throbber
REVIEW
`
`Engineered antibodies now represent over 30% of biopharmaceuticals in clinical trials, as highlighted by recent
`approvals from the US Food and Drug Administration. Recombinant antibodies have been reduced in size, rebuilt
`into multivalent molecules and fused with , for example radionuclides, toxins, enzymes, liposomes and viruses. The
`emergence of recombinant technologies has revolutionized the selection, humanization and production of
`antibodies, superseding hybridoma technology and allowing the design of antibody-based reagents of any
`specificity and for very diverse purposes.
`
`Engineered antibodies
`
`The discovery of hybridoma technology
`by Kohler and Milstein1 in 1975 heralded
`a new era in antibody research and clini-
`cal development. Mouse hybridomas were
`the first reliable source of monoclonal antibodies and were de-
`veloped for a number of in vivo therapeutic applications (Table 1;
`see also FDA product approval information at http://www.
`fda.gov/cber/efoi/approve.htm). Throughout the 1990s, innova-
`tive recombinant DNA technology, including chimerization and
`humanization, enhanced the clinical efficiency of mouse anti-
`bodies and led to the recent wave of approvals by the FDA for
`therapeutic immunoglobulin (Ig) and Fab molecules (monova-
`lent antibody fragment produced by proteolysis) (Table 1 and
`Fig. 1; refs. 2,3). These developments have continued, and in
`2002 the FDA approved the first radiolabeled antibody for cancer
`immunotherapy (Zevalin)4.
`The list of approved antibody therapeutics against cancer
`and against viral and inflammatory diseases is growing
`rapidly (Table 1), with more than 30 antibodies in late-phase
`clinical trials2,3. Most recently, innovative structural designs
`have improved in vivo pharmacokinetics, expanded immune
`repertoires and permitted screening against refractory targets
`and complex proteome arrays, while new molecular evolu-
`tion strategies have enhanced affinity, stability and expres-
`sion levels. This review describes these emerging technologies
`and discusses the creation of a vast range of engineered, anti-
`body-based reagents that specifically target biomarkers of
`human health and disease1–3. We review antibodies designed
`as intact molecules and recombinant fragments and then
`focus on the latest technologies for attaching additional ther-
`apeutic payloads, such as radionuclides, drugs, enzymes and
`vaccine-inducing epitopes.
`
`Intact antibodies, humanization and de-immunization
`Intact antibodies provide high-specificity, high-affinity target-
`ing reagents and are usually multivalent (Fig. 1). Their simulta-
`neous binding to two adjacent antigens increases functional
`affinity and confers high retention times, for example, on cell
`surfaces. In addition, intact antibodies comprise Fc domains,
`which can be important for cancer immunotherapy through
`their abilities both to recruit cytotoxic effector functions2,5 and
`to extend the serum half-life, mediated by the neonatal Fc re-
`ceptor6. Unmodified mouse monoclonal antibodies formed the
`first wave of FDA-approved immunotherapeutic reagents
`(Table 1), although their in vivo applications were limited be-
`cause repeated administrations provoked an anti-mouse im-
`mune response2. Simple strategies have been developed to
`avoid, mask or redirect this human immune surveillance; these
`strategies include fusion of mouse variable regions to human
`constant regions as ‘chimeric’ antibodies, ‘de-immunization’
`by removal of T-cell epitopes and ‘humanization’ by grafting
`
`©2003 Nature Publishing Group http://www.nature.com/naturemedicine
`
`PETER J. HUDSON
`& CHRISTELLE SOURIAU
`
`mouse surface residues onto human ac-
`ceptor antibody frameworks (Table 1; see
`also Supplementary Note online)2.
`Modern alternative strategies now allow
`selection of fully human antibodies directly from natural or
`synthetic repertoires, including live transgenic mice producing
`purely human antibodies7. Human antibody-display libraries
`were used to transform a mouse antibody in vitro into a fully
`human derivative (D2E7), which is likely to be the first FDA-ap-
`proved fully human anti-inflammatory antibody. Many other
`fully human antibodies, including Efalizumab for the treat-
`ment of psoriasis, are in clinical evaluation (see http://www.
`fda.gov/cber/efoi/approve.htm).
