throbber

`
`IMMUNOGEN 2045, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2045, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Papers
`
`A specific and potent immunotoxin composed
`of antibody ZME-018 and the plant
`toxin gelonin
`
`Michael G. Rosenblum, James L. Murray,* Lawrence Cheung,* Robert Rifkin, t
`Sydney Salmon, t and Richard Bartholomew+
`*Department of Clinical Immunology and Biological Therapy, M.D. Anderson Cancer Center,
`Houston, TX, USA; tDepartment of Hematology and Oncology, Arizona Cancer Center, Tucson, AZ,
`USA; and :f:HYBRITECH, Inc., Lalolla, CA, USA.
`
`Murine monoclonal antibody ZME-OI8 recognizes a 240 Kda glycoprotein present on the surface of
`most human melanoma cells and on over 80% of human biopsy specimens tested. Gelonin is a ribo(cid:173)
`some-inactivating plant toxin similar in nature and rivaling the activity of ricin A chain. ZME-OI8 was
`coupled to purified gelonin using the reagents SPDP and 2-iminothiolane. The ZME-gelonin conjugate
`was purified by S-300 Sephacryl and Blue Sepharose chromatography, removing unreacted gelonin
`and antibody, respectively. PAGE analysis showed that ZME was coupled to I, 2, or 3 gelonin mole(cid:173)
`cules. The ZME-gelonin conjugate was IfJ6-fold more active than gelonin itself in inhibiting the growth
`of log-phase human melanoma cells in culture. The immunoconjugate was not cytotoxic to antigen
`negative T-24 (human bladder carcinoma) cells. Treatment of melanoma cells with recombinant IFN-a.
`or TNF substantially augmented the cytotoxicity of the immunoconjugate while treatment with IFN--y
`had a minor effect. Using the human tumor colony assay of melanoma cells obtained from fresh
`biopsy specimens, >90% growth suppression was observed in 2 of 4 samples tested at a concentration
`of 250 nglml. In addition, 25% growth suppression was observed with a third sample tested, and no
`growth suppression was observed in I sample. Thus, clonogenic melanoma cells are sensitive in vitro
`to the cytotoxic activity of this immunotoxin at concentrations which we presume are pharmacologi(cid:173)
`cally relevant.
`
`Keywords: Human melanoma; immunotoxins; gelonin; human tumor colony assay; cytokines.
`
`Introduction
`Since the introduction of monoclonal antibody tech(cid:173)
`nology, numerous efforts have been made to exploit
`the specificity of these reagents for cancer therapy. 1- 5
`As a first step, radiolabeled monoclonal antibodies to
`tumor cell surface antigens have been utilized suc(cid:173)
`cessfully to image tumors in patients by external scin(cid:173)
`tigraphy.6·7 Extensive studies with antibodies to mela(cid:173)
`noma antigens8- 10 and to CEA11 •12 among others 13•14
`have demonstrated specific tumor localization in man
`after systemic and intraperitoneal administration. Un(cid:173)
`fortunately, the accumulation of antibody by normal
`organs (i.e., non-tumor) remains a key problem.
`Because of their unique ability to localize within
`human tumors after systemic administration, antibod(cid:173)
`ies have the potential to serve also as targeting vehi-
`
`This research was conducted, in part, by the Clayton Foundation
`for Research.
`Address reprint requests to Dr. Rosenblum at the Department of
`Clinical Immunology and Biological Therapy, M.D. Anderson
`Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA.
`Accepted for publication October 1990.
`
`cles for specific delivery of cytotoxic chemothera(cid:173)
`peutic agents, toxic peptides, biological response
`modifiers, and radionuclides. Antibody ZME-018
`(subclass IgG2A) is reactive with epitope "a" of a
`240,000 molecular weight antigen (gp240) found on the
`surface of over 80% of melanoma cell lines and fresh
`tumor samplesY Labeled with (1 11In) and adminis(cid:173)
`tered systematically to patients with melanoma, anti(cid:173)
`body ZME-018 was found to localize in 77% of soft
`tissue melanoma nodules. 16
`Several investigators have been interested in utiliz(cid:173)
`ing monoclonal antibodies as carriers of extremely ac(cid:173)
`tive protein toxins. 17- 19 In the case of these immuno(cid:173)
`toxins, the antibody serves as a vehicle for delivery of
`the toxin to the tumor. The binding of the antibody to
`the cell-surface target and pinocytosis or internaliza(cid:173)
`tion of the antibody also serves as the mechanism for
`specific intracellular entry of the toxin. 20 The first
`clinical trials with immunotoxins utilized an antibody
`against colorectal carcinoma cells and ricin A chain
`(RTA) or diphtheria toxin A chain. 21·22 Since then, a
`variety of immunotoxins have been developed utiliz(cid:173)
`ing toxins such as abrin, ricin, RTA, diphtheria toxin,
`gelonin, and pseudomonas exotoxin. 23- 26
`
`6 © 1991 Butterworth- Heinemann
`
`Mol. Biother., 1991, vol. 3, March
`
`IMMUNOGEN 2045, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`ZME-gelonin conjugate as an immunotherapeutic agent: Rosenblum eta!.
