throbber
Protein Cell 2010, 1(4): 319–330
`DOI 10.1007/s13238-010-0052-8
`
`Protein & Cell
`
`REVIEW
`Monoclonal antibodies – a proven and rapidly
`expanding therapeutic modality for human
`diseases
`
`Zhiqiang An ✉
`
`Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030,
`USA
`✉ Correspondence: Zhiqiang.an@uth.tmc.edu
`Received March 10, 2010 Accepted March 31, 2010
`
`ABSTRACT
`
`The study of antibodies has been a focal point in modern
`biology and medicine since the early 1900s. However,
`progress in therapeutic antibody development was slow
`and intermittent until recently. The first antibody therapy,
`murine-derived murononab OKT3 for acute organ rejec-
`tion, was approved by the US Food and Drug Adminis-
`tration (FDA) in 1986, more than a decade after César
`Milstein and Georges Köhler developed methods for the
`isolation of mouse monoclonal antibodies from hybri-
`the scientific,
`doma cells in 1975. As a result of
`technological, and clinical breakthroughs in the 1980s
`and 1990s, the pace of therapeutic antibody discovery
`and development accelerated. Antibodies are becoming
`a major drug modality with more than two dozen
`therapeutic antibodies in the clinic and hundreds more
`in development. Despite the progress, need for improve-
`ment exists at every level. Antibody therapeutics pro-
`vides fertile ground for protein scientists to fulfill the
`dream of personalized medicine through basic scientific
`discovery and technological innovation.
`
`KEYWORDS
`monoclonal antibodies, personalized
`medicine, therapeutic antibodies
`
`INTRODUCTION
`
`The pioneering research by Robert Koch, Kitasato Shibasa-
`buro, Emil von Behring, and Paul Ehrlich in late 19th and the
`early 20th centuries on the treatment of infectious diseases
`with serum from patients who had recovered from the same
`disease was the first use of antibodies as therapeutics. The
`
`active components in the serum were described as “anti-
`bodies” “antitoxins” and “magic bullets” (Ehrlich, 1908; Winau
`et al., 2004). This crude “serum therapy” was later modified by
`isolating antibodies from the serum for the treatment of
`infectious and immune diseases, known as intravenous
`immune globulin (IVIG) (Stangel and Pul, 2006). Despite the
`early success of serum therapy and IVIG treatment, no
`significant progress was made in therapeutic antibody
`discovery and development until César Milstein and Georges
`Köhler developed methods for isolating mouse monoclonal
`antibodies (mAbs) from hybridoma cells in 1975 (Köhler and
`Milstein, 1975). Since then, mAbs have not only fueled
`breakthrough discoveries in basic research, but have also
`been developed as clinical diagnostics, reagents for high
`throughput drug screening, and more importantly, life-saving
`medicines. The first therapeutic mAb murononab, a murine-
`derived antibody for acute organ rejection, was approved by
`the US Food and Drug Administration (FDA) in 1986, a
`decade after the discovery of the mouse hybridoma technol-
`ogy (Thistlethwaite et al., 1987). As a result of technological
`breakthroughs in the 1980s and 1990s, progress in ther-
`apeutic mAbs field has been accelerated. Therapeutic
`antibodies have shown desirable safety profiles, high target
`specificity and affinity, and efficiency in disrupting protein/
`protein interactions. They are becoming a major drug
`modality with more than 25 therapeutic antibodies in clinical
`use and hundreds more in development (Reichert and Valge-
`Archer, 2007; An, 2009).
`
`ANTIBODY STRUCTURE
`
`An antibody of the IgG isotype is a homodimer composed of
`two heterodimers of one light chain and one heavy chain.
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`319
`
`1
`
`EX2091
`Eli Lilly & Co. v. Teva Pharms. Int'l GMBH
`IPR2018-01427
`
`