`
`Design of ‘antibody fragments’ for unique clinical applications
`For cytokine inactivation, receptor blockade or viral neutraliza-
`tion, the Fc-induced effector functions are often unwanted and
`can be simply removed by proteolysis of intact antibodies to
`yield monovalent Fab fragments (ReoPro, Remicade; Table 1).
`Proteolysis, however, does not easily yield molecules smaller
`than a Fab fragment, and microbial expression of single-chain
`Fv (scFv) is currently the favored method of production (Fig. 1).
`In scFvs, the variable (VH and VL) domains are stably tethered
`together with a flexible polypeptide linker (Fig. 1)2,3. In com-
`parison with whole antibodies, small antibody fragments such
`as Fab or scFv exhibit better pharmacokinetics for tissue pene-
`tration and also provide full binding specificity because the
`antigen-binding surface is unaltered. However, Fab and scFv
`are monovalent and often exhibit fast off-rates and poor reten-
`tion time on the target8,9. Therefore, Fab and scFv fragments
`have been engineered into dimeric, trimeric or tetrameric con-
`jugates to increase functional affinity through the use of either
`chemical or genetic cross-links (Fig. 1)2,3,10,11. Various methods
`have been devised to genetically encode multimeric scFvs, of
`which the most successful design was the simple reduction of
`scFv linker length to direct the formation of bivalent dimers
`(diabodies, 60 kDa), triabodies (90 kDa) or tetrabodies (120
`kDa) (Fig. 1)11. Indeed, the first clinical trials of scFv fragments
`are likely to be as multivalent reagents, because they exhibit
`high functional affinity and have been very successful in pre-
`clinical studies8,12–14.
`
`Pharmacokinetics of intact antibodies versus fragments
`The efficiency of antibodies in vivo, for example in cancer ther-
`apy, lies in their capacity to discriminate among tumor-associ-
`ated antigens at low levels. Immunotherapy has been more
`successful against circulating cancer cells than solid tumors be-
`cause of better cell accessibility. This is illustrated by the FDA
`approval of intact antibodies: Rituxan for the treatment of non-
`Hodgkin lymphoma and Campath and Mylotarg for the treat-
`ments of leukemia (Table 1)15. Only two monoclonal antibodies
`
`NATURE MEDICINE • VOLUME 9 • NUMBER 1 • JANUARY 2003
`
`129
`
`Lassen - Exhibit 1046, p. 1
`
`

`

`REVIEW
`
`Fig. 1
`Schematic representation of an intact
`Ig together with Fab and Fv fragments and
`single V (colored ovals; dots represent anti-
`gen-binding sites) and C domains (uncol-
`ored). Engineered recombinant antibodies are
`shown as scFv monomers, dimers (diabodies),
`trimers (triabodies) and tetramers (tetrabod-
`ies), with linkers represented by a black line.
`Minibodies are shown as two scFv modules
`joined by two C domains. Also shown are Fab
`dimers (conjugates by adhesive polypeptide
`or protein domains) and Fab trimers (chemi-
`cally conjugated). Colors denote different
`specificities for the bispecific scFv dimers (dia-
`bodies) and Fab dimers and trimers.
`
`©2003 Nature Publishing Group http://www.nature.com/naturemedicine
`
`have been approved for the treatment of
`solid tumors: Herceptin for the treatment
`of breast carcinoma and PanoRex for
`colon cancer (in Germany). Although the
`mechanisms of action are still under in-
`vestigation, Herceptin appears to utilize
`Fc receptors and angiogenesis16, whereas
`Rituxan activates apoptosis through re-
`ceptor dimerization17.
`Radiolabeled antibodies are important clinical reagents for
`both tumor imaging and therapy and also provide an effective
`evaluation of pharmacokinetics18. The choice of radionuclide
`dictates the application. For example, Zevalin is approved for
`lymphoma therapy as a rapidly cleared, intact mouse antibody
`to match the clearance rates of yttrium-90 (ref. 4). Therapeutic
`administration requires a balance between long dissociation
`rates at the target site and slow blood clearance, which can lead
`to accumulation in the liver and high radiation exposure of
`other tissues. Biodistribution studies in solid tumors have also
`revealed that whole IgG molecules are too large (150 kDa) for
`rapid tumor penetration. The best tumor-targeting reagents
`comprise an intermediate-sized multivalent molecule, provid-
`ing rapid tissue penetration, high target retention and rapid
`blood clearance8,12–14. For example, diabodies (60 kDa) are effi-
`cacious with short-lived radioisotopes for clinical imaging as a
`result of the fast clearance rates12–14. Larger molecules, such as
`minibodies (90 kDa), are used with long-lived radioisotopes
`and are suitable for tumor therapy because they achieve a
`higher total tumor ‘load’14. Fab dimers (110 kDa) have also
`been effective in preclinical studies19.
