throbber
R E V I E W S
`
`THERAPEUTIC ANTIBODIES FOR
`HUMAN DISEASES AT THE DAWN OF
`THE TWENTY-FIRST CENTURY
`
`Ole Henrik Brekke* and Inger Sandlie ‡
`
`Antibodies are highly specific, naturally evolved molecules that recognize and eliminate
`pathogenic and disease antigens. The past 30 years of antibody research have hinted at
`the promise of new versatile therapeutic agents to fight cancer, autoimmune diseases and
`infection. Technology development and the testing of new generations of antibody reagents
`have altered our view of how they might be used for prophylactic and therapeutic purposes.
`The therapeutic antibodies of today are genetically engineered molecules that are designed
`to ensure high specificity and functionality. Some antibodies are loaded with toxic modules,
`whereas others are designed to function naturally, depending on the therapeutic application.
`In this review, we discuss various aspects of antibodies that are relevant to their use as as
`therapeutic agents.
`
`IMMUNOGLOBULIN DOMAIN
`Compactly folded globular units
`of approximately 110 amino
`acids that comprise immuno-
`globulin heavy and light chains.
`
`CYTOKINES
`A class of small proteins
`released by one cell that affects
`the physiology of other cells
`locally and systemically in a
`particular fashion through
`binding to a specific receptor.
`
`Antibody structure
`The typical antibody — or immunoglobulin (Ig) —
`consists of two antigen-binding fragments (Fabs),
`which are linked via a flexible region (the hinge) to a
`constant (Fc) region (FIG. 1). This structure comprises
`two pairs of polypeptide chains, each pair containing a
`heavy and a light chain of different sizes. Both heavy
`and light chains are folded into IMMUNOGLOBULIN DOMAINS.
`The ‘variable domains’ in the amino-terminal part of the
`molecule are the domains that recognize and bind anti-
`gens; the rest of the molecule is composed of ‘constant
`domains’ that only vary between Ig classes (BOX 1). The
`Fc portion of the Ig serves to bind various effector mole-
`cules of the immune system, as well as molecules that
`determine the biodistribution of the antibody.
`
`*Affitech AS, Gaustadalleen
`21, N-0349 Oslo, Norway.
`‡Department of Biology,
`University of Oslo, N-0349,
`Norway. Correspondence
`to O.H.B. e-mail:
`o.h.brekke@affitech.com
`doi:10.1038/nrd984
`
`Mechanisms of in vivo action
`In antibody-based therapies, the goal is to eliminate or
`neutralize the pathogenic infection or the disease target,
`for example, bacterial, viral or tumour targets.
`Therapeutic antibodies can function by three principal
`modes of action: by blocking the action of specific mole-
`cules, by targeting specific cells or by functioning as
`
`signalling molecules. The blocking activity of therapeutic
`antibodies is achieved by preventing growth factors,
`CYTOKINES or other soluble mediators reaching their tar-
`get receptors, which can be accomplished either by the
`antibody binding to the factor itself or to its receptor.
`Targeting involves directing antibodies towards specific
`populations of cells and is a versatile approach; anti-
`bodies can be engineered to carry effector moieties,
`such as enzymes, toxins, radionuclides, cytokines or
`even DNA molecules, to the target cells, where the
`attached moiety can then exert its effect (for example,
`toxins or radionuclides can eliminate target cancer
`cells). The natural EFFECTOR FUNCTIONS of antibodies are
`associated with binding to Fc receptors or binding
`to complement proteins and inducing COMPLEMENT-
`DEPENDENT CYTOTOXICITY (CDC). Targeting antibodies can
`retain such effector functions intact or they can be
`abolished during the design of the antibody, depending
`on the therapeutic strategy. The signalling effect of anti-
`bodies is predicated on either inducing crosslinking of
`receptors that are, in turn, connected to mediators of
`cell division or PROGRAMMED CELL DEATH, or directing them
`towards specific receptors to act as agonists for the
`
`52 | JANUARY 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2002 Nature Publishing Group
`
`Pfizer v. Genentech
`IPR2017-01488
`Genentech Exhibit 2053
`
`

`

`R E V I E W S
`
`that are responsible for antigen binding are inserted into
`the human variable-domain framework. The ability to
`manipulate antibodies into more human variants
`finally made antibodies useful for clinical use. With the
`isolation of genes encoding human variable regions,
`their successful expression in Escherichia coli 8,9 and the
`introduction of phage-display technology10, the task of
`selecting fully human variable domains has been
`greatly simplified.