`
`The plant toxin gelonin is extracted from the seeds
`of the Gelonium multiflorum plant and exists in nature
`as a single polypeptide chain with a molecular weight
`of 29 KDa. 27 While the A chain of ricin (RTA) has
`been popular for use in immunotoxins, 28 •29 gelonin,
`one of the 60 S ribosome inactivating hemitoxins, has
`several advantages over RTA. Gelonin appears to be
`more stable to chemical and physical treatment than
`RTA. 3° Furthermore, gelonin itself does not bind to
`cells and is, therefore, non-toxic (e,Z(cept in high con(cid:173)
`centrations) and is safe to manipulate in the laborato(cid:173)
`ry. While the primary sequence of gelonin is un(cid:173)
`known, mechanistic studies indicate that it operates
`identically to that of RTA. 31,32
`We have utilized
`the antimelanoma antibody
`ZME-018 as a model delivery vehicle for the plant
`toxin gelonin. The ZME-gelonin conjugate was syn(cid:173)
`thesized, purified, and tested in a variety of in vitro
`human tumor models to determine its potential utility
`as an immunotherape'tttic agent.
`
`Materials and Methods
`Materials
`
`Antibody ZME-018 was prepared at Hybritech, Inc.
`(LaJolla, CA, USA), using salt fractionation and
`DEAE chromatography and was judged homogenous
`by SDS PAGE. 33 SPDP reagent (N-succinimidyl 3:[2-
`pyridyldithio] propionate), Sephacryl S-300 gel perme(cid:173)
`ation resin and Blue Sepharose CL-6B resin were
`purchased from Pharmacia, Piscataway, NJ, USA.
`2-Iminothiolane HCl was purchased from Pierce
`Chemical Co., Rockford, IL, USA. All materials were
`of reagent grade or higher. For estimation of the cell(cid:173)
`free protein synthesis inhibitory activity of the toxin,
`a rabbit reticulocyte translation kit was purchased
`from Bethesda Research Labs (Bethesda, MD, USA).
`
`Methods
`
`Coupling of ZME-018 to gelonin. A stock solution of
`SPDP (6 mg/ml) in dry DMF was prepared. To 1 ml of
`a PBS solution containing 1 mg of ZME-018 in a 12 x
`75 mm glass test tube, SPDP was slowly added to a
`5-fold molar excess (approx. 10 J.Ll of stock solution).
`The mixture was vortexed every 5 minutes for 30 min(cid:173)
`utes at room temperature.
`Excess unreacted SPDP was removed from the
`sample by gel filtration chromatography on a Sepha(cid:173)
`dex G-25 column (1 x 24 em) pre-equilibrated in 100
`mM sodium phosphate buffer pH 7.0 containing 0.5
`mM EDTA (Buffer A). Fractions (0.5 ml) were col(cid:173)
`lected and analyzed for protein content using the
`Bradford dye binding assay. 34 Absorbance (600 nm)
`was monitored in a 96-well plate using a Bio-TEK Mi(cid:173)
`croplate autoreader. Antibody eluted at the void vol(cid:173)
`ume (fractions 14-20) and these fractions were pooled
`and kept at 4°C.
`For these studies, gelonin toxin was extracted from
`the seeds of Gelonium multiflorum and purified to ho-
`
`mogeneity utilizing the method of Stirpe et alP One
`milligram of purified gelonin (2 mg/ml PBS) was added
`to triethanolamine hydrochloride (TEA/HCl) buffer to
`a final concentration of 60 mM TEA/HCl and adjusted
`to pH 8.0. The solution was made 1 mM EDTA. 2-
`Iminothiolane stock solution (0.5 M in 0.5 M TEA/
`HCl pH 8.0) was added to a final concentration of 1
`mM and the sample was incubated for 90 minutes at
`4°C under nitrogen gas.
`Excess 2-iminothiolane reagent was removed by gel
`filtration on a column of Sephadex G-25 (1 x 24 em)
`pre-equilibrated with 5 mM bis-tris acetate buffer pH
`5.8 containing 50 mM NaCl and 1 mM EDTA. Frac(cid:173)
`tions were analyzed for protein content in 96 well mi(cid:173)
`crotiter plates using the Bradford dye binding assay.
`Gelonin eluted at the void volume (fractions 14-20).