`The short half-life of antibody fragments can also be ex-
`tended by ‘pegylation’, that is, a fusion to polyethylene glycol
`(PEG)20. Renal and hepatic localization of intact radiolabeled
`antibody fragments constitutes a major problem. An impor-
`tant study demonstrated that a previously undescribed radio-
`iodination reagent could liberate radionuclide from the anti-
`body fragment before incorporation into renal cells21. The ra-
`dionuclide is excreted rapidly, thus decreasing the total renal
`radiation dose21. The development of new metabolizable
`chelates will further improve the pharmacokinetics of recom-
`binant antibodies for cancer targeting. Modifications to sur-
`face charge designed to alter the isoelectric point (pI), such as
`glycolation, can also reduce the tissue (kidney) uptake22. The
`improved functional affinity, tumor penetration and biodistri-
`bution of these engineered antibody fragments will stimulate
`the development of a new generation of reagents for imaging
`and therapy.
`
`Intact IgG
`
`Monovalent
`fragments
`
`Diabodies
`(~60 kDa)
`
`Fab
`(~55 kDa)
`
`Fv
`
`scFv
`(~30 kDa)
`
`Bivalent
`
`Bispecific
`
`Triabodies (~90 kDa)
`
`V Domain
`(~15 kDa)
`
`C Domain
`(~15 kDa)
`
`Fab conjugates: dimers and trimers
`
`Minibody
`
`Adhesive
`domains
`or
`helices:
`dimeric
`Fab
`
`Chemical
`conjugate:
`trimeric Fab
`
`Trivalent
`
`Tetrabodies (~120 kD)
`
`Tetravalent
`
`Engineering multiple specificity in antibody fragments
`Bispecific antibodies contain two different binding specificities
`fused together and, in the most simple example, bind to two
`adjacent epitopes on a single target antigen, thereby increasing
`the avidity. Alternatively, bispecific antibodies can cross-link
`two different antigens and are powerful therapeutic reagents,
`particularly for recruitment of cytotoxic T cells for cancer treat-
`ment23,24. Bispecific antibodies can be produced by fusion of
`two hybridoma cell lines into a single ‘quadroma’ cell line;
`however, this technique is complex and time-consuming, and
`it produces unwanted pairing of the heavy and light chains. Far
`more effective methods to couple two different Fab modules
`incorporate either chemical or genetic conjugation or fusion to
`adhesive heterodimeric domains, including designed CH3 do-
`mains23,24. Bispecific diabodies provide an innovative alterna-
`tive therapeutic25 (Fig. 1).
`
`Bifunctional antibodies. The original ‘magic bullet’ concept is
`still alive: antibodies have been fused to a vast range of mole-
`cules that provide important ancillary functions after target
`binding. These include radionuclides (discussed earlier) and
`also cytotoxic drugs, toxins, peptides, proteins, enzymes and
`viruses, the latter for targeted gene therapy26–30. For cancer ther-
`apy, bifunctional antibodies are engineered to effectively target
`tumor-associated antigens at low levels and then deliver a cyto-
`toxic payload to tumor cells. The latest antibody-toxin conju-
`gates are stable in vivo and minimally immunogenic28,29.
`Antibodies have also been fused to lipids and PEGs20, both to
`enhance in vivo delivery and pharmacokinetics and to direct
`drug-loaded liposomes31,32. As immunoliposomes, anti-transfer-
`rin receptor antibodies have been used to deliver drugs to the
`brain, passing through the blood-brain barrier32. Antibody-en-
`zyme fusions have also been developed for prodrug activation,
`primarily for cancer therapy33.
