throbber
EXHIBIT /7
`WIT. kR -ov
`DATE 3.tya-
`KRAMM COURT REPORTING
`
`Therapeutics, Targets, and Chemical Biology: Mather,,.,,-
`
`Increase of Plasma VEGF after Intravenous Administration
`of Bevacizumab Is Predicted by a Pharmacokinetic Model
`
`Marianne 0. Stefanini 1, Florence T. H. Wu', Feilirn Mac Gabhann2, and Aleksander S. PopeI 1
`
`(A) Check for updates
`
`Cancer
`Research
`
`Downloaded from hltp://aactioumal
`
`rljde-pdf!70123/98&6!2644719/9386pdf by guest on 03 Marelt 2022
`
`Abstract
`Vascular endothelial growth factor (VEGF) is one of the most potent cytokines targeted in antiangiogenic
`therapies. Bevacizumab, a recombinant humanized monoclonal antibody to VEGF, is being used clinically in
`combination with chemotherapy for colorectal, non-small cell lung and breast cancers, and as a single agent for
`glioblastoma and is being tested for other types of cancer in numerous clinical trials. It has been reported that
`the intravenous injection of bevacizumab leads to an increase ofplasma VEGF concentration in cancer patients.
`The mechanism responsible for this counterintuitive increase has not been elucidated, although several
`hypotheses have been proposed. We use a multiscale systems biology approach to address this problem.
`We have constructed a whole-body pharmacokinetic model comprising three compartments: blood, normal
`tissue, and tumor tissue. Molecular interactions among VEGF-A family members, their major receptors, the
`extracellular matrix, and an anti-VEGF ligand are considered for each compartment Diffusible molecules
`extravasate, intravasate, are removed from the healthy tissue through the lymphatics, and are cleared from the
`blood. Cancer Res 7q'23); 9886-94. ©2010 zIACR.
`
`Major Findings
`
`Our model reproduces the experimentally observed
`increase of plasma VEGF following intravenous adminis-
`tration of bevacizumab and predicts this increase to be a
`consequence of intercompartmental exchange of VEGF,
`the anti-VEGF agent and the VEGF/anti-VEGF complex.
`Our results suggest that a fraction of the anti-VEGF drug
`extravasates, allowing the agent to bind the interstitial
`VEGF. When the complex intravasates (via a combination
`of lymphatic drainage and microvascular transport of
`macromolecules) and dissociates in the blood, VEGF is
`released and the VEGF concentration increases in the
`plasma. These results provide a new hypothesis on the
`kinetics of VEGF and on the VEGF distribution in the body
`caused by antiangiogenic therapies, as well as their
`mechanisms of action and could help in designing anti-
`angiogenic therapies.
`
`Authors' Affiliations: 'Department of Biomedical Engineering, and 2lnsti-
`tute for Computational Medicine and Department of Biomedical Engineer-
`ing, Johns Hopkins University, Baltimore, Maryland
`Note: Supplementary data for this article are available at cancer Research
`Online lhttp:/Jcancen'es.aacrjournals.org.
`Corresponding Author. Aleksander S. Popel, Department of Biomedical
`Engineering, Johns Hopkins University School of Medicine, 720 Rutland
`Avenue, 611 Traylor Research Building, Baltimore, MD 21205. Phone:
`410-955-6419; E-mail; apopel@jhu.edu.
`doi: 10.115B10008-5472.cAN-1O-1419
`C201 American Association for cancer Research.
`
`9886
`
`Cancer Res; 70(23) December 1, 2010
`
`Introduction
`
`VEGF is a key factor in tumor angiogenesis, and it has
`become a major target of antiangiogenic cancer therapy (1). A
`large body of evidence suggests that the free plasma VEGF
`concentration is elevated several fold in cancer patients
`compared to healthy subjects (2). Therapies targeting VEGF
`have shown promising results in cancer. Bevacizumab (Avas-
`tin, Genentech Inc.), a recombinant humanized monoclonal
`antibody to VEGF, has demonstrated efficacy in colorectal
`cancer, non-small cell lung cancer, breast cancer, renal cell
`carcinoma, and glioblastoma. The drug has been approved by
`the Food and Drug Administration (FDA) for these indications
`under certain conditions in combination with chemothera-
`peutic agents and is being tested for other types of cancer and
`other conditions in numerous clinical trials.