`Selection from phage-display libraries of human
`antibody fragments is today the most used and well-
`established technology for the development of new
`human antibodies (FIG. 3a). Another approach is to use
`mice that are transgenic for the human Ig locus11.
`Immunization of such a transgenic mouse results in a
`human antibody response, from which hybridomas that
`produce human antibodies can be generated (FIG. 3b).
`Today there are approximately 200 antibodies in
`clinical trials and the US Food and Drug Admin-
`istration has approved several antibodies against
`cancer12,13, transplant rejection14, rheumatoid arthritis
`and Crohn’s disease15,16, and antiviral prophylaxis17
`(TABLE 1). So far, 20% of all biopharmaceuticals in clini-
`cal trials are monoclonal antibodies (examples are
`shown in TABLE 2,and see clinical trials web site in online
`links box), making this the second largest biopharmaceu-
`tical product category after vaccines. As the development
`of potential new therapeutic agents into commercial
`products takes about 10 years, the FDA-approved anti-
`bodies, and some of those in the end-stages of develop-
`ment pipelines, are chimeric or humanized antibodies
`that were developed with early antibody engineering
`technologies. The more recently developed reagents,
`on the other hand, are completely human antibodies
`that are derived from phage antibody libraries and
`transgenic mice17–20.
`
`Antibodies against infectious agents
`In the pre-antibiotic era, SERUM THERAPY was widely used
`as treatment for infectious diseases such as anthrax,
`smallpox, meningitis and the plague. With the introduc-
`tion of vaccines and antibiotics, the use of serum therapy
`declined. However, with the reduction and eradication
`of certain diseases (for example, smallpox), which has
`had the result that populations are no longer routinely
`vaccinated against these infectious agents, and the
`increasing emergence of antibiotic-resistant bacteria,
`humans are susceptible to acute outbreaks of disease or
`to biological terrorism. Antibiotics are obviously ineffec-
`tive at eliminating viral infections, and the antiviral
`drugs in use today are often associated with a short
`serum half-life and resistance often emerges after
`repeated use21. Passive antibody serum therapy is today
`used merely in replacement therapy for patients with
`immune disorders, for POST-EXPOSURE PROPHYLAXIS against
`several viruses (for example, rabies, measles, hepatitis A
`and B, varicella and respiratory syncitial virus (RSV))
`and for toxin neutralization (for example, diphtheria,
`botulism and tetanus). Passive immunization has sub-
`stantial advantages over the administration of anti-
`microbial agents, including low toxicity and high specific
`
`Antigen binding
`
`VH
`
`VL
`
`CH
`1
`
`CL
`
`Fab
`
`CDR loops
`
`1
`
`3
`
`2
`
`N
`
`s
`
`s
`
`Complement and
`Fc receptor binding
`
`C
`
`CH
`
`2 C
`
`H
`3
`
`Fv
`
`Fc
`
`Figure 1 | The modular structure of immunoglobulins. This figure shows a single
`immunglobulin (Ig) molecule. All immunoglobulin monomers are composed of two identical light
`(L) chains and two identical heavy (H) chains. Light chains are composed of one constant domain
`(CL) and one variable domain (VL), whereas heavy chains are composed of three constant
`domains (CH1, CH2 and CH3) and one variable domain (VH). The heavy chains are covalently linked
`in the hinge region and the light chains are covalently linked to the heavy chain. The variable
`domains of both the heavy and light chains compose the antigen-binding part of the molecule,
`termed Fv. Within the variable domains there are three loops designated complementarity-
`determining regions (CDRs) 1, 2 and 3, which confer the highest diversity and define the
`specificity of antibody binding. The Fc portion is glycosylated and contains the sites for interaction
`with effector molecules, such as the C1 complex of the complement system and a variety of Fc
`receptors including the neonatal Fc receptor (FcRn).
`
`activation of specific cell populations. Another
`approach is to use antibodies as delivery vehicles for
`DNA and, more recently, to deliver antigens to certain
`immune cells that present processed antigenic pep-
`tides, or epitopes, to T cells, to activate a specific
`immune response against that antigen.
