throbber
JOURNAL OF HEMATOTHERAPY 1:85-94 (1992)
`Mary Ann Liebert, Inc., Publishers
`
`Initial Trial of Bispecific Antibody-Mediated
`Immunotherapy of CD15-Bearing Tumors:
`Cytotoxicity of Human Tumor Cells Using a
`Bispecific Antibody Comprised of Anti-CD 15
`(MoAb PM81) and Anti-CD64/Fc7RI (MoAb 32)
`
`EDWARD D. BALL,1 PAUL M. GUYRE,3 LETHA MILLS,1 JAN FISHER,4
`NATHAN B. DINCES,4 and MICHAEL W. FÄNGER3
`
`ABSTRACT
`The high-affinity receptor for IgG, Fc-yRI, expressed on monocytes and interferon-7
`(IFN-7)-stimulated neutrophils, is a trigger molecule for cell-mediated cytotoxicity. We have
`prepared murine monoclonal antibodies (MoAb 22 and MoAb 32) that bind to Fc^RI outside
`the ligand binding site and thus bind to and trigger cytotoxicity that is not competed by other
`immunoglobulins. Because of these properties, it seemed that these MoAbs would be very
`useful for the development of bispecific antibodies (BsAb) for targeting normal cellular
`immune defense mechanisms as a new form ofimmunotherapy for treatment ofcancer. BsAbs
`incorporate into a single molecule the binding specificities of two different antibodies, and,
`thus, can be used to target myeloid cells to tumors, ensure activation of cellular cytotoxic
`mechanisms, and target cell lysis and/or phagocytosis. BsAbs were prepared using anti-FcyRI
`MoAb and an anti-myeloid cell MoAb, PM81, reactive with the CD15 antigen, for studies of
`antibody-dependent cellular cytotoxicity. Conjugates were made by cross-linking sulfhydryl
`groups of Fab fragments of MoAb 32 or 22 (both IgG,) and sulfhydryl groups added to intact
`PM81 (an IgM) using A^-succinimdyl-acetyl-S-thioacetate (SATA). The resulting product was
`purified by high-performance size-exclusion chromatography. The ability of the BsAbs to
`mediate attachment of human monocytes to tumor target cells was confirmed in a microtiter
`well assay of binding of MTT-labeled U937 cells (a human Fc7RI-bearing cell line) to SKBR-3
`(PM81-reactive breast carcinoma) target cells. The ability of the BsAbs to mediate killing of
`HL-60 promyelocytic leukemia cells was studied using a 6-hour Chromium-51 release assay.
`Effector cells were monocytes obtained by cytopheresis and cultured for 18 hours with IFN-7.
`Monocytes alone caused minimal killing (5-20%), monocytes plus BsAb caused moderate
`killing (20-50%), and monocytes plus BsAb plus human serum resulted in maximal killing
`(50-80%). Experiments were performed to test the ability of the BsAb to purge bone marrow
`
`Departments of 'Medicine, 2Microbiology, and 3Physiology, Dartmouth Medical School, Hanover, NH.
`4Medarex, Inc., West Lebanon, NH.
`
`85
`
`1 of 10
`
`BI Exhibit 1013
`
`

