throbber

`
`
`
`
`
`
`PHIGENIX
`PHIGENIX
`Exhibit 1015
`Exhibit 1 0 1 5
`
`

`

`From (613) _991—2760_
`
`0rd_er'I# 05421267DP04474253
`
`Mon Apr 18 23:24:30 2005
`
`Page 2 of 17
`
`1.. Lafleur. Y. Fmdct.
`vating proteins-con-
`:lder cztrcinorrta 601]
`l.
`aslan. Pseudnmouas
`iody against ovarian
`arian cancer cells in
`:i. USA 84 (I987)
`
`thlingham. RJl.
`Chemoimmunotoxin
`HT-29) xenografted
`t 3562—3567.
`:, R.C. Young. LW.
`:Clay. S. Howell. E.
`l. I. Pastan. Clinical
`aortas cxotoxitt
`im-
`vithiovan'an cancer,
`
`I.
`in", S. Ktntwnr,
`that
`is active on
`:ed of the Fv region
`truncated form of
`land. Sci. USA 90
`
`1, K. Covalcuic, l).
`I vitro and in viva
`tmunotoxin contain—
`er Res. 55 “995)
`
`hire, R. Aoki. M.
`of gclonin immuno—
`N—l
`tn cndomefrinl
`unto: cells in yitro.
`
`argcting — a new
`, Pharmuc. ’icr. 64
`
`If large solid tumors
`:ted against
`tumor
`\ 90 (1993) 8996—
`
`L. Watkins. T.S.
`'ction in mice by
`tctor to tumor vas-
`
`Advanced Drug Delivery ReVicws 31 (1998) 89-104
`
`
`
`advanced
`drug delivery
`reviews
`
`
`
` Targeted delivery of chemotherapeutics: tumor-activated prodrug therapy
`
`Ravi VJ. Chari’k
`
`ImmmmGrm, nun, I48 Sidney Street, Cambridge, MA 02139. USA
`
`
`
`Abstract
`
`‘
`
`The potential of targeted delivery of chemotherapeutic drugs for the treatment of cancer has not yet been realized owing to
`the difficulty of delivering therapeutic concentrations to the target site. While in vivo studies in animal tumor models have
`produced very encouraging results, clinical studies with antibody—drug conjugates have been less successful. This paper will
`review the current status of the targeted delivery approach and analyze some of the reasons for the lack of success so far.
`Starting with a historical perspective,
`this review will end .with a description of newer, more potent and specific
`antibody—drug conjugates, which behave like tumor-activated prodrugs that may yet fulfil the promise of the targeted
`delivery approach for the treatment of cancer. © 1998 Elsevier Science B.V.
`
`Keywords: Antibody—drug conjugates; Cancer; Prodrugs; Anti-cancer agents
`
`Contents
`
`
`
`1. Introduction ............................................................................................................................................................................
`2. ’l‘umor-activated prodrug (TAP) therapy ..........................................................
`
`3. Tumor-specific agents .....................................................................................
`4. Early antibody—drug conjugates ...............................................................................................................................................
`4TI. Design of conjugates ...................................................................................
`In vin‘o evaluation.
`4.2.
`
`4.3.
`in vivo cfficacy..
`
`4.4. Clinical evaluation
`
`5. Factors influencing the effectiveness of antibody—drug conjugates
`..........
`
`6. Antibody conjugates willt more potent drugs ...........................
`
`6.1. Design of conjugates ................................................
`
`.
`..
`6.2. in vitm evaluation.................................................
`6.3. Anti-tumor efficacy in vivu ...............................................................................................................................................
`7. Conclusion ...... . ......................................................................................................................................................................
`Acknowledgements ......................................................................................................................................................................
`References ..................................................................................................................................................................................
`
`89
`90
`91
`92
`93
`94
`95
`96
`96
`97
`97
`99
`loo
`[01
`l0'2
`l 03
`
`1. Intruduction
`
`Cancer chemotherapy today relies on the expecta—
`
`‘Tet; V+l rm 4971113; Fax: +1 617 4975406; email:
`raViuchari@immunogen.cemaiLeompuservecom
`
`
`
`
`
`
`
`
`0l69—409X/98/51900 © 1998 Elsevier Science B.V. All rights reserved.
`PII 30169-409X(97)0l)095-l
`
`tion that anti-cancer drugs will preferentially kill
`rapidly proliferating tumor cells rather than normal
`cells. Typically, cancer patients with disseminated
`disease present themselves with approximately 10”
`tumor cells, and it
`is well establislted'that at least
`99% of these cells have to be killed (Le. a two-log or
`
`--
`'
`PAGE 2/17 " RCVD AT 4/18I2005 8:28:24 PM [Paclt‘lc Dayllght Tlme] " SVR:SVCSO1I0 ‘ DNI
`
`or
`6034 " CEI
`
`"3)991-2150
`
`" DURATION mm-ss 218-10
`
`PHIGENIX
`
`Exhibit 1015-01
`
`