`
`The development of therapeutic antibodies
`Target specificity in the treatment and prophylaxis of
`diseases such as infection, cancer and autoimmune dis-
`orders has become more viable through the develop-
`ment of monoclonal antibodies. The mouse HYBRIDOMA
`technology described by Köhler and Milstein was an
`important step in the development of antibody technol-
`ogy and paved the way for the emergence of therapeutic
`monoclonal antibodies1. During the 1980s, resources
`were directed towards the evaluation of the in vivo use
`of mouse monoclonal antibodies in humans, aimed at
`both imaging and therapy2. Mouse monoclonal anti-
`bodies were shown to have limited use as therapeutic
`agents because of a short serum half-life, an inability to
`trigger human effector functions and the production of
`human antimouse-antibodies3 (the HAMA response).
`In an attempt to reduce the immunogenicity of mouse
`antibodies, genetic engineering was used to generate
`chimeric antibodies, that is, antibodies with human
`constant regions and mouse variable regions4,5.
`However, although chimeric antibodies were perceived
`as less foreign, and therefore less immunogenic, than
`mouse monoclonal antibodies, human anti-chimeric
`antibody responses (HACAs) have nonetheless been
`observed6. Further minimization of the mouse com-
`ponent of antibodies was achieved through CDR
`(complementarity-determining region) grafting7 (FIG. 2).
`In such ‘humanized’ antibodies, only the CDR loops
`
`EFFECTOR FUNCTIONS
`The antigen-elimination
`processes mediated by
`immunoglobulins and initiated
`by the binding of effector
`molecules to the Fc part of the
`immunoglobulin. The common
`effector functions are
`complement-dependent
`cytotoxicity (CDC), phagocytosis
`and antibody-dependent cellular
`cytotoxicity (ADCC).
`
`COMPLEMENT-DEPENDENT
`CYTOTOXICITY
`Once bound to antigen, both IgM
`and IgG can trigger a sequence of
`reactions by which serum
`proteins called complement
`factors are cleaved. One of the
`results is destruction of the target
`cell through complement-
`dependent cytotoxicity.
`
`PROGRAMMED CELL DEATH
`Programmed cell death infers
`that cells are determined to die at
`a specific stage of development
`or having received a specific
`signal. The process is known as
`apoptosis. The cells shrivel and
`are engulfed by nearby
`phagocytic cells without eliciting
`any inflammatory response.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 2 | JANUARY 2003 | 5 3
`
`© 2002 Nature Publishing Group
`
`

`

`R E V I E W S
`
`Box 1 | Immunoglobulin classes
`
`Antibodies belong to either one of five immunoglobulin (Ig) classes: IgA, IgD, IgE, IgG
`or IgM. Each class has a distinct structure and biological activity. Some of the classes
`are further divided into subclasses — for example, there are four IgG subclasses and
`two IgA subclasses. IgM is the first antibody to be produced in an immune response
`and forms a pentameric complex comprised of Ig monomers. IgA is the main class of
`antibody in external secretions, where it is found as a dimer that protects the body’s
`mucosal surfaces from infection; it is also found as a monomer in serum. IgD is the
`main antibody on the surface of B cells. IgE is found bound to cells that secrete
`histamines after antigen binding. IgG is the main antibody in serum. The IgG class is
`the most stable and has a serum half-life of 20 days, whereas IgM and IgA persist for
`only 5–8 days. Both IgM and IgG can mediate complement fixation, whereas only IgG
`can promote antibody-dependent cellular cytotoxicity (ADCC). IgA, and to a certain
`degree IgM, can mediate trancytosis to mucosal surfaces, whereas only IgG can be
`transported across placenta for fetal protection.
`
`activity, as well as an immediate effect compared with
`vaccines and even antibiotics. Given these issues,
`biotechnology companies and institutions working
`within the field of infectious disease protection are
`certain to direct efforts towards developing highly
`effective and functional antibody candidates against
`specific disease targets.
`The isolation of protective toxin-neutralizing
`human monoclonal antibodies was described in 1993
`in a study in which several human monoclonal anti-
`bodies against tetanus toxin were isolated and a pro-
`tective effect against tetanus toxin was observed22.