`
`Antibody libraries: construction, display and selection. Library
`display has superseded hybridoma technology for the selec-
`tion of human antibodies through the creation of large nat-
`ural and synthetic immune repertoires in vitro34–36. From these
`
`130
`
`NATURE MEDICINE • VOLUME 9 • NUMBER 1 • JANUARY 2003
`
`Lassen - Exhibit 1046, p. 2
`
`

`

`REVIEW
`
`libraries, specific high-affinity antibodies can be selected by
`linking phenotype (binding affinity) to genotype, thereby al-
`lowing simultaneous recovery of the gene encoding the se-
`lected antibody. Antibodies are usually displayed as
`monovalent Fab or scFv fragments and then, as required, re-
`assembled into intact Ig or multivalent variants after selec-
`tion34,37. If the repertoire is sufficiently large, a high-affinity
`Fab or scFv can be selected directly or, more frequently, the
`recovered gene can be subjected to cycles of mutation and
`further selection to enhance affinity (Fig. 2). Furthermore,
`new methods of selection and screening have been designed
`to specifically isolate antibodies with desired characteristics,
`such as enhanced stability, high expression or capacity to ac-
`tivate receptors38,39.
`
`Bacteriophage display. Fd phage and Fd phagemid technolo-
`gies are currently the most widely used in vitro methods for
`the display of large repertoires and for the selection of high-
`affinity recombinant antibodies against a range of clinically
`important target molecules35,37,40,41. Innovative selection
`methods have proved powerful for isolating antibodies
`against previously refractory antigens, such as new tumor-as-
`sociated antigens, cell surface receptors and HLA-A1-pre-
`sented peptides35,42. Important improvements in selection
`technology have included array screening for high-avidity
`antibodies43 and recovery of internalized phage from live cells
`to select against internalizing (human) receptors38. Phage
`technology has been applied to complete proteome analysis
`using membrane-based screening41. The latest methods for
`generating large phage libraries and avoiding the limitations
`imposed by bacterial cell transformation are discussed in the
`Supplementary Note online.
`
`Libraries of mRNA-protein complexes. Ribosome display relies
`on stabilized complexes of antibody, ribosome and mRNA to
`replace bacteriophage as the display platform39,44,45. Ribosome
`complexes are constructed totally in vitro, thereby eliminating
`the need for cell transformation and allowing the production
`of large libraries, ≤1014 members. The system is limited only by
`the requirement of a ribonuclease-free environment for selec-
`tion and buffer compositions suitable for antibody folding.
`Indeed, picomolar affinity antibodies have been selected and
`rapid affinity maturation cycles carried out using this innova-
`
`tive in vitro method39,45. Covalent display using puromycin-sta-
`bilized mRNA-protein complexes is an alternative strategy to ri-
`bosome display46,47.
`
`Cell surface libraries. Before the advent of bacteriophage sys-
`tems, antibodies had been displayed on or in bacterial cells,
`although replica plating had limited screening to libraries of
`<108. The recent development of high-speed flow cytometers
`has re-activated the efforts in cell surface display, and several
`high-affinity antibodies have been
`isolated by
`this
`method48,49.
`
`Transgenic mice. Transgenic mice have been produced that
`lack the native mouse immune repertoire and instead harbor
`most of the human antibody repertoire in the germline.
`Injection of antigens into these mice leads to the development
`of human antibodies that have undergone mouse somatic hy-
`permutation and selection to relatively high affinity7.
`Antibodies can be recovered by classic hybridoma technology
`or, for more efficient affinity enhancement, by in vitro display
`and selection technologies (Fig. 2).
`
`Production, stability and expression levels
`Production of antibodies for preclinical and clinical trials has
`been evaluated in numerous expression systems, including
`bacteria, yeast, plant, insect and mammalian cells. Bacteria
`are favored for expression of small, non-glycosylated Fab and
`scFv fragments, usually with terminal polypeptides such as c-
`Myc, His or FLAG, for affinity purification11. Mammalian or
`plant cells are favored for intact antibodies and, occasionally,
`also for expression of scFvs, diabodies and minibodies14,50
`There is still hope that eukaryotic cell cultures, such as those
`of the yeast Pichia pastoris, will allow efficient production of
`fully processed scFvs, albeit with high-mannose oligosaccha-
`rides51. Additional expression methods are discussed in the
`Supplementary Note online.
