throbber
jnm.snmjournals.org Downloaded from Downloaded from by on October 28, 2014. For personal use only. by on October 28, 2014. For personal use only. jnm.snmjournals.org
`
`
`
`
`
`
`
`
`
`Radioimmunotherapy Targeting of HER2/neu Oncoprotein on Ovarian Tumor Using
`Lead-212-DOTA-AEl
`(cid:160)
`Eva Horak, Frank Hartmann, Kayhan Garmestani, Chuanchu Wu, Martin Brechbiel, Otto A. Gansow, Nicholas F. Landolfi
`and Thomas A. Waldmann
`J Nucl Med.(cid:160)(cid:160)
`
`1997;38:1944-1950.
`
`This article and updated information are available at:
`
` http://jnm.snmjournals.org/content/38/12/1944
`
`Information about reproducing figures, tables, or other portions of this article can be found online at:
`
` http://jnm.snmjournals.org/site/misc/permission.xhtml
`(cid:160)
`Information about subscriptions to JNM can be found at:
` http://jnm.snmjournals.org/site/subscriptions/online.xhtml
`(cid:160)
`
`
`
` is published monthly.The Journal of Nuclear Medicine
`
`SNMMI | Society of Nuclear Medicine and Molecular Imaging
`1850 Samuel Morse Drive, Reston, VA 20190.
`(Print ISSN: 0161-5505, Online ISSN: 2159-662X)
`
`© Copyright 1997 SNMMI; all rights reserved.
`
`IMMUNOGEN 2086, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Downloaded from
`
`
`
`by on October 28, 2014. For personal use only. jnm.snmjournals.org
`
`from preexisting
`
`variant
`
`cells within a
`
`results
`22. Fidler U. Kripkc ML. Metastasis
`malignant
`tumor. Science 1977:197:893-895.
`tumor
`relationships of intravaseular
`23. Liotta LA, Kleineman J. Saidcl GM. Quantitative
`cells,
`tumor vessels and pulmonary métastasesfollowing tumor
`implantation. Cancer
`Res 1974:34:997
`1004.
`24. Terranova VP. Hujanen ES. Martin CiR. Basement membrane and the invasive activity
`of metastatic tumor cells. J Mall Cancer
`Insl 1986:77:311-316.
`25. Fishman AJ. Khaw BA. Strauss HW. Quo vadis radioimmune
`1989:30:1911-1915.
`Inhibition of
`Ito Y. Tanaka K. Sugioka Y.
`Iwamoto Y. Nomizu M. Yamada-Y.
`angiogenesis.
`(umour growth and experimental metastasis of human tibrosarcoma
`cells
`
`26.
`
`imaging. J NucÃ(cid:173)Med
`
`(YIGSR).
`
`form of the laminin sequence Tyr-lle-Gly-Ser-Arg
`HT1080 by a multimene
`Br./ Cancer 1996:73:589-595.
`27. Hill RP. Metastasis.
`In: Tannock IF. Mill RP. eds. The basic science of oncologi; 2nd
`ed. New York: McGraw-Hill;
`1992:178-195.
`In:
`28. Hanna N. Role of natural killer cells in host defense against cancer metastasis.
`Nicolson GL. Milas L, eds. Cancer
`invasion and metastasis:
`hiologic and therapeutic
`aspects. New York: Raven Press; 1984:309-319.
`29. Greenberg AH. Egan SE. Jarolin L. et al. Natural killer cell regulation of implantation
`and early lung growth of H-/-«.v-transformed10Tl/2fibroblasts
`in mice. Cancer Rex
`1987:47:4801-4805.
`
`Radioimmunotherapy Targeting of HER2/neu
`Oncoprotein on Ovarian Tumor
`Using Lead-212-DOTA-AEl
`
`Eva Horak, Frank Hartmann, Kayhan Garmestani, Chuanchu Wu, Martin Brechbiel, Otto A. Gansow, Nicholas F. Landolfi
`and Thomas A. Waldmann
`Metabolism Branch and Inorganic and Radioiinmune Chemistry Section, Radiation Oncology Branch, National Cancer
`Institute. National
`Institutes oj Health, Bethesda, Maryland; and Protein Design Labs, Inc., Mountain View, California
`
`radioimmuno-
`lead f12Pb)
`toxicity and efficacy of
`The specificity,
`therapy were evaluated in nude mice bearing the SK-OV-3 human
`ovarian tumor cell
`line expressing the HER2/neu proto-oncogene.
`Methods: The therapeutic agent used was the tumor-specific anti-
`HER2/neu monoclonal antibody AE1 conjugated to 212Pb, 212Bi
`being the daughter and thus the source of the alpha-particle and
`beta emissions. A bifunctional derivative of
`tetraazacyclodode-
`canetetraacetic acid (p-SCN-Bz-DOTA) was used to couple 212Pb
`to the anti-HER2/neu monoclonal antibody AE1. The chelating agent
`did not alter
`the binding affinity to its antigenic target or
`the
`pharmacokinetics and tissue distribution of the AE1 antibody. Tox
`icity and therapeutic efficacy of 212Pb-AE1 were evaluated in nude
`mouse ascites or solid tumor models, wherein SK-OV-3 cells were
`administered i.p. or s.c., respectively. Results: The dose-limiting
`acute toxicity after i.v. administration of 212Pb-AE1 was bone mar
`row suppression, which was observed at doses above 25 ^Ci.
