throbber
nature publishing group
`
`Eli Lill & c
`EX2268
`y
`o. v. Teva Pharms. lnt'l GMBH
`IPR2018-01422, ·01423, 01424 -01425

`·01426, •01427
`
`I
`
`Monoclonal Antibody Pharmacokinetics and
`Pharmacodynamics
`W Wang1, EQ Wang2 and JP Balthasar3
`
`More than 20 monoclonal antibodies have been approved as therapeutic drugs by the US Food and Drug Administration,
`and it is quite likely that the number of approved antibodies will double in the next 7-10 years. Antibody drugs show
`several desirable characteristics, including good solubility and stability, long persistence in the body, high selectivity
`and specificity, and low risk for bioconversion to toxic metabolites. However, many antibody drugs demonstrate
`attributes that complicate drug development, including very poor oral bioavailability, incomplete absorption following
`intramuscular or subcutaneous administration, nonlinear distribution, and nonlinear elimination.ln addition, antibody
`administration often leads to an endogenous antibody response, which may alter the pharmacokinetics and efficacy of
`the therapeutic antibody. Antibodies have been developed for a wide range of disease conditions, with effects produced
`through a complex array of mechanisms. This article attempts to provide a brief overview of the main determinants of
`antibody pharmacokinetics and pharmacodynamics.
`
`INTRODUCTION
`Antibodies, which are also called immunoglobulins (Igs), are
`large proteins used by the immune system to identify and neu(cid:173)
`tralize foreign objects such as bacteria and viruses. All Ig mol(cid:173)
`ecules are composed of a basic unit of two identical heavy chains
`and two identical light chains, held together by a number of
`disulfide bonds. In humans, there are two types oflight chains
`(KandA.) and five types oflg heavy chains (a., 0, E, y, and fl). 1 Igs
`are grouped into five classes according to the structure of their
`heavy chains: IgA, IgD, IgE, IgG, and IgM. Among these, IgG
`is the predominant class, comprising -80% of the Igs in human
`serum. All of the approved therapeutic antibodies are IgGs, and
`this review focuses on this class.
`Intact IgGs have a molecular weight of -150 kDa and a valence
`of 2 (meaning that each molecule of IgG contains two identical
`antigen-binding domains). The antigen-binding sites are located
`in the complementarity determining regions (CDRs) within the
`Fab portion of the antibody (Figure 1). Fab, which refers to the
`fragment of antigen binding, is composed of domains associated
`with the light chain (VL, CL) and domains associated with the
`heavy chain (VH, CHI). The stem, or Fe, portion oflgG con(cid:173)
`tains the CH2 and CH3 domains of the heavy chains, and this
`region of the antibody is involved with binding to a wide range
`of cell-associated receptors (i.e., Fe receptors). The IgG family of
`
`antibodies may be further divided, again based on the structure
`oftheir heavy chains, into four subclasses: IgG 1, IgG2, IgG3, and
`IgG4. Structural differences among IgG heavy chains lead to dif(cid:173)
`ferences in subclass binding to Fe receptors and, consequently,
`to subclass-specific differences in processes mediated by Fe
`receptors (e.g., activation of complement or antibody-dependent
`cell-mediated cytotoxicity). For example, antibody-dependent
`cell-mediated cytotoxicity by mononuclear cells is more efficient
`for IgG 1 and IgG3 than for IgG2 and IgG4. On the other hand,
`IgG4 is much more active in recruiting the alternative comple(cid:173)
`ment pathway than are the other three IgG subclasses. 1
`Antibody drugs typically possess several desirable pharma(cid:173)
`cological characteristics, such as long serum half-lives, high
`potency, and limited off-target toxicity. Initial antibody thera(cid:173)
`pies were prepared from hyperimmune sera, collected follow(cid:173)
`ing immunization of animals. The resulting antibody product,
`which is derived from a large number of genetically distinct
`cells, contains a distribution oflg isotypes and affinities. In 1975,
`Kohler and Milstein demonstrated that antibody-producing
`B lymphocytes may be fused with myeloma cells to generate
`hybrid cells (hybridomas) that propagate indefinitely in culture
`and secrete antibody. 2 Cloning the hybridoma cells enabled effi(cid:173)
`cient production of antibody derived from a single progenitor,
`and the resulting monoclonal antibody (mAb) preparations are
`
`1 Department of Drug Metabolism and Pharmacokinetics, Merck Research Laboratories, West Point, Pennsylvania, USA; 2Department of Pharmacokinetics, Dynamics,
`and Drug Metabolism, Pfizer Global Research and Development, Groton Laboratories, Groton, Connecticut, USA; 3Department of Pharmaceutical Sciences, Center for
`Protein Therapeutics, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA.
