcutaneous

    interleukin 2. Br. J. Cancer 70, 652661

    67 Wild, M.K. et al. (1999) Tumor therapy with bispecific antibody: thetargeting and triggering steps can be separated employing a CD2-

    based strategy. J. Immunol. 163, 20642072

    68 Segal, D.M. et al. (1995) Alternative triggering molecules and single

    chain bispecific antibodies. J. Hematother. 4, 377382

    69 Edberg, J.C. et al. (1989) Immune adherence and the processing of

    soluble complement-fixing antibody/DNA immune complexes in mice.

    Clin. Immunol. Immunopathol. 51, 118132

    70 Reinagel, M.L. and Taylor, R.P. (2000) Transfer of immune complexes

    from erythrocyte CR1 to mouse macrophages. J. Immunol. 164,

    19771985

    71 Blok, V.T. et al. (1998) A bispecific monoclonal antibody directed

    against both the membrane-bound complement regulator CD55 and

    the renal tumor-associated antigen G250 enhances C3 deposition and

    tumor cell lysis by complement. J. Immunol. 160, 34373443

    72 French, R.R. et al. (1995) Delivery of the ribosome-inactivating

    protein, gelonin, to lymphoma cells via CD22 and CD38 using

    bispecific antibodies. Br. J. Cancer 71, 986994

    73 Ford, C.H. et al. (2001) Bispecific antibody targeting of doxorubicin to

    carcinoembryonic antigen-expressing colon cancer cell lines in vitro

    and in vivo. Int. J. Cancer 92, 851855

    74 Zhu, H. et al. (1998) Tumor pretargeting for radioimmunodetection

    and radioimmunotherapy. J. Nucl. Med. 39, 6576

    75 Dorvillius, M. et al. (2002) Targeting of human breast cancer by a

    bispecific antibody directed against two tumour-associated antigens:

    ErbB-2 and carcinoembryonic antigen. Tumour Biol. 23, 337347

    76 Loffler, A. et al. (2000) A recombinant bispecific single-chain antibody,

    CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity

    by unstimulated T lymphocytes. Blood 95, 20982103

    77 Kufer, P. et al. (1997) Construction and biological activity of a

    recombinant bispecific single-chain antibody designed for therapy of

    minimal residual colorectal cancer. Cancer Immunol. Immunother. 45,

    193197

    78 Mack, M. et al. (1997) Biologic properties of a bispecific single-chain

    antibody directed against 17-1A (EpCAM) and CD3: tumor cell-

    dependent T cell stimulation and cytotoxic activity. J. Immunol. 158,

    39653970

    79 Irvine, D.J. (2003) Function-specific variations in the immunological

    synapses formed by cytotoxic Tcells.Proc.Natl. Acad. Sci. U. S. A.100,

    1373913740

    80 Dreier, T. et al. (2002) Extremely potent, rapid and costimulation-

    independent cytotoxic T-cell response against lymphoma cells cata-

    lyzedby a single-chain bispecific antibody.Int. J. Cancer 100, 690697

    81 Dreier, T. et al. (2003) T cell costimulus-independent and very

    efficacious inhibition of tumor growth in mice bearing subcutaneous

    or leukemic human B cell lymphoma xenografts by a CD19-/CD3-

    bispecific single-chain antibody construct. J. Immunol. 170,43974402

    82 Loffler, A. et al. (2003) Efficient elimination of chronic lymphocytic

    leukaemia B cells by autologous T cells with a bispecific anti-CD19/

    anti-CD3 single-chain antibody construct. Leukemia 17, 900909

    83 Baeuerle, P.A. et al. (2003) Bispecific antibodies for polyclonal T-cell

    engagement. Curr. Opin. Mol. Ther. 5, 413419

    84 Garrido, F. and Algarra, I. (2001) MHC antigens and tumor escape

    from immune surveillance. Adv. Cancer Res. 83, 117158

    TIBTEC making the most of your personal subscription

    High-quality printouts (from PDF files)Links to other articles, other journals and cited software and databases

    All you have to do is:

    Obtain your subscription key from the address label of your print subscription.Then go to http://www.trends.com, click on the Claim online access button and select

    Trends in Biotechnology.

    You will see a BioMedNet login screen.Enter your BioMedNet username and password. If you are not already a BioMedNet member, please click on the Register button.

    Once registered, you will be asked to enter your subscription key. Following confirmation, you will have full access toTrends in Biotechnology.

    If you obtain an error message please contact Customer Services ([email protected]) stating your subscription key andBioMedNet username and password. Please note that you only need to enter your subscription key once; BioMedNet remembersyour subscription. Institutional online access is available at a premium. If your institute is interested in subscribing online, please

    ask them to contact [email protected].

    Review TRENDS in Biotechnology Vol.22 No.5 May 2004244

    www.sciencedirect.com

    http://www.sciencedirect.com/http://www.sciencedirect.com/