`
`Affinity maturation
`Both transgenic mice and display libraries typically produce
`human antibodies with binding affinities (KD) ranging from
`10–7 to 10–9 M. Obtaining higher-affinity antibodies is impor-
`tant for efficient binding to the antigenic target for in vitro di-
`agnosis, viral neutralization, cell targeting and in vivo
`
`Table 1 FDA-approved therapeutic antibodies
`
`Product namea
`
`Orthoclone OKT3
`ReoPro
`Rituxan
`Zenapax
`Remicade
`Simulect
`Synagis
`Herceptin
`Mylotarg
`CroFab
`DigiFab
`Campath
`Zevalin
`
`Specificity
`
`CD3
`GpIIb/gpIIa
`CD20
`CD25
`TNF-α
`CD25
`RSV
`Her-2
`CD33
`Snake venom
`Digoxin
`CD52
`CD20
`
`Product type
`
`Mouse
`Chimeric Fab
`Chimeric
`Humanized
`Chimeric
`Chimeric
`Humanized
`Humanized
`Humanized
`Ovine Fab
`Ovine Fab
`Humanized
`Mouse
`
`Indication
`
`Transplant rejection
`Cardiovascular disease
`Non-Hodgkin lymphoma
`Transplant rejection
`Crohn disease, rheumatoid arthritis
`Transplant rejection
`Respiratory syncytial virus
`Metastatic breast cancer
`Acute myeloid leukemia
`Rattlesnake antidote
`Digoxin overdose
`Chronic lymphocytic leukemia
`Non-Hodgkin lymphoma
`
`Year
`
`1986
`1994
`1997
`1997
`1998, 1999
`1998
`1998
`1998
`2000
`2000
`2001
`2001
`2002
`
`aProduct names are registered trademarks. Recent developments in FDA approvals can be obtained from http://www.fda.gov/cber/efoi/approve.htm. Updates on products
`relevant to lymphoma immunotherapy can be obtained from http://www.lymphomainfo.net/therapy/immunotherapy/. The latest product developments, antibody formula-
`tion and prescribing details can usually be obtained from http://www.productname.com or the manufacturer’s Internet site.
`
`NATURE MEDICINE • VOLUME 9 • NUMBER 1 • JANUARY 2003
`
`131
`
`©2003 Nature Publishing Group http://www.nature.com/naturemedicine
`
`Lassen - Exhibit 1046, p. 3
`
`

`

`REVIEW
`
`a
`
`c
`
`1. Display
`
`4. Mutation
`
`2. Selection
`
`3. Amplification
`
`1. B-cell surface display
`
`In vivo:
`4.
`B cell germline
`Somatic recombination
`Somatic mutation
`Class switching
`
`2. Antigen stimulation:
`T-helper cell binds to
`activated B cell
`
`3. Clonal expansion of B cells
`
`b
`
`d
`
`Fig. 2 Affinity maturation cycles.
`a, General strategy of protein dis-
`play, selection (gene recovery), am-
`plification and gene mutation. For
`the specific affinity maturation cy-
`cles (b–d), alternative technologies
`are listed for each component of the
`cycle. b, Mammalian in vivo process of
`antibody maturation52. c, Bacterio-
`phage display of engineered anti-
`body repertoires (library) using an in
`vitro cycle of mutation and selec-
`tion. d, Ribosome display of engi-
`neered antibody repertoires (library)
`with an in vitro cycle of mutation
`and selection.
`
`1. Phage library
`
`1. Ribosome library
`
`In vitro:
`4.
` Error-prone PCR
`Mutating enzyme
`
`3. PCR
`
`2. Panning
`on antigen;
`recovery
`of binders
`by RT-PCR.
`
`Cloning for high-level expression
`
`Alternative scaffolds
`Intact antibodies, Fab and scFv
`fragments provide an antigen-
`binding surface comprising six
`CDR loops; these can be mu-
`tated, sequentially or collec-
`tively, to bind to a vast array of
`target molecules. Some target
`molecules are refractory to the
`immune repertoire, however,
`particularly those with cavities
`or clefts that require a small penetrating loop for tight binding.