`

`Order . # 05421267DP04474253
`
`Mon Apr 18 23:24:30 2005
`
`Page 3 of 17
`
`R.V.J. Clittri / Advanced Drug Delivery Reviewr 3] (I993) 89— 704
`
` From (613) 91—2760,
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`radioisotope or a small cytotoxic drug. This chapter
`will review the progress made in the area of targeted
`delivery of small chemotherapeutic drugs, discuss
`some of the shortcomings of the earlier approaches,
`and provide potential. solutions that may help restore
`the promise of the targeted delivery approach for
`cancer therapy.
`
`2. Tumor-activated prodrug (TAP) therapy
`
`The basic premise of the targeted delivery ap-
`proach is that conjugation of drug to a tumor-specific
`molecule renders the drug inactive until
`it reaches
`the target site. Once at the tumor site. the conjugated
`drug binds to the surface of tumor cells and is further
`processed (internalized, released from the carrier
`
`molecule) to restore its original potency. Thus, drug
`conjugates can he considered as
`tumor-activated
`
`prodrugs (TAPS). While most conventional prodtugs
`are converted to active drugs by mechanisms such as
`chemical or enzymatic hydrolysis.
`restoring the
`activity of TAPS should ideally be dependent on
`interaction with antigens or receptors specifically
`found on the surface of tumor cells. Historically,
`conventional prodrugs have been designed to over-
`come a physiological barrier, such as poor oral
`bioavailability or rapid metabolism 13,4]. Often, the
`oral delivery of a drug is
`improved by merely
`converting it into a water soluble prodrug. Conven-
`tional pnodrugs are designed with the expectation
`that improving the pharmacokinetic properties of a
`parent drug will result in increased levels in circula-
`tion and thus greater levels at the target site. In TAP
`therapy. the specific affinity of the tumor-associated
`antigen or receptor for the targeting component of
`the drug conjugate results, in addition, in a greater
`uptake and retention of the TAP at
`the targeted
`tumor site and,
`therefore,
`in increased selectivity.
`This is followed by liberation of the active drug
`resulting in high local concentration at the target site.
`Ideally, TAPS would be stable during circulation-
`such that no conversion of the prodrug to the active
`form occurs outside the targeted tumor. Also, TAPs
`would not bind to non-target tissues and thus will be
`non—toxic while in circulation in vivo.
`
`The principle of drug conjugates as tumor-acti—
`vated prodrugs
`is
`illustrated with antibody—drug
`conjugates as an example in Fig. 2. In an ideal
`
`greater cell kill) to achieve a complete remission.
`Continued treatment during remission is required to
`achieve complete
`eradication of
`the
`tumor. A
`schematic
`representation of
`the drug treatment
`categories for cancer as originally described by Frei
`[.11 is shown in Fig. 1. Unfortunately, clinically used
`anti—cancer drugs have limited selectivity for the
`tumor. The levels of drug required to kill sufficient
`number of tutuor cells to achieve and maintain a
`
`state of complete remission in patients causes signifi-
`cant
`toxicity towards actively proliferating non-
`' malignant cells, such as normal cells of the gastroin-
`testinal tract and bone marrow. Thus, a continuing
`challenge in cancer treatment
`is
`to develop new
`cytotoxic agents with greater selectivity for
`the
`tumor. To achieve this goal,
`it is first necessary to
`identify inherent differences between normal and
`
`cancer cells that can be potentially exploited.
`The discovery that. tumor cells expressed specific
`dcwrminants on their cell surface that were not found
`on normal cells suggested that this distinction could
`form the basis for the selective targetingof tumors.
`The advent of monoclonal antibody technology [2]
`led to the development of a myriad of monoclonal
`antibodies, each with its own binding specificity for
`novel tumor-specific antigens. The logical outcome
`was to exploit the binding specificity of the antibody
`to deliver a cytotoxic agent selectively to the tumor
`site with the hope of delivering a ' high,
`lethal
`concentration of drug to the target cells. The cytotox-
`ic agent could be in the form of a protein toxin, a
`
`Complete remission
`
`
`
`
`
`
`
`
`
`“ Treatment
`lnductlontreatment \durlng remissim
`
`8
`
`Eradicatlon of
`neoplastic cells
`
`T?
`Immune Response
`
`Time ( from start of treatment)
`
`Fig. I. Chemotherapy of cancer (reproduced with permission from
`Frei [1]).
`"
`
`
`
`
`
`
`
`(Log,0)
`CancerCells
`
`
`PAGE 3/17 ' RCVD AT 4/18/2005 8:28:24 PM [Paclflc Dayllght Tlme] " SVR:SVCSOl/0 ‘ D
`
`
`
`PHIGENIX
`
`Exhibit 1015-02
`
`