`

`Protein & Cell
`
`Zhiqiang An
`
`Both the heterodimers and homodimers are linked by inter
`chain disulphide bonds (Stanfield and Wilson, 2009) (Fig. 1A).
`The light and heavy chains each contain variable and
`constant
`regions. The antigen binding complementarity
`determining regions (CDRs) are short hypervariable amino
`acid sequences found in the variable domains of both light
`(variable light or VL) and heavy (variable heavy or VH) chains.
`Each VH and VL contains three pairs of non-identical CDRs
`(CDR1, CDR2 and CDR3). CDRs are termed hypervariable
`domains because the majority of the sequence variations
`associated with antibodies is found in the CDRs. Among the
`six CDRs in an IgG molecule, CDR3s have the greatest
`variability. The Fc-region (fragment crystalizable region) of a
`mAb, residing in the constant regions of the heavy chains, can
`recruit effector cells such as natural killer cells, macrophages
`or neutrophils to activate the complement system to destroy
`the target-associated cells. These functions are referred to as
`antibody-dependent cellular cytotoxicity (ADCC) and comple-
`ment-dependent cytotoxicity (CDC). Four additional antibody
`isotypes are found in humans, IgA, IgD, IgE, and IgM. All five
`isotypes share a common theme of a core heterodimer
`building unit of a heavy and light chain. In IgG, IgA and IgD
`antibody isotypes, the Fc region is composed of two identical
`protein fragments, derived from the second (CH2) and third
`(CH3) constant domains of the antibody's two heavy chains.
`The Fc regions in IgM and IgE contain three heavy chain
`
`constant domains in each polypeptide chain. The IgG isotype
`is most commonly used in therapeutic applications.
`
`ANTIBODY THERAPEUTIC HISTORY
`
`The progress of antibody therapeutics is driven by both
`scientific and technological breakthroughs (Fig. 2). Thera-
`peutic antibody development also parallels the desire of the
`industry to reduce immunogenicity.
`Immunogenicity can
`reduce the efficacy of therapeutic mAbs. In severe cases,
`immunogenicity can cause anaphylaxis and hypersensitivity
`reactions. Soon after the approval of
`the murine-derived
`monoclonal antibody murononab for acute organ rejection in
`1986 (Thistlethwaite et al., 1987), it was realized that murine-
`derived monoclonal antibodies are less than ideal therapeu-
`tics due to their high immunogenicity in humans. Several
`strategies to make antibodies more human, such as chimeric
`mAb (Morrison et al., 1984) and CDR grafting (Kettleborough
`et al., 1991), were devised to reduce the human anti-mouse
`antibody (HAMA) responses. It took a decade for the first
`chimeric mAb, abciximab for hemostasis, to be approved by
`FDA in 1994 (Faulds and Sorkin, 1994). The first humanized
`mAb, Zenapax for kidney transplant rejection, was approved
`for clinical use by FDA in 1997 (Vincenti et al., 1998).
`Humanization alleviated the HAMA response to various
`degrees, but many other drawbacks became evident. For
`
`Figure 1. Diagrams of various antibody structures. (A) A generic IgG molecule. (B) A scFv fragment. (C) A Fab fragment. (D) A
`F(ab’)2 fragment. (E) A mouse IgG molecule. (F) A murine:human chimeric IgG molecule. (G) A humanized IgG molecule. (H) A
`human IgG molecule.
`
`320
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`2
`
`

`

`Therapeutic monoclonal antibodies
`
`Protein & Cell
`
`example, the humanization process is technically demanding
`and the process may result in reduced antigen binding affinity
`and decreased efficacy. To avoid the human immune
`response to murine-derived mAbs and to overcome the
`technical challenges associated with humanizing murine
`mAbs, two major approaches were developed for generating
`fully human mAbs. The first approach was to express human
`antibody fragments on bacteriophage surfaces. The resulting
`libraries contain billions of unique human antibody fragments
`which can be screened for leads (Vaughan et al., 1996).
`Humira, the first fully human mAb derived from a bacterioph-
`age displayed antibody library, was approved by the FDA in
`2003 for the treatment of rheumatoid arthritis (Weinblatt et al.,
`2003). The second approach was to use transgenic mice to
`produce fully human antibodies (Russell et al., 2000; Lonberg,
`2005). This is achieved by replacing the mouse native antibody
`genes with their human counterparts. Vectibix, an anti-EGFR
`antibody approved for colorectal cancer therapy in 2006, was
`the first
`fully human antibody therapeutic derived from a
`transgenic mice system (Chua and Cunningham, 2006). The
`industry trend is to develop more human like antibodies for
`clinical use. However, immunogenicity is a complex biological
`
`process and it cannot be predicted solely on human content of
`an antibody. For example, Humira, a fully human antibody, has
`a relatively high incidence of immunogenicity (Bender et al.,
`2007). Surprisingly, there is little difference in immunogenicity
`(anti-antibody response) between humanized and chimeric
`mAbs in clinical use today (Table 1). Clearly more basic and
`clinical research is needed to develop reliable parameters to
`predict immunogenicity of therapeutic antibodies prior to their
`reaching the clinic.
`
`SOURCES OF THERAPEUTICS ANTIBODIES
`
`Accessing diversified antibody sources are paramount to the
`success in the discovery and development of antibody
`therapies. Most therapeutic antibodies in the clinic today are
`of murine origin largely due to the early availability of the
`mouse hybridoma technology; however, entirely mouse
`antibodies have poor pharmacokinetics in humans due to
`human anti-mouse antibody immune responses (Fig. 1E). To
`reduce immunogenicity, murine antibodies are commonly
`modified to murine/human chimeric antibodies or humanized
`antibodies for therapeutic applications (Carter, 2006; Reichert
`
`Figure 2. History of antibody therapeutics. Green boxes represent scientific and technology milestones. Blue boxes are
`antibody therapeutics developed as a result of the scientific and technology breakthroughs.
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`321
`
`3
`
`