`Correspondence: JP Balthasar (jb@buffalo.edu)
`
`Received 17 July 2008; accepted 30 July 2008; advance online publication 10 September 2008. doi: 1 0 1 038/clpt.2008.170
`
`548
`
`VOLUME 84 NUMBER S i NOVEMBER 20081 www.nature.com/cpt
`
`

`

`ANTIBODY ABSORPTION
`The majority of marketed antibodies are labeled for intravenous
`(IV) administration; however, several antibodies have been
`approved for extravascular administration. For example, cer(cid:173)
`tolizumab pegol, adalimumab, efalizumab, and omalizumab are
`all approved for subcutaneous (SC) administration. Palivizumab
`is approved for intramuscular (IM) administration, and ranibi(cid:173)
`zumab is administered by intravitreal injection. Antibodies have
`not been successfully developed for oral administration, as oral
`absorption of antibody is limited by presystemic degradation in
`the gastrointestinal tract and by inefficient diffusion or convec(cid:173)
`tion through the gastrointestinal epithelium. With the exception
`of ranibizumab, where intravitreal administration is employed to
`promote a regional effect, efficacy of mAbs following extravascu(cid:173)
`lar administration is dependent on systemic absorption.
`Primary pathways for systemic absorption include convec(cid:173)
`tive transport of antibody through lymphatic vessels and into
`the blood, and diffusion of antibody across blood vessels dis(cid:173)
`tributed near the site of injection. Based on work conducted
`by Supersaxo et at.S that investigated the lymphatic uptake of a
`variety of proteins following SC injection in sheep, it has been
`suggested that the majority of antibody administered via SC
`or IM injection is absorbed via convection through lymphatic
`vessels. However, recent investigations conducted in rats suggest
`that the role of diffusion into blood vessels may be underesti(cid:173)
`mated by the sheep studies.6 Using insulin, bovine serum albu(cid:173)
`min, and erythropoietin as model proteins, Kagan et al. found
`that <3% of the administered dose of each protein was absorbed
`via the lymph. Neither Kagan et al. nor Supersaxo et al. have
`thoroughly investigated the fate oflgG following SC injection
`and, consequently, there is substantial uncertainty regarding the
`primary determinants of antibody absorption. The kinetics of
`antibody absorption, however, has been well described. After
`IM or SC injection, absorption proceeds slowly, and the time
`to reach maximal plasma concentrations (tmax) typically ranges
`from 2 to 8 days. Absolute bioavailability is generally reported
`between 50 and 100%.3
`In practical terms, bioavailability is determined by the rela(cid:173)
`tive rates of presystemic catabolism and systemic absorption.
`Presystemic catabolism may be dependent on rates of extra(cid:173)
`cellular degradation (e.g., via proteolysis), rates of antibody
`endocytosis (e.g., receptor-mediated, fluid phase), and rates
`of recycling through interaction with the Brambell receptor
`(FeRn). FeRn protects IgG from intracellular catabolism, and
`FeRn has been shown to be capable of transporting IgG across
`cell monolayers in both the apical-to-basolateral and basolateral(cid:173)
`to-apical directions. Work from the Balthasar Laboratory
`(A. Garg, P.J. Lowe, and J.P. Balthasar, unpublished data) has
`indicated that the systemic bioavailability of7E3, a monoclonal
`IgG 1 antibody, was threefold higher in wild-type mice vs. FeRn(cid:173)
`deficient mice (82.5 ± 15.6% vs. 28.3 ± 6.9%, P < 0.0001). It
`is not yet known whether the effects of FeRn on SC bioavail(cid:173)
`ability are primarily related to FeRn-mediated protection from
`catabolism or from FeRn-mediated transport across the vascu(cid:173)
`lar endothelium (from interstitial fluid to the blood); however,
`the former mechanism is considered to be more plausible.