`

`BALL ET AL.
`
`of small numbers of leukemia cells using bone marrow mononuclear phagocytes treated for 18
`hours with IFN-7 prior to adding target cells. Without the addition of human serum as a
`source of complement, a 90% depletion of clonogenic HL-60 cells could be demonstrated.
`With human complement, up to 95 % depletion was seen. Thus, this BsAb possessed the ability
`to lyse tumor cell targets by two different mechanisms, complement and cell-mediated lysis. In
`a Phase I clinical trial, 4 patients with CD15+ tumors were treated with up to 48 mg of this
`BsAb with no toxicity. Of this group, the patient with acute myelogenous leukemia experi-
`enced a transient 30-60% reduction in circulating leukemic blast cells during each of six
`infusions over a 2-week period. Although in vitro assays indicated maximal effectiveness
`between 1 and 10 |xg/ml of this BsAb, in vivo reduction in circulating cell counts was observed
`when peak serum concentrations were as low as 50 ng/ml. As such, this BsAb may be useful for
`in vivo therapy of high-risk tumors, especially after induction of remission or after bone
`marrow transplant, i.e., for treatment of minimal residual disease in patients with CD15-
`expressing tumors, including acute myeloid leukemia, small cell carcinoma of the lung, and
`colon and breast carcinoma.
`
`INTRODUCTION
`
`Although MURiNE monoclonal antibodies (MoAb) directed to tumor-associated antigens have consid-
`erable therapeutic potential, there are also several limitations to their use in vivo (Dillman, 1989). These
`include lack of host cellular or humoral effector mechanisms, immune response to the foreign protein, and
`delivery of adequate amounts of the MoAb to the tumor. For a MoAb to be an effective anticancer agent, it is
`necessary that it work in concert with an effector mechanism such as complement (Stepan et al, 1984) or a
`cellular effector, as in antibody-dependent cellular cytotoxicity (ADCC)' (Ortaldo et al, 1987). Many
`potentially useful MoAbs are not able to use these effector mechanisms because they are not of the appropriate
`subclass of immunoglobulin. It has become clear that murine Ig of the IgG2a and IgG3 subclasses are the most
`effective at mediating ADCC by virtue of their ability to bind to the high-affinity Fc receptor, FC7RI (Lübeck
`et al, 1985). Murine IgG, and IgG2b MoAb mediate ADCC by human effector cells far less efficiently, if at
`all (Van de Winkel et al, 1991). In addition, human complement is not as capable of mediating cytotoxicity
`with murine MoAb as guinea pig or rabbit complement. Thus, only MoAbs of the IgG2a and IgG3 subclasses
`have shown activity during in vivo therapy, presumably through ADCC mediated by FcyRI-bearing cells
`(Steplewski et al, 1988).
`In this paper, we describe an approach to overcoming some of the limitations of certain MoAbs for in vivo
`therapy. We have prepared murine MoAbs (MoAb 22 and MoAb 32) that bind to FC7RI outside the ligand
`binding site, and thus their ability to bind to and trigger cytotoxicity through this receptor is not competed by
`other immunoglobulins. Bispecific antibodies (BsAbs) were previously shown to mediate ADCC of red blood
`cells that was not inhibited by nonimmune human IgG (Shen et al, 1986a). We now report use of BsAbs
`linkage using Fab fragments of anti-Fc7RI MoAb 32 or 22 and a whole IgM
`prepared by chemical
`anti-myeloid cell MoAb, PM81, reactive with the CD 15 antigen, for studies of ADCC in vitro and therapy.
`This BsAb was able to mediate ADCC of HL-60 leukemia cells. Moreover, when testing for the ability of
`human plasma to block this effect, we found an enhancement of killing mediated at least partly by human
`complement. Thus, this BsAb lysed tumor cells by two different mechanisms, complement and cellular-
`mediated lysis. In a Phase I clinical trial, 4 patients with CD 15 + tumors were treated with up to 48 mg of this
`BsAb with no toxicity, and in one of these patients who had acute myelogenous leukemia and could be
`evaluated, there was a transient 30-60% reduction in circulating leukemic blast cells during each of six
`infusions over a 2-week period. Thus, this BsAb may be useful for treatment of minimal residual disease in
`patients with CD15-expressing high-risk tumors.
`
`86
`
`2 of 10
`
`BI Exhibit 1013
`
`