`However, when combinations of these specific mono-
`clonal antibodies were administered, an extra potent
`(that is, synergistic) effect was observed. Such an effect
`has also recently been described in the neutralization
`
`Mouse hybridoma
`
`In vitro antibody libraries
`Transgenic mouse
`Human hybridomas
`
`Mouse
`
`Chimeric
`
`Humanized
`
`Human
`
`Genetic engineering
`V gene cloning
`CDR grafting
`Eukaryotic expression
`
`Figure 2 | Antibody engineering. Mouse hybridoma technology generates mouse monoclonal
`antibodies. Genetic engineering has fostered the generation of chimeric, humanized and human
`antibodies. Cloning of mouse variable genes into human constant-region genes generates
`chimeric antibodies. Humanized antibodies are generated by the insertion of mouse
`complementarity-determining regions (CDRs) onto human constant and variable domain
`frameworks; however, additional changes in the framework regions have, in several cases, been
`shown to be crucial in maintaining identical antigen specificity75,76. Fully human antibodies can be
`generated by the selection of human antibody fragments from in vitro libraries (see BOX 2 and
`FIG. 3a), by transgenic mice (FIG. 3b) and through selection from human hybridomas.
`
`of botulinum neurotoxin. The anti-botulinum toxin
`antibodies were derived from different phage-display
`libraries obtained from humans or immunized mice
`(J. Marks, presented at Cambridge Healthtech
`Institute conference on Recombinant Antibodies,
`Cambridge, Massachusetts, USA, 24–25 April 2002).
`Recently, Maynard and colleagues described high-
`affinity antibodies against Bacillus anthracis23. These
`antibodies were fragments derived from variable-
`chain genes of a mouse monoclonal antibody and
`expressed in E. coli as single-chain Fv fragments
`(scFvs). By administering the antibody fragments to
`mice before injection of anthrax toxin, protection
`against the toxin was observed. Protective human
`antibodies against Shiga-toxin1 were recently isolated
`by the immunization of transgenic mice20. The panel
`of ten different antibodies of the IgM and IgG1 class
`that showed specificities to different subunits of the
`toxin effectively neutralized the toxin.
`Compared with toxin neutralization, the use of
`antibodies in the prevention of viral disease is a more
`complex prospect. Palivizumab (Synagis; MedImmune
`Inc) is a humanized IgG1 monoclonal antibody
`approved for the prevention of RSV infections in high-
`risk infants17,24. Palivizumab was the first monoclonal
`antibody approved for an infectious pathogen and is,
`so far, the only antiviral monoclonal antibody in clini-
`cal use. The development of sevirumab (Protovir;
`Protein Design Labs) — a human anti-cytomegalo-
`virus (CMV) antibody — was, by contrast, halted in
`Phase III clinical trials as a supplemental treatment for
`CMV-induced retinitis because of a lack of evidence of
`efficacy. The elimination of a viral infection requires
`that a number of events occur, including inhibition of
`cell infection, mediation of cell killing of infected cells,
`inhibition of viral replication, inhibition of viral
`release and inhibition of cell–cell transmission25,26.
`Within the multitude of antibody specificities gener-
`ated in the typical human polyclonal response, it is
`likely that one or several effective antibodies against
`one or more of the particular processes listed will be
`found. Some of these antibodies bind neutralizing epi-
`topes, whereas others bind non-neutralizing epitopes.
`The overall outcome might indeed be effective protec-
`tion due to the combination of the blocking, neutral-
`izing and eliminating effect of human antibodies.
`Recently, XTL Pharmaceuticals reported on Phase I/II
`clinical trials with two human monoclonal antibodies
`against hepatitis B virus28. The two monoclonal anti-
`bodies were combined with lamivdudine (Epivir-
`HBV; GlaxoWellcome) — an antiviral drug that
`inhibits DNA replication — and showed significant
`reduction in serum viral titre. If monoclonal antibod-
`ies are to be used as prophylactics or therapeutics
`against infectious diseases, it is likely that their efficacy
`will be increased when a polyclonal passive human
`serum therapy is mimicked — that is, when a pool of
`highly specific and high-affinity monoclonal antibod-
`ies are administered. There are, however, limitations
`to this approach, including the production costs asso-
`ciated with manufacturing intact human antibodies.