`

`Protein & Cell
`
`Zhiqiang An
`
`Ferrajolietal.,2001
`
`Sorokin,2000
`
`1999
`
`AlbanellandBaselga,
`
`Storch,1998
`
`OnrustandLamb,1998
`
`Nashanetal.,1997
`
`Vincentietal.,1998
`
`Maloneyetal.,1997
`
`1994
`
`FauldsandSorkin,
`
`Cohenetal.,1989
`
`IV
`
`IV
`
`IV
`
`IM
`
`IV
`
`IV
`
`IV
`
`IV
`
`IV
`
`IV
`
`humanizedIgG1
`
`CD52
`
`leukemia
`
`chroniclymphocytic
`
`oncology
`
`2001
`
`gatedwithozogamicin
`humanizedIgG4conju-
`
`CD33
`
`leukemia
`
`acutemyelogenous
`
`oncology
`
`2000
`
`humanizedIgG1
`
`Her2
`
`breastcancer
`
`oncology
`
`chimericIgG1
`
`RSVF-protein
`
`virus
`
`respiratorysyncytial
`
`ID
`
`chimericIgG1
`
`TNFalpha
`
`rheumatoidarthritis
`
`AIID
`
`chimericIgG1
`
`CD25
`
`transplantrejection
`
`AIID
`
`humanizedIgG1
`
`CD25
`
`transplantrejection
`
`AIID
`
`chimericIgG1
`
`CD20
`
`phoma
`
`Non-Hodgkin’slym-
`
`oncology
`
`chimericFab
`
`CD41
`
`cardiovasculardisease
`
`CV
`
`murineIgG2a
`
`CD3
`
`transplantrejection
`
`AIID
`
`1998
`
`1998
`
`1998
`
`1998
`
`1997
`
`1997
`
`1995
`
`1986
`
`Bayer-Schering
`Campath
`Alemtuzumab
`
`Wyeth
`Mylotarg
`cin
`Gemtuzumab/ozogami-
`
`Genentech
`Herceptin
`Trastuzumab
`
`MedImmune
`Synagis
`Palivizumab
`
`Centocor
`Remicade
`Infliximab
`Novartis
`Simulect
`Basiliximab
`
`Roche
`Zenapax
`Daclizumab
`
`Genentech/Roche
`Rituxan/MabThera
`Rituximab
`
`EliLilly
`ReoPro
`Abciximab
`
`Johnson&Johnson
`Orthoclone/OKT3
`Muromonab
`
`reference
`
`delivery
`
`proteinform/isotype
`
`target
`
`majorindication
`
`manufacturer
`tradename
`genericname
`Table1Monoclonalantibodytherapeuticsapprovedforclinicaluse
`
`therapyarea
`
`launchdate
`
`322
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`4
`
`

`

`Therapeutic monoclonal antibodies
`
`Protein & Cell
`
`Paul-Pletzer,2006
`
`2004
`
`RudickandSandrock,
`
`Kerr,2004
`
`Chenetal.,2005
`
`KiesandHarari,2002
`
`Weinblattetal.,2003
`
`Davies,2004
`
`Gauvreauetal.,2003
`
`Davis,2004
`
`2001
`
`KrasnerandJoyce,
`
`reference
`(Continued)
`
`IV
`
`IV
`
`IV
`
`IV
`
`IV
`
`SC
`
`IV
`
`SC
`
`SC
`
`IV
`
`humanizedIgG1
`
`IL-6R
`
`Castleman'sdisease
`
`AIID
`
`humanizedIgG1
`
`VLA4
`
`multiplesclerosis
`
`CNS/AIID
`
`humanizedIgG1
`
`VEGF
`
`smallcelllungcancer
`colorectalandnon-
`
`oncology
`
`gatedwithiodine-131
`chimericIgG1conju-
`
`tumors
`
`intracellularDNAin
`
`advancedlungcancer
`
`oncology
`
`chimericIgG1
`
`EGFR
`
`colorectalcancer
`
`oncology
`
`humanIgG1
`
`TNFalpha
`
`rheumatoidarthritis
`
`AIID
`
`gatedwithiodine-131
`murineIgG2aconju-
`
`CD20
`
`phoma
`
`Non-Hodgkin’slym-
`
`oncology
`
`humanizedIgG1
`
`CD11A
`
`psoriasis
`
`AIID
`
`2005
`
`2004
`
`2004
`
`2003
`
`2003
`
`2003
`
`2003
`
`2003
`
`humanizedIgG1
`
`IgE
`
`asthma
`
`respiratory
`
`2003
`
`gatedwithYttrium90
`murineIgG1conju-
`
`CD20
`
`phoma
`
`Non-Hodgkin’slym-
`
`oncology
`
`2002
`
`delivery
`
`proteinform/isotype
`
`target
`
`majorindication
`
`therapyarea
`
`launchdate
`
`Roche/Chugai
`Actemra
`Tocilizumab
`
`BiogenIDEC/Elan
`Tysabri
`Natalizumab
`
`Genentech
`Avastin
`Bevacizumab
`
`BiotechCo.
`ShanghaiMedipharm
`I-131ch-TNT
`
`ImClone/BMS
`Erbitux
`Cetuximab
`
`Abbott
`Humira
`Adalimumab
`
`GSK
`Bexxar
`Tositumomab
`
`Genentech
`Raptiva
`Efalizumab
`
`Genentech/Novartis
`Xolair
`Omalizumab
`
`Biogen/Idec
`Zevalin
`Ibritumomabtiuxetan
`
`manufacturer
`tradename
`genericname
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`323
`
`5
`
`