`Despite the growing clinical applications of bevacizumab,
`the mechanism of action of this anti-VEGF agent and that of
`other anti-VEGF large molecules is not sufficiently understood
`(3). Specifically, two important questions remain: whether the
`drug acts by sequestering VEGF in the blood, tumor inter-
`stitium or both; and whether, as a result, the VEGF concen-
`tration in these compartments is reduced to 'normal' levels.
`Answering these questions would significantly contribute to
`understanding the mechanism of action not only at the
`molecular level, but also at the levels of tissue, organ, and
`whole body and would help in the design of anti-VEGF agents.
`Gordon and colleagues reported that the intravenous injection
`of bevacizumab led to an increase in serum total VEGF in
`clinical trials whereas free VEGF concentration was reduced
`(4). Since then, other groups have reported counterintuitive
`increases in the plasma VEGF level following bevacizumab
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 1
`
`

`

`Downloaded from htlp:f/aacsoumalsorg/canceres/arlide-pdf/7W23!98S6Q6447l9!9886pdf by guest on 03 March 202
`
`Increased Plasma VEGF Predicted by a Phanmacokinetic Model
`
`Quick Guide to Equations and Assumptions
`
`Key Equations
`
`The molecular detailed compartmental model is described by nonlinear ordinary differential equations on the basis of the
`principles of chemical kinetics andbiological transport (summarized in Supplement l).The following example equation describes
`the changeover time of the concentration of vascular endotlielial growth factor VEGF 121 isoform in the interstitial space of the
`normal tissue, denoted by the subscript N. The blood compartment is denoted by the subscript B.
`
`
`
`dt
`
`- N
`Will - krn,v121,Rl [VI2flv[RI] -+k011 y121R1 [VI2IRI]N
`
`- k00vi2ie2IVi2i],.IR2]+k0ijvi2iit2[Vi2iR2]
`- k0n,vl2,,,jNI [V12d [R 1 .WI] v+kofLvl2rnm-1 [V111 R1 A'i1
`
`-
`
`(kL + k9SNs\ Vi2iJç
`UN
`)CAr..v +kvUu[l2I)B
`
`The right-hand side terms represent: secretion of VEGF 121 isoform (qv)2,); binding to VEGF 12 , to its receptors (VEGFRI and
`VEGFR2) and to the complex VEGFRl/NRPt; binding of VEGI? i21 to the anti-VEGE agent A; and the intercompartmental
`transport of VEGF, 2, by lymphatics (1<,) and microvascular permeability to macromolecules (kr). S 8 and KAvNrepresent the
`total surface of microvessels at the normal tissue/blood interface and the available volume fraction for VEGF III in the total
`volume U, respectively. The total volumes are denoted by U. The subscript p in tJ denotes plasma as distinct from blood.
`Note that, with this nomenclature, the ratio (4,/UB represents the available fluid volume fraction for VEGF, 21 in the blood.
`The injection ofthe anti-VEGI'agent occurs after establishment of a physiologic steady state (t<0).At t = 0, the anti-VEGF agent is
`administered intravenously at a rate q,, for a duration at
`00 (typically in minutes). The subscript T represents the tumor. The
`equation governing the change of the antiVEGF agent concentration in the blood over time reads:
`
`-
`
`'lA cA[A]n_kPV &iA]n+(j) .
`
`-
`
`B STD [Al T
`A
`pv L/ [ lR + k pv
`p
`LiE "MIT
`
`- km,v121,A[VIIIID[A]a+koff,vIIIA[VIIIA]a
`
`-
`
`and q,, = 0 for all other times in = number of
`where q,, = total dose/(n x a• nr,aon) during the duration of each treatment
`injections). The first two terms on the right-hand side are the intravenous infusion of ar,ti-VEGF at rate q4 and the clearance of anti-
`VEGF from the blood at a rate CA. The next terms represent. drug extravasation; removal of anti-VEGF agent bylymphatics; and drug
`intravasation (when the intercompartnient transports are included). The last two terms describe the binding of the anti-VEGF agent
`to both VEGE isoforms.