`

`From (513) 991—2760 ,
`
`Order .# 05421267DP04474253
`
`Mon Apr 18 23:24:30 2005
`
`Page 4 of 17
`
`-
`
`.
`
`It.V..l. Chari / Advanced Drug Delivery Reviews 31 (I998) 89- 104
`
`91
`
`1g. This chapter
`area of targeted
`dings, discuss
`lier approaches,
`nay help restore
`'y approach for
`
`') therapy
`
`3d delivery ap—
`a tumor-specific
`until it reaches
`
`., the conjugated
`lls and is further
`70111
`the. carrier
`
`ncy. Thus, drug
`tumor-activated
`
`ational prodrugs
`hanisms such as
`
`restoring the
`,
`2 dependent on
`tors specifically
`lls. Historically,
`:signcd to over-
`h as poor oral
`[3,4]. Often, the
`ved by merely
`fodrug. Conven-
`the expectation
`properties of a
`evels in circula-
`
`‘gct‘site. In TAP
`armor-associated
`g component of
`on, _in a greater
`at
`the targeted
`ased selectivity.
`the active drug
`at the target site.
`tring circulation
`rug to the active
`nor. "Also, TAPS
`and thus will be
`'0.
`
`s as tumor-acti-
`
`antibody—drug
`2.
`In an ideal
`
`1000
`
`900
`
`800
`
`>.
`2 700
`E 600
`2
`m 500
`E
`< 400
`ii
`m 300
`
`200
`100
`
`°__
`
`
`
`Drug
`
`Conjugate Drug
`
`__
`___
`Conjugate
`
`TARGET CELL
`
`NON-TARGET CELL
`
`Fig. 2. Relative potency of free drug and antibody—drug conju-
`gates (tumor—activated prodrugs. TAPS) towards target and non«
`target cells.
`
`situation, for an antigen-negative cell which does not
`bind conjugate, conversion of the free drug into :1
`TAP by conjugation to an antibody results in in-
`activation of the drug. For an antigen-positive cell,
`binding to the TAP is foll0wed by internalization and
`release of the free drug in its fully active form. Thus
`the free drug and TAP have equal potency for the
`target cell. The therapeutic window is determined by
`the difference in cytotoxicity of the TAP for the
`target versus the non~target cell. As we will see later,
`the effectiveness of TAP therapy depends on several
`factors-including choice of the targeting molecule,
`the potency of the drug and the nature of the release
`mechanism for conversion of the prodrug into the
`active drug.
`
`3. Tumor-specific agents
`
`The success of the targeted drug delivery approach
`for the treatment of cancer relies to a great extent on
`the tumor-specificity of the targeting agent. The cell
`
`surface molecrrle which the targeting agent recog-
`nizes can be a tumor-specific. antigen (typically a
`glycoprotein, carbohydrate or oncoprotein), a growth
`factor receptor, or a receptor for a hormone. Ideally,
`the cell surface molecule would have the following
`properties:
`(a) well defined molecule expressed ex-
`clusively on tumor
`tissue,
`(b) not expressed on
`normal
`tissues (c) binds to the targeting molecule
`with high affinity,
`((1) expressed homogeneously on
`all target tumor cells, (e) present on the tumors of all
`patients with the same type of cancer (f) not shed
`into the serum of patients.
`In the early phase of the targeted therapy ap-
`preach, monoclonal antibodies were heralded as
`ideal
`targeting agents that hound exclusively to
`antigens expressed on tumor cells. However, the use
`of more sensitive analytical
`techniques
`such as
`immunofluoresccnce
`and
`imtnunohistochcmical
`
`staining revealed that most antibodies bound to
`tumor-associated antigens that were only preferen-
`tially expressed on the surface of tumor cells.
`In
`most cases,
`the antibodies also bound to varying
`extent.
`to antigens found on a limited number of
`normal tissues. In fact, the target antigens for most
`antibodies [5,6! developed against solid tumors were
`selected mainly on the basis of the higher expression
`of the antigen on tumors in comparison with normal
`tissues. The only truly tumor-specific antigens appear
`to be those found in hematopoietic tumors, such as
`idiotypes present on the surface of B-cell tumors {7],
`and the T-cell receptor expressed in T—cell leukemia
`and lymphOma. Although the cross-reactivity of
`antibodies with normal tissues is a matter of concern,
`
`the benefit potentially gained from the improvement
`in the therapeutic window of cytotoxic drugs by
`conjugation to antibodies often outweigh the toxicity
`concerns. Of course, selection of antibodies with an
`
`In
`acceptable cross-reactivity profile is important.
`addition,
`thorough pre—clinical toxicology studies in
`animals that bear the same antigenic determinants
`and show similar crossvreactivity patterns to that
`found with human tissues is critical.
`Monoclonal antibodies are also attractive as target—
`
`ing agents because of their high binding affinity for
`their respective antigens. This should allow for the
`localization and retention of high concentrations of
`drug at the tumor site. In addition, the long circula-
`tion time of antibodies also allows for a greater
`probability that the. drug will reach the tumor site.
`
` PAGE 4/17 " RCVD AT 4/18/2005 8:28:24 PM [Paclflc Dayllght Tlme] " SVR:SVCSO1
`
`PHIGENIX
`
`Exhibit 1015-03
`
`