`
`54 | JANUARY 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2002 Nature Publishing Group
`
`

`

`R E V I E W S
`
`Figure 3 | In vitro and in vivo human antibody techniques exemplified by phage display and transgenic mouse
`technologies. a | The in vitro process is based on panning the library of antibodies against an immobilized target. The non-
`binding phage antibodies are washed away and the recovered antibodies are amplified by infection in Escherichia coli. The
`selection rounds are subsequently repeated until the desired specificity is obtained. The antibody format for screening is either
`Fab or single-chain Fv. The expression of antibodies in E. coli and recent developments in screening technologies77 have made
`it possible to screen tens of thousands of clones for specificity. The antibody fragments themselves can be used as therapeutic
`agents as discussed in this review, but they can also be converted into intact immunoglobulins by the cloning of the variable
`genes into plasmids incorporating the constant-region genes of immunoglobulins. The genes are transfected into cell lines and
`therefore produce fully human immunoglobulins. b | The in vivo process is based on the immunization of a transgenic mouse.
`The mouse has been genetically engineered and bred for the expression of human immunoglobulins. The B cells harvested after
`immunization can be immortalized by fusion with a myeloma cell line, as in traditional hybridoma technology. The hybridomas
`can then be screened for specific antibodies.
`
`A possible solution to this problem is to instead use
`blocking antibody fragments synthesized in E. coli,
`which could be used to combat, for example, viral
`infections. A further difficulty in developing such
`multi-antibody therapeutics might arise from regula-
`tory concerns regarding the administration of multiple
`monoclonal antibodies. The experience obtained
`from XTL Pharmaceuticals’ use of combined antibod-
`ies might be an important step in resolving — or
`exacerbating — these concerns.
`
`Anti-inflammatory antibodies
`Antibodies with high specificity and affinity can be
`developed to bind specific cytokines or their recep-
`tors. In both cases, the purpose is to inhibit the detri-
`mental effect of the cytokine. Cytokines associated
`
`with inflammation and autoimmunity include
`tumour-necrosis factor-α (TNF-α), interleukins and
`complement proteins. Modulation of
`immune-
`responses, such as immune-cell depletion by the target-
`ing of antibodies to cell-surface receptors, for example,
`CD20 and CD4 on B or T cells, has also been shown as
`a viable therapeutic strategy in autoimmune diseases.
`Together with cancer, inflammatory and autoimmune
`diseases are an important focus for companies devel-
`oping antibody therapies. In patients with rheumatoid
`arthritis (RA), TNF-α accumulates in the joints and
`contributes to the inflammation and joint destruction
`that is associated with the disease. Marketed products
`directed towards the regulation of TNF-α include
`the soluble TNF-α receptor eternacept (Enbrel;
`Amgen Inc/Wyeth) and the antibody infliximab
`
`HYBRIDOMA
`An antibody-secreting B-cell line
`that is generated by fusing
`splenic-derived B cells with a
`plasmacytoma. A hybridoma
`produces the same antibody as
`the parent B cell and divides and
`grows in culture like the parent
`cancer cell. The antibody
`produced is monoclonal.
`
`SERUM THERAPY
`The treatment of an infectious
`disease with the serum from an
`immunized animal or individual,
`and which contains antibody.
`
`POST-EXPOSURE PROPHYLAXIS
`A treatment that is designed
`to protect an individual against
`a disease agent to which the
`individual has been recently
`exposed.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 2 | JANUARY 2003 | 5 5
`
`© 2002 Nature Publishing Group
`
`

`

`R E V I E W S
`
`Table 1 | Approved monoclonal antibodies
`Product
`Year
`Type of molecule
`approved
`1986
`1994
`
`OKT-3
`ReoPro
`
`Panorex
`(Germany only)
`Rituxan
`Zenapax
`
`Herceptin
`
`Remicade
`Simulect
`
`Synagis
`
`Mylotarg
`
`Campath
`
`Zevalin
`
`1995
`
`1997
`1997
`
`1998
`
`1998
`1998
`
`1998
`
`2000
`
`2001
`
`2002
`
`Murine (anti-CD3)
`Chimeric
`(anti-platelet gpIIb/IIIa)
`Murine (anti-EpCAM)
`
`Chimeric (anti-CD20)
`Humanized
`(anti-IL-2 receptor)
`Humanized
`(anti-ERBB2)
`Chimeric (anti-TNF-α)
`Chimeric
`(anti-IL-2 receptor)
`Humanized
`(anti-F-protein)
`Humanized
`(anti-CD33)
`Humanized
`(anti-CD52)
`Mouse (anti-CD20)
`
`2002
`
`Xolair
`(Australia only)
`Adapted and updated from REF. 81.