`

`THERAPEUTIC BISPECIFIC ANTIBODY TO Fc-yRI AND CD 15
`
`MATERIALS AND METHODS
`
`Cells
`
`HL-60 and U937 cells (American Type Culture Collection (ATCC), Rockville, MD) were cultured in
`RPMI-1640 (GIBCO, Grand Island, NY) containing 10% fetal bovine serum (FBS) (Hyclone, Logan, UT) as
`previously described (Ball et al, 1983). SKBR-3 cells (ATCC) were cultured in DMEM + 10% FBS.
`Monocytes were purified as previously described (Shen et al, 1986b). Briefly, mononuclear cells were
`harvested from the peripheral blood of normal donors by leukapheresis. The monocytes were further purified
`by separation over Histopaque (Sigma Chemical Co., St. Louis, MO), and incubation with rotation at 4°C
`(Shen et al, 1986b). The resulting aggregates were allowed to sediment and then sedimented again through
`FBS. The resulting preparations were >95% monocytes as assessed by morphological examination of
`Wright's-Giemsa-stained cytospins and expression of CD14.
`The hybridomas PM81 (Bell etal, 1983), 32 (Anderson et al, 1986), and 22 (Guyre era/, 1989) were tested
`for mycoplasma, sterility, and murine viruses by the MAP test and found to be free of contamination. The
`hybridomas were cultured in MF-1 medium (modified Excell-300, J.R. Scientific) containing 0.5% FBS.
`
`Production of MoAbs
`Antibodies were produced using hollow-fiber technology, with a serum-free,
`low-protein media feed
`stream. Each MoAb was purified from harvests of antibody-rich supernatant from a hollow-fiber cartridge
`using high-performance liquid chromatography (HPLC). The MoAb was purified using a semipreparative
`anion exchange (DEAE) column with a combined pH and salt gradient. The gradient was optimized for each
`MoAb to achieve a product of greater than 90% purity, with extremely low endotoxin (0.48-0.96 EU/ml or
`0.42-0.84 EU/mg antibody) and DNA contamination levels (<5 pg/mg).
`
`Flow cytometry
`Briefly, cells (106) were incubated for 1 hour with varying concentrations of MoAb or BsAb (in the
`presence of normal human IgG ( 10^5 moles/liter), Sigma), washed with phosphate-buffered saline containing
`0.1% bovine serum albumin and 0.05% sodium azide (PBA), and further incubated with fluorescein
`isothiocyanate (FITC)-coupled, affinity-purified F(ab')2 goat anti-mouse (GAM) Ig (Caltag, Inc., S. San
`Francisco, CA) for 1 hour at 4°C. Cells were then washed once in PBA and resuspended in PBA containing 1 %
`paraformaldehyde (Sigma). The cells were analyzed on the Ortho Systems 50H flow cytometer (Ortho
`Diagnostics, Westwood, MA) interfaced to a 2150 computer using a logarithmic amplification scale.
`
`Bispecific antibody preparation and purification
`BsAbs composed of PM81 conjugated to 32 Fab (or 22 Fab) were prepared by cross-linking the sulfhydryl
`of the 32 Fab fragment to sulfhydryl groups added to the PM81 MoAb. Sulfhydryl groups were added to PM81
`using A'-succinimidyl-acetyl-S-thioacetate (SATA). 32 was digested with pepsin to produce the F(ab')2
`fragment, which was then purified by high-performance size-exclusion chromatography. Reduction with
`mercaptoethylamine and reaction with 5,5'-dithio-bis(2-nitrobenzoic acid) converted the F(ab')2 fragment to
`a Fab fragment with endogenous sulfhydryl groups activated with thionitrobenzoic acid (Fab-TNB). The
`Fab-TNB was purified by gel filtration and assayed to make sure that there were enough TNB groups attached
`to the Fab to make a good coupling partner. An excess of 32 Fab-TNB was added to the PM81 SATA to drive
`the reaction to complete conversion to bispecific antibody. The bispecific antibody was then purified using
`high-performance size-exclusion chromatography. Endotoxin levels, as determined by the Limulus ameboe-
`cyte lysate assay, were =£1.74 EU/mg.
`
`87
`
`3 of 10
`
`BI Exhibit 1013
`
`