`

`From (613) 991—2760.»
`:-
`
`Order.# 05421267DP04474253
`
`Mon Apr 18 23:24:30 2005
`
`Page 5 of 17
`
`
`
`
` 92 .
`
`R.V..l. Cliari / Advanced Drug Delivery Reviews .31 {1998’} 89—104
`
` poorly vascularized. The use of smaller antibody
`
`The therapeutic potential of conjugates of cytotox-
`ic drugs with monoclonal antibodies derived front
`murine hybridomas is dampened by the development
`of a predictable anti—globulin immune response in
`humans. The generation of a human anti-mouse
`antibody (HAMA) response leads to rapid neutraliza-
`tion and clearance of the itnmunoconjugate from the
`blood stream,
`thus limiting its therapeutic utility.
`Recent advances in recombinant DNA technology,
`and knowledge of antibody gene structure have been
`applied to the engineering of rodent antibodies to
`make them less immunogenic. A- ‘humanized’ anti-
`body is constructed by transferring the murine
`complementarity determining regions (CDRs) on to
`an appropriate human framework region. Since
`CDRs form the antigen combining site, a humanized
`or CDR-grafted antibody preserves the murine an-
`tigen specificity, but because most of the antibody
`structure is human,
`it is likely to be less immuno—
`genic in patients than the parent mouse antibody.
`Recent clinical studies [8] with humanized antibodies
`
`fragments instead of whole lgG molecules may be
`advantageous in sonte cases. Three comparative
`studies [ll—13] of the tumor localization of radio-
`labeled intact IgGs and smaller fragments [Fv, Fab'.
`F(ab')2] in mice have shown that smaller fragments
`penetrate the tumor faster (maximum tumor penetra-
`tion of Fv is at 0.5 h) than intact IgG. which showed
`an equivalent degree of tumor penetration only at 48
`h post-injection. However,
`the smaller fragments
`displayed faster clearance and delivered lower over—
`all tumor doses than the intact IgG, suggesting that
`conjugates with intact lgG molecules may be prefer-
`able for the specific application.
`Although monoclonal antibodies probably provide
`the greatest binding selectivity for cancer cells, other
`targeting agents that preferentially bind to tumor cell
`surface markers may provide distinct advantages
`such as smaller size. rapid internalization and non-
`intnnmogcnicity. For example, the epidermal growth
`factor receptor (EGFR) gene is amplified in a high
`proportion of human squamous carcinoma cell lines _
`[l4]. Levels of EGFR are three to sixty—four—fold
`higher in several tumor types, such as lung. breast
`and head and neck, as compared to that found on
`normal keratinocytes [15]. Human EGF is a single
`polypeptide of 53 amino acids and is
`specially
`attractive as a targeting agent because of its small
`size (M,=6201), high binding affinity for its re-
`ceptor (apparent K‘1 = 2*4 X ill—:0 M) and its rapid
`internaligation upon binding to the receptor [16]. In
`addition'human EGF will not be immunogenic. An
`example of another polypeptide that can also be lused
`to target EGFR-exprcssing carcinomas is transform-
`ing growth factor alpha (TGl"'a) []7|. Other exam-
`ples of low molecular weight targeting agents that
`appear
`to bind preferentially to tumors
`include
`melanocyte
`stimulating hormone (MSH) against
`melanomas [18], thyroid stimulating hormone (TSH )
`and thyrotropin against
`thyroid cancers [19], and
`interleukin 2 (1L2) for T—cell leukemias [’20].
`
`:
`
`'
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`in 46 patients have demonstratedthat unlike human-
`
`mouse chimeric antibodies, COR-grafted antibodies
`
`were found not
`to induce a primary immune re-
`
`sponse, even after several courses of treatment.
`
`However, humanization by CDR-grafting often re-
`
`sults in an antibody with a lower binding affinity
`
`than the parent murine antibody. Further amino acid
`
`substitutions in the framework region are usually
`
`required to maintain the correct conformation of the
`
`CDRs. Even with this improvement, COR-grafted
`
`antibodies with somewhat
`lower affinity than the
`
`parent. antibodies are often produced. A newer
`
`technique called variable domain resurfacing 19]
`
`takes advantage of the generally accepted view that
`
`the antigenicity of proteins is determined solely by
`
`surface epitopes. In this approach, the binding affini-
`
`ty is maintained by retaining the CDRs and the core
`
`of the murine variable region framework. Only the
`
`surface residues in the tnurine variable region are
`
`replacedby' those front a human variable region.
`
`This technique was applied to two murine antibodies
`
`and,
`in both cases, affinity was fully preserved
`
`[10].
`
`Although monoclonal antibodies have been the
`
`most commonly used targeting agent
`for chemo—
`
`therapeutics,
`the pharmacodynamics of these large
`Early antibody—ding conjugates were comprised
`
`immunoglobulin molecules may impede their ability
`of a monoclonal antibody covalently linked to sever-
`
`al molecules'of a clinically used anti-cancer drug.
`to access or penetrate solid tumors which are often
`
`
`4. Early antibody—drug conjugates
`
`
`
` PAGE 5/17 . RCVD AT 4I18l2005 8:28:24 PM [Paclt'lc Daylight Tlme] ‘ svnfiycgggq;3N1
`
`PHIGENIX
`
`Exhibit 1015-04
`
`