`Therefore, doses of 10 and 20 ¿¿Ciwere used in efficacy trials. The
`i.p. administration of 212Pb-AE1 3 days after i.p. tumor inoculation
`led to a significant (p2 = 0.015) prolongation of tumor-free survival.
`In a second model,
`i.v. treatment with 212Pb-AE1 3 days after s.c.
`tumor inoculation prevented subsequent
`tumor development
`in all
`animals treated with 10 or 20 ¿¿Ciof 212Pb-AE1 (p2 = 0.002
`compared to control groups). This efficacy in the adjuvant setting
`was antibody specific because treatments with equivalently labeled
`control antibody or unlabeled AE1 antibody or no treatment were
`less effective. The rate of growth of small (mean tumor volume, 15
`mm3) SK-OV-3 tumors was modestly inhibited. However,
`tumor
`growth was not inhibited in mice bearing larger (mean tumor volume,
`146 mm3) SK-OV-3 tumors by the administration of a single dose of
`10 or 20 juCi of 212Pb-AE1. Conclusion: Lead-212-AE1 as an intact
`radiolabeled monoclonal antibody may be of only modest value in
`the therapy of bulky solid tumors due to the short physical half-life of
`212Pb and time required to achieve a useful tumor-to-normal
`tissue
`ratio of radionuclide after administration. However, the radiolabeled
`monoclonal antibody may be useful
`in therapy of
`tumors in the
`adjuvant setting. Furthermore, 212Pb may be of value in select
`situations,
`including treatment of leukemia, intercavitary therapy or
`strategies that target vascular endothelial cells of tumors.
`
`Received Jul. 23, 1996; revision accepted Feb. 3, 1997.
`For correspondence
`or reprints contact: Thomas A. Waldmann, MD. Metabolism
`Branch, National Cancer
`Institute, National
`Institutes of Health, Building 10. Room
`4N115, Bethesda. MD 20892.
`
`Key Words: HER2; neu proto-oncogene; alpha-emitting radionu-
`clides; monoclonal antibody therapy; ovarian tumor
`J NucÃ(cid:173)Med 1997; 38:1944-1950
`
`targeted to cell surface antigens have
`Lonoclonal antibodies
`been used to treat patients with a variety of cancers. However,
`unmodified monoclonal antibodies have been relatively ineffec
`tive (7). One of the factors
`in this low therapeutic
`efficacy is
`that most of the monoclonal
`antibodies used are not effective
`cytocidal agents against human neoplastic cells. Furthermore,
`in
`most cases,
`the antibodies were not directed
`against
`a vital
`structure on the surface of malignant
`cells, such as a growth
`factor receptor
`involved in tumor cell proliferation and survival.
`In an attempt
`to circumvent
`these problems,
`researchers
`have
`augmented
`the cytotoxic
`action of monoclonal
`antibodies
`by
`arming them with toxins or radionuclides
`(2-6). Furthermore,
`cell surface antigenic
`targets, especially receptors
`for growth
`factors,
`have been defined
`for more
`effective monoclonal
`antibody
`action.
`In particular,
`the HER2/neu
`receptor,
`the
`product of the c-erbB-2 proto-oncogene,
`has been the target of
`monoclonal
`antibody therapeutic
`trials (7-12). The HER2/neu
`oncogene
`(erbB-2)
`encodes
`a Mr 185,000
`transmembrane
`phosphoglycoprotein.
`The \\ER2lneu
`gene is overexpressed
`in
`20%-30% of adenocarcinomas
`of the breast, ovary,
`lung and
`stomach and has been linked to poor prognosis
`(13-15). A
`series of anti-HER2/neu monoclonal
`antibodies has been gen
`erated that
`includes antibodies without biological
`activity,
`as
`well as those with agonist action in terms of tyrosine phosphor-
`ylation
`of HER2/neu
`(7-12).
`Preclinical
`efficacy
`in mice
`bearing HER2/neu-expressing
`human
`tumor
`xenografts
`has
`been demonstrated with certain antibodies directed toward this
`receptor.
`to the HER2/neu
`antibodies
`unmodified murine
`Although
`oncoprotein inhibited tumor growth in certain cases,
`in general
`it was not sufficient
`to cure animals of established tumors. This
`limited
`efficacy
`led to alternative
`approaches
`that
`include
`the humanization
`of an anti-HER2/neu
`antibody (12,16),
`the
`development
`of a humanized
`bispecific F(ab')2 fragment
`for
`
`1944
`
`Tin-:JOURNALOFNUCLEARMKDICINK•Vol. 38 •No. 12 •December 1997
`
`IMMUNOGEN 2086, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Downloaded from
`
`jnm.snmjournals.org
`
`by on October 28, 2014. For personal use only.
`
`retargeting cytotoxic T cells (17,18), a disulfide stabilized
`anti-HER2/neu Pseitdomonas immunotoxin (//)
`and a disul-
`fide-stabilized anti-HER2/neu Fv-beta-lactamase fusion protein
`for the activation of a cephalosporin doxorubicin prodrug (¡9).