`

`BALL ET AL.
`
`Cytotoxicity assays
`HL-60 cells expressing CD 15 on their surface were labeled for 1 hour at 37°C with 200 mCi of5 ' Cr sodium
`chromate in normal saline (New England Nuclear, Boston, MA) and used as target cells. Effector cells were
`normal monocytes isolated from a leukopack as described and cultured overnight with 50 units/ml of IFN-7.
`To quantify cytotoxicity of HL-60 cells, equal volumes of test reagents (control antibodies and conjugates),
`51Cr-labeled HL-60 cells, and effector cells were mixed in round-bottomed microtiter wells. Plates were
`incubated for 6 hours at 37°C, after which half of the supernatant was removed and counted for release of 51Cr.
`Maximal lysis was obtained by addition of 2% sodium dodecyl sulfate in water. Percent cytotoxicity was
`— spontaneous lysis) + (maximal
`lysis — spontaneous
`calculated as lOOx (counts released with effectors
`lysis). In all experiments, tests were conducted in triplicate and the results are expressed as the mean ± SD.
`
`MTT cell binding assay
`An assay to determine bispecificity was developed that relies on the binding of a BsAb to CD 15 on a target
`cell (SKBR-3 cells, American Type Culture Collection, Rockville, MD) and subsequently to FC7RI on model
`effector cells (U-937 cells, American Type Culture Collection, Rockville, MD)
`labeled with MTT
`(3-[4,5-dimethylthiazol-2-yl]2,5-diphenyltetrazolium bromide) (Green et al, 1984). This binding induces
`stable association between effector and target cells. SKBR-3 cells were plated at 4 x 104 cells per well into
`96-well flat-bottomed plates (Costar) in DMEM + 50 u.g/ml gentamicin + 10% FBS, and cultured for 24-48
`hours. Culture media was then removed by rapid inversion, and antibodies (PM81 X 32, PM81, 32,
`PM81 + 32 or P3, a nonspecific murine IgG[ MoAb) were added in a volume of 50 p.1 at the concentrations
`shown. The plate was rotated at 4°C for 30 minutes, and then each well was washed twice with 200 u.1 of
`PBS-BSA at 4°C. After washing, 4 X 105 MTT-labeled U-937 cells were added and the plate rotated for an
`additional 30 minutes at 4°C. MTT labeling of U-937 cells was accomplished by adjusting cells to 2 x 106
`cells/ml in RPMI-1640 + 10% FBS and incubating at 37°C, 5% C02 for 1 hour with 0.5 mg/ml MTT. After
`incubation of MTT-labeled U-937 cells with the SKBR-3 monolayer for 30 minutes, each well was washed
`five times very gently with 200 jjlI of PBS-BSA. The plate was forcefully inverted between washes, without
`centrifugation, to remove nonadherent cells. After the last wash, 100 (¿1 of 95% isopropanol + 0.04 N HC1
`was added to each well, the cells were thoroughly mixed by pipetting to dissolve the MTT reaction product,
`and the plate was read in an ELISA reader (Dynatech, MR650). Results are reported as O.D. units, and
`represent absorbance at 570 nm.
`
`Colony-forming assay
`Peripheral blood mononuclear cells from a normal donor were cultured for 18 hours in IFN-7 ( 100 units/ml)
`prior to addition of HL-60 cells (to 1% final concentration of HL-60 cells). This mixture of cells was then
`cultured for another 18 hours under a variety of conditions (with or without autologous plasma as a source of
`complement; with or without bispecific or monospecific MoAb, including the negative control MoAb, Thy-1 )
`and then seeded into methylcellulose cultures with HL-60 conditioned media (as a source ofgrowth factors for
`HL-60 cells) (Howell & Ball, 1985).
`
`In vivo administration of bispecific antibody
`With the approval of the DHMC Committee for the Protection of Human Subjects, and after informed
`consent, 4 patients with CD15+ tumors were treated in a Phase I study of BsAb infusions. The first 2 patients
`were treated with 0.065 mg/kg per dose and the second 2 patients with 0.125 mg/kg dose. The antibody dose
`was diluted in 500 ml of normal saline. Each patient was treated six times (three times per week over 2 weeks)
`with 6-hour infusions of antibody at a single dose level per patient.
`
`88
`
`4 of 10
`
`BI Exhibit 1013
`
`

`

`THERAPEUTIC BISPECIFIC ANTIBODY TO Fc-yRI AND CD 15
`
`-A— 32 F(ab')2
`PM81
`PM81x32Fab
`
`30000 -
`
`25000 -
`
`20000 -
`
`15000 -
`
`10000 -
`
`5000 -
`
`01U
`
`
`oB3OM 3U0
`
`>
`"o
`
`s <
`
`0<
`.001
`
`r i ifii|
`100
`mAb or BsAb Concentration ((ig/ml)
`FIG. 1. Bindingof BsAb to normal human monocytes. The binding of BsAb PM81 x 32 Fab, orthe component MoAb,
`to freshly isolated human leukocytes was analyzed by flow cytometry (gated on light scatter to include only the monocytes)
`as described in Materials and Methods. Values are the mean for duplicate measurements of the number of second Ab
`molecules bound per cell.
`
`i
`
` r iiimq
`1000
`
`RESULTS
`
`Characteristics of BsAb
`The ability of the BsAb to bind to antigen-positive target cells was assessed by flow cytometry. As shown
`in Fig. 1, the BsAb recognized approximately the same number ofantigenic sites on normal monocytes as the
`the avidity of binding may have been slightly reduced by the chemical
`constituent MoAb. However,
`conjugation. The MTT cell-binding assay was used to demonstrate that the antibody was functioning as a true
`BsAb. This assay indicated that maximal attachment of the CD64+ U937 cells to the CD15+ SKBR-3 cells
`occurred at 5 p,g/ml of BsAb and that both the PM81 x 32andthePM81 x 22 BsAbs were similarly effective
`in mediating conjugate formation between these cells (Fig. 2).
`
`BsAbPM81X32
`BsAbPM81X22
`
`80 -i
`
`60
`
`4<H
`
`20 H
`
`QZ Os
`
`a V
`
`iJ
`-J
`
`W «oHOwb
`
`.
`i*,
`w
`t£
`
`0
`
`5
`25
`10
`15
`20
`BsAb Concentration (|ig/mL)
`FIG. 2. Demonstration of the ability of the BsAb to mediate attachment of U937 cells (Fc7RI+) to SKBR-3 cells
`(CD15+). MTT-labeled U937 cells were added to monolayers of SKBR-3 cells in the presence or absence of two different
`BsAb. Cell attachment was measured by spectrophotometry as described in Materials and Methods. Values are
`mean ± SD for triplicate measures of bound U937 cells. The broken line indicates the maximum binding in the absence
`of antibody or in the presence of the uncoupled component MoAb.
`
`89
`
`5 of 10
`
`BI Exhibit 1013
`
`