`As a final example, the change over time of the corresponding VEGF/anti-VEGF concentration in the normal tissue when the
`extravasation of the anti-VEGE agent is governed by
`
`d[V]IIA]N
`dt
`
`IN
`
`(/c. + k?'V2SNfl) 1Vi214]N + kEN SND U3
`
`-
`
`UN
`
`KAVN
`
`pV
`
`[V 2 A] 8
`
`
`and is dependent on: VEGF, 21 binding to the anti'VEGF agent and transport of the VEGF/anti-VEGF complex between the
`compartments.
`
`Major Assumptions of the Model
`
`Our model does not represent a particular stage or type of cancer to keep the model general in light of the fact that
`bevacixumab is administered in primary and metastatic diseases and in adjuvant or neaadjuvant settings. Therefore, our tumor
`compartment can either be a primary tumor or the aggregate of metastases in tissue.
`
`www.aacrjoumals.org
`
`Cancer Res; 70(23) December 1, 2010
`
`9887
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 2
`
`

`

`Stefanini at al.
`
`Because the simulation results for a smaller tumor (half the diameter of the tumor considered in this study) were not
`significantly different (both qualitatively and quantitatively--data not shown), our model does not consider the possible change
`in tumor volume that may result from the injection of the anti-VEGF agent for the duration of our simulations.
`The degradation of VEGF by proteases is not currently included in the model. Effects of platelets and leukocytes as potential
`sites for sequestering VEGF, antiVEGF, and their products are not considered and should also be added in the future. We assume
`that only endothelial cells express VEGF receptors. Our model does not include the presence of receptors on the luminal surface
`of endothelial cells and the quantification of abluminal receptors has been estimated from previous studies.
`The model does not include multimeric binding of the anti-VEGF or the ability of the anti-VEGF to bind to matrix-bound
`VEGF. We assume that the anti-VEGF has a half-life of 21 days. Its complexes formed by the binding of VEGF,,, or VEGF, are
`assumed to have the same half-lives because hound and free bevacizumab exhibit the same pharmacokinetic profile. The binding
`and unbinding rates of the anti-VEGF to VEGF are taken from the literature to be 9.2 x iO4 (mol/L)'-C' and 2.0 x 10 s'
`respectively leading to a dissociation constant Kdof 2.2 nM. The above assumptions can be relaxed, if warranted by experimental
`data, within the framework of the model that is generally suitable for simulating anti-VEGF therapeutics.
`
`aded from htp:!/aactjournaIsorg/canrresJartide-pdfI7OI23I9886a64471I9886.pdf by guest on 03 M
`
`isoforms and placental growth factor (PIGF). This fusion
`protein serves as a soluble decoy receptor and is currently in
`clinical trials.
`Our model includes two VEGF-A isoforms (VEGF 121 and
`VEGF 165), as well as VEGF receptors (VEGFI11 and VEGFR2)
`and the coreceptor neuropilin-1 (NRPI). In this study, we
`assume that VEGFRI, VEGFR2, and NEPI are present only on
`the abluminal surface of the endothelial cells. The transcap-
`illary microvascular permeability for the diffusible molecules
`(VEGF, anti-VEGF, and the VEGF/anti-VEGF complex) is
`included, as well as lymphatic drainage from the interstitial
`space into the blood compartment. The model equatiOns are
`presented in the Supplemental Information (Supplement I).
`
`Materials and Methods
`
`Most of the parameters for the anti-VEGF agent were taken
`from published data on hevacizumab. We assume a half-life of
`21 days (4) for the anti-VEGF whether unbound or bound to
`VEGF J2, or VEGF 165, as bound and free bevacizumab exhibit
`the same pharmacokinetic profile (9). Kinetic parameters (k0 ,
`k0ff) for the binding and unbinding of the anti-VEGF to the
`vascular endothelial growth factor were taken to be 9.2 >< io
`(molfL)'-s' and 2.0 x iO 4 s- ' respectively, leading to a
`dissociation constant K,, of 2.2 nM (17).