`SPDP-modified antibody ZME was mixed with an
`equal weight of 2-iminothiolane modified gelonin. This
`proportion corresponded to a 5-fold molar excess of
`gelonin as compared to antibody. The pH of the mix(cid:173)
`ture was adjusted to 7.0 by the addition of0.5 M TEA/
`HCl buffer (pH 8.0), and the mixture was incubated
`for 20 hours at 4°C under nitrogen. Iodoacetamide (0.1
`M in H20) was added to a final concentration of 2 mM
`to block any remaining free sulfydryl groups, and in(cid:173)
`cubation was continued for an additional hour at 25°C.
`
`Purification of ZME-gelonin complexes. To remove
`low molecular weight products and non-conjugated
`gelonin, the reaction mixture was applied to a Sepha(cid:173)
`cryl S-300 column (1.6 x 31 em) previously equilibrat(cid:173)
`ed with PBS. Fractions (1.0 ml) were collected and 50
`J.Ll aliquots were analyzed for protein content using the
`Bio-Rad dye binding assay. To remove unconjugated
`ZME-018, the high molecular peak (Fractions 28-41)
`from the S-300 column was applied to an affinity chro(cid:173)
`matography column of Blue Sepharose CL-6B (1 x 24
`em) pre-equilibrated with 10 mM phosphate buffer
`(pH 7.2) containing 0.1 M NaCl. After sample loading,
`the column was washed with 50 ml of buffer to elute
`completely non-conjugated antibody. The column was
`eluted with a linear salt gradient of 0.1 to 2M NaCl in
`10 mM phosphate buffer pH 7.2. Protein content of
`the eluted fractions was determined by the dye-bind(cid:173)
`ing assay described previously. 34
`
`Cell culture methods. Human bladder carcinoma
`(T-24), human cervical carcinoma, or human metastat(cid:173)
`ic melanoma tumor cells A375M or AAB-527 were
`maintained in culture using minimal essential medium
`(MEM) supplemented with 10% heat-inactivated fetal
`bovine serum plus 100 J.LM non-essential amino-acids,
`2 mM L-glutamine, 1 mM sodium pyruvate, vitamins,
`and antibiotics. Cultured cells were screened routine(cid:173)
`ly and found free of mycoplasma infection.
`
`Cell proliferation assay. Cell lines were maintained in
`culture in complete medium at 37°C in a 5% C02-hu(cid:173)
`midified air incubator. For assays with combinations
`ofTNF, immunotoxins, riFNaA, and riFN'Y, cultures
`
`Mol. Biother., 1991, vol. 3, March
`
`7
`
`IMMUNOGEN 2045, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`ZME-oiS + ZHE-saONIN
`
`Papers
`
`1.0
`
`E' 0.8
`c
`0
`"<;)" 0.6
`.!£l
`w
`u :z: 0.4
`<C rn
`0:
`0 en
`rn 0.2
`
`<C
`
`FLOW THRU
`
`ZME-GELONIN
`
`.30
`
`.25
`
`.20
`
`.15
`
`.10
`
`.05
`
`E'
`c
`0
`"<;)"
`.!£l
`w
`u
`:z:
`<C rn
`0:
`0 en
`rn
`<C
`
`0.0
`0
`
`20
`
`60
`40
`(1 ml)
`FRACTION
`Figure 1. Purification of ZME-gelonin by S-300 gel permeation
`chromatography. High molecular weight material consisting of
`unmodified ZME and ZME-gelonin conjugate was eluted in
`fractions 28-41. Free gelonin eluted in fractions 45-65.
`
`80
`
`100
`
`were washed, detached using versene, and resuspend(cid:173)
`ed in complete medium at a density of 25 x 103 cell/
`mi. Two hundred ,_..1 aliquots were dispensed into 96-
`well microtiter plates and the cells were then allowed
`to adhere. This results in a sparsely seeded population
`of cells. Mter 24 hours, the media were replaced with
`media containing different concentrations of either
`immunotoxin, gelonin, TNF, IFN'Y, or IFNa. The
`cells were incubated for 72 hours and analyzed for rel(cid:173)
`ative cell proliferation by crystal violet staining.