`
`Humanized
`(anti-IgE Fc)
`
`(Remicade; Centocor Inc). Infliximab is a chimeric
`antibody with mouse variable domains and human
`constant domains of the IgG1 subclass. There are a
`number of anti-TNF-α antibodies in clinical trials,
`including CDP571 and CDP870 (Celltech Plc) and
`adalimumab (D2E7; Abbot Laboratories/Cambridge
`Antibody Technology). Adalimumab is the first
`phage-display-derived human antibody brought into
`the clinic, and was generated by ‘guided selection’
`using a mouse monoclonal antibody28. The method is
`based on the selection of a human variable-domain
`repertoire coupled to one of the original mouse vari-
`able domains, so as to ‘guide’ the human variable-
`domain repertoire towards the same specificity as the
`original mouse variable domains. The antibody is
`affinity optimized by iterative rounds of selection and
`mutagenesis (FIG. 3; BOX 2, 3). Adalimumab has com-
`pleted Phase III clinical trials and is currently in regis-
`tration for FDA approval. Recently, the anti-CD20
`antibody rituximab (Rituxan; IDEC Pharmaceuticals;
`see below) completed Phase II clinical trials in RA and
`showed promising results compared with the anti-
`TNF-α antibodies adalimumab and infliximab29, indi-
`cating that depletion of B cells might be an effective
`treatment in RA and other autoimmune diseases30.
`Eculizumab (5G1.1; Alexion Pharmaceuticals) is a
`humanized monoclonal antibody that prevents the
`cleavage of human complement component C5 into
`its pro-inflammatory components31, whereas J695
`(Cambridge Antibody Technology) is a human anti-
`body derived from a phage-display library against
`interleukin-2 (see online links box). Both antibodies
`
`Disease indication
`
`Company
`
`Organ transplant rejection
`Coronary intervention
`and angioplasty
`Colorectal cancer
`
`Ortho Biotech Products LP
`Centocor Inc
`
`Centocor Inc
`
`Non-Hodgkin’s lymphoma
`Refractory unstable angina
`
`IDEC Pharmaceuticals Corp
`Centocor Inc
`
`Metastatic breast cancer
`
`Genentech Inc
`
`Crohn’s disease
`Kidney transplant rejection
`
`Centocor Inc
`Novartis AG
`
`Respiratory syncitial viral
`disease
`Chemotherapy for acute
`myeloid leukemia
`B-cell chronic lymphocytic
`leukemia
`B-cell non-Hodgkin’s
`lymphoma
`Allergy
`
`MedImmune Inc
`
`Celltech Group plc/Wyeth
`
`Millennium Pharmaceuticals/
`Ilex Oncology Inc
`IDEC Pharmaceuticals Corp
`
`Tanox Inc/Genentech Inc/
`Novartis AG
`
`are in clinical trials at present and have shown poten-
`tial in the treatment of inflammatory diseases such as
`RA and nephritis.
`
`Antibodies targeting cancers
`In cancer therapy, the purpose of antibody adminis-
`tration is to induce the direct or indirect destruction of
`cancer cells, either by specifically targeting the tumour
`or the vasculature that nourishes the tumour. Indeed,
`new technologies for the panning of antibody libraries
`on intact cells have made it possible to isolate antibod-
`ies against novel and promising cancer-associated
`antigens32. However, the most common cancer targets
`are the carcinoembryonic antigen (CEA), which is
`associated with colorectal cancers, MUC1, epidermal
`growth factor receptor (EGFR) and ERBB2 (also
`known as HER2/neu, associated with lung and breast
`cancer) and CD20 on B cells, which is a marker for
`non-Hodgkin’s lymphoma (NHL). Examples of
`approved therapeutic antibodies directed towards
`ERBB2 and CD20 include the humanized IgG1 anti-
`body trastuzumab (Herceptin; Genentech) for breast
`cancer and the chimeric IgG1 antibody rituximab
`(Rituxan; IDEC Pharmaceuticals) for NHL.