`In addition, anti-HER2/neu antibodies have been used as
`carriers of toxic agents, including radionuclides. De Santéset al.
`(10) demonstrated that animals treated with 400-700 ju,Ciof
`131I-anti-HER2/neu monoclonal
`antibodies manifested
`a
`marked inhibition of the growth of large tumors. In general,
`131I-labeledmonoclonal antibodies have been relatively inef
`fective due to limitations of this radionuclide as a therapeutic
`agent.
`In light of the limitations of radioiodine, metallic radionu
`clides that can be linked to antibodies may provide a better
`choice. Antibodies may be armed with beta- or alpha-emitting
`radionuclides. Future development of radionuclide-conjugated
`monoclonal antibodies may focus on radionuclides that emit
`alpha particles, which may be the most effective agents at
`killing tumor targets without damaging distant normal tissues
`(2-6).
`Our present studies focused on the radionuclide 2l2Pb, which
`has a 10.6-hr half-life. Lead-212 is the parent of 212Bi,which is
`the source of an alpha particle as well as beta and gamma
`emissions.
`In this study, we used 2l2Pb-tetraazacyclodode-
`canetetraacetic acid-AEl
`(2l2Pb-DOTA-AEl ) directed toward
`SK-OV-3 tumors in our therapeutic trials. The goals of this
`study were:
`to determine specificity and pharmacokinetics,
`including biodistribution of the DOTA chelate AE1 anti-HER2/
`neu monoclonal antibody in our experimental tumor model; to
`define the toxicity of the 2l2Pb-DOTA-AEl monoclonal anti
`body conjugate and, thereby, establish the maximum tolerated
`dose; and to define the efficacy, specificity and toxicity of 2l2Pb
`radioimmunotherapy of a human HER2/neu-expressing tumor
`SK-OV-3 in nude mice.
`
`MATERIALS AND METHODS
`Cell Lines and Monoclonal Antibodies
`The SK-OV-3 cell line (ATCC HTB 77), derived from a human
`ovarian adenocarcinoma, was used. AE1 is an IgG2a murine
`monoclonal antibody directed against the extracellular domain of
`the HER2/neu receptor. The previously described anti-Tac, an
`IgG2a murine monoclonal antibody that binds to the human
`IL-2Ralphasubunit, but not to murine SK-OV-3cells, was used as
`a negative control antibody (20).
`Radioiodination of Monoclonal Antibodies
`Radioiodinations of AE1 for bindability studies with I25I were
`performed using a modification of the chloramine-T method;
`the
`resultant
`specific
`activity was 4.0-6.5
`/u,Ci//xg. Ninety-seven
`percent of the iodinated AE1 was precipitable with 20% trichloro-
`acetic acid.
`Conjugation of the Chelate to Monoclonal Antibodies
`To permit labeling with 2l2Pbor SXY,the monoclonal antibody
`AE1, as well as anti-Tac, which served as the control antibody for
`all studies, was first conjugated with 2-(p-SCN-Bz)-DOTA
`(21,22). Forconvenience, the chelate will be referredto as DOTA.
`Typically, 8-10 mg of antibody at a concentration of about 5
`mg/ml were dialyzed against bicarbonate buffer, pH 8.6, for 6 hr.
`The antibody preparation was then conjugated to l4C-(p-SCN-Bz)-
`DOTA as described (21,22). The average number of DOTA
`chelates per molecule of antibody was 1.2.determinedas described
`previously (22). Each lot of chelated AE1 antibody was compared
`with the unmodified antibody and was shown to have an unaltered
`binding capacity using a competitive binding assay (Fig. 1).
`Briefly, 5 X IO5 SK-OV-3 cells were incubated on ice with
`
`AE-1[j
`
`1 B eft
`
`1
`
`
`
`DDOTA-AE-1aDD
`
`eoOxEa
`
`u600-500-400-300-200-100-0_•
`
`10.1
`
`•1
`
`1
`
`10100000antibody
`1000
`concentration
`
`•
`
`(ng)
`
`FIGURE 1. Assay of the effect of AE1 and DOTA-AE1 on the binding of
`125I-AE1 to SK-OV-3 cells. •unmodified competitor; D, DOTA-AE1 com
`petitor.
`
`increasing concentrations of unmodified (AE1) or chelated
`(DOTA-AE1) antibody (ranging from 0 to 75,000 ng/vial or from
`0 to 2,688 ng/vial, respectively) in the presence of a constant
`nonsaturatingamount of 125I-AE1antibody (6.25 ng/vial). After 3
`hr of incubation,cells were centrifugedthrough an oil cushion, and
`cell-bound radioactivity was determined.
`
`Procedure for Radiolabeling of Chelated Antibody
`by
`Carrier-free SXY was purified from metal contaminants
`column extraction chromatography and incorporated into the che
`lated antibody as described (23).