`

`From (613) 991—2760 .
`we
`
`0 rder‘ ,# 05421267DP044742 5 3
`
`Mon Apr 18 23:24:30 2005
`
`Page 6 of 17
`
`RM]. C/mri / Advanced Drug Delivery Reviews 3] ( I 998'} 6‘9— /04
`
`93
`
` l l
`
`smaller antibody
`molecules may be
`nee comparative
`lization of radio—
`
`gments [Fv. Fab’,
`unaller fragments
`m tumor penetra-
`G, which showed
`
`nation only at 48
`mallet
`fragments
`Iered lower over-
`
`}, suggesting that
`:5 may be prefer-
`
`probably provide
`aancer cells, other
`tirtd to tutnor cell
`
`:tinct advantages
`lizalion and non
`
`epidermal growth
`tplified in a high
`cinoma cell lines
`
`:o sixty-four—fold
`h as lung, breast
`to that found on
`
`EGF is a single
`and is
`specially
`lust: of its small
`
`finity 'for its re-
`M) and its rapid
`receptor [16}. in
`.nmunogenic. An
`can also be used
`nas is transform-
`
`.7}. Other exam-
`:ting agents that
`tumors
`include
`
`against
`(MSH)
`' hormone (’I‘SH)
`tncers
`[l9], and
`mias [20].
`
`is
`
`were comprised
`' linked to sever-
`
`uni—cancer drug.
`
`The linker connecting the antibody and the drug was
`either non-cleavable or cleavable upon entry into the
`cell. Representative examples of types of linkers and
`drugs that were used for the generation of drug
`conjugates are shown in Fig. 3.
`
`4.1. Design of conjugates
`
`in the early development phase of antibody-drug
`conjugates. it was believed that the tumor specificity
`of anti-cancer drugs could be improved merely by
`linking these drugs directly to antibodies via amide
`bonds. Thus, several anti-cancer drugs [21—26] such
`as methotrexate. daunorubicin,
`the Virrca alkaloids,
`mitomycin C,
`idarubicin’ and N-acetyl melphalan
`were linked via non-cleavable bonds to a multitude
`
`of murine monoclonal antibodies [27—29]. Typically,
`an average of 4—8 molecules of drug werelinked per
`antibody molecule.
`in most cases.
`the conjugates
`lacked cytotoxic potency and were less potent than
`the unconjugated drugs. Attempts were then made to
`increase their potency by linking a larger number of
`drug, molecules either directly to the antibody or
`through the intermediaey of a macromolecular car-
`rier,
`such as poly-L-lysine, poly-I,«glutainic acid,
`dextran or human serum albumin [30,31]. However,
`these conjugates displayed impaired binding affinity
`in vitro, and unfavorable pharmacokinetics in ani-.
`mals deterring further development.
`Since the full potency of the drug could not be
`observed when such non—cleavable
`linkers were
`
`used. the emphasis shifted towards the development
`of linkers that wbuld be cleaved inside the cell to
`release active drug. An approach, that was explored
`by Trouet et al. and Umemoto et at. [32.33]. consists
`of linking daunorubicin or methotrexate to an anti-
`body via a tetrapeptide spacer arm under the premise
`that free‘drug can be released from the conjugate by
`the action of lysosomal peptidases. One of the first
`cleavable linkers to be widely employed was an
`acid-labile linker based on cis-aconitic acid,
`that
`takes advantage of the acidic environment of the
`endosomes
`that might be encountered by these
`conjugates after receptor mediated endocytosis. Shen
`and Ryse'r' used acid labile linkers for the preparation
`of conjugates of daunorubicin with macromolecular
`carriers [34]. Subsequently, Yang and Reisfeld [351
`and Dillman et ali [36] used a similar approach to
`
`LlNKEFl
`
`Non-Cleavable
`
`Amide
`O
`a) RtNJk
`H
`
`R'
`
`Succinimide
`
`0
`
`H
`
`a) R‘OJWN‘R'
`
`0
`
`Cleavable
`
`DRUGS
`
`Methotrexate [21]
`Daunorubicin (22]
`Vinca Alkaloids [23]
`Mitomycin C {24]
`Idarubicin [25]
`N-Ac-Melphalan [26] .
`
`Peptldase—Labile
`Fl-Ala-Leu-Ata—Ala-R'
`
`Other tri- and tetra- peptides
`
`Doxorubicin {32]
`Methotrexate [33]
`
`Acld Lablle
`
`a) Cis-Aconitic
`
`o
`
`OH
`
`b) Hydrazide
`
`RI
`
`/
`Fl/
`
`N‘N’
`*5
`
`Re
`
`c) Thiocarbamoyl
`S
`NJLNH o
`HoH
`R.N,
`0
`H
`
`Doxorubicin [34)
`Daunorubicin {35,36}
`
`Vlnblastine [37]
`Doxorubicin {38]
`
`Daunorubicin [39]
`
`Fig. 3. Linkers and drugs used in early antibody—drug conjugates.
`
`conjugate daunorubicin to an anti-melanoma anti—
`body and an anti-T cell antibody, respectively. A
`more popular method of conjugating drugs to anti—
`bodies uses acid—labile hydrazone bonds. The hy—
`drazone linkage has been used to conjugate vin-
`blastine and doxoruhiein to antibodies {37,38l. An
`thiocarbamoyl
`acid-labile
`linker
`for
`coupling
`
`
`
`PHIGENIX
`
`Exhibit 1015-05
`
`