`

`Protein & Cell
`
`Zhiqiang An
`
`Pappasetal.,2009
`
`SC
`
`humanIgG1
`
`AIID,arthritis,immuneandinflammatorydisorders;CNS,centralnervoussystem;CV,cardiovascular;ID,infectiousdisease;IM,intramuscular;IV,intravenous;SC,subcutaneous
`Johnson&Johnson
`Simponi
`Golimumab
`
`TNFalpha
`
`rheumatoidarthritis
`
`AIID
`
`2009
`
`Keatingetal.,2010
`
`Rotheretal.,2007
`
`Rutgeertsetal.,2007
`
`2007
`
`CohenuramandSaif,
`
`IV
`
`IV
`
`SC
`
`IV
`
`humanIgG1
`
`CD20
`
`leukemia
`
`chroniclymphocytic
`
`oncology
`
`2009
`
`hybrid
`
`humanizedIgG2/IgG4
`
`C5a
`
`sis)
`
`PNH(chronichemoly-
`
`hematology
`
`2007
`
`PEGylatedfragment
`
`TNFalpha
`
`rheumatoidarthritis
`
`AIID
`
`humanIgG2
`
`EGFR
`
`colorectalcancer
`
`oncology
`
`2007
`
`2006
`
`2006
`
`KennethandKertes,
`
`injectionintotheeye
`
`mentofAvastin
`
`humanizedmabfrag-
`
`VEGF
`
`lardegeneration
`
`wetage-relatedmacu-
`
`ophthalmology
`
`2006
`
`reference
`(Continued)
`
`delivery
`
`proteinform/isotype
`
`target
`
`majorindication
`
`therapyarea
`
`launchdate
`
`GSK
`Arzerra
`Ofatumumab
`
`Alexion
`Soliris
`Eculizumab
`
`UCB-Schwarz
`Cimzia
`Certolizumabpegol
`
`Amgen
`Vectibix
`Panitumumab
`
`Genentech/Novartis
`Lucentis
`Ranibizumab
`
`manufacturer
`tradename
`genericname
`
`324
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`6
`
`