`Lead-212 was eluted from a 224Ra/2l2Pbgenerator with 1 ml of
`2.0 M HC1. The solution was passed through a column (2 X 20
`mm) of MP-50 resin, pre-equilibrated with 2.0 M HC1 to remove
`any breakthrough of 224Ra. The solution was then evaporated to
`dryness with the addition of 0.5 ml of concentrated HNO,. The
`2l2Pb activity was dissolved in 0.1 M HNO,, with the pH adjusted
`to —4.0with 3 M NH4OAc. Chelated antibody was added to the
`2l2Pb solution and allowed to react at 35°Cfor 45 min. Five
`microliters of 0.1 m EDTA were added to scavenge any free
`radionuclide. Radiolabeled antibody was purified by high-perfor
`mance liquid chromatography using a size-exclusion column and
`4-morpholinepropanesulfonic
`acid/Cl buffer as the mobile phase ( 1
`ml/min flow rate). Radiolabeling yields were 65%-75%, and more
`than 99% of the 2l2Pb was protein-bound before administration.
`
`in
`
`Animals
`Female 6-8-wk-old athymic-NCR-nude mice were kept
`microisolation units on sterilized water and food.
`Pharmacokinetics and Tissue Distribution of
`Radiolabeled AE1
`To define the rate of entry of AEl antibody into a HER2/neu-
`expressing tumor, mice bearing established s.c. injected SK-OV-3
`tumors on their shoulder were injected i.v. with the 2-ju.Ci dose of
`ssY-labeled monoclonal antibody. Seventeen mice were injected
`with the specific anti-HER2 antibody (K8Y-DOTA-AE1), and two
`mice were injected with an irrelevant (*sY-DOTA-anti-Tac) anti
`body. Three of the mice injected with specific antibody were killed
`at 2, 6, 10 and 24 hr, and two mice were killed at 48 hr following
`infusion; their organs were removed for weighing and radioactivity
`determination using a gamma ray counter.
`Toxicity of Lead-212
`Lead-212 was used as the radionuclide in the immunotherapeutic
`studies. The emissions of 2l2Pb, the decay product daughters and
`the details of the decay events as they relate to radioimmuno
`therapy have been described in the literature and are outlined in
`Figure 2 (24).
`tolerated dose of the
`To establish the toxicity and maximal
`2l2Pb-DOTA-AEl,
`healthy,
`tumor-free mice were injected with
`
`RADIOTHERAPYOFHER2/Ntu-ExpRESsiNGTUMOR•Horak et al.
`
`1945
`
`IMMUNOGEN 2086, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`
`
`jnm.snmjournals.org Downloaded from
`
`by on October 28, 2014. For personal use only.
`
`tumor occur
`was tumor-free survival (time from treatment until
`rence). Data were evaluated according to the methods of Kaplan
`and Meier (25) and Mantel and Haenszel (26). For the third and
`fourth studies,
`the observed parameter was the change in tumor
`volume calculated as (L X W2)/2, where L is length and W is
`width. The tumor volume for each animal at the time indicated was
`normalized to the tumor volume of the animal that was present on
`the day of therapy.
`
`RESULTS
`SK-OV-3 Murine Model of a HER2/neu-Expressing Human
`Solid Tumor Model
`In our initial studies, we established a murine tumor model
`for human HER2/neu-directed
`alpha particle-emitting
`radioim
`munotherapy
`using the SK-OV-3
`human ovarian tumor cell
`line. The SK-OV-3
`cell
`line expressed
`approximately
`10h
`HER2/neu receptors per cell, as defined by Scatchard analysis
`using the '25I-anti-HER2/neu
`antibody AEl. With this cell line,
`after administration
`of 2 X IO'1cells, tumor occurrence rate was
`over 95%, with a median time to tumor appearance of 8 days
`after s.c.
`injection,
`and the occurrence
`rate was 60%, with a
`median time to appearance of 10-13 days after i.p. inoculation.
`The s.c. administered
`tumor
`remained localized, but grew to a
`diameter of 1 cm in 15-71 days
`(mean, 38 days) and to a
`diameter of 2 cm in slightly over 100 days.
`and to
`cells
`To define
`the radiosensitivity
`of SK-OV-3
`tumor models
`provide
`a baseline
`for comparison with other
`used to evaluate
`radioimmunotherapeutic
`agents, we quanti-
`tated tritiated thymidine uptake and defined the percentage of
`remaining cells still capable of colony formation after external
`irradiation of SK-OV-3 cells at different dose levels (0-10,000
`rad). A dose of 520 rad reduced the colony-forming
`ability of
`irradiated cells to 2% of that observed with nonirradiated cells.
`Biological Activity of DOTA-AE1
`of HER2/neu-ex-
`To be effective
`in radioimmunotherapy
`pressing tumors,
`the chelating agent used to link the radionu-
`clide to the AEl
`antibody should not alter
`the specificity
`or
`binding affinity of the monoclonal
`antibody
`to its antigenic
`target. The binding integrity of DOTA-AEl was assessed by
`comparing it to unlabeled AEl
`in a competitive
`binding assay
`(Fig.
`I).
`In this assay, SK-OV-3 cells were incubated with a
`nonsaturating
`amount of l25I-AEl, along with increasing quan
`tities of either unmodified
`or DOTA-AEl.
`The chelated anti
`body manifested identical binding activity as unmodified anti
`body when evaluated in this competition assay.