`Experiments have shown that bevacizumab may have mul-
`timeric binding to VEGF (9, 18) and can bind to extracellular
`matrix-sequestered VEGF (19). For simplicity purposes, we
`limit our model to monomeric binding to VEGF and neglect
`binding to VEGF sequestered by the extracellular matrix; these
`can be included when quantification of binding sites and the
`kinetics become available. Bevacizumab has also been
`reported to alter the VEGF-dependent microvascular perme-
`ability to soluble molecules (20). As a first approximation, we
`assume that the geometry of each tissue and the capillary
`density remains constant in the course of our simulations, that
`is, we do not include tissue remodeling after the injection of
`the anti-VEGF agent. Although it may be important, the
`inclusion of tissue remodeling would take the model beyond
`the scope of this study but could be of interest for further
`studies. This model does not include VEGF receptors on the
`luminal side of endothelial cells that have not been experi-
`
`administration (5-7). In the ocular setting, Campo and col-
`leagues reported that intravitreal bevacizumab injection
`increased the VEGF concentration in the aqueous humor
`(8). Several hypotheses have been formulated to explain this
`phenomenon. l-lsei and colleagues have suggested that the
`clearance of complexed VEGF is lower than that of free VEGF
`in rats and hypothesized that this lower clearance could
`explain the accumulation of total VEGF in serum (9). Other
`groups have suggested alternate pathways activated by the
`injection of bevacizumab, such as: accumulation of hypoxia-
`inducible factor leading to an increase of VEGF in serum, or
`secondary macular edema for the eye (8, 10, 11). Loupakis and
`colleagues immunodepleted plasma to remove bevacizumab
`and bevacizumab-VEGF complexes and found that plasma-
`free VEGF was significantly reduced after bevacizumab
`administration (12); this methodology helps to circumvent
`the problem that the ELISA method used in a number of
`studies cannot distinguish between free and total (including
`bevacizurnah-bound) VEGF. The results of the study corro-
`borate an earlier proposal by Christofanilli and colleagues (13)
`that free VEGF can serve as a surrogate marker.
`Systems biology approaches, specifically computational and
`mathematical modeling, are emerging as powerful tools in
`fundamental studies of cancer and design of therapeutics (14,
`15). To better understand VEGF distribution in the body, we
`have built a three-compartment model composed of normal
`(healthy) tissue, blood, and tumor (16). In this study, we have
`extended our computational model by including an anti-VEGF
`agent delivered by the intravenous infusion (i.e., into the blood
`compartment). The model describes the effect of such admin-
`istration on the VEGF distribution in the blood, normal, and
`diseased tissues. Our goal is to understand how the distribu-
`tion of VEGF, anti-VEGF agent, and their products changes
`following the agent administration: in particular, we will
`investigate whether the plasma VEGF level increases or
`decreases following an intravenous injection of the anti-VEGF
`agent.
`Even though the results are presented using the para-
`meters for bevacizumab, the model can be applied to other
`anti-VEGF agents. One such agent is aflibercept or VEGF
`Trap (Regeneron Pharmaceuticals Inc.), a soluble huma-
`nized VEGF receptor protein designed to bind all VEGF-A
`
`9888
`
`Cancer Pea; 70(23) December 1, 2010
`
`Cancer Research
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 3
`
`

`

`Downloaded trot,, hllp:/IaacrJournalsorg)cancorres/artde-pdf/7W239886)2644719/9886.pdt by guest on 03 Moron 2022
`
`Increased Plasma VEGF Predicted by a Pharrnacokinetic Model
`
`If the anti-VEGF agent is confined to the blood compart-
`ment, a single injection causes the concentration of free VEGF
`(i.e., not bound to anti-VEGF) in plasma to decrease precipi-
`tously by 98.4% (Fig. IA, dashed line; minimum as the infusion
`ends), as the anti-VEGF agent binds to VEGF available in
`plasma. However, this is not predicted to significantly affect
`the free VEGF level in the healthy tissue (maximum 0.1% drop
`at 9 hours (solid line)] or the free VEGF level in the tumor
`compartment [maximum 0.2% drop at 30 hours (dotted line)].