`
`In some cases, an inverse relationship between SC bioavail(cid:173)
`ability and antibody dose has been noted? Such relationships
`are suggestive of saturable endocytosis and/or saturable deg(cid:173)
`radation processes. Degradation at the injection site is likely to
`account for some presystemic loss of antibody, but the quantita(cid:173)
`tive significance is uncertain. Charman et al. have demonstrated
`that the major determinant of the SC bioavailability of human
`growth hormone in sheep is presystemic catabolism during the
`course oflymphatic transport. 8 The role oflymphatic catabo(cid:173)
`lism on the bioavailability of other proteins, including mAbs,
`is not known.
`As a result of limited solubility of antibodies in solution
`( -100 mg/ml) and limitations on the volume of fluid that may
`be tolerated with IM or SC injection ( -5 and 2.5 ml, respec(cid:173)
`tively), IM and SC administration are feasible only for anti(cid:173)
`bodies that demonstrate relatively high dose potency. Use of
`multiple injections may help to overcome this limitation, at least
`to some extent. For example, doses of 375 mg of omalizumab
`are routinely administered clinically, via three separate 1-ml SC
`injections.
`Although they have not yet been employed in routine clinical
`use, there is substantial interest in the development of antibodies
`and Fc-fusion proteins for pulmonary delivery.9 The lungs have
`a very large surface area and high perfusion rate. In addition,
`pulmonary epithelial cells are known to express FeRn, which
`may facilitate efficient systemic absorption of antibody delivered
`to the lung. As discussed with SC and IM administration, the
`feasibility of pulmonary delivery of antibodies is likely limited to
`those antibodies associated with very high dose potency, as only
`small volumes of fluid may be delivered to the lung.
`
`ANTIBODY DISTRIBUTION
`The distribution of mAbs is determined by the rate of extra vasa(cid:173)
`tion in tissue, the rate of distribution within tissue, the rate and
`extent of antibody binding in tissue, and the rates of elimination
`from tissue. For large, polar substances such as mAbs, diffusion
`across vascular endothelial cells is very slow, and convection
`is believed to be the primary mechanism responsible for the
`transport of antibody from blood fluid to interstitial fluids of
`tissue. Of note, physiologically based analyses of antibody dis(cid:173)
`position in mice suggest that >98% of antibody enters tissue via
`convection. 10 The rate of extravasation by convective transport,
`or the movement of antibody into tissue by "solvent drag;' is
`determined by the rates of fluid movement from blood to tissue
`and by the sieving effect of paracellular pores in the vascular
`endothelium. Sieving is thought to be largely determined by
`the size and tortuosity of the pores and by the size, shape, and
`charge of the solute (i.e., the antibody). Most physiologically
`based models of antibody disposition describe the uptake clear(cid:173)
`ance for antibody extravasation as a product of the lymph flow
`rate (L) and an efficiency term ( 1 - cr). The reflection coefficient,
`0", represents the fraction of solute sieved during the movement
`of solvent through a pore. In the case of mAbs, tissue reflection
`coefficients are often assumed to be equal in all tissues, with
`values in the range of 0.95-0.98. 10- 12 However, it is likely that
`reflection coefficients may be much lower in tissues such as the
`
`550
`
`VOLUME 84 NUMBER Si NOVEMBER 20081 www.nature.com/cpt
`
`

`

`substances with several repeated epitopes, may bind with two
`or more antibodies, leading to the formation oflarge complexes
`that may be rapidly eliminated by phagocytosis. Elimination of
`large immune complexes may explain, in part, the nonlinear
`elimination kinetics of omalizumab and denosumab, which are