`
`Crystal violet staining. Cells were washed 3 times with
`PBS containing calcium and magnesium fixed and
`stained with 20% (v/v) methanol containing 0.5% (w/v)
`crystal violet. Bound dye was eluted with 150 ,....I of
`Sorensen's citrate buffer (0.1 M sodium citrate, pH
`4.2-50% (v/v) ethanol) for 1 hour at room tempera(cid:173)
`ture. The absorbance was measured at 600 nm using a
`Bio-Tek microplate reader. Relative cell proliferation
`(RCP) was calculated as follows:
`
`Mean Absorbance (Drug Treated)
`OOM
`RCP = - - - - - - - - - - - - - - - - X 1
`/0
`Mean Absorbance (Non-drug Treated)
`
`[eq 1]
`
`Human tumor colony assay. Thmor biopsy specimens
`were obtained from melanoma patients during clini(cid:173)
`cally indicated biopsy procedures. Portions of tumor
`not required for standard diagnostic evaluation were
`transferred promptly to the human tumor cloning lab(cid:173)
`oratory, wherein tumor cell suspensions were pre(cid:173)
`pared aseptically. 35 Additionally, the A375P melanoma
`and the CEM leukemia cell lines from the American
`Type Culture collection (Rockville, MD, USA) were
`also studied. Testing for the effects of ZME-gelonin on
`the fresh melanoma cell suspensions and cell lines was
`assessed in the HTCA using standardized procedures
`for tumor cell plating in semi-solid medium (agarose)
`in the presence of complete medium containing 10%
`fetal calf serum. Each 0.5 ml culture plate contained
`100,000 cells from fresh tumors and 10,000 cells from
`
`80
`
`100
`
`0
`0
`
`20
`
`60
`40
`FRACTION ( 1 ml)
`Figure 2. The high-molecular weight material from S-300 chro(cid:173)
`matography was applied to a column of Blue Sepharose and
`eluted with a linear salt gradient (0-300 mM NaCI). Two protein
`peaks were demonstrated: a flow-through peak (fractions
`11-25) and a bound peak eluted with high salt (fractions
`44-80).
`
`the celllines. 36- 38 ZME-gelonin prepared as described
`above was tested by addition to the culture plates
`shortly after tumor cell plating. ZME-gelonin was
`added to triplicate plates at each of four concentra(cid:173)
`tions 0.025 ng/ml to 250 ng/ml. In addition to untreated
`control plates, unconjugated ZME-018 monoclonal
`antibody and free gelonin were tested in parallel. Cell
`lines and tumor cell cultures were incubated for an
`average of 10 days at 37°C in 5% C02 in air in a humid(cid:173)
`ified incubator. Colony formation was evaluated with
`a viability stain39 and an automated image analysis in(cid:173)
`strument optimized for colony counting.40 Percent sur(cid:173)
`vival of ZME-018-treated cultures in relation to un(cid:173)
`treated controls were determined
`in
`the same
`experiments. Dose-response curves were then plotted
`graphically.
`
`Results
`Conjugation and purification of
`ZME-gelonin immunotoxin
`Both ZME-018 and gelonin were modified by expo(cid:173)
`sure to the reagents SPDP and 2-iminothiolane, re(cid:173)
`spectively. Modification of ZME with 2 to 3 molecules
`of SPDP was accomplished by addition of a 6-fold
`molar excess of SPDP. Analysis of the number of
`SPDP molecules was accomplished spectrophotomet(cid:173)
`rically.41 The derivitization of ZME-018 and gelonin
`and the coupling were varied to provide optimal yield
`of ZME-gelonin conjugate (data not shown). Figure 1
`demonstrates separation of the ZME-gelonin conju(cid:173)
`gate from gelonin. Blue sepharose CL-6B chromatog(cid:173)
`raphy (Figure 2) was effective at separating free ZME
`from ZME-gelonin conjugate. As demonstrated in
`Figure 3, the final eluent from Blue Sepharose was
`free of unreacted gelonin and ZME-018. The final
`product contained ZME-coupled to 1, 2, and 3 gelonin
`molecules. Average gelonin content was 1.5 molecules
`per antibody molecule. Average yield of purified im-
`
`8
`
`Mol. Biother., 1991, vol. 3, March
`
`IMMUNOGEN 2045, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`ZME-gelonin conjugate as an immunotherapeutic agent: Rosenblum et at.
`
`~
`"'
`~
`
`2.0
`
`1.5
`
`w
`(._)
`:z:
`<C
`aJ 1.0
`cr::
`Cl
`(f)
`aJ
`<C
`
`0.5~
`
`0.0 '---'='-~~._,_,_[_~~~o.L_~~ ........... ~~~_._,J
`10 1
`10 2
`10 3
`10 4
`1QO
`CONCENTRATION (ug/ml)
`Figure 4. Comparative ELISA Assay of ZME (0) and ZME ge(cid:173)
`lonin (•). Various concentrations of either ZME or ZME-gelonin
`were added to antigen-positive (AAB-527) human melanoma
`cells and incubated at room temperature for 60 minutes. The
`cells were washed and a standard ELISA was performed for mu(cid:173)
`rine antibody.
`
`negative T-24 cells as estimated by ELISA assay (data
`not shown). Therefore, the apparent change in anti(cid:173)
`body affinity resulting in the coupling of ZME to ge(cid:173)
`lonin does not appear to reduce selectivity of the anti(cid:173)
`body.