`The free anti-VEGF agent saturated the blood (Supplementary
`Figure SM) and reached a maximum of -4.7 Ismol/L in
`plasma (-8S ig/mL plasma) at the end of the infusion, which
`corresponds to the total injected amount of the 150-kDa agent
`distributed in the volume of plasma for a 70-kg patient. The
`VEGF/anti-VEGF complex reached its maximum concentra-
`tion in the blood (--'2.1 nmol/L) alter about 12 days (Supple-
`mentary Figure S2A). The total (free and bound to the anti-
`VEGF agent) VEGF concentration is typically what is mea-
`sured by VEGF ELISA methods (see Supplement 2 for a
`compilation of experimental data on free/total VEGF changes
`following bevacizuniab administration). Our results show a
`100- to a 1,000-fold difference between free VEGF concentra-
`tion (Fig. I) and the concentration of VEGF bound to the anti-
`VEGF (Supplementary Figure 82). Because of this difference in
`magnitude, the unbound VEGF concentration represents only
`a small percentage of the total VEGF concentration, and thus
`Supplementary Figure 52 also illustrates the total VEGF con-
`centration profile.
`For metronomic therapy (lower daily dose of 1 mg/kg over
`10 days), the free VEGF in plasma declines 86.8% following the
`first infusion, but is predicted to reach a pseudo-steady state
`after multiple infusions (Fig. IC, dashed line). The concentra-
`tion of free VEGF returned to its baseline level within 3 weeks
`once the treatment was stopped. Metronomic therapy showed
`delayed and lowered maximum levels of anti-VEGF compared
`to the single-dose regimen (Supplementary Figure SIC versus
`Supplementary Figure SIA); although the half-life of the anti-
`VEGF agent is relatively long, it is being cleared from plasma
`continuously. The VEGF/anti-VEGF complex (and therefore
`the total VEGF concentration) reached its maximum about a
`week later than for the single dose (Supplementary Figure S2C
`versus Supplementary Figure S2A).
`
`For an anti-VEGF agent that extravasates, plasma-free
`vIcl: is predicted to first decrease and then increase
`above the baseline level
`As for a nonextravasating anti-VEGF agent (Fig. IA),
`plasma-free VEGF decreased (97.0% drop in the first 45
`minutes) following administration of anti-VEGF that can
`extravasate (Fig, 18, dashed line), as the agent binds to the
`available free VEGF. In this case, however, VEGF concentra-
`tion then rebounded to 41.1 pmol/l. (a 9.1-fold increase over
`baseline) after about I week. Unlike the no-extravasation case
`where the concentration returned to baseline after 3 weeks,
`the free VEGF concentration in plasma was predicted to
`remain significantly elevated after 3 weeks (40.5 pmol/L, 9-
`fold the baseline level). The free VEGF concentration in the
`normal (solid line) and tumor (dotted line) tissues both also
`
`mentally characterized, but we have recently shown how such
`expression would alter the VEGF distribution (21); we do not
`expect that any qualitative conclusions of the study would be
`affected by the presence of luminal receptors.
`Note that the simulations are not aimed at representing a
`particular type or stage of cancer, recognizing that VEGF-
`neutralizing agents may be administered in cases of both
`metastatic and primary tumors. Thus, in the model, the tumor
`compartment can represent either an aggregate volume of
`metastases or a primary tumor. Due to the wide range of
`possibilities that could be represented for different types and
`stages of cancer, we adopt the parameters for this compart-
`ment from our previous study (16) and conduct a sensitivity
`study to ascertain that our qualitative conclusions are not
`dependent on the choice of parameters.
`For each simulation, the system was first equilibrated at a
`baseline for a cancer patient with tumor before the injection of
`the VEGF-neutralizing agent. At time zero, intravenous infu-
`sion of the anti-VEGF agent begins and delivery to the blood
`compartment continues as a slow infusion for 90 minutes. We
`considered two treatment regimens: a single-dose treatment
`of 10 mg/kg or 10 consecutive daily doses of I mg/kg (metro-
`nomic therapy).
`The parameters and their assigned numerical values are
`summarized in Supplement 3. The equations governing the 3-
`compartment VEGF transport system have been described in
`our previous papers (16,21) and can be found in Supplement I.
`We have also added equations to describe the interactions and
`intercompartmental transport of the anti-VEGF molecule
`(Equations S.30-S.38).