`thought to interact with soluble targets (IgE and receptor acti(cid:173)
`vator of nuclear factor-KB ligand). The majority of marketed
`antibodies demonstrate dose-dependent elimination consistent
`with target-mediated elimination, where clearance decreases as
`a function of dose (e.g., trastuzumab, rituximab, gemtuzumab,
`and panitumumab). Examples of state-of-the-art mathemati(cid:173)
`cal modeling of target-mediated antibody elimination include
`reports by Ng et al., describing the nonlinear disposition of
`TRX1, an anti-CD4mAb; 16 by Hayashi et al., describing the
`nonlinear disposition of omalizumab, an anti-IgE mAbY and
`by Lammerts van Bueren et al., presenting an interesting con(cid:173)
`ceptual model of target-mediated antibody elimination from a
`peripheral distribution compartment. 18
`IgG antibodies may also interact with Fe-y-receptors (FcyR),
`and IgG-FcyR complexes may trigger endocytosis and catabo(cid:173)
`lism. Considering the relatively high affinity ofigG for FcyR
`and the high endogenous concentrations of IgG in plasma
`( -65 f!mol/1), it has been argued that FcyR-mediated elimination
`is unlikely to be important for monomeric IgG. 3 It is possible
`that FcyR-mediated elimination is significant, and perhaps dom(cid:173)
`inant, in cases where antibody is able to form soluble immune
`complexes containing three or more IgG molecules, as well as
`in cases where antibody binds to cells suspended in blood or
`other body fluids (perhaps including viruses, bacteria, plate(cid:173)
`lets, erythrocytes, and leukocytes). IgG "opsonized" particles are
`rapidly engulfed following engagement ofFcyR on macrophages
`and on other phagocytic cells. This mechanism of elimination
`is well supported by the immunology literature; however, little
`work has been performed to link FcyR-mediated phagocytosis
`to the systemic pharmacokinetics of therapeutic antibodies.
`Additional study is required to allow meaningful discussion of
`the role ofFcyR-mediated endocytosis in the elimination of such
`antibodies.
`IgG, like other proteins found in plasma and interstitial
`fluid, may enter cells in all tissues via fluid-phase endocytosis.
`Interestingly, however, IgG differs from most proteins in that
`a significant fraction of endocytosed IgG is not sorted to the
`lysosome but is redirected to the cell surface and released into
`plasma or interstitial fluids. The recycling of IgG is mediated
`by the Brambell receptor, FeRn, which binds to IgG with pH(cid:173)
`dependent affinity. 19•20 Within the acidified environment of the
`early endosome, IgG binds tightly to FeRn. The IgG-FcRn com(cid:173)
`plexes are not delivered to the lysosome for catabolism but rather
`are sorted to the cell surface for fusion with the cell membrane.
`The receptor shows virtually no affinity for IgG at physiological
`pH and, upon fusion of the sorting vesicle with the cell mem(cid:173)
`brane, IgG dissociates from the receptor and is rapidly released
`into extracellular fluid.
`FeRn-mediated recycling of IgG appears to be quite effi(cid:173)
`cient based on studies conducted with knockout mice. In ani(cid:173)
`mals lacking expression of FeRn, IgG clearance is increased
`
`by approximately tenfold,20 which would be consistent with a
`recycling efficiency of90% (i.e., in wild-type animals express(cid:173)
`ing FeRn). Because FeRn expression is limited, FeRn-mediated
`recycling is capacity limited. The average concentration of IgG
`in plasma in humans is -10 mg/ml. At this concentration, IgG
`has a half-life of-25 days21 and a plasma clearance of -10 ml!h
`( -3.5ml!kg/day). High concentrations ofigG are able to saturate
`the recycling system, decreasing recycling efficiency and leading
`to an increase in the fractional catabolic rate ofigG. For exam(cid:173)
`ple, in myeloma patients, where IgG concentrations in plasma
`may approach 100mg/ml, IgG half-life decreases to 8-10 days.