`
`Cytotoxicity of ZME-gelonin
`Cytotoxicity studies of the ZME-gelonin conjugate
`were performed on antigen-positive cells after contin(cid:173)
`uous (72-hour) exposure to the immunotoxin or native
`gelonin. As shown in Figure 5, 50% cell death by the
`immunotoxin occurred at a concentration of approxi(cid:173)
`mately 0.02 f.Lg/ml. A similar effect (i.e., 50% cell
`death) was observed for native gelonin at a concentra(cid:173)
`tion of 5 f.Lg/ml. These concentrations are equated to a
`dose of 0.1 nM for ZME-gelonin and a dose of 100 nM
`of free gelonin to achieve reduction of the number of
`viable cells to 50% of control value.
`The activity of both ZME-gelonin immunotoxin and
`free gelonin were determined using a cell-free transla(cid:173)
`tion assay system. 42 One unit of activity in this assay
`was defined as the amount of protein required to pro(cid:173)
`vide 50% inhibition of protein synthesis compared to
`untreated controls. Utilizing this assay, the specific
`activity of both the native gelonin and the ZME(cid:173)
`gelonin conjugate were determined to be 2 x 108 f.L/mg
`and 8.2 x 105 f.L/mg, respectively. Target cells then
`were treated with various concentrations of ZME-ge(cid:173)
`lonin and gelonin alone. As shown in Figure 6, a 50%
`inhibitory concentration was obtained using 50 units/
`ml of ZME-gelonin conjugate while 1 x 107 units/ml
`of the free gelonin were required to achieve the same
`effect.
`The effect of ZME-gelonin was determined against
`antigen-negative T-24 cells in log-phase culture. Ge(cid:173)
`lonin alone produced 50% cytotoxicity in these cells at
`a concentration of 10 f.Lg/ml; similar to that found on
`AAB-527 melanoma cells (data not shown). However,
`the ZME-gelonin immunotoxin was not active against
`
`Figure 3. Silver stained PAGE analysis of ZME-gelonin conju(cid:173)
`gation and purification procedure.
`LANE 1-ZME-018 antibody standard.
`LANE 2-Native gelonin.
`LANE 3-Unpurified ZME-gelonin reaction mixture showing
`the presence of unreacted ZME antibody and unreacted gelon(cid:173)
`in. Also shown are high molecular weight conjugate bands cor(cid:173)
`responding to ZME + 1 gelonin molecule, ZME + 2 gelonin
`molecules and ZME + 3 gelonin molecules.
`LANE 4-High molecular weight peak from S-300 chroma(cid:173)
`tography demonstrating removal of low molecular weight com(cid:173)
`ponents including most of the free gelonin.
`LANE 5--Fiow-through fraction from application of the con(cid:173)
`jugate to a Blue Sepharose column. All of the unreacted anti(cid:173)
`body eluted from the column in the low-salt wash.
`LANE 6--Final purification product eluted from the Blue
`Sepharose column. As shown, there was no remaining free an(cid:173)
`tibody and only small amounts of unreacted gel-onin in the elut(cid:173)
`ed fraction.
`
`munotoxin was approximately 20% of expected maxi(cid:173)
`mal value.
`
`Binding of ZME-gelonin to cells in culture
`The binding of ZME-gelonin immunotoxin to antigen
`positive (AAB-527 cells) or antigen negative (T-24
`cells) was tested by ELISA assay. As shown in Figure
`4, both native ZME and the ZME-gelonin conjugate
`bound well to target cells after 60-minute exposure.
`Surprisingly, the ZME-gelonin conjugate bound target
`cells better than did the native antibody. This increase
`was not due to modification of the antibody by SPDP
`since SPDP-modified ZME behaved identically to that
`of native ZME. The increase was also not due to bind(cid:173)
`ing of target cells to the gelonin portion of the mole(cid:173)
`cule since pre-treatment of target cells with native
`gelonin had no effect on either antibody or immuno(cid:173)
`toxin binding. Thus, these results suggest that cou(cid:173)
`pling of ZME to gelonin may affect antibody affinity
`for the gp240 antigen.
`Neither ZME nor ZME-gelonin bound to antigen
`
`Mol. Biother., 1991, val. 3, March
`
`9
`
`IMMUNOGEN 2045, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Papers
`
`100
`
`75
`
`50
`
`25
`
`__j
`
`C) a:
`1-:z:
`8
`6
`
`o GELONIN
`e ZME-GELONIN
`
`o~~~~~~~~~~~~~~~~
`
`10- 3
`
`10- 2
`
`10 1
`10°
`10- 1
`(nM)
`CONCENTRATION
`Figure 5. Cytotoxicity of ZME-gelonin and free gel on in on log(cid:173)
`phase AAB-527 cells. Cells were plated for 24 hours and various
`doses of either gelonin or ZME-gelonin were added in complete
`media and incubated for 72 hours. Cell viability was determined
`by crystal violet staining.