`
`Results
`
`Experiments demonstrate an inverse relationship between
`microvascular permeability and the size of a molecule (mole-
`cular weight or Stokes-Einstein radius; refs. 22-24). Therefore,
`in the absence of active transport, large proteins, such as anti-
`VEGF agents (150 kDa for bevacizumab and 110 kDa for
`ailibercept), should extravasate relatively slowly. In apparent
`agreement with this, the level of bevacizumab following an
`intravenous injection has been observed to be several times
`lower in normal tissues (25) and in tumors (19) than in the
`blood. However, little is known about what role, if any, the
`extravasation of an anti-VEGF agent may play in the ther-
`apeutic mechanism. To address this issue, we considered two
`computational scenarios: in the first, the anti-VEGF agent is
`constrained in the blood compartment (negligible extravasa-
`tion); in the second, the extravasation of the anti-VEGF agent
`is included.
`
`Plasma-free VEGF is predicted to decrease following
`intravenous injection of an anti-VEGF agent confined
`to the blood compartment (no extravasation)
`Changes in plasma- and tissue-free VEGF are summarized
`in Fig. 1A for a single-injection (10 mg/kg) and Fig. IC for
`metronomic therapy (1 mg/kg daily for 10 days), that is,
`repeated lower doses over a longer period of time (26). Total
`amount of drug injected is the same in both scenarios.
`
`www.aacrjournals.org
`
`Cancer Res; 70(23) December 1, 2010
`
`9889
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 4
`
`

`

`0
`
`7
`
`14
`
`21
`
`Time (days)
`
`50
`
`- -n
`
`(V
`
`0
`
`0 -
`
`Stefanini at al.
`
`C 200
`
`150
`
`100
`E
`0. 50
`
`Single injection (10mg/kg)
`
`Metonomic therapy (1 mg/kg)
`
`No extravasation of the anti-VEGF agent
`
`axnvasation of the anti-VEGF agent included
`
`200
`
`150
`
`too
`
`50
`
`0
`
`0 1
`
`B 200
`
`150
`
`100
`
`50
`
`D 200
`
`ISO.
`
`Figure 1. Free VEGF concentration profiles following the intravenous injection of an anti-VEGF agent. A and B, single injection (10 mg/kg). C and D,
`daily injection of 1 mg/kg for 10 days (metronomic therapy). One pmot'L of VEGF equivalent to 24 pg/mL of total blood. Solid line, normal tissue; dashed line,
`blood; dotted line: tumor.
`
`showed an initial transient decrease (58.596 and 88.9%, respec-
`tively), followed by a slight rebound, reaching steady states
`6S% and 69.7% below baseline, respectively. This could be due
`to the long half-life of the anti-VEGF agent (21 days) as
`compared to the characteristic times of clearance, binding
`affinities, and internalization rates of VEGF receptors. This
`may suggest that an important whether not the primary action
`of the anti-VEGF agent is to deplete the tumor VEGF after the
`anti-VEGF extravasation. Interestingly, the increase of free
`VEGF in plasma is also predicted even in the absence of a
`tumor compartment (data not shown).
`Supplementary Figure SIB shows the dynamic response of
`the free anti-VEGF agent concentration. Upon injection, the
`free anti-VEGF concentration at first increases but then
`decreases rapidly within the next 12 hours as it travels to
`the normal and tumor tissues. Interestingly, the free anti-
`VEGF concentrations in the blood and in the tumor were
`almost identical. This was mainly due to the higher micro-
`vascular permeability and the absence of functioning lympha-
`tics in the tumor. The formation of the VEGF/anti-VEGF
`
`complex (and the total VEGF concentration) reached a max-
`imum after about 4 days and was significantly higher in the
`tumor than in the other compartments due to higher VEGF
`concentrations (Supplementary Figure S2B).
`In metronomic therapy (Fig. 1D), similar results were
`observed. The free VEGF concentration decreased in the
`plasma upon the anti-VEGF injection then rebounded and
`increased further after each injection (dashed line). In the
`healthy and tumor compartments (solid and dotted lines,
`respectively), a decrease in the free VEGF concentration
`was observed, followed by a rebound effect without exceeding
`their respective baseline levels. In all three compartments, the
`free VEGF concentrations are predicted to reach a steady state
`at the end of the 10 days of treatment and then remain almost
`constant (varying within a small range) over the duration of
`the experiment: the free VEGF level in the tumor was sig-
`nificantly decreased ('-.70.1%), whereas that in the plasma was
`significantly increased (by 8-fold) as compared to the baseline.