`

`From (613) 99172760 ..
`
`0 rder‘ .# 05421267DP044742 5 3
`
`Mon Apr 18 23:24:30 2005
`
`Page 7 of 17
`
`
`
`
`
`
`
`
`
`
`
`
`
`R.V.J. Chari I Advanced Drug Delivery Reviews 31 (I998) 89-104
`
`daunorubicin to a polymeric carrier has also been
`described [39]. Structures of representative examples
`of acid—cleavable conjugates are provided in Fig. 4.
`
`4.2,
`
`In vitro evaluation
`
`Evaluation of antibody—drug conjugates for bind-
`ing to antigen-positive cells indicated that.
`in most
`cases,
`the affinity of the antibody was either fully
`preserved or only slightly diminished upon conjuga-
`tion of up to eight molecules of drug. In many cases.
`the results of in vitro cytotoxicity evaluation of the
`conjugates have not been reported. In the remaining
`cases, evaluation of in vitro cytotoxicity was general-
`ly performed using 3H-thymidine incorporation as a
`measure of cell growth inhibition. Antibody conju—
`
`gates of methotrexate, the Vina: alkaloids or doxont-
`bicin had [C50 values in the range of 10"b M to 10-7
`M [40—42]. The efficiency and extent of cell kill in
`vitro is difficult
`to gauge because the cytotoxicity
`assays are limited in their capability of detecting low
`survival numbers (lower limit of detection is about
`5%). Thus, these assays do not provide a measure of
`the steepness of the killing curve or the number of
`logs of cells killed. Often, conjugation appeared to
`diminish the cytotoxicity of the drug as compared to
`the parent unconjugated drug. For example, conju-
`gates of doxorubicin with the antibodies BR96 and
`BR64. directed against. the Lewis Y antigen found on
`several
`tumors, were eight-fold and live—fold less
`active respectively in vitro than free doxombicin
`[42.43]. Similarly. free doxorubicin is fivc-fold more
`
`0
`
`o
`
`iW
`
`l1
`
`"
`
`"l/N
`
`
`
`0 u
`
`se
`
`0
`
`H36
`
`0
`
`c
`
`
`
`OH H"
`