`
`100
`
`80
`
`60
`
`40
`
`20
`
`.......
`0
`_,_,
`'-
`c
`0
`LJ
`.......
`0
`
`~
`
`•••
`•,.
`'
`\
`•
`•
`'
`\
`•
`' •
`• \ •
`
`\
`
`0-o-o-o-o
`\
`0.0
`
`0
`
`0
`
`\
`\
`\
`
`0
`
`o~~~~~~~~~~~=L~=L~~
`
`10°
`
`10 1
`
`10 7
`
`108
`
`10 4
`10 3
`10 2
`10 6
`105
`Concentration (u/ml)
`Figure 6. Cytotoxicity of ZME-gelonin and free gelonin on log(cid:173)
`phase AAB-527 cells. As in Figure 6, log-pha~e AAB-527 cells
`were treated with various doses of gelonin or ZME-gelonin. The
`biological activity of gelonin or ZME-gelonin was determined by
`a cell-free translation inhibition assay. One unit activity was de(cid:173)
`fined as the amount of toxin required to inhibit translation activ(cid:173)
`ity by 50%.
`
`100
`
`80
`
`60
`
`40
`
`20
`
`__j
`
`C) a:
`I -
`:z:
`C)
`LJ
`
`l.J_
`C)
`
`~
`
`<)
`
`0
`
`0
`
`0
`
`0
`
`0
`
`0
`
`0 0-0
`
`8 TARGET
`
`(AAB-527)
`
`<>NON-TARGET
`
`IT -24)
`
`0
`10- 2
`
`10 1
`10°
`10- 1
`CONCENTRATION (ug/ml)
`Figure 7. Cytotoxicity of ZME-gelonin on antigen positive mel(cid:173)
`anoma cells (AAB-527) and antigen negative T-24 cells in cul(cid:173)
`ture. As in Figure 6, log-phase AAB-527 or T-24 cells were treat(cid:173)
`ed for 72 hours with various concentrations of ZME-gelonin
`immunotoxin. Cell viability was then determined by crystal vio(cid:173)
`let staining.
`
`100
`
`80
`
`60
`
`40
`
`>-
`I -
`>--<
`LJ
`>--<
`><
`0
`I -
`C)
`I -
`>-
`LJ
`
`l.J_
`C)
`
`~ 20
`
`o 15A8
`e ZME-018
`
`0
`10-1
`
`10 3
`
`10 2
`10 1
`10°
`CONCENTRATION (ug/ml)
`Influence of free antibody on ZME-gelonin cytotoxicF
`Figure 8.
`ty. A fixed concentration of ZME-gelonin was mixed with in(cid:173)
`creasing concentrations of ZME-018 or irrelevant antibody
`15A8. The mixture was then added to log-phase AAB-527 mela(cid:173)
`noma cells for 72 hours. As shown, increasing amounts of
`ZME-018 antibody prevented ZME-gelonin cytotoxicity. This ef(cid:173)
`fect was specific since irrelevant 15A8 antibody had no effect.
`
`these cells even at the highest concentration tested
`(Figure 7).
`If the cytotoxic effect of ZME-gelonin is mediated
`through recognition of a specific cell-surface antigen,
`addition of free ZME antibody should reduce cytotox(cid:173)
`icity of the immunotoxin. To demonstrate this, a fixed
`immunotoxin designed
`to
`dose of ZME-gelonin
`achieve 80% cytotoxicity was added to log-phase mel(cid:173)
`anoma cells in culture. Simultaneously, various doses
`of ZME or irrelevant antibody were added. As shown
`in Figure 8, high concentrations of ZME-018 antibody
`substantially reduced the cytotoxicity of ZME-gelonin
`conjugate, while addition of irrelevant antibody had
`no effect.
`
`Modulation of ZME-gelonin cytotoxicity with
`IFNa, IFNy, and TNF
`To determine the effects of treatment with various bio(cid:173)
`logical response modifiers on immunotoxin cytotoxici-
`
`ty, log-phase melanoma cells were treated for 24 hours
`with fixed doses of IFNaA (200 f.L/ml), IFN-y (20,000
`f.L/ml), or rTNF-a (20,000 f.L/ml). These doses were de(cid:173)
`termined previously to have minimal (~20%) cytotox(cid:173)
`ic effect against these cells. The cells then were
`washed and treated for 72 hours with various doses of
`ZME-gelonin. As shown in Figure 9, treatment with
`riFN-y resulted in a 2-fold increase in sensitivity to the
`immunotoxin. However, pre-treatment with both
`riFNaA and TNF both resulted in a 100-fold increase
`in sensitivity to the immunotoxin.