`Although the rebound in free VEGF in plasma occurred after
`45 minutes, the rebound still happens if we limit the duration
`
`9890
`
`Cancer Res; 70(23) December 1, 2010
`
`Cancer Research
`
`Downloaded From htlp:llaacrioumals.orgfcancerres/arlide-pdtI7DQ3I9886I2644719f9886.pdf by guest on 03 March 2022
`
`Time (days) 1
`
`7
`
`14
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 5
`
`

`

`Downloaded from htlp:/!aaclioumals.orglcancerreslarlide-pdt/70123198861264471919886pdl by guest on 03 Marth 202
`
`Increased Plasma VEGF Predicted by a Pharniacokinetic Model
`
`VEGF agent followed by subsequent binding to VEGF once in
`the normal or tumor tissues. For example, within 4 to S hours,
`58% of VEGF in the healthy tissue has become complexed with
`the anti-VEGF agent (Fig. 113). This is mainly due to the fact
`that the dose of the anti-VEGF agent injected is several orders
`of magnitude higher than the free VEGF concentration in the
`blood and, therefore, the anti-VEGF saturates the plasma
`upon injection. This concentration difference is counteracted
`by the microvascu1ar permeability of the high molecular
`weight (150 kDa) anti-VEGF being lower than that of VEGF
`(-'45 kDa).
`The net intercompartmental flow of the VEGF/anti-VEGF
`complex reveals that effectively, there is a net flow of complex
`intravasating from the normal tissue. This result suggests that
`the role of the anti-VEGF is to deplete VEGF from the
`interstitial spaces of the normal tissue and the tumor in
`the form of a complex.
`
`The VEGF distribution is modified by the injection of
`the anti-VEGF agent
`Relative VEGF distribution changes in normal tissue, blood,
`and tumor upon administration of the anti-VEGF agent
`(Fig. 3). In the blood (middle graph), there is no significant
`amount of free VEGF because most of it is complexed with the
`anti-VEGF agent. In the normal and tumor tissues (top and
`bottom graphs, respectively), most VEGF is complexed with
`the anti-VEGF agent (light gray region). The decrease in the
`relative amount of unbound VEGF in each compartment
`(black region), however, is mostly due to increase in total
`VEGF, due to the formation of VEGF/anti-VEGF complex
`(Supplementary Figure S3). In normal tissue, unbound VEGF
`declines only transiently; in tumors, there is a steady-state
`decline, where VEGF is much less bound to its receptors (drop
`by 48% - dark gray region) and less sequestered in the matrix
`(drop by 69% - white region) than before injection.
`
`The administration of the anti-VEGF agent significantly
`modifies the VEGFRI and Mfl'I occupancies in the
`tumor
`In keeping with the predicted effect on unbound VEGF
`(Fig. I), occupancy of the receptors in normal tissue was only
`transiently altered by the administration of the anti-VEGF
`agent (Fig. 4. solid line), whereas the VEGFRI and NRPI
`occupancies (top and bottom graphs) in the tumor were
`significantly decreased (from 31% to about 10%, and 35% to
`about 18% for VEGFR1 and NRPI, respectively) and remained
`fairly unchanged over the course of the experiment (dotted
`lines). Changes in VEGFR2 occupancy appeared less signifi-
`cant (middle graph), due to the saturation of tumor VEGFR2
`by cell-surface association of VEGF-bound NRPI; we assume
`10-fold higher NRPI expression on tumor endothelial cell
`surfaces than that of VEGFR2 in our model and 10-fold higher
`than on normal tissue endothelial cells. The effect of para-
`meters on the qualitative results of the study is discussed
`below. VEGFR2 occupancy after the anti-VEGF administration
`does not significantly change in the tumor compartment as
`compared to baseline (50% and 100% ligated in single and
`metronomic therapies, respectively).
`
`of the treatment to 45 minutes instead of 90 minutes (data not
`shown). The free anti-VEGF concentration peaked following
`each injection, reaching an overall maximum after 10 days of
`about half that for the single-dose treatment (Supplementary
`Figure S1D versus Supplementary Figure SIB). Interestingly,
`more VEGF/anti-VEGF complex was formed in the tumor
`(dotted line) than in the blood (dashed line) or in the normal
`tissue (solid line), regardless of the regimen (Supplementary
`Figures S2B and S2D) for single-injection and metronomic
`therapy, respectively). This also means that the total VEGF
`concentration was higher in the tumor than in the blood or the
`normal tissue.