`The natural mammalian antibody repertoire simply does not en-
`code penetrating loops, and only rarely has this type of antibody
`been selected57. Unexpectedly, both camelids (camels, llamas
`and related species) and sharks produce natural, single V-like do-
`main repertoires displaying cavity-penetrating CDR loops that
`complement the repertoire of conventional antibodies58–60. This
`theory has led to a number of attempts to design single-domain
`display libraries in vitro, based on V domains61 and other Ig-like
`scaffolds60,62. These small molecules complement both antibody
`and peptide libraries and are expected to have improved phar-
`macokinetics for several clinical applications, including those
`that require access to buried (immunosilent) sites or clefts in en-
`zymes, receptors and viruses62. Many important diagnostic tar-
`gets, notably prions63, have also been refractory to conventional
`antibodies, and we expect that new molecular libraries and scaf-
`folds will be required to provide the required binding reagents.
`
`4. In vitro
`Error-prone PCR
`DNA shuffling
`Chain shuffling
`Site-directed
`mutagenesis
`
`2. Panning
`on antigen;
`recovery
`of binders
`
`3. Transform
`into E. coli
`
`High-level expression
`
`©2003 Nature Publishing Group http://www.nature.com/naturemedicine
`
`imaging. To improve antibody affinity, various in vitro strate-
`gies have recently been optimized to mimic the mammalian
`in vivo process of somatic hypermutation and selection52 (Fig.
`2). These include site-specific mutagenesis based on structural
`information, combinatorial mutagenesis of complementar-
`ity-determining regions (CDRs), random mutagenesis of the
`entire gene or chain shuffling39,48,53,54.
`After a decade of developing library display strategies, it is
`now obvious that the most successful methods rely on several
`cycles of mutation, display, selection (recovery) and gene am-
`plification (Fig. 2). These cycles of mutation and selection can
`be carried out using either in vitro or in vivo strategies and
`have been far more effective than precisely designed alter-
`ations for affinity enhancement39,48. Even with the most de-
`tailed structural information, the techniques for design of
`precisely complementary surfaces through interface muta-
`tions remain in their infancy.
`Using the cycles depicted in Figure 2, affinity enhance-
`ment can be restricted to mutations in the antigen-binding
`surface (CDR loops). Importantly, mutations in the underly-
`ing framework regions have frequently provided large in-
`creases in affinity, stability and expression39,45,48. Random
`mutations over the entire V-domain genes can be derived
`from Escherichia coli mutator cells, homologous gene re-
`arrangements or error-prone PCR. Sequential ‘chain shuf-
`fling’ of the two V genes in the Fv module is also ‘random’
`but offers the advantage that only one V domain is altered
`at a time, while the other domain is kept constant to pro-
`vide a defined specificity55. Recent advances include the in-
`corporation of highly mutagenic enzymes such as mRNA
`reverse transcriptase and DNA polymerase with no proof-
`reading activity to achieve a high gene mutation rate56. The
`integration of such polymerases into the ribosome display
`and selection process could rapidly generate large libraries
`of mutants (Fig. 2).
`
`Clinical applications
`Aside from radioimmunotherapy (discussed earlier), there are
`a variety of other clinical applications of engineered antibod-
`ies for viral infection, cancer, autoimmune disease, allograft
`rejection, asthma, stroke and glaucoma surgery. Specific clin-
`ical applications are discussed below.
`
`Pathogen neutralization and antiviral therapy. Antibody
`binding can directly and effectively block the activity of many
`pathogens, often without requiring Fc-mediated cytotoxicity.
`Indeed, this has always been the promise of antibody-mediated
`viral neutralization. The first monoclonal antibody for the
`treatment of viral disease, Synagis, was approved by the FDA in
`1998 (Table 1). Synagis is a humanized antibody used for the
`prevention of severe respiratory syncytial virus (RSV) disease64.
`Despite this success, and the wide range of antibodies available
`against human immunodeficiency type 1 (HIV) and herpes sim-
`
`132
`
`NATURE MEDICINE • VOLUME 9 • NUMBER 1 • JANUARY 2003
`
`Lassen - Exhibit 1046, p. 4
`
`

`

`REVIEW
`
`plex virus (HSV), the use of recombinant antibodies as thera-
`peutics for viral infection has been limited65. Only a few rare an-
`tibodies have exhibited potent neutralization in vitro and
`antiviral efficacy in animal models57. This is probably due to
`viral efficiency both in producing escape mutants and in evolv-
`ing immunosilent receptor-binding surfaces65. For neutraliza-
`tion of other pathogenic molecules, monomeric Fab molecules
`have recently been approved by the FDA as antivenenes
`(CroFab; Table 1), and both scFv fragments and oligoclonal an-
`tibody mixtures have been effective against bacterial toxins66,67.