`

`BALL ET AL.
`
`BsAbPM81x32
`-A-
`PM81
`
`-
`
`— BsAb PM81x22
`PM81x32 + PM81x22
`
`.05
`.5
`5
`.1
`1
`mAb or BsAb Concentration (¡ig/ml)
`FIG. 3. ADCC of HL-60 cells by BsAb. 5lCr-labelled HL-60 cells were cultured for 6 hours at 37°C in the presence of
`MoAb PM81, BsAb PM81 x 32, BsAb PM81 x 22, or a combination of the two BsAb. Specific cytotoxicity was
`calculated as described in Materials and Methods.
`
`ADCC mediated by BsAb
`Both the PM81 x 32 Fab BsAb, and a similar BsAb (PM81 x 22 Fab) which targets to a separate epitope
`on Fc-yRI, were capable of mediating ADCC of HL-60 cells (Fig. 3). Using the chromium release assay and
`correcting for background lysis, 30-35% killing was demonstrated using 0.5-1.0 |xg/ml of BsAb, with lesser
`killing at both higher and lower concentrations. Monospecific PM81 had little activity in this assay in the
`absence of serum, and combining two bispecifics made from the same tumor-specific MoAb did not
`significantly increase cytotoxicity. Thus, these BsAbs mediate ADCC of HL-60 target cells whereas PM81
`alone does not. However, in the presence of fresh human serum, collected with precautions to keep the serum
`chilled, both the BsAb (Fig. 4) and PM81 alone (not shown) lysed up to 80% of chromium-labeled HL-60
`target cells. When monocytes were labeled with chromium and subjected to incubation with BsAb plus
`
`PM81
`PM81 x32Fab
`PM81
`PM81 x32
`
`100
`
`xoo O u
`
`IX
`
`.01
`
`1 0
`
`mAb or BsAb Concentration (|j.g/ml)
`FIG. 4. Augmented ability of BsAb to mediate ADCC in the presence of human serum. The PM81 x 32Fab BsAb and
`PM81 alone were tested by the 5lCr-release assay for cytotoxicity of HL-60 cells in the absence (closed symbols) or
`presence (open symbols) of fresh human serum as a source of complement.
`
`90
`
`6 of 10
`
`BI Exhibit 1013
`
`

`

`THERAPEUTIC BISPECIFIC ANTIBODY TO Fc-yRI AND CD 15
`
`unlabeled monocytes as effectors, no chromium release over background levels was observed (data not
`shown), indicating that the BsAb does not cause lysis of the effector cells.
`Mixing experiments
`As seen in Table 1, when evaluated in a colony formation assay, both unconjugated PM81 and PM81 x 32
`Fab eliminated 92% of HL-60 cells that had been mixed with normal bone marrow cells plus autologous
`plasma as a source ofcomplement. BsAbPM81 x 32 Fab eliminated 85% of HL-60 colonies via ADCC and
`in the absence of autologous plasma.
`In vivo therapy
`Four patients were treated with BsAb infusions. Their diagnoses were acute myelogenous leukemia
`lung cancer (SCCL), breast cancer, and pancreatic islet cell carcinoma. All were in
`(AML), small cell
`advanced stages of disease and had chemotherapy-resistant disease at the times of treatment. No acute
`toxicities were observed other than a transient period (several minutes) of substernal discomfort in 1 patient.
`The infusion was interrupted until the symptoms resolved and then completed without further incident.
`During the infusion in the patient with AML, the circulating blast cells decreased by about 50% for several
`hours after each infusion, but returned to baseline or greater levels by the next morning. The reduction in blast
`cells was somewhat surprising since less than 2% of the available binding sites were occupied by BsAb in vivo
`(Fig. 5). Peak serum levels of free BsAb were achieved at the end of the infusion period in all patients, and
`ranged from 0.08 to 0.27 p,g/ml.
`In the patients with the solid tumors, it was not possible to monitor the effects of BsAb on the tumor
`population. However, the function of the antibody was indicated by the fact that the circulating neutrophils
`decreased dramatically after each infusion. Despite this phenomenon, there were no associated signs or
`symptoms of acute reaction or of infection.
`
`DISCUSSION
`
`Redirected cellular cytotoxicity mediated by BsAb is a new form of immunotherapy with potential
`advantages over unmodified antibody therapy and other immunoconjugates such as immunotoxins. These
`advantages include a probable lack of toxicity, since no inherently toxic compounds would be introduced into
`in vivo cytotoxic mechanisms, and the ability to recruit very large numbers of
`the host, the use of normal
`cytotoxic cells to the target.
`We are particularly interested in the role of mononuclear phagocytes in mediating cytotoxicity of tumor
`cells through FC7RI (CD64) (Anderson etal, 1986; Guyre et al, 1989). Using bispecific antibodies comprised
`
`Table 1. Effect of MoAb PM81 and BsAb in the Presence or Absence
`of Human Complement on the Killing of HL-60
`Colony-Forming Cells"
`
`Without human
`complement"
`
`With human
`complement
`MoAb or BsAb
`Thy-1
`540
`PM81
`405
`40
`PM81 x 32
`60
`45
`"HL-60 cells were incubated with IFN-7-activated monocytes and MoAb
`PM81, BsAb, or a control IgM MoAb (Thy-1), in the presence or absence of
`human serum as a source of complement. This mixture was then seeded into
`methylcellulose cultures as described in Materials and Methods.
`"Colonies/200,000 cells plated.
`
`•
`
`91
`
`7 of 10
`
`BI Exhibit 1013
`
`