`
`Formation of the VEGF/anti-VEGF complex mediates
`the depletion of VEGF from the tumor
`The changes in VEGF concentrations induced by the anti-
`VEGF agent can be interpreted by a detailed study of the
`movement of VEGF and anti-VEGF between the three com-
`partments. We define the net flow for each molecule as the
`difference between the intercompartmental flows of that
`molecule (in moles per unit time) entering and leaving the
`compartment. For example, the net intercompartmental flow
`of VEGF in the normal tissue is the difference between VEGF
`influx (by extravasation) and VEGF leaving the compartment
`(by intravasation and lymphatic drainage). With this metric,
`any negative net flows represent flows of diffusible molecules
`traveling from the blood into the normal tissue or the tumor
`compartment, whereas positive net flow illustrateg the flow of
`molecules entering the blood compartment. To visualize the
`relative effects, Ave plotted the net intercompartmental flows
`on the same graph. Figure 2 illustrates the net flows for the
`free VEGF, free anti-VEGF agent, and VEGF/anti-VEGF com-
`plex for the few hours following the anti-VEGF agent intra-
`venous injection.
`The anti-VEGF net flows (light gray) increased significantly
`during the first 6 to 12 hours in the normal (solid line) and
`tumor tissues (dotted line), showing extravasation of the anti-
`
`VEGFIanTi.VEG ti0L VEGF(anti-VEGF(No,msl tissue)
`
`0
`
`-2 1'
`
`am;-VEGF
`(Tumurl
`
`/
`VEGF
`Both tissucal
`
`0
`S
`
`ii Aso
`
`anl,-VEGF (No,mal tissue
`
`0
`
`12
`
`24
`
`48
`
`Time
`
`Figure 2. Net intercompartmental flows of VEGF, anti-VEGF, and VEGF/
`anti-VEGF complex. Solid line, normal tissue; dotted line, tumor. Black,
`Free VEOF; light gray, anti-VEGF agent; dark gray, VEGF/anti-VEGF
`complex (also total VEGF).
`
`v,w.aacdoumals.org
`
`Cancer Res; 70(23) December 1, 2010
`
`9891
`
`Mylan Exhibit 1105
`Mylan v. Regeneron, IPR2021-00880
`Page 6
`
`

`

`-pdfI7O23988&26447l9/9886.pdf by guest on 03 March 2022
`
`Stefanini et S.
`
`- srnØe irecton
`- Meeor,om,c therapy
`
`NormS issue
` Tunic,
`
`-
`
`
`
`7
`
`10
`
`e
`
`40%
`
`20%
`
`0%
`
`eo%
`
`20%
`
`0%
`
`80%
`
`60%
`
`40%
`
`20%
`
`LI.
`'3
`
`0.
`
`Figure 3. Relative VEGF distribution profiles in normal (top), blood (middle),
`and tumor (bottom) tissues. Percentage of VEGF sequestered in the anti-
`VEGF agent, free, sequestered In the receptors, and sequestered in the
`extracellular matrix. From top to bottom: normal tissue, blood, and tumor.
`Light gray, VEGF sequestered in anti-VEGF (VEGF/anti-VEGF complex);
`white, WOE sequestered in the extracellular matrix; dark gray, VEGF
`sequestered in receptors; black, unbound VEGF.
`
`Discussion and Conclusion
`
`The mechanism of action of bevacizumab has been com-
`monly accepted as its binding to the VEGF protein resulted
`in inhibition of angiogenesis. However, recent analysis
`points out that "whereas the molecular targets for anti-VEGF
`therapy with large molecules are identified, the mechanism
`of action, that is, how an anti-VEGF approach can exert
`single-agent activity in some cancers and augment the
`efficacy of conventional chemotherapy, is not well under-
`
`Time (days)
`
`Figure 4. VEGF receptor occupancy profiles. Percentage of ligated
`and unligated receptors. P VEGFR1. B, VEGFR2. 0, NRPI. Solid
`line, normal tissue; dotted line, tumor, Black,

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