`Conversely, in patients with very low plasma concentrations of
`IgG, the half-life ofigG antibody may be> 70 days.21
`IgG affinity for FeRn is species specific. Human FeRn shows
`high affinity for human IgG and also for IgG from guinea pigs
`and rabbits; however, the human receptor shows very little affin(cid:173)
`ity for IgG derived from most other species, including mice and
`rats.22 The low affinity of human FeRn for mouse IgG helps to
`explain the very rapid elimination of murine mAbs in humans.
`Approved murine monoclonal IgGs (e.g., muromononab-CD3,
`ibritumomab) demonstrate half-lives of -1 day in patients,
`whereas human IgG is typically associated with a half-life of
`-25 days.
`Although FeRn recycling is capacity limited, significant altera(cid:173)
`tion in the efficiency of FeRn recycling is not typically achieved
`with therapeutic doses of mAbs. Most mAbs are administered at
`doses of< 10 mg/kg, which will increase the total IgG "body load"
`by< 1-2%, as humans typically possess 50-100 g of endogenous
`IgG. However, high-dose intravenous immunoglobulin (IVIG)
`therapy, which calls for the administration of 2 g/kg of pooled
`human IgG, increases IgG plasma concentrations sufficiently to
`increase IgG clearance approximately threefold. 23 This increase
`in IgG clearance leads to a decrease in endogenous antibody
`concentrations; consequently, IVIG therapy for the treatment of
`autoimmune conditions may achieve effects by decreasing the
`plasma concentrations of endogenous, pathogenic autoantibod(cid:173)
`ies. Although IVIG therapy is an effective treatment of a variety
`of autoimmune conditions, it is very expensive because of the
`high doses of antibody required. Of note, preclinical experiments
`have demonstrated that anti-FeRn antibodies are able to achieve
`effects similar to those ofiVIG therapy, at dose levels that are
`-100-fold lower than those required for use in IVIG therapy.24
`There is significant interest in the development of specific FeRn
`inhibitors for use in the treatment of autoimmunity.25
`
`IMMUNOGENICITY
`Any exogenous protein may be viewed by the body as foreign
`and trigger immune responses that lead to the generation of
`endogenous antibodies against the protein. Therapeutic anti(cid:173)
`bodies are no exception. mAb drugs may be categorized as
`(i) rodent antibodies, which are typically obtained from murine
`or rat hydridomas; (ii) chimeric antibodies, which are derived
`from chimeras that have been engineered to express IgG antibod(cid:173)
`ies with human constant regions and rodent variable regions; (iii)
`CDR-grafted antibodies, which contain specific regions within
`rodent variable domains, the CDRs, grafted onto a human IgG
`
`552
`
`VOLUME 84 NUMBER Sl NOVEMBER 20081 www.nature.com/cpt
`
`

`

`function was employed to describe the formation of antibody(cid:173)
`ligand complexes, and complexes dissociated via a first-order
`process. The models are nonlinear with respect to antibody(cid:173)
`ligand binding because of the second -order nature of the binding
`process, as well as capacity limitations associated with the avail(cid:173)
`able concentrations ofligand and antibody. The models relate
`unbound ligand concentration to the effect of interest; as such,
`the models link the PK effects of the anti -ligand antibody to the
`pharmacodynamics of the ligand. Of note, the models differ sub(cid:173)
`stantially in terms of their characterization of the fate of the anti(cid:173)
`body-ligand complex. In the infliximab model, it was assumed
`that the antibody-ligand complex was eliminated with the same
`fractional catabolic rate as the unbound ligand, tumor necrosis
`factor-a. Fitting the model parameters to the data resulted in an
`estimation of a 30-40-day half-life for tumor necrosis factor-a,
`which is considerably different from the known value ( < 1 h).
`The omalizumab model, which is much more plausible, does not
`assume an equivalent elimination rate constant for the complex
`and the ligand (IgE) but allows for kinetically distinct elimination
`ofigE, omalizumab, and the IgE-omalizumab complex.