`
`Human tumor colony assay (HTCA) of
`ZME-gelonin immunotoxin
`An alternative method of assessing in vitro cytotoxici(cid:173)
`ty against human cells in culture is the soft-agar
`HTCA. 39 Various doses of ZME-gelonin immunotoxin
`were added to an antigen positive (A-375 melanoma)
`
`10
`
`Mol. Biother., 1991, vol. 3, March
`
`IMMUNOGEN 2045, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`ZME-gelonin conjugate as an immunotherapeutic agent: Rosenblum eta/.
`
`·- '
`
`-.. __
`-.
`---._ ' '
`•--.. ___
`
`1000
`
`100
`
`10
`
`_j
`<C
`>
`>-<
`>
`a:
`::::J
`(f)
`
`1-:z:
`w
`u
`a:
`w
`0...
`
`,--<
`0
`c_
`_,__,
`c:
`0
`u
`
`"---
`0
`
`;,-E!
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`0
`10-2
`
`10 1
`10-1
`100
`Concentration (ug/ml)
`Figure 9. Effect of IFN-a, IFN--y, and TNF on ZME-gelonin cyto(cid:173)
`toxicity. Log-phase AAB-527 cells were treated with various
`doses of ZME-gelonin alone (-•), or ZME-gelonin in the pres(cid:173)
`ence of fixed concentrations of IFN--y (<>), IFN-a (6), or TNF
`(•· · ·•). Doses of IN F-a, IFN--y, or TNF alone demonstrated mini(cid:173)
`mal cytotoxicity (<20%) against this cell line.
`
`10 2
`
`and antigen negative CEM cell lines. Survival of colo(cid:173)
`nies was assessed 72 hours after addition. As shown in
`Figure 10, doses of immunotoxin between 0.25 and
`2500 ng/ml resulted in almost complete suppression of
`colony survival of the antigen-positive line. Unconju(cid:173)
`gated ZME-018 and free gelonin alone or combined
`together were not cytotoxic. There was no effect
`against the antigen-negative line (CEM) even at the
`highest concentration of immunotoxin tested.
`The effect of ZME-gelonin against four different
`fresh biopsy specimens is shown in Figure 11. Eighty
`to 90% reduction in survival of melanoma colony
`forming cells was found in two specimens at the high(cid:173)
`est immunotoxin dose tested (250 ng/ml). Only a mod(cid:173)
`est (25%) reduction in colony number was noted with
`a third specimen. Growth enhancement·was noted in
`the fourth sample at the highest immunotoxin dose. In
`addition, growth enhancement was observed in one
`specimen at low doses, while higher doses produced
`substantial cytotoxicity. As in the cell line experi(cid:173)
`ments, addition of unconjugated ZME-018 and free
`gelonin were not cytotoxic at the doses tested. The
`surface antigen expression of gp240 and the internal(cid:173)
`ization rates for ZME-gelonin conjugate were not ex(cid:173)
`amined in these four lines and are currently under in(cid:173)
`vestigation.
`
`Discussion
`
`The necessity for precisely targeting cancer therapy is
`critical since adequate tumor response is dependent
`upon delivery and maintenance of intratumor thera(cid:173)
`peutic concentrations of drugs. Site-directed therapy
`has become a goal of several investigators utilizing
`monoclonal antibodies as specific carriers of thera(cid:173)
`peutic agents.
`The cytotoxic agents frequently utilized for anti(cid:173)
`body conjugates primarily fall into three classes of
`agents: toxins, radionuclides, and chemotherapeutic
`agents. Antibody conjugates with each of these types
`
`1L-~~~~~--~~~~~~~~~~
`
`.01
`
`.1
`
`100
`10
`CONCENTRATION (ng/ml)
`Figure 10. Effect of ZME-gelonin on antigen positive (A-375 e)
`and antigen negative (CEM D) cells in a human tumor stem cell
`assay. There was no effect of ZME-gelonin on antigen negative
`cells. In contrast, there was only 2% survival of antigen positive
`cells exposed to 20 ng/ml of immunotoxin.
`
`1000 10000
`
`10110r
`
`100
`
`10
`
`_j
`<C
`>
`>-<
`> a:
`
`::::J
`(f)
`
`1-:z:
`w
`u
`a:
`w
`0...
`
`1~~~L-~~~~~~~~~~~~
`
`100
`
`1000
`
`.01
`
`.1
`
`10
`CONCENTRATION (ng/ml)
`Figure 11. Cytotoxic effect of ZME-gelonin on stem cell surviv(cid:173)
`al of different lines obtained from fresh biopsy specimens of 4
`different patients. As shown by the various symbols, two line
`exhibited marked growth inhibitory effects with the ZME-gelon(cid:173)
`in conjugate (•,•). Approximately 25% growth inhibition was
`obtained with one line (6) and slight growth enhancement was
`observed with another line tested (A).