`In Vivo Pharmacokinetics and Tissue Distribution of
`Radiolabeled DOTA-AE1
`antibody using
`The stability of a 21l3Pb-labeled monoclonal
`DOTA as a chelating agent has been examined previously and
`shown to be stable under in vivo conditions
`(24). To test the in
`vivo stability of the particular DOTA-AEl
`antibody used in this
`study, we used 88Y rather
`than 2l2Pb,
`in light of
`its longer
`physical
`half-life
`and gamma
`emission. The yttrium-DOTA
`complex has been shown to be and is also generally accepted to
`be inert and stable
`in vitro and in vivo,
`thus providing
`an
`unequivocal
`biodistribution
`standard
`for a radiolabeled
`anti
`body using a chelated metal. We and others have noted that the
`indium complex formed with DOTA was not identical with the
`yttrium complex and,
`therefore, 88Y was used as an accurate
`tracer
`isotope.
`In these pharmacokinetic
`studies, animals with
`established
`s.c. SK-OV-3
`tumors
`(~l
`g) were given
`i.v.
`injections of X8Y-DOTA-AE1 (2 juCi per mouse), serial blood
`samples were collected after
`injection and the fraction of the
`dose remaining
`in the plasma was determined. The terminal
`
`1.52(21%)
`1.80(50%)
`EY=0.511
`(23%)
`0.583 (86%)
`0.860(12%)
`2.614(100%)
`
`FIGURE 2. Decay series of 224Rafor generating therapeutic doses of 212Pb
`and 212Bi, with description of the radioactivity emissions of 212Pb and its
`daughters. The emissions are expressed in MeV.
`
`unmodified AEl or different doses of 2'2Pb-DOTA-AEl. Group I
`received only unmodified AEl antibody, while groups 2, 3 and 4
`received 10, 25 and 40 /¿Ciof 2l2Pb-DOTA-AEl,
`respectively.
`
`Radioimmunotherapy of HER2/neu-Expressing Tumors
`with Lead-212 AE1
`In all radioimmunotherapy experiments, we used female athymic
`nude mice (10-12 wk old). In the initial preliminary therapeutic
`trial, mice were given i.p. injections of 2 X IO6 SK-OV-3 cells.
`Three days later, groups of mice were given i.p.
`injections of
`2l2Pb-AEl
`at dose levels of 10 and 20 /nCi (specific activity,
`0.6-1.5
`/u.Ci//ig), the control
`radiolabeled monoclonal antibody
`(2l2Pb-anti-Tac) at the same dose levels (specific activity, 1.0-2.5
`jj-Ci/iJLg),unlabeled AEl
`(10 jug) or no treatment.
`In the second
`trial, animals were inoculated with 2 X IO6 SK-OV-3 cells s.c. in
`the back of the neck. Three days later, groups of five mice were
`treated i.v. with either 212Pb AEl or 2l2Pb-anti-Tac at dose levels
`of 10 and 20 /¿Ci.Control groups again received either
`i.v.
`unlabeled anti-Tac or no treatment. In the third trial, groups of five
`mice with established small-sized s.c. tumors (studied 14 days after
`s.c.
`tumor
`inoculation when mean tumor volume was 15 mm3)
`were treated with the same agents under the same i.v. protocol,
`whereas
`in the fourth group animals with larger
`(mean tumor
`volume, 146 mm3) tumors were studied with the same agents as
`used in trials 2 and 3. The primary end point in the initial two trials
`
`1946
`
`Tin: JOURNALOFNUCLKARMEDICINE•Vol. 38 •No. 12 •December 1997
`
`IMMUNOGEN 2086, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Downloaded from
`
`
`
`by on October 28, 2014. For personal use only. jnm.snmjournals.org
`
`TABLE 1
`Tissue Distribution of Yttrium-88-AE1 and Yttrium-88-Anti-Tac
`
`in Percentage of Injected Dose per Gram of Tissue
`
`Yttrium-88-AE1 specific antibody
`
`Yttrium-88-anti-Tac
`control antibody
`
`2hr
`
`6hr
`
`10 hr
`
`24 hr
`
`48 hr
`
`24 hr
`
`Specific-to-
`control ratio
`of incorporation
`
`24 hr
`
`
`
`Kidney
`IntestineHeartLung
`
`Femur3.70
`
`SK-OV-3tumorBloodLiverSpleen
`
`±0.8835.57
`
`±5.0511.57
`±0.346.45
`±0.63
`9.57 ±0.35
`
`2.88 ±0.1013.82
`±0.5611.
`
`0.69
`48 ±0.70
`2.31 ±0.207.7030.189.906.02
`
`2.05±±0.2410.35
`
`1.10±0.53
`
`±6.1723.81
`
`0.28±4.13±±26.65
`
`±8.42
`
`±6.019.1
`±6.07
`
`5 ±1.336.13
`±5.70
`±
`±
`±1.46
`6.08 ±
`7.25 ±
`5.79
`±0.35
`0.17
`8.13 ±1.00
`8.44
`±1.02
`0.53
`2.26±9.17
`
`±0.26±0.72±0.622.24±8.50
`0.191.010.37
`
`2.42 ±0.3610.80
`
`2.5810.5410.38±0.14±0.48±
`0.001.720.81
`1.838.806.98
`±9.14
`±3.0510.53
`±8.50
`±
`±
`±3.06
`±0.106.40
`2.25 ±2.350.840.000.20
`1.90 ±0.574.341.301.25
`2.37 ±0.4424.6717.386.425.45
`1.05
`0.2026.05
`
`1.94±3.88±3.19±0.73±0.420.004.071.111.391.01
`
`±21.50±6.56
`
`1.34
`1.081.271.15
`
`half-life (beta) of these studies was 86 hr; the fraction intravas-
`cular was 56% with 33% of the i.v. pool catabolized per day as
`assessed by the method of Matthews
`(27).