`The recent work ofMarathe et al., which describes the PK/
`PD of denosumab, a monoclonal IgG2 antibody directed
`against the receptor activator of nuclear factor-KB ligand, rep(cid:173)
`resents the state of the art in modeling immunotoxicothera(cid:173)
`pies (Figure 2). 40 The receptor activator of nuclear factor-KB
`ligand is thought to be a soluble ligand, but there is some pos(cid:173)
`sibility that the protein is also expressed on the cell surfaces.
`Denosumab pharmacokinetics were captured with a target(cid:173)
`mediated disposition model, and denosumab pharmacody(cid:173)
`namics were described with a model that relates the unbound
`concentrations of denosumab to the inhibitory effect of the
`antibody on the receptor activator of nuclear factor-KB ligand
`binding. This mechanistic model provided an excellent descrip(cid:173)
`tion of the pharmacokinetics and pharmacodynamics of deno(cid:173)
`sumab in multiple myeloma patients.
`Several antibodies, including rituximab, cetuximab, and tras(cid:173)
`tuzumab, are designed to bind to cell-surface proteins to mediate
`
`Denosumat>-RANKL
`
`II
`~
`
`Active _L__ RANKL
`losreoblaSIS)
`
`OPG-RANKL
`
`rc;;,;oclas~
`r~u~
`
`l
`
`RANK-RANKL
`
`"A;;;
`(osteoclasiS 1
`Serum NXT ~~~~- -
`..
`
`such as volume of each compartment (organ) and blood flow
`rate, are used to build the model and predict human pharma(cid:173)
`cokinetics. A significant advantage of the PBPK approach is that
`it allows prediction of antibody levels in many tissues, including
`tumor. PBPK models are ideally suited to the consideration of
`effects of saturable processes (e.g., target binding, FeRn process(cid:173)
`ing) on antibody pharmacokinetics, and these models are also
`well suited to predict the influence of a variety of factors (e.g.,
`antigen expression, antibody affinity) on the tissue selectivity of
`antibody disposition. Recent PBPK models have incorporated
`FeRn -antibody binding, allowing consideration of the effects
`of FeRn on antibody catabolism and distribution. 11 •12 The limi(cid:173)
`tations for use of PBPK models to predict the disposition of
`antibodies in humans are significant, however. PBPK models
`are complex, mathematically difficult to construct, poorly suited
`to population analyses, and often limited because of a lack of
`tissue concentration data, parameter availability, or parameter
`identifiability.
`Despite the large number of antibodies in development, only
`a handful of reports using preclinical data to predict the clinical
`pharmacokinetics of antibodies have been published, perhaps
`indicating the difficulties associated with the interspecies scaling
`of antibody disposition. Any effort to predict human pharmacoki(cid:173)
`netics based on preclinical disposition data should consider pos(cid:173)
`sible species differences in the expression or turnover of the target
`receptor, antibody affinity for the target, antibody-FeRn binding,
`endogenous IgG concentrations (i.e., as a determinant of FeRn
`saturation), and potential effects of host anti-drug antibodies.
`
`PHARMACODYNAMICS
`mAbs have been marketed for use in the treatment of a wide
`range of conditions, including cancer, autoimmunity, and
`inflammatory diseases. It is convenient to discuss antibody
`pharmacodynamics relating to four main categories of applica(cid:173)
`tions: (i) immunotoxicotherapy, where antibody is employed to
`alter the pharmacokinetics and pharmacodynamics of soluble
`ligands (e.g., drugs, xenobiotics, and cytokines); (ii) elimination
`of target cells; (iii) alteration of cellular function (e.g., receptor
`blockade); and (iv) targeted drug delivery.3
`Antibodies used for immunotoxicotherapy include bevacizu(cid:173)
`mab, adalimumab, ranibizumab, omalizumab, and infliximab.
`Each of these antibodies binds to a soluble ligand (e.g., vascular
`endothelial growth factor or tumor necrosis factor) and alters
`the pharmacokinetics and pharmacodynamics of the ligand.