`
`of agents offer substantial promise as therapeutic
`agents as well as unique problems. 43 •44 Immunoconju(cid:173)
`gates containing plant toxins offer a unique advantage
`to other types of antibody conjugates because:
`
`1) Doses of immunotoxins required for antitumor ac(cid:173)
`tivity are, in general, much lower than that re(cid:173)
`quired for antibody-drug conjugates.
`2) The conjugation of toxins to antibodies does not
`appear to affect antibody affinity.
`
`Previous studies have described a number of anti(cid:173)
`body-toxin conjugates containing gelonin. 45- 51 Re(cid:173)
`cently, Ozawa et al. 50 constructed a gelonin immuno(cid:173)
`toxin comprised of antibody B4G7 which binds to the
`cellular receptor for epidermal growth factor (EGF).
`This B4G7-gelonin conjugate was highly cytotoxic for
`EGF receptor expressing cells but was non-cytotoxic
`
`Mol. Biother., 1991, vol. 3, March
`
`11
`
`IMMUNOGEN 2045, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Papers
`
`for receptor-deficient cells. Sivam et al. 51 have made
`a conjugate of the antimelanoma antibody 9.2.2.7 with
`gelonin and compared in vitro and in vivo cytotoxic
`activity with a 9.2.2.7 conjugate of abrin and ricin A
`chain. These studies demonstrate that gelonin conju(cid:173)
`gates demonstrate substantial cytotoxic effects selec(cid:173)
`tively against antigen-positive cells in vitro. In vivo
`experiments demonstrated that gelonin conjugates are
`not toxic up to 2 mg total antibody dose/mouse and
`that multiple I. V. administration of gelonin immuno(cid:173)
`toxin significantly retarded the growth of an estab(cid:173)
`lished S.C. human tumor xenograft in nude mice.
`Compared to conjugates with abrin and ricin, gelonin
`conjugates appeared to have similar potency, better
`selectivity and tumor localization with more signifi(cid:173)
`cant in vivo therapeutic properties.
`In the current study, we have demonstrated that the
`ZME-gelonin immunoconjugate appears in vitro to be
`a highly cytotoxic reagent. Moreover, the cytotoxicity
`of the immunotoxin appears to be specific for antigen(cid:173)
`bearing cells.
`Substantial augmentation of ZME-gelonin cytotox(cid:173)
`ic effect was observed with pre-treatment of riFNaA
`and rTNF but not with riFN--y. While previous stud(cid:173)
`ies by our group52 and by others53- 57 have demonstrat(cid:173)
`ed that IFNa and IFN--y can up-regulate some mela(cid:173)
`noma surface antigens such as P-97, there was little
`effect of these biological response modifiers on the
`high molecular weight antigen (gp240) recognized by
`ZME. Therefore, the mechanism ofTNF-a and IFN-a
`induced augmentation of ZME-gelonin activity is not
`clear but could involve changes in the antibody inter(cid:173)
`nalization rate, changes in the cellular processing of
`the immunotoxin, or a modulation of any one of sever(cid:173)
`al interferon-mediated enzymes. 57
`The evaluation of ZME-gelonin using the HTCA is
`of potential clinical interest. In other studies, the
`HTCA has been used as a predictive model for clinical
`response of a given tumor to a given potential thera(cid:173)
`peutic agent. 37•58 The results indicate that ZME-gelon(cid:173)
`in was active against 3 of 4 fresh human tumor lines
`tested and against all antigen positive melanoma cell
`lines. These findings are important since most human
`tumors are composed of heterogeneous populations of
`malignant cells with varying antigen expression. Anti(cid:173)
`genic modulation by tumor cells may result in the de(cid:173)
`velopment of resistance to ZME-gelonin, and such re(cid:173)
`sistance might be predicted utilizing the HTCA. Phase
`I clinical evaluation and in vivo pharmacokinetics will
`be required to determine if the doses of ZME-gelonin
`required for cytotoxic effect against fresh human
`tumors in culture will be achievable without unaccept(cid:173)
`able toxicity.
`While site-directed therapy with immunotoxins,
`specifically ZME-gelonin, appears to be feasible, at
`least in vitro, there still remain substantial practical
`barriers to the optimal delivery of immunotoxins and
`maintenance of therapeutic concentrations of drug at
`the tumor sites. Further studies with immunotoxins
`will explore in vivo behavior of ZME-gelonin and
`
`evaluation of this reagent as a potential and practical
`tool for clinical use.
`
`Reference

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