`tumors with
`The effective
`treatment
`of
`large established
`2l2Pb-AEl
`is dependent
`in part on the physical half-life of 2l2Pb
`(10.6 hr) and the time required to achieve an optimum tumor-
`to-normal
`tissue ratio of radioactivity
`after i.v. administration.
`To address
`this issue, animals with established
`s.c. SK-OV-3
`tumors were given
`i.v.
`injections
`of either
`tumor-specific
`antibody (SXY-AE1 or of an irrelevant antibody X8Y-anti-Tac).
`In Table 1, we show the tissue distribution of XKY-AE1and the
`control antibody KXY-anti-Tac expressed as a percentage of the
`injected dose per gram of tissue in these tumor-bearing mice, as
`well as the ratio of specific-to-control
`antibody incorporation
`into different organs 24 hr after the i.v. infusion. The values are
`expressed as the mean ±s.d. of three mice killed at each time
`point. Within the first 6 hr, there was essentially no enrichment
`of AE1 in the SK-OV-3 tumors compared with normal
`tissues.
`However, by 24 hr, SK-OV-3 tumors manifested an enrichment
`of 88Y-DOTA-AE1,
`yielding tumor-to-normal
`solid tissue ra
`tios of 2.85-11 to 1. Such enrichment was not observed with the
`control monoclonal
`antibody anti-Tac. Furthermore,
`the ratio of
`incorporated specific antibody (AE-1) to control antibody (anti-
`Tac) at 24 hr was 4.07 for the tumor, whereas
`there was no
`enrichment
`(ratios of 1.01-1.39) with the normal organs.
`Radioactivfty-Toxicity Relationship and Maximum
`Tolerated Dose of Lead-212-AE1
`for 2l2Pb-AEl,
`relationship
`To define
`an activity-toxicity
`increasing
`doses
`(10, 25 and 40 /nCi) of 2'2Pb-AEl were
`administered
`i.v.
`into tumor-free
`athymic nude mice, and the
`effects on hematological
`and serum chemistry parameters were
`defined over a period of 180 days. The activity-limiting
`toxicity
`was bone marrow suppression in animals receiving a more than
`25-/u,Ci dose of lead-radiolabeled AE1 (Fig. 3). Specifically, all
`animals receiving the 40-jnCi dose of 2l2Pb-labeled monoclonal
`antibody died within 6-10 days of therapy, manifesting a mean
`weight
`loss of 25.8%. Pathological
`examination
`of the tissues
`revealed significant
`lymphoid depletion and a marked reduction
`in the size of the spleen (which, on histological
`examination,
`was associated with decreased extramedullary
`hematopoiesis),
`an acellular bone marrow and pulmonary congestion. Four of
`the five animals
`in this group developed
`septicemia, with
`bacterial
`infections affecting the various tissues. All 16 animals
`receiving 25- and 10-/H.CÃ(cid:141)doses of 212Pb monoclonal
`antibody
`i.v. survived, but manifested
`transient granulocytopenia,
`lym-
`phocytopenia
`and modest
`thrombocytopenia
`(Fig. 3). The
`animals
`receiving the 25-/U.CÃ(cid:141)dose developed a nadir
`in their
`hematopoietic
`elements
`in the period spanning days 4-11, with
`
`range in
`leukocyte
`a leukocyte nadir of 400 cells/ju.1 (normal
`mice, 2,600-10,700
`/nl), lymphocyte
`levels of approximately
`250 (normal
`lymphocyte
`range in mice, 1,430-9,940
`jul) and
`thrombocytopenia with a nadir of approximately
`170,000 ¡JL\
`(normal platelet
`count
`range, 592,000-2.97
`X 106//Ltl). The
`platelet and total white blood cell levels returned to the normal
`range by day 20, whereas
`the lymphocyte
`levels were normal
`
`Platelets
`
`ilì*
`
`20
`
`40
`
`100 120 140 160 180
`80
`60
`Days post I.v. therapy
`
`WBC
`
`-20
`
`20
`
`40
`
`100 120 140 160 180
`80
`60
`Days post i.v. therapy
`
`Lymphocytes
`
`-20
`
`:
`40
`
`20
`
`I
`I
`I
`I
`I
`l
`i
`100 120 140 160 180
`80
`60
`Days post i.v. therapy
`
`unlabeled AE1
`
`lOuCi AE1
`
`25uCiAE1
`
`FIGURE 3. The effect of i.v. administered unlabeled AE1 (•).10 ¿¿Ciof
`212Pb-AE1 (O) and 25 /xCi of 212Pb-AE1 (*) on platelet, white blood cell
`(WBC) and lymphocyte levels.