`

`BALL ET AL.
`
`Ab Bound in vitro
`
`Ab Bound in vivo
`
`58247
`
`116
`
`731
`
`827
`
`96
`
`80000-f
`
`60000 H
`
`40000 H
`
`20000
`
`o.
`S»
`W3
`
`A<
`
`6
`2
`4
`.2
`2
`20
`.02
`PM81x32 Fab (u.g/ml)
`Treatment Time (hours)
`FIG. 5. Binding of BsAb to AML patient blast cells in vitro and in vivo. Peripheral leukocytes (>96% blast cells) were
`isolated from patient 1 and examined by indirect immunofluorescence plus flow cytometry to determine their maximum
`binding capacity for BsAb (left panel). Cells were also isolated, kept at 0°C, and rapidly assayed by the same method for
`the amount of BsAb that had bound in vivo (right panel).
`
`8
`
`0
`
`of anti-Fc7RI MoAb (32 and 22) and antitumor-associated antigens, it may be possible to lyse tumor cells in
`vivo specifically with little toxicity. Moreover, the anti-Fc7RI mAb 32 and 22 bind to the FC7RI outside the
`domain occupied by the Fc portion of human IgG, so that the presence of physiological levels of human IgG
`does not block binding of the these MoAbs nor of BsAb made from these MoAbs to this cytotoxic trigger
`molecule (Guyre et al, 1989).
`We have prepared bispecific antibodies comprised of whole IgM against the CD 15 antigen and Fab
`fragments of 32 and 22. These BsAb are capable of mediating cellular cytotoxicity by IFN-7-activated
`monocytes against CD15+ target cells. In addition, the conjugation of Fab to the IgM anti-CD 15 did not
`abrogate its ability to mediate human complement-dependent cytotoxicity. Thus, in the presence of normal
`human serum, or plasma containing high concentrations of IgG, the cxytotoxicity of this BsAb is actually
`augmented. Moreover, the dose-response curves of the two mechanisms demonstrate that the two activities
`are optimal at different concentrations. Complement-mediated killing was more efficient in the range of 10
`u-g/ml BsAb or more, whereas ADCC was optimal at about 1 p-g/ml. Since intratumoral concentrations of
`antibody are likely to be lower than plasma concentrations, the ADCC activity curve suggests that ADCC may
`be the predominant mechanism of killing in tumors.
`We envisage two therapeutic applications of this BsAb. One is in vivo therapy of patients with CD15 +
`tumors such as AML (Ball et al, 1983), SCCL (Huang et al, 1983), colorectal cancer (Brockhaus et al, 1982),
`and breast carcinoma (Vredenburgh et al, 1991). It is likely that the BsAb will be most useful to treat minimal
`disease, such as in patients with high-risk breast cancer following primary surgical therapy or patients with
`AML after remission is induced by chemotherapy. Another application is as an adjunct to chemotherapy in
`vivo, either for remission-induction or as part of a preparative regimen used for bone marrow transplantation
`(Ball etal, 1990).
`There are theoretical drawbacks to the particular combination of antibodies we have chosen because the
`tumor-associated antigen is expressed on both the target and effector cells. Thus, the possibility exists that
`monocytes can kill monocytes by engaging the CD15 antigen on one cell and FC7RI on an adjacent monocyte.
`Our experiments with 51Cr-labeled monocytes show that this does not appear to occur. Another consequence
`of the use of the PM81 x 32 F(ab')2 construct is that normal neutrophils and monocytes that express CD15
`may interfere with binding of the bispecific antibody to tumor target cells in vivo. Binding of the antibody to
`neutrophils may also produce neutropenia and/or the release of inflammatory mediators leading to toxicity.
`However, the preliminary data from a Phase I clinical trial of this antibody has not been associated with
`toxicity, though transient neutropenia has been observed. Nevertheless, it would seem most efficacious to use
`
`92
`
`8 of 10
`
`BI Exhibit 1013
`
`