`

`Therapeutic monoclonal antibodies
`
`Protein & Cell
`
`and Valge-Archer, 2007) (Fig. 1F–G). Later, transgenic mice
`and in vitro phage display were employed to generate fully
`human therapeutic antibodies to circumvent the immunogeni-
`city issue associated with murine sequences (Fig. 1H)
`(Hoogenboom, 2005; Lonberg, 2005; Jakobovits et al.,
`2007; Lee et al., 2007). In addition to phage display, antibody
`fragments can also be displayed on yeast (Feldhaus et al.,
`2003), bacteria (Harvey et al., 2004), mammalian cells (Smith
`and Zauderer, 2009) and other in vitro systems such as
`ribosomes (Hanes et al., 1998). The pros and cons of the
`various antibody platforms have been broadly reviewed
`recently (An, 2009). The ever
`increasing demand for
`improved tools for antibody drug discovery will lead to new
`platforms and technologies. For example, humanized rabbit
`mAbs are being developed as therapeutics (News, 2010; Yu
`et al., 2010).
`Another important source of antibodies is the human
`antibody B cell repertoire. The isolation of human mAbs has
`been a labor-intensive endeavor, either through EBV immor-
`talization or hybridoma fusion, or by constructing phage-
`displayed antibody libraries (Vaughan et al., 1996; Traggiai
`et al., 2004; Li et al., 2006b; Rothe et al., 2007). Significant
`technological breakthroughs in B lymphocyte culture and
`cloning were reported recently including the analysis of HIV
`and flu mAbs in naturally infected or vaccinated humans
`(Wrammert et al., 2008; Jin et al., 2009; Ogunniyi et al., 2009;
`Scheid et al., 2009; Walker et al., 2009; Kwakkenbos et al.,
`2010). It is now possible to isolate human memory B cells
`(CD27 + , sIgG + , IgD-) from peripheral blood mononuclear
`cells (PBMC), and more importantly, culture them where they
`proliferate and differentiate to IgG secreting cells (ISC) (Smith
`et al., 2009; Walker et al., 2009). Single cell culturing vessels
`have been engineered,
`thus enabling for high-throughput
`screening of functional mAbs (Jin et al., 2009; Ogunniyi et al.,
`2009). New methods of B cell immortalization other than EBV
`infection or hybridoma, such as Bcl-6/Bcl-xL, or hTERT, have
`been reported (Kwakkenbos et al., 2010).
`In addition,
`methods for cloning of IgG encoding genes from single B
`cells have been optimized (Wrammert et al., 2008; Jin et al.,
`2009; Ogunniyi et al., 2009; Scheid et al., 2009; Walker et al.,
`2009; Kwakkenbos et al., 2010). These technical and
`engineering accomplishments make it
`feasible to isolate
`human mAbs with broad coverage of therapeutic targets.
`
`FORMATS OF ANTIBODY THERAPEUTICS
`
`Most therapeutic antibodies are full length IgG molecules and
`IgG1 is the most commonly used sub-isotype (Table 1). This
`is because IgG1 molecules possess several
`favorable
`characteristics: they are structurally stable; they have a long
`in vivo half life; and IgG1 confer Fc-mediated biological effects.
`In designing antibody therapeutics, it is sometimes desirable
`to diminish or abolish the ADCC and CDC functions while
`retaining its pharmacokinetic profile, in the case of a “benign
`
`blocker” antibody. For this purpose, both IgG2 and IgG4 have
`been used in antibody therapeutics (Table 1). Protein
`engineering has been applied to create Fc with altered
`properties. For example, IgG2m4, a novel engineered IgG
`isotype with reduced Fc functionality was recently reported
`(An et al., 2009). The engineered IgG2m4 is based on the
`IgG2 isotype with four key amino acid residue changes
`derived from IgG4 (H268Q, V309L, A330S and P331S). An
`IgG2m4 antibody has an overall reduction in complement and
`Fcg receptor binding in in vitro binding analyses while
`in vivo serum half-life in rhesus
`maintaining the normal
`monkeys.
`In addition to IgG molecules, antibody fragments (e.g.,
`Fab) have also been developed as therapeutics (Sandborn
`et al., 2007). Relative to IgG molecules, antibody fragments
`have more extensive penetration of tissues (particularly of
`solid tumors) due to their smaller size. The smaller size of
`antibody fragments has the advantage of accessing ther-
`apeutically important epitopes that may be sterically hindered.
`In addition, antibody fragments may be manufactured more
`cost effectively in a microbial
`fermentation system. The
`shorter half life of antibody fragments can be extended by
`modifying the molecules such as through PEGylation. The
`absence of the Fc region in an antibody fragment may lessen
`side effects caused by the interaction between Fc and the
`immune system. ReoPro, an anti-GPIIb/IIa chimeric Fab for
`the prevention of blood clots in angioplasty, was the first
`antibody fragment approved for clinic use in the US (Faulds
`and Sorkin, 1994). Lucentis, a Fab fragment of Avastin, is
`used for the treatment of wet age-related macular degenera-
`tion (Kenneth and Kertes, 2006). More recently, Certolizumab
`pegol
`(Cimzia), a PEGylated antibody fragment, was
`approved for the treatment of rheumatoid arthritis in Europe
`(Rutgeerts et al., 2007). Currently, about 19 antibody
`fragment based therapeutics are in active clinical develop-
`ment (Nelson and Reichert, 2009).
`Other antibody formats such as domain antibodies and
`single chain antibodies are also being explored for diagnostic
`and therapeutic applications (Holt et al., 2003; Holliger and
`Hudson, 2005; Enever et al., 2009). A PEGylated human anti-
`IL-1R domain antibody is in clinical testing for the treatment of
`rheumatoid arthritis (Vk or VH dABs) (Enever et al., 2009). A
`llama nanoantibody targeting the von Willebrand factor is
`being developed for
`the treatment of
`thrombosis (Van
`Bockstaele et al., 2009).
`Antibodies can also be used as carrier agents of small
`molecule toxins or radiolabeled isotopes, guiding drugs to
`specific disease sites and limiting undesired effects on
`healthy cells. This application is most commonly employed
`in oncology. At least two radiolabeled antibodies, Zevalin and
`Bexxar, are approved for clinical use (Table 1). These drugs
`are difficult
`to administer because a radiologist and an
`oncologist are needed to oversee the administration. Mylo-
`targ, a humanized anti-CD33 IgG4 antibody conjugated to
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`325
`
`7
`
`

