throbber
11Q)Am:S,~"1ff,Q ~'lf'lllliES~ ~ ll!ES !E~~ S BMID,~lU IEt:j
`UNITED STATES DEPARTMENT OF COMMERCE
`United States