`These "neutralizing" antibodies act as competitive inhibitors of
`ligand-receptor binding, shifting ligand concentration-effect
`relationships. In addition, by binding to soluble ligand, immu(cid:173)
`notoxicotherapies often produce dramatic alterations in ligand
`pharmacokinetics. In most cases, the anti-ligand antibody will
`decrease the unbound fraction of ligand in plasma, decrease the
`ligand volume of distribution and clearance, and increase the half(cid:173)
`life of the ligand. For example, omalizumab, an anti-IgE mAb,
`dramatically decreases the clearance of its target ligand, leading
`to a fivefold increase in the plasma half-life ofigE.
`PK/PD models for omalizumab and infliximab have been pub(cid:173)
`lished recently. 38•39 In each model, a second-order association
`
`Figure 2 Pharmacodynamic model for denosumab. Marathe eta/. provide
`an excellent example of a pharmacodynamic model for an antibody acting
`as an antagonist of a soluble ligand.40 Denosumab, like other antibodies
`used for immunotoxicotherapy, binds to a soluble ligand (receptor activator
`of nuclear factor-KB ligand, RANKL), preventing the ligand from binding to
`its endogenous receptor (receptor activator of nuclear factor-KB, RANK),
`and antagonizing the effect of the ligand (i.e., inhibiting RANKL stimulation
`of osteoclast maturation). The Marathe eta/. model, which has been
`simplified herein, employs equilibrium binding functions to relate plasma
`concentrations of denosumab, RANK, and the natural RANKL antagonist
`(osteoprotegrin, OPG) to unbound concentrations of RANKL, and to the
`measured biomarker (serum N-telopeptide, NTX).
`
`554
`
`VOLUME 84 NUMBER Si NOVEMBER 20081 www.nature.com/cpt
`
`

`

`V m • efalizumab.CD11 a
`Vc • Km + elalizumab
`
`CD11a
`production
`
`Figure 4 Pharmacodynamic model forefalizumab. The Ng eta/. model,
`selected as an example model for antibodies that alter cellular function,
`describes the effects of efalizumab on CD11 a expression, elimination, and
`the psoriasis area and severity index (PAS I). The model allowed excellent
`characterization of efalizumab pharmacodynamics in a large, population
`pharmacokinetic and pharmacodynamic analysis. The figure shown is a
`simplified version of the model presented by Ng eta/.48
`
`the psoriasis area and severity index, an efficacy end point for
`psoriasis (Figure 4).48
`Preclinical modeling examples include a report by Luo et al.
`that describes the development of a model of cetuximab PK/
`PD using data collected from studies conducted with a murine
`colon carcinoma xenograft model. 49 Although the antibody
`demonstrates nonlinear, target-mediated disposition in humans,
`cetuximab pharmacokinetics were dose-proportional in the
`mouse model and were well characterized with a linear, one(cid:173)
`compartment model. Cetuximab's effects on the phosphoryla(cid:173)
`tion of the epidermal growth factor receptor were captured with
`an indirect effect model, which allowed comparison between
`estimated values of EC50 and EC90 (half maximal effective
`concentration and 90% effective concentration, respectively),
`plasma concentrations of cetuximab achieved in patients, and
`the efficacy of cetuximab in clinical trials.
`In comparison with the other main categories of antibody
`usage, relatively little success has come from the development
`of antibodies for targeted drug delivery. Most of the interest
`in this area has centered on the development of conjugates
`of antibodies and toxic agents (e.g., chemotherapeutic drugs,
`radioisotopes, and biological toxins), with the intent of using
`the high specificity and selectivity of antibodies to mediate
`targeted delivery of toxins. The antibody-toxin conjugates, or
`immunotoxins, carry the complexities shared by other types of
`antibody drugs (e.g., potential for nonlinear target-mediated
`disposition, immunogenicity) . In addition, off-target toxic(cid:173)
`ity is often a greater concern for immunotoxins because of
`the potential for dissociation, in vivo, of the toxin from the
`antibody and because of the high potency of toxins employed.