`

`Protein & Cell
`
`Zhiqiang An
`
`calicheamicin, is an example of antibody used to carry a
`cytotoxic payload (Table 1). Many challenges still exist in
`designing antibody:drug conjugates such as choice of linker,
`stoichiometry, and conjugation chemistry. Recent advances
`have resulted in linkers having increased stability in the
`bloodstream while allowing efficient payload release within
`the tumor cell (Ducry and Stump, 2010). Increasing evidence
`suggests that conjugated antibodies remain an effective
`alternative to mAb, small molecule or radiolabeled isotope
`monotherapies.
`It is interesting to note that the early antibody therapeutics
`started as crude polyclonals (serum therapy and IVIG) and
`the majority of today’s antibody therapeutics is target-specific
`monoclonals. Progress is being made in developing recom-
`binant polyclonal antibodies (rpAb) for the treatment or
`prophylaxis of human diseases (Pedersen et al., 2010). The
`difference is between IVIG and rpAbs is that rpAbs are
`mixtures of carefully selected monoclonal antibodies. Recom-
`binant polyclonal antibodies (rpAb) mimic the natural human
`immune response in which the human body produces
`different types of antibodies targeting different epitopes of
`an antigen. This polyclonal response may have a better
`chance to neutralize the disease target than a single antibody
`does. One of the major challenges facing this approach is to
`manufacture the antibody cocktail consistently in both
`quantity and quality.
`Today’s monoclonal antibody therapeutics functions on a
`single disease target.
`It
`is advantageous if one antibody
`molecule can bind to two or more different targets since many
`complex diseases are the result of multiple mediators.
`Multispecificity has shown in naturally isolated antibodies,
`for example, the monoclonal IgE antibody SPE7 binds not
`only to its intended antigen 2,4-dinitrophenyl (DNP) hapten,
`but it also binds to several unrelated compounds with a broad
`range of affinities (James et al., 2003). More recently,
`antibodies that binds to both HER2 and VEGF were reported
`(Bostrom et al., 2009). In these cases, the multispecificity is
`conferred by a single binding pocket (James et al., 2003;
`Bostrom et al., 2009). Multispecifity antibodies are not
`common in nature, but they can be constructed by recombi-
`nant DNA methods (Kufer et al., 2004; Wu et al., 2007).
`Design of bispecifc antibodies is an active research area and
`the anti-IL-12/IL-18 dual-variable-domain immunoglobulin
`DVD-Ig molecule is an example of
`the many designs of
`bispecific antibodies (Wu et al., 2007).
`
`IMPACT OF POST-TRANSLATIONAL
`MODIFCATION ON THE PHYSICAL AND BIOLOGICAL
`PROPERTIES OF THERAPEUTIC ANTIBODIES
`
`Antibodies are large proteins which are subjected to
`extensive and complex posttranslational modifications, such
`as deamidation, glycosylation, N-terminal pyroglutamation,
`C-terminal lysine truncation, and methionine oxidation; and
`
`these posttranslational modifications profoundly impact the
`physical, chemical, and pharmacological properties of ther-
`apeutic antibodies (Wang et al., 2009). Oxidation of methio-
`nine residues is one of the most common protein degradation
`pathways including antibodies.
`In addition, methionine
`oxidation of recombinant monoclonal antibodies can alter
`their interaction with protein A and protein G resulting in a
`decrease in binding affinity (Gaza-Bulseco et al., 2008). The
`spontaneous nonenzymatic deamidation of glutaminyl and
`asparaginyl residues can alter the structure and function of
`therapeutic antibodies, potentially resulting in decreased
`bioactivity, as well as alterations in pharmacokinetics and
`antigenicity of antibody therapeutics (Huang et al., 2005).
`Among the various posttranslational processes, glycosylation
`has the broadest effect on biologic activity, protein conforma-
`tion, stability, solubility, secretion, pharmacokinetics and
`immunogenicity of
`therapeutic antibodies (Arnold et al.,
`2007). For example, differential IgG sialylation may provide
`a switch from innate anti-inflammatory activity in the steady-
`state to generating adaptive pro-inflammatory effects upon
`antigenic challenge (Kaneko et al., 2006). Low fucose levels
`on antibodies enhance neutrophil- and mononuclear cell-
`mediated ADCC (Peipp et al., 2008). Antibody glycoengineer-
`ing is one of most active areas of research in therapeutic
`antibody discovery and development today (Mimura et al.,
`2009).
`
`MANUFACTURING
`
`Manufacturing of mAbs is expensive. A large scale facility can
`take multiple years and hundreds millions of dollars to build.
`Mammalian cell culture is the dominant production platform
`for mAb therapeutics. About half of the current marketed
`mAbs are expressed in Chinese hamster ovary (CHO) cell
`lines. Recombinant myelomas or hybridomas are still being
`used for antibody production, but their utility as a production
`platform is limited due to the low expression titer and
`instability of the cell
`lines. To reduce the cost of antibody
`production, other methods of antibody expression, such as
`bacteria, plants, transgenic animals (milk), eggs, and yeast,
`are being developed. Certolizumab pegol is an example of
`mAb (fragment) therapeutic made in a bacteria (Rutgeerts
`et al., 2007). However, antibodies produced in E. coli are not
`glycosylated and this severely limits its use as a manufactur-
`ing platform. Antibodies with specific human N-glycan
`structures have been expressed in glycoengineered lines of
`the yeast Pichia pastoris and its utility as a general platform
`for producing recombinant antibodies with human N-glycosy-
`lation is being developed (Li et al., 2006a; Lin et al., 2010).
`Antibody has been expressed in engineered chicken eggs
`and in plants (Zhu et al., 2005; Cox et al., 2006). Despite
`significant effort, cost saving alternative antibody manufactur-
`ing platforms is still lacking. This is in part due to the effect of
`the various expression hosts on antibody posttranslational
`
`326
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`8
`
`