`

`THERAPEUTIC BISPECIFIC ANTIBODY TO Fc^RI AND CD 15
`
`this antibody for minimal residual disease, such as after bone marrow transplant
`for AML, or after
`myelosuppressive chemotherapy, at a time when there are few circulating neutrophils to interfere with
`binding. The in vitro and in vivo studies reported here indicate that such an approach will mediate killing of
`significant numbers of tumor cells, will have little if any toxicity, and, as an adjunct to other therapies, could
`increase the proportion of patients with tumor or leukemia who achieve long-term disease-free survival. From
`the standpoint of clinical research, one complication of studies using the BsAb is that dissociating the effects
`of the monospecific PM81 (which mediates complement-dependent killing) from those of the BsAb
`PM81 x 32 (which mediates both complement dependent killing and ADCC), may require a comparison of
`therapy using PM81 alone versus therapy with the BsAb.
`Although a very recent development, BsAbs have been used in several other Phase I clinical trials with
`In one trial, 12 patients with lung, ovarian, or breast carcinoma were treated with
`encouraging results.
`intraperitoneal or intrapleural infusions of activated T lymphocytes targeted with an anti-CD3 x anti-tumor
`(MOC31 ) BsAb (deLeij et al, 1991 ). These targeted cells induced considerable local lysis of tumor cells and
`a mild inflammatory reaction, but no toxic side effects or anti-mouse antibody responses.
`In a very
`preliminary trial of an anti-CD3 x anti-CD19 BsAb in a patient with a B-cell malignancy, significant
`reduction in peripheral tumor cell counts was also achieved (Clark et al, 1991). In the most promising and
`extensive series of studies to date, IL-2-activated lymphocytes and an anti-CD3 x anti-glioma BsAb were
`injected intracranially in 20 patients with malignant glioma (Nitta et al, 1990). After 2 years, 76% of patients
`treated with BsAb and lymphocytes were tumor free, as compared with 33% of patients given IL-2-activated
`lymphocytes and no BsAb.
`In summary, our results and those cited above indicate that BsAb may be most efficacious for high-risk
`tumors after induction of remission, after bone marrow transplant, and for treatment of minimal residual
`disease.
`
`REFERENCES
`
`Anderson, C.Guyre, P., Whitin, J., Ryan, D.H., Looney, R.J., and Fanger, M.W. (1986): Monoclonal antibodies to Fe
`receptors for IgG on human mononuclear phagocytes. J. Biol. Chem. 261: 12856.
`Ball, E., Graziano, R., and Fänger, M. (1983): A unique antigen expressed on myeloid cells and acute leukemia blast cells
`defined by a monoclonal antibody. J. Immunol. 130: 2937.
`Ball, E.D., Mills, L.E., Cornwell, G.G., Davis, B.H., Coughlin, CT., Howell, A.L., Stukel, T.A., Dain, B.J.,
`McMillan, R., Spruce, W., Miller, W.E., and Thompson, L. (1990): Autologous bone marrow transplantation for
`acute myeloid leukemia using monoclonal antibody-purged bone marrow. Blood 75: 1199.
`Brockhaus, M., Magnani, J., and Herlyn, M. (1982): Monoclonal antibodies directed against the sugar sequence of
`lacto-N-fucopentaose III are obtained from mice immunized with human tumors. Arch. Biochem. Biophys. 217: 647.
`Clark, M., Bolt, S., Tunnacliffe, A., and Waldman, H. Use of bispecific monoclonal antibodies to treat hematological
`malignancies: a model system using CD3 transgenic mice. In Bispecific Antibodies and Targeted Cellular Cytotoxicity,
`eds. Romet-Lemonne, J.L., Fanger, M.W., and Segal, D.M.: 1991, pp243-247.
`deLeij, L., de Jonge, M., Ter Haar, A., Spakman, H., The, H., de Vries, L., Mulder, N., Berendsen, H., Elias, M., and
`Smit-Sibinga, C. Intrapleural and intraperitoneal application of bispecific antibody retargeted lymphocytes to cancer
`patients. In Bispecific Antibodies and Targeted Cellular Cytotoxicity, eds. Romet-Lemonne, J.L., Fänger, M. W., and
`Segal, D.M.: 1991, pp249-253.
`Dillman, R. (1989): Monoclonal antibodies for treating cancer. Ann. Int. Med. Ill: 592.
`Green, L., Reade, J., and Ware, C. (1984): Rapid colorimetric assay for cell viability: application to the quantitation of
`cytotoxic and growth inhibitory lymphokines. J. Immunol. Meth. 70: 257.
`Guyre, P., Graziano, R., Vance, B., Morganelli, P., and Fänger, M. (1989): Monoclonal antibodies that bind to distinct
`epitopes on FcyRI are able to trigger receptor function. J. Immunol. 143: 1650.
`Howell, A. and Ball, E. (1985): Monoclonal antibody mediated cytotoxicity of human myeloid leukemia cells: an in vitro
`model for estimating efficiency and optimal conditions for cytolysis. Blood 66: 649.
`
`93
`
`9 of 10
`
`BI Exhibit 1013
`
`