`
`RADIOTHERAPYOKHER2/NEU-ExpRESSiNGTUMOR•Horak et al.
`
`1947
`
`IMMUNOGEN 2086, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Downloaded from
`
`jnm.snmjournals.org
`
`by on October 28, 2014. For personal use only.
`
`90-1
`
`60-«
`50-
`
`0)1
`
`30-,2
`
`20-
`20o
`10-V—
`
`•— n
`
`—O—Aj,
`
`therapy
`
`E1
`12Pb-iOn
`
`2pb-20u
`
`AE1
`
`2Pb-10M
`
`Z\ anti-irTac
`
`12Pb-20nl
`
`TacIII
`
`notherapy-0-
`
`s?3(AikOE90-80-70-60-50-30-10-•
`AE1-»-
`2'2pb-10uCiAEl—O-
`
`212Pb-20uCiAElI'
`
`anti-Tacu
`
`-*-212Pb-10uCianti-Tac_û^
`
`2i2Pb.20uc¡
`
`50Ci
`
`100200Days
`150
`
`after treatment
`
`i
`
`I
`50
`
`0
`
`i
`
`i
`I
`I
`100200Days
`150
`
`After Treatment
`
`i •
`
`l
`
`FIGURE 4. Tumor-free survivalafter i.p. treatment with 20 /¿Ciof 212Pb-AE1
`(O; n = 4), 10 iiCi of 212Pb-AE1 (*; n
`4), 20 /xCi of ?12Pb-anti-Tac (A; n =
`5), 10 /¿Ciof 212Pb-anti-Tac (A; n •5), unlabeled AE1 (D; n = 5) or no
`treatment (•;n
`10) 3 days after i.p. inoculation with 2 x 106 SK-OV-3 cells.
`f>2 = 0.015, 10- and 20-jiCi AE1 treatment groups compared with the two
`control groups not treated with a radiolabeled monoclonal antibody. p2 =
`0.06, comparison of 212Pb-AE1 to 212Pb-anti-Tac groups.
`
`FIGURE 5. Tumor-free survivalafter i.v. treatment with 20 /¿Ciof 2l2Pb-AE1
`(O; n = 4), 10 /¿Ciof 2l2Pb-AE1 (*; n
`4), 20 /¿Ciof 212Pb-anti-Tac (A; n =
`5), 10 /¿Ciof 212Pb-anti-Tac (A; n
`5), AE1 (D; n = 5) or no therapy (•;n =
`10) 3 days after s.c.
`inoculation with 2 x 106 SK-OV-3 cells. p? •0.002,
`comparison of 10- and 20-itCi AE1 treatment groups with the two control
`groups not treated with a radioactive monoclonal antibody.
`
`irradiation because
`of tumors was not due solely to nonspecific
`four of five animals
`receiving
`10 juCi of 212Pb-anti-Tac
`and
`three of
`five receiving
`20 juCi of
`this control
`radiolabeled
`monoclonal
`antibody developed tumors by day 22 (p2 = 0.003,
`animals receiving 10 or 20 /u.Ciof AEl compared with animals
`receiving the same doses of radiolabeled anti-Tac). The animals
`surviving after
`i.p. and s.c.
`inoculation of the SK-OV-3 tumor
`were killed on day 180, and the tissues obtained at necropsy
`were submitted for histológica! evaluation. At that
`time,
`there
`was no evidence of residual
`tumor or bone marrow abnormality.
`A third trial group of mice with small-sized established s.c.
`tumors
`(studied
`14 days after
`tumor
`inoculation when mean
`tumor volume was 15 mm'1) was treated with the same agents
`using the same protocol
`(Fig. 6). The rate of tumor growth was
`inhibited in the period after
`therapy with 20 ¿z.Ciof 212Pb-
`spccific
`antibody. However,
`no complete
`remissions were
`observed. A fourth group of animals had large (mean tumor
`volume,
`146 mm')
`s.c. SK-OV-3
`tumors when they were
`
`no Tx
`
`AE1
`
`20nCi212PbAE1
`
`lOnCi 212Pb anti-Tac
`
`2CHiCi212Pb anti-Tac
`
`30.0-1
`
`25.0-
`
`15.0
`
`10.0-
`
`5.0-
`
`o 3
`
`10
`
`20
`
`60
`50
`40
`30
`Days after treatment
`
`70
`
`80
`
`90
`
`FIGURE 6. Tumor growth of small s.c. SK-OV-3 tumors (mean tumor
`volume, 15 mm3) after i.v. treatment with 20 /¿Ciof ?12Pb-AE1 (.:*), 10 /¿Ciof
`2l2Pb-AE1 (*), 20 /¿Ciof 212Pb anti-Tac (A), 10 ¿iCiof 212Pb anti-Tac (A),
`unlabeled AE1 (O and no therapy (•).The tumor volume for each animal at
`the time indicated was normalized to the tumor volume for that animal that
`was present on the day of treatment. Mean values for the five animals in each
`group are shown.