`

`Therapeutic monoclonal antibodies
`
`Protein & Cell
`
`modifications and the low production titers. There is a clear
`need for innovation and technology breakthroughs in redu-
`cing the manufacturing cost of therapeutic antibodies. This is
`not limited to the choice of expression hosts. Purification,
`formulation, storage, and other steps in the entire manufac-
`turing process need to be improved to bring antibody
`therapeutics in a more cost competitive position against
`small molecule drugs.
`
`ANTIBODY THERAPEUTIC TARGETS
`
`Antibodies can engage a wide range of extracellular drug
`targets such as membrane bound proteins or circulating
`ligands and cytokines (Table 1). Even though antibodies do
`not readily cross cell membranes or the brain blood barrier
`(BBB), about 80% of
`the current druggable targets are
`accessible to antibodies (Strohl, 2009). Extracellular signaling
`(ECS) drug targets generally are not modulated by small
`molecules as ECS targets typically function through protein-
`protein interactions. ECS proteins have been successfully
`targeted by antibodies. The therapeutic areas in which
`antibodies have the strongest presence, in terms of marketed
`products and developmental research, are oncology and
`Arthritis, immune and inflammatory disorders (AIID). Of the
`more than 200 monoclonal antibodies in clinical use and
`development
`today, about half are being developed for
`oncology. The second largest therapeutic category is in the
`AIID area, and infectious disease is fast becoming a major
`disease area for antibody therapeutics. While the emphasis
`on oncology and AIID therapeutic areas will continue,
`antibody therapeutics are being developed in almost all
`disease areas such as central nervous system, cardiovas-
`cular, women's health, diabetes/endocrinology, hematology,
`ophthalmology, and respiratory diseases (Strohl, 2009).
`
`SUMMARY
`
`Antibody therapeutics represent a major breakthrough in
`combating human diseases and the improvement of human
`health. This is reflected by the recent trend in drug discovery
`and development. In 2000, nine of the top 10 medicines were
`small molecules while only one was a recombinant protein but
`by 2008, a short eight years later, half of the top 10 medicines
`are recombinant proteins and antibodies. This trend will
`continue as about 50% of the new drugs in various stages of
`clinical development are antibodies. Despite the remarkable
`progress, many scientific, technological, and clinical chal-
`lenges remain in the area of therapeutic antibody discovery
`and development. Opportunities for innovation exist at every
`level: accessing difficult antibody targets (such as G protein-
`coupled receptors), novel antibody sources and formats,
`crossing the BBB and cell membranes, modified effector
`functions, improved formulation and delivery methods, and
`lower cost manufacturing, to name a few.
`
`ABBREVIATIONS
`
`AIID, arthritis, immune and inflammatory disorders; ADCC, antibody-
`dependent cellular cytotoxicity; BBB, brain blood barrier; CDC,
`complement-dependent cytotoxicity; CDRs, complementarity deter-
`mining regions; DNP, 2,4-dinitrophenyl; ECS, extracellular signaling;
`FDA, Food and Drug Administration; HAMA, human anti-mouse
`antibody; IM, intramuscular; ISC, IgG secreting cells; IV, intravenous;
`IVIG,
`intravenous immune globulin; mAbs, mouse monoclonal
`antibodies; PBMC, peripheral blood mononuclear cells;