`

`BALL ET AL.
`
`Huang, L., Brockhaus, M., Magnani, J., Cuttitta, F., Rosen, S., Minna, J., and Ginsburg, V. (1983): Many monoclonal
`antibodies with an apparent specificity for certain lung cancers are directed against a sequence found in lacto-N-
`fucopentaose III. Arch. Biochem. Biophys. 220: 318.
`Lübeck, M., Steplewski, Z., Baglia, F., Klein, M., Dorrington, K., andKoprowski, H. (1985): The interaction of murine
`IgG subclass proteins with human monocyte Fc receptors. J. Immunol. 135: 1299.
`Nitta, T., Sato, K., Yagita, H., Okumura, K., and Ishii, I. (1990): Preliminary trial of specific targeting therapy against
`malignant glioma. The Lancet 335: 368.
`Ortaldo, J., Woodhouse, C, Morgan, A., Herberman, R., Cheresh, D., and Reisfeld, R. (1987): Analysis of effector
`cells in human antibody-dependent cellular cytotoxicity with murine monoclonal antibodies. J. Immunol. 138: 3566.
`Shen, L., Guyre, P.M., Anderson, C.L., and Fanger, M.W. (1986a): Heteroantibody-mediatedcytotoxicity: antibody to
`the high affinity Fc receptor for IgG mediates cytotoxicity for human monocytes that is enhanced by interferon-g and is
`not blocked by human IgG. J. Immunol. 137: 3378.
`Shen, L., Guyre, P., Ball, E., and Fänger, M. (1986b): Glucocorticoid enhances gamma interferon effects on human
`monocyte antigen expression and ADCC. Clin. Exp. Immunol. 65: 387.
`Stepan, D., Bartholomev/, R., and LeBien, T. (1984): In vitro cytodestruction of human leukemia cells using murine
`monoclonal antibodies and human complement. Blood 63: 1120.
`Steplewski, Z., Sun, L., Shearman, C, Ghrayeb, J., Daddona, P., and Koprowski, H. (1988): Biological activity of
`human-mouse IgGl, IgG2, IgG3, and IgG4 chimeric monoclonal antibodies with antitumor specificity. Proc. Nati.
`Acad. Sei. USA. 85: 4852.
`Van de Winkel, J.G.J. and Anderson, C.L. (1991): Biology of human immunoglobulin Fc receptors. J. Leukocyte Biol.
`49:511.
`Vredenburgh, J., Simpson, W., Memoli, V., and Ball, E. (1991): Reactivity of anti-CD15 monoclonal antibody PM81
`with breast cancer and elimination of breast cancer cells from human bone marrow by PM81 and immunomagnetic
`beads. Cane. Res. 51: 2451.
`
`Address reprint requests to:
`Edward D. Ball, M.D.
`Division ofHematologylBone Marrow Transplantation
`Montefiore University Hospital
`University of Pittsburgh Medical Center
`3459 Fifth Ave.
`Pittsburgh, PA 15213
`
`94
`
`10 of 10
`
`BI Exhibit 1013
`
`

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