`
`Intracellular antibodies. Antibody fragments can be expressed
`as intracellular proteins, typically as scFvs termed intrabodies,
`and equipped with targeting signals either to neutralize intra-
`cellular gene products or to target cellular pathways. For exam-
`ple, expression of p21ras, erbB2, huntingtin and MHC have all
`been
`individually downregulated using
`antibodies68–71.
`Intrabodies also have important antiviral potential, particularly
`through their targeting of intracellular action to mandatory
`viral proteins such as the Vif, Tat or Rev components of HIV72.
`Antibody frameworks have been adapted that substantially im-
`prove expression levels and solubility in the intracellular reduc-
`ing environment73. Direct in vivo selection from large libraries
`will greatly facilitate the isolation of many previously unknown
`intracellular antibodies or ‘intrabodies’74,75. Obviously, the ex-
`pression of intrabodies in vivo can be encoded into gene therapy
`vectors, and this could ultimately be their most powerful clini-
`cal application.
`
`Cancer therapy and cell recruitment strategies. The promise of
`engineered antibodies for effective cancer therapy, especially ra-
`dioconjugates, has been described earlier. Blocking angiogene-
`sis to prevent the establishment and growth of tumors is
`becoming an important strategy76,77. Cancer cells can be de-
`stroyed by cell recruitment of cytotoxic T cells, natural-killer
`(NK) cells or macrophages that can be targeted by encoding cell
`surface antibodies (usually scFv)78. Alternative cell recruitment
`strategies include bifunctional antibodies, fused to cytokines for
`T-cell stimulation and proliferation at the tumor site79.
`
`Innovative vaccine applications. Troybodies are engineered
`vaccine antibodies containing cryptic T-cell epitopes to enhance
`antigen presentation80. Troybodies effectively target antigen-pre-
`senting cells (APCs) and, after processing, expose cryptic T-cell
`epitopes to direct T-cell activation. In the preferred format, the
`Fv domain provides APC specificity and the C domains encode
`the cryptic T-cell epitopes. These new vaccines can be redesigned
`to target many different APCs and enhance immunity to many
`different T-cell epitopes. Alternative vaccine strategies include
`the use of engineered APC-targeted antibodies that direct aden-
`oviruses to deliver vaccine-inducing epitopes as a gene therapy
`capsule30 and B7-targeted scaffolds (scFv and VL domains) that
`enable antigen-loading of dendritic cells61.
`
`Biosensors and microarrays: the future of diagnosis.
`A likely prediction is that biosensing devices and microarrays
`will dominate the in vitro diagnostic market by 2005. Antibodies
`currently provide high-sensitivity reagents for a huge range of
`diagnostic kits, accounting for approximating 30% of the $20
`billion per year diagnostic industry. It is therefore not surprising
`that antibodies are the paradigm for proof-in-principle of new
`biosensing devices, focused initially on glass-surface microar-
`
`rays81. Already in 2002 we have seen more protein-friendly sur-
`faces being developed as array platforms for antibody-based di-
`agnosis (Triage from Biosite, San Diego, California; Protein
`Profiling Biochip from Zyomyx, Hayward, California; Hydrogel
`from Perkin-Elmer, Boston, Massachusetts). These platforms
`will become increasingly available over the next few years, dri-
`ven by the demand for new reagents to diagnose the vast array
`of biomarkers stemming from proteomics discovery programs.
`These platforms will also be developed for robust ex vivo appli-
`cations, including the detection of microbial contaminants,
`pesticides and biological (warfare) pathogens82.
`
`Conclusion
`During the past few years, there has been growing excitement in
`both scientific and commercial communities in engineered anti-
`bodies. Scientific interest has stemmed from the elucidation of
`the key elements required for antibody fragment design, effi-
`cient expression and desired pharmacokinetics. Commercial in-
`terest is driven by increasing sales (Table 1). Many of the
`antibody fragments and fusion proteins discussed in this review
`are now undergoing scale-up production. It is likely that we will
`soon witness development of display and screening technologies
`incorporating nanoarray robotics. By providing a highly stable,
`protease-resistant scaffold, engineered recombinant antibody
`fragments will continue to be the model for selection of high-
`affinity clinical targeting reagents.