`rpAb,
`recombinant polyclonal antibodies; SC, subcutaneous; VL, variable
`light; VH, variable heavy
`
`REFERENCES
`
`Albanell, J., and Baselga, J. (1999). Trastuzumab, a humanized anti-
`HER2 monoclonal antibody, for the treatment of breast cancer.
`Drugs Today (Barc) 35, 931–946.
`An, Z. (2009). Therapeutic monoclonal antibodies: from bench to
`clinic, Hoboken, NJ: John Wiley and Sons.
`An, Z., Forrest, G., Moore, R., Cukan, M., Haytko, P., Huang, L., Vitelli,
`S., Zhao, J.Z., Lu, P., Hua, J., et al. (2009). IgG2m4, an engineered
`antibody isotype with reduced Fc function. MAbs 1, 572–579.
`Arnold, J.N., Wormald, M.R., Sim, R.B., Rudd, P.M., and Dwek, R.A.
`(2007). The impact of glycosylation on the biological function and
`structure of human immunoglobulins. Annu Rev Immunol 25,
`21–50.
`Bender, N.K., Heilig, C.E., Dröll, B., Wohlgemuth, J., Armbruster, F.P.,
`and Heilig, B. (2007). Immunogenicity, efficacy and adverse events
`of adalimumab in RA patients. Rheumatol Int 27, 269–274.
`Bostrom, J., Yu, S.F., Kan, D., Appleton, B.A., Lee, C.V., Billeci, K.,
`Man, W., Peale, F., Ross, S., Wiesmann, C., et al. (2009). Variants
`of the antibody herceptin that interact with HER2 and VEGF at the
`antigen binding site. Science 323, 1610–1614.
`Carter, P.J. (2006). Potent antibody therapeutics by design. Nat Rev
`Immunol 6, 343–357.
`Chen, S., Yu, L., Jiang, C., Zhao, Y., Sun, D., Li, S., Liao, G., Chen, Y.,
`Fu, Q., Tao, Q., et al. (2005). Pivotal study of iodine-131-labeled
`chimeric tumor necrosis treatment radioimmunotherapy in patients
`with advanced lung cancer. J Clin Oncol 23, 1538–1547.
`Chua, Y.J., and Cunningham, D. (2006). Panitumumab. Drugs Today
`(Barc) 42, 711–719.
`Cohen, D.J., Benvenisty, A.I., Cianci, J., and Hardy, M.A. (1989).
`OKT3 prophylaxis in cadaveric kidney transplant recipients with
`delayed graft function. Am J Kidney Dis 14, 19–27.
`Cohenuram, M., and Saif, M.W. (2007). Panitumumab the first fully
`human monoclonal antibody: from the bench to the clinic. Antic-
`ancer Drugs 18, 7–15.
`Cox, K.M., Sterling, J.D., Regan, J.T., Gasdaska, J.R., Frantz, K.K.,
`Peele, C.G., Black, A., Passmore, D., Moldovan-Loomis, C.,
`Srinivasan, M., et al. (2006). Glycan optimization of a human
`monoclonal antibody in the aquatic plant Lemna minor. Nat
`Biotechnol 24, 1591–1597.
`Davies, A.J. (2004). Tositumomab and iodine [131I] tositumomab in
`the management of follicular lymphoma. An oncologist's view. Q J
`Nucl Med Mol Imaging 48, 305–316.
`Ducry, L., and Stump, B. (2010). Antibody-drug conjugates: linking
`
`© Higher Education Press and Springer-Verlag Berlin Heidelberg 2010
`
`327
`
`9
`
`

`

`Protein & Cell
`
`Zhiqiang An
`
`cytotoxic payloads to monoclonal antibodies. Bioconjug Chem 21,
`5–13.
`Ehrlich, P. (1908). Partial cell functions—Nobel lecture, December 11,
`1908 in Physiology or Medicine: including presentation speeches
`and laureates' biographies. Amsterdam, 1967: Elsevier Publisher.
`Enever, C., Batuwangala, T., Plummer, C., and Sepp, A. (2009). Next
`generation immunotherapeutics—honing the magic bullet. Curr
`Opin Biotechnol 20, 405–411.
`Faulds, D., and Sorkin, E.M. (1994). Abciximab (c7E3 Fab). A review
`of its pharmacology and therapeutic potential in ischaemic heart
`disease. Drugs 48, 583–598.
`Feldhaus, M.J., Siegel, R.W., Opresko, L.K., Coleman, J.R.,
`Feldhaus, J.M., Yeung, Y.A., Coc

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