`on
`
`|
`
`0
`
`o
`
`2
`
`Ah
`
`on
`
`on HN
`
`H
`N
`"\n’ V
`5
`
`n
`
`0
`
`R = Polymeric backbone
`
`a
`
`Fig. 4. Stnictures of representative acid-labile ding conjugates: hydrazone-linked doxorubicin conjugate, 1
`daunorubicin conjugate. 2 [35.36], daunombicin conjugate containing a thiocarbamoyl link. 3 [39].
`
`
`
`[38,42], cis-aconitate linked
`
`mess
`034 ' C
`
`PAGE 7/17 ‘ RCVD AT 4/18/2005 8:28:24 PM [Paclflc Daylight Tlme] SVR.SVCSO1I
`
`PHIGENIX
`
`Exhibit 1015-06
`
`

`

`From (613) 991—2760 .
`
`,
`
`0 rder ,# 054212670 P0447425 3
`
`Mon Apr 18 23:24:30 2005
`
`Page 8 of 17
`
`
`
`RM]. Char} I Advanced Drug Delivery Reviews 31 (I993) 89— 104
`
`95
`
`:aloids. or doxoru—
`.r 10'“ M to to"
`eat of cell kill in
`
`the cytotoxicity
`:
`y of detecting low
`ietection is about
`vide a measure of
`
`or the number of
`ation appeared to
`tg as compared to
`' example, conju—
`bodies BR96 and
`
`' antigen found on
`.nd fivefold less
`free doxorubicin
`. is live-fold more
`
`iK.
`
`i;
`:5.
`
`ii;
`i;
`j
`.'
`l:
`
`
`
`.,..-.._..___...,._.._._._.
`
`, cis-aconitate linked
`
`potent than doxorubicin conjugated to the antibody
`SEN‘7 directed against small cell
`lung cancer [44].
`Although conjugation of antibodies
`to drugs
`is
`purported to increase their specificity of cell kill, in
`vitro cytotoxicity data with antibody conjugates of
`anti-cancer drugs have been somewhat disappointing.
`‘ In conjugates where antibody and doxorubicin were
`linked via acid-labile hydrazone bonds, antigen-
`negative cells were only five to ten-fold less sensitive
`to conjugate (24 h exposure) than antigen-positive
`cells, suggesting that this type of link is quite labile
`[42-44]. Even when stored at optimal pH and
`temperature, significant hydrolysis of the hydrazonc
`link to release active drug has been reported I45].
`Conjugates of daunorubicin and doxorubicin linked
`via an acid labile cis-aconitate linker were also
`cytotoxic in the 10"6 to 10 7 M range, but showed a
`greater degree of antigen-specificity Suggesting that
`this link was more stable than the hydrazone link.
`[35].
`
`4.3.
`
`In. vivo efficacy
`
`New anticancer agents are gcncrally assessed for
`in vivo anti-tumor efficacy in animal tumor models
`to determine their potential for further evaluation in
`patients. Antibody—drug conjugates have been no
`exception and several of them have been evaluated
`for anti-tumor activity in human tumor xcnograft
`models in immunodeficient mice. Most of these
`conjugates showed greater
`therapeutic efficacy in
`vivo than the corresponding unconjugated drugs or
`isotype-matched
`but
`non-binding
`antibody—drug
`conjugates. in somc cases, biodisl‘ribution studies in
`tumor-bearing mice indicated that the antibody~drug
`conjugate was indcul localized at the tumor site. For
`example, when lymphoma-bearing mice were treated
`with a methotrcxatc conjugate of the anti-EL4 anti—
`body, after 3 11 as much as 15% of the administered
`dose was localized per gram of .tumor
`[46].
`In
`another experiment, comparative studies
`in mice
`with free doxorubicin or a conjugate of doxorubicin
`with the anti-melanoma antibody 9.2.27 showed that
`there was a preferential accumulation of cloxorubicin
`at
`the tumor when delivered as ‘a conjugate as
`compared to'that of free doxorubicin [47]. After'48
`11, there was a 45-fold greater amount (if conjugated
`doxorubicin at the tumor as compared to free dom-
`rubicin. Experiments in tumor—bearing mice indi-
`
`cated that greater tumor localization translated into
`better anti-tumor efficacy, with 9.2.27-Doxombicin
`being significantly more efficacious than free dom-
`rubicio. Another doxorubicin conjugate (BR96-Dox)
`with the human-mouse chimeric monoclonal anti-
`
`body BR96 has been shown to completely Cure
`athymic mice that had been implanted subcutaneous-
`ly with human lung or colon tumor xenografts [42].
`The anti-tumor effect observed is especially impres-
`sive as the tumors were well—established prior to
`commencement of therapy, and the treated animals
`were tumor-free for up to 200 days. However,
`the
`dose of conjugate necessary to achieve 100% cures
`was quite large (2.1 to 2.7 g/kg conjugate, equiva-
`lent to 60 mg/kg doxorubicin). Doaorubicin conju-
`gates of other monoclonal antibodies have also been
`shown to induce tumor regressions or cause signifi—
`cant delay in tumor growth in mice, while unconju—
`gated doxorubicin or non-binding antibody-doxoru-
`bicin only showed a slight therapeutic effect in these
`models {48,491.
`Several conjugates of the Vinca alkaloids linked
`via acid-labile hydrazonc bonds to different mono-
`clonal antibodies have been tested for anti-tumor
`
`tumor models. The best efficacy
`efficacy in animal
`was observed with a conjugate of the vinblastinc
`derivative DAVLBl-IYD with the murine monoclonal
`
`antibody K5114 which reacts with human adeno-
`carcinomas. Significant suppression of tumor growth
`in a well-established subcutaneous lung xcnograft
`model
`in nude mice was demonstrated [50]. Al-
`though complete tumor regression was not observer],
`the conjugate showed much greater efficacy than -
`both the unconjugated vinblastine derivative or the.
`non-binding antibody conjugate. The same conjugate
`was also tested in a human ovarian carcinoma
`
`survival mode] in athymic mice and showed a five-
`fold increase in survival when compared with un-
`
`the
`In this model,
`conjugated DAVLBHYD [51].
`also
`non-binding antibodyvvinblastine
`conjugate
`showed some therapeutic effect causing a 25-fold
`increase in life span as compared to frcc drug. 'l‘his
`non-specific anti-tumor effect might be explained by
`the fact. that the non-binding conjugate lasts longer in
`circulation than free vinblastine,
`thus acting as a
`‘slow-release’ formulation offtlie drug.
`in vivo tumor efficacy has also been demonstrated
`with conjugates of monoclonal
`antibodies with
`milomycin C [24] and idaiubicin [26].
`
`
`
`PAGE 8/17 ' RCVD AT 4/18/2005 8:28:24 PM [Paclflc Dayllght Tlme] ‘ SVR:SVCSD1IO ' DNI
`
`PHIGENIX
`
`Exhibit 1015-07
`
`