`
`after day 60. Seventy-five percent of the mice receiving 25 ju,Ci
`of 2l2Pb-AEl
`or 2l2Pb anti-Tac
`developed
`long-term renal
`toxicity with elevated mean blood urea nitrogen or creatinine.
`The
`levels of blood
`urea nitrogen
`and creatinine
`for all
`surviving mice were 39 and 0.8 mg/dl,
`respectively,
`compared
`with the upper limits of the normal range for mice of 28 and 0.7
`mg/dl,
`respectively.
`Lead-212-AE1 Radioimmunotherapy: Therapeutic Efficacy
`We first evaluated the therapeutic
`efficacy of 2l2Pb-AEl
`in
`an SK-OV-3 tumor model
`featuring direct access of the thera
`peutic agent
`to the tumor-containing
`compartment.
`For
`this
`group,
`I0-l2-wk-old
`female nude mice were given i.p.
`injec
`tions of 2 X IO6 SK-OV-3 cells. Three days later, different
`groups of five mice each received i.p. administered ZI2Pb-AEl
`at doses of IO and 20 juCi, the control
`radiolabeled monoclonal
`antibody (2l2Pb-anti-Tac)
`at
`the same dose levels, unlabeled
`AEl
`(20 fig) or no treatment
`(Fig. 4). Lead-2l2-AEl
`treatment
`resulted in a statistically significant delay in the appearance or
`prevention
`of tumor occurrence when mice received
`10- and
`20-jnCi doses; considered together,
`these mice were compared
`with the two groups of mice receiving either no treatment or
`unlabeled AEl
`(p2 = 0.0 15). In interpreting these exploratory
`studies,
`it should be noted that only 60% of the animals
`in the
`control
`group
`injected
`i.p. with SK-OV-3
`cells developed
`tumors.
`group of animals was used to obtain a more
`A second
`evaluation of the effectiveness
`of 2l2Pb-AEl
`ther
`meaningful
`apy in the adjuvant
`setting.
`In this study trial, nude mice were
`injected with 2 X 10" SK-OV-3 cells s.c.
`in the back of the
`neck. Three days later, mice were treated i.v. with 212Pb-AEl or
`control 2l2Pb-anti-Tac
`at dose levels of 10 and 20 /nCi, treated
`with unlabeled AEl or
`received no treatment. The Kaplan-
`Meier plot depicting
`tumor-free
`survival
`after
`treatment
`re
`vealed no differences
`between the group of animals
`receiving
`no therapy (n = 10) and that receiving unlabeled AEl
`(n = 5);
`all animals
`in both groups developed a tumor by day 20.
`In
`contrast, all animals receiving cither 10 or 20 /iCi of 21"Pb-AEl
`remained tumor-free throughout
`the 180-day study period (Fig.
`5; p2 = 0.002,
`animals
`receiving
`low and high doses of
`2l2Pb-AEl
`compared with those in the nontreated group). We
`conclude that
`this effectiveness
`in preventing the development
`
`1948
`
`Tui JOURNALOF NUCLKARMEDICINE•Vol. 38 •No. 12 •December
`
`1997
`
`IMMUNOGEN 2086, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Downloaded from
`
`
`
`by on October 28, 2014. For personal use only. jnm.snmjournals.org
`
`-D-AE-1
`-•-10mCi212PbAE1
`-O- 25(iCi212PbAE1
`-O- 25tiCi212PbAE1
`-•-anti-Tac
`-A-
`10nCi212Pb anti-Tac
`-û- 25nC¡212Pbanti-Tac
`
`30.0-,
`
`<S25.0
`
`_2
`> 20.0
`kO
`2 15.0
`i== 10.0
`
`I
`
`5.0 H
`
`5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80
`Days after treatment
`
`FIGURE 7. Tumor growth of larger (mean tumor volume, 146 mmr!)estab
`lished s.c. SK-OV-3 tumors after i.v. treatment with 25 ^Ci of 21?Pb-AE1 (O),
`25 ,iCi of ?l:>Pb-AE1 (- ), 10 juCi of ?1?Pb-AE1 (»),25 /iCi of pl;;Pb-anti-Tac
`(i,), 10 P.CÃ(cid:141)of ?1?Pb-anti-Tac (A), unlabeled AE1 (IT) and unlabeled anti-Tac
`(•).The tumor volume for each animal at the time indicated was normalized
`to the tumor volume for that animal that was present on the day of treatment.
`Mean values for the five animals in each group are shown.
`
`(Fig. 7). Tumor growth was not
`treated with the same agents
`inhibited in these mice bearing large SK-OV-3 tumors by the
`administration
`of a single doses of 25 /u.Ci of 2l2Pb-specific
`antibody
`as compared
`to those
`receiving
`the nonspccified
`radiolabeled
`anti-Tac
`antibody.
`In particular,
`the size of the
`tumors
`increased
`in all
`treatment
`groups and there were no
`partial or complete
`remissions. Thus,
`Pb-AEl
`did not pro
`vide effective therapy for large established tumors.
`
`DISCUSSION
`armed with toxins or
`antibodies
`The use of monoclonal
`target
`these cytotoxic
`agents
`to
`radionuclides
`to specifically
`tumor cells provides a valuable alternative
`for cancer
`therapy.
`Factors that appear critical
`in developing a

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