`Today, specific antibody therapy is used in combi-
`nation with classical chemotherapy, but the remaining
`challenge is to develop specific and highly cytotoxic
`drugs against cancer cells. Recently, the FDA approved
`two new antibody chemotherapies: ibritumomab
`tiuxetan (Zevalin; IDEC Pharmaceuticals) and gem-
`tuzumab ozogamicin (Mylotarg; Wyeth). Ibritu-
`momab tiuxetan is a mouse anti-CD20 antibody
`
`56 | JANUARY 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2002 Nature Publishing Group
`
`

`

`R E V I E W S
`
`attached to the radioisotope 90yttrium that targets the
`surface of the mature B cells and B-cell tumours,
`inducing cellular damage in the target and neighbour-
`ing cells. Ibritumomab tiuxetan is the first radio-
`immunotherapeutic antibody approved by the FDA
`and is approved for use in the treatment of NHL. The
`antibody reacts with the same antigen as rituximab,
`but is a mouse antibody, which results in beneficial
`rapid clearance to decrease the undesired effect of total
`body irradiation. Gemtuzumab ozogamicin is a
`humanized monoclonal antibody that is linked to the
`antitumour agent calicheamicin, a bacterial toxin33.
`The antibody is targeted to CD33, which is expressed
`in about 90% of all acute myelogenic leukaemia
`(AML) cases, and has been approved for administra-
`tion to patients who have relapsed AML. Both ibritu-
`momab tiuxetan and gemtuzumab ozogamicin are
`examples of antibodies designed to specifically deliver
`their toxic load directly to cancer cells. Antibodies
`directed against major histocompatibility complex
`(MHC) class II proteins specifically target and elimi-
`nate cancer cells. The mouse monoclonal antibody
`Oncolym (Lym-1; Peregrine Pharmaceuticals)34 and
`the antibody Hu1D10 (Remitogen; Protein Design
`Labs)35 are two examples of such antibodies. Whereas
`
`one form of Oncolym is radiolabelled with 131iodine
`(REF. 36), the humanized IgG1 Hu1D10 eliminates cancer
`cells by virtue of its natural effector functions (FIG. 4 and
`see below), and also induces apoptosis. Recently, an anti-
`MHC class II human antibody derived from an antibody
`phage-display library was shown to induce apoptosis of
`activated lymphoid cells19. Vascular endothelial growth
`factor (VEGF) is a potent cytokine for angiogenesis (the
`formation of blood vessels). Inhibitory agents, including
`specific antibodies, have been developed to block VEGF-
`stimulated angiogenesis as a strategy to inhibit tumour
`growth. One example of such an antibody is the human-
`ized anti-VEGF antibody bevacizumab (Avastin; Genen-
`tech), which is in Phase III clinical trials for the treatment
`of breast cancer and colorectal cancer. Monoclonal anti-
`bodies are being continuously developed for cancer ther-
`apy, and most new antibody formats have demonstrated
`utility within this field.