`

`From (613) 991—2760 ..
`
`‘
`
`Order .# 0 54212 67DP0447425 3
`
`Mon Apr 18 23:24:30 2005
`
`Page 9 of 17
`
`
`
`R.V.J. (.‘liari I Advanced Drug Delivery Reviews 3] (19.98) 8.9- I04
`
`4.4. Clinical evaluation
`
`The pre-Iclinical studies with antibody conjugates
`of anti—cancer drugs demonstrated that the conjugates
`generally displayed greater anti—tumor efficacy than
`the respective free drugs, suggesting that
`targeted
`delivery of drugs was indeed improving the tumor
`localization and selectivity of . the dl'ngS. These
`encouraging results led to the further evaluation of
`some of these agents.
`in humans. A conjugate of .
`methotrexate and the monoclonal antibody, KSl/4.
`was evaluated in two different Phase I human
`
`trials in patients with non-smallcell lung
`clinical
`cancer. A majority of patients elicited a human
`anti-mouse antibody (HAMA)
`response
`[52,53].
`Although, immunopemxidase staining of carcinoma
`samples provided evidence of post-treatment locali-
`zation of the conjugate,
`there was no therapeutic
`effect and clinical responses were not observed in
`either study. The same KSl/4 antibody was also
`linked to the vinblastine derivative DAVLBl-IYD.
`
`trials in patients with
`and evaluated in clinical
`adenocarcinomas. Again. a majority of the patients
`elicited a HAMA response resulting in rapid clear-
`ance of the conjugate. and no clinical restionses were
`observed in these studies [54,55]. Further human
`studies with these immunoeoniugales appear to have
`been discontinued. Recently, a phase I human clini~
`cal
`trial with BR96-Dox (chimeric monoclonal anti-
`body BR96 linked to the anti-cancer drug doxorubi—
`cin) has been completed in patients with colon
`cancer
`[56]. Preliminary indications are that
`this
`conjugate was also not
`therapeutically effective.
`Thus,
`in clinical
`trials conducted so far, early
`antibody

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