`Considerable toxicity often results from "nonspecific" distribu(cid:173)
`tion of the immunotoxin to off-target sites. Bone marrow stem
`cells are particularly susceptible to toxicity from immunoto(cid:173)
`xins because of their rapid growth rate and high sensitivity to
`chemotherapy, along with the leaky vasculature of the bone
`marrow, which allows relatively efficient convective uptake of
`immunotoxins.
`
`Most of the work associated with the use of antibodies for
`targeted drug delivery has been focused on the treatment of
`solid tumors. Solid tumors are problematic targets for anti(cid:173)
`body drugs, as tumor growth often leads to the collapse oflym(cid:173)
`phatic vessels within the tumor, which leads to an increase in
`the tumor interstitial pressure. High interstitial pressure mini(cid:173)
`mizes the blood-to-tumor hydrostatic pressure gradient, and
`this decreases the driving force for antibody uptake into tumor
`by convection. Once antibody extravasates, distribution may be
`limited by the binding-site barrier (discussed above), further
`reducing the effectiveness of antibody-directed delivery of toxins
`to solid tumors.
`For chronic immunotoxin therapy, it may be important to
`select a cellular target that is easily accessed by antibody in blood
`(i.e., hematological cells, cells in tissues with "leaky" vascula(cid:173)
`ture), antibodies with little risk for immunogenicity, toxins with
`little risk for immunogenicity (e.g., protein toxins such as ricin
`would not be desired), and conjugation chemistry that allows for
`little off-target release of toxin, but where there is efficient release
`of toxin within target cells (i.e., in cases where this is required for
`efficacy). Successfully marketed antibodies include gemtuzumab
`ozogamicin, tositumomab, and ibritumomab tiuxetan. In each
`case, the antibodies target hematological cells. Tositumomab
`and ibritumomab utilize radioisotope toxins, where dissociation
`from the antibody is not required for the desired cytotoxic effect.
`Gemtuzumab ozogamicin employs a calicheamicin derivative
`toxin that is released in target cells after binding of gemtuzu(cid:173)
`mab to the target receptor (CD33) and after receptor-mediated
`endocytosis of the immunotoxin. The toxin migrates to the
`nucleus and binds DNA, leading to double-strand breaks and
`cell death.
`There are few publications of PK/PD models for immuno(cid:173)
`toxin therapies. Ideally, mathematical models of immunotoxin
`pharmacokinetics and pharmacodynamics should account for
`the intact immunotoxin, "naked" antibody (i.e., antibody alone,
`following release of the toxin), and "free" toxin. In an interest(cid:173)
`ing example, Zhu et al. applied physiologically based modeling
`and simulation to investigate relationships between the dose of
`radioimmunotoxins and uptake of the conjugates into tissue. 50
`Their modeling led to the conclusion that Fab fragments would
`be preferred for use in detection of tumors, whereas Fab2 frag(cid:173)
`ments were predicted to be more effective for use in radioimmu(cid:173)
`notherapy. 50 The structure of their PBPK model may be easily
`adapted to the prediction and characterization of the PK/PD of
`additional immunotoxins.
`
`CONCLUSIONS
`Antibody drugs demonstrate unique, complex PK characteristics.
`Absorption following IM or SC administration is slow and, for
`some antibodies, dose dependent. Antibody distribution kinetics
`is influenced by rates of convective transport, binding to tissue
`sites, and rates of catabolism within tissue. Traditional noncom(cid:173)
`partmental analyses and mammillary models may underestimate
`the steady-state distribution volume of many antibodies, partic(cid:173)
`ularly those associated with substantial elimination from tissue
`sites .. Antibodies often demonstrate target-meditated disposition,
`
`556
`
`VOLUME 84 NUMBER 5 I NOVEMBER 20081 www.nature.com/cpt
`
`

`

`42. Meijer, R.T., Koopmans, R.P., ten Berge, IJ. & Schellekens, P.T.
`Pharmacokinetics of murine anti-human CD3 antibodies in man are
`determined by the disappearance of target antigen.). Pharmacal. Exp. Ther.
`300, 346-353 (2002).
`43. Mould, D.R. eta/. A p

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