`
`Note: Supplementary information is available on the Nature Medicine website.
`
`Acknowledgments
`We thank many colleagues at CSIRO and the CRC for their helpful contribu-
`tions, and especially B. Mason and K. Wark for their assistance with the
`manuscript.
`
`1. Milstein, C. With the benefit of hindsight. Immunol. Today 21, 359–364 (2000).
`2. Carter, P. Improving the efficacy of antibody-based cancer therapies. Nat. Rev.
`Cancer 1, 118–129 (2001).
`3. Hudson, P.J. & Souriau, C. Recombinant antibodies for cancer diagnosis and
`therapy. Expert Opin. Biol. Ther. 1, 845–855 (2001).
`4. Wiseman, G.A. et al. Radiation dosimetry results for Zevalin radioimmunotherapy
`of rituximab-refractory non-Hodgkin lymphoma. Cancer 94, 1349–1357 (2002).
`5. Clynes, R.A., Towers, T.L., Presta, L.G. & Ravetch, J.V. Inhibitory Fc receptors
`modulate in vivo cytoxicity against tumor targets. Nat. Med. 6, 443–446 (2000).
`6. Ober, R.J., Radu, C.G., Ghetie, V. & Ward, E.S. Differences in promiscuity for an-
`tibody–FcRn interactions across species: implications for therapeutic antibodies.
`Int. Immunol. 13, 1551–1559 (2001).
`7. He, Y. et al. Efficient isolation of novel human monoclonal antibodies with neu-
`tralizing activity against HIV-1 from transgenic mice expressing human Ig loci.
`J. Immunol. 169, 595–605 (2002).
`8. Goel, A. et al. 99mTc-labeled divalent and tetravalent CC49 single-chain Fvs: novel
`imaging agents for rapid in vivo localization of human colon carcinoma. J. Nucl.
`Med. 42, 1519–1527 (2001).
`9. Adams, G.P. et al. High affinity restricts the localization and tumor penetration of
`single-chain Fv antibody molecules. Cancer Res. 61, 4750–4755 (2001).
`10. Tomlinson, I. & Holliger, P. Methods for generating multivalent and bispecific an-
`tibody fragments. Methods Enzymol. 326, 461–479 (2000).
`11. Todorovska, A. et al. Design and application of diabodies, triabodies and tetra-
`bodies for cancer targeting. J. Immunol. Methods 248, 47–66 (2001).
`12. Nielsen, U.B., Adams, G.P., Weiner, L.M. & Marks, J.D. Targeting of bivalent anti-
`ErbB2 diabody antibody fragments to tumor cells is independent of the intrinsic
`antibody affinity. Cancer Res. 60, 6434–6440 (2000).
`13. Tahtis, K. et al. Biodistribution properties of indium-111-labeled C-functionalized
`trans-cyclohexyl diethylenetriaminepentaacetic acid humanized 3S193 diabody
`and F(ab′)2 constructs in a breast carcinoma xenograft model. Clin. Cancer Res. 7,
`1061–1072 (2001).
`14. Yazaki, P.J.et al. Tumor targeting of radiometal-labeled anti-CEA recombinant T84.66
`diabody and t84.66 minibody: comparison to radioiodinated fragments. Bioconjug.
`Chem. 12, 220–228 (2001).
`15. Gura, T. Therapeutic antibodies: magic bullets hit the target. Nature 417, 584–586
`(2002).
`16. Izumi, Y., Xu, L., di Tomaso, E., Fukumura, D. & Jain, R.K. Tumour biology: herceptin
`
`NATURE MEDICINE • VOLUME 9 • NUMBER 1 • JANUARY 2003
`
`133
`
`©2003 Nature Publishing Group http://www.nature.com/naturemedicine
`
`Lassen - Exhibit 1046, p. 5
`
`

`

`REVIEW
`
`acts as an anti-angiogenic cocktail. Nature 416, 279–280 (2002).
`17. Cardarelli, P.M. et al. Binding to CD20 by anti-B1 antibody or F(ab′)2 is sufficient for
`induction of apoptosis in B-cell lines. Cancer Immunol. Immunother. 51, 15–24 (2002)

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