`
`Naked antibodies
`Both trastuzumab and rituximab are ‘naked antibodies’,
`meaning that they do not have a radioisotope or toxin
`attached to them. The elimination of the target of these
`antibodies depends entirely on the recruitment of the
`body’s own effector mechanisms, namely complement
`
`Table 2 | A selection of antibodies in clinical development
`Product
`Stage
`Type of molecule
`Disease indication
`(2002)
`Phase II
`Phase II
`
`ABX-EGF
`ABX-IL8
`
`Human (anti-EGF-R)
`Human (anti-IL-8)
`
`Eculizumab
`(5G1.1)
`D2E7
`
`Phase IIb
`
`Humanized (anti-C5)
`
`Phase III
`
`Human (anti-TNF-α)
`
`CAT-152
`
`Phase II/III
`
`Human (anti-TGF-β2)
`
`J695
`
`Phase II
`
`Human (anti-IL-2)
`
`Antegren
`(natalizumab)
`
`Phase III
`
`Humanized
`(anti-α-4 integrin)
`
`Phase III
`(anti-VEGF)
`
`Humanized
`
`Non-small-cell lung cancer
`Pulmonary disease, chronic
`obstructive bronchitis
`Rheumatoid arthritis,
`lupus nephritis
`Rheumatoid arthritis
`
`Scarring following glaucoma
`surgery
`Autoimmune diseases
`including rheumatoid arthiritis
`
`Crohn’s disease,
`multiple sclerosis,
`inflammatory bowel disease
`Metastatic breast cancer,
`non-small-cell lung cancer
`
`Company
`
`Abgenix Inc/Immunex Corp
`Abgenix Inc/Immunex Corp
`
`Alexion Pharmaceutical Inc
`
`Cambridge Antibody
`Technology/
`Abbot Laboratories
`Cambridge Antibody
`Technology
`Cambridge Antibody
`Technology/Abbot/
`Wyeth Genetics Institute
`Elan Pharmaceuticals
`Corp/Biogen Inc
`
`Genentech Inc
`
`Avastin
`(rhuMAb-
`VEGF)
`IDEC-151
`(Clenoliximab)
`MEDI-507
`(Siplizumab)
`XTL 001
`(pairs of
`monoclonals)
`CDP870
`
`Phase II
`
`Primatized (anti-CD4)
`
`Rheumatoid arthritis
`
`IDEC Pharmaceuticals Corp
`
`Phase II
`
`Phase I/II
`
`Humanized
`(anti-CD2 Receptor)
`Human (anti-HBV)
`
`Suppresses NK and T-cell
`function
`Hepatitis B virus neutralization
`
`MedImmune Inc
`
`XTL Bioharmaceuticals Ltd
`
`Phase II
`
`Fab fragment
`(anti-TNF-α)
`Humanized
`(anti-TNF-α)
`Humanized
`Hu1D10
`(anti-MHC class II)
`(Remitogen)
`Adapted from REFS 34, 81 and selected company web sites.
`
`CDP571
`
`Phase III
`
`Phase II
`
`Rheumatoid arthritis
`
`Celltech Group plc
`
`Crohn’s disease
`
`Celltech Group plc
`
`Non-Hodgkin’s Lymphoma
`
`Protein Design Labs Inc
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 2 | JANUARY 2003 | 5 7
`
`© 2002 Nature Publishing Group
`
`

`

`R E V I E W S
`
`Box 2 | In vitro human antibody libraries
`
`The strategy of selecting antibodies from large repertoires is dependent on the coupling between genotype and
`phenotype — that is, the displayed protein (the antibody) carries its own encoding gene. As a result, the process
`enables the easy recovery of the DNA encoding the selected protein. The power of such antibody libraries lies in the
`fact that selection takes place in vitro, thereby making them excellent systems for isolating antibodies to certain drugs,
`potent toxins and haptens that would normally be impossible to raise within in vivo systems such as the mouse
`hybridoma technology.
`In phage display, the variable genes encoding the antibody variable domains are fused to genes encoding
`bacteriophage coat proteins. The most commonly used phage protein is the pIII protein located at the tip of the long,
`thin filamentous phage M13. The system is highly effective and is used to isolate single-chain Fv or Fab fragments with
`specificity to almost any kind of antigen. The plasmid encoding the variable genes is packaged within the viral capsid
`and the expressed antibody is presented on the bacteriophage surface. Phage libraries are made from the human naive
`repertoire (that is, the IgM and IgD class) from pre-immunized donors or totally synthetic variable genes. As the
`repertoire is created in vitro, the technology also generates high-affinity human antibodies against human proteins. The
`process is based on panning the library against an immobilized target in a test tube (FIG. 3a). The non-binding phage
`antibodies are washed away and the recovered antibodies are amplified by infection in E. coli. The selection rounds are
`subsequently repeated until the desired specificity is obtained.
`In messenger RNA (mRNA)-display technologies, such as the ribosomal display technology, antibody domains are
`linked to ribosomes attached to the mRNA that they are translating. This process takes place entirely in vitro by
`transcripton of a DNA library followed by in vitro translation of the mRNA library. The subsequently recovered mRNA
`transcripts are reverse transcribed and amplified by polymerase chain reaction (PCR) for the subsequent round of
`selection. The method has been successfully used for the affinity maturation and stability engineering of antibody
`fragments58,70. A similar technology called ProFusion is based on the covalent linkage of the synthesized proteins to the
`mRNA template via a puromycin linker71. Another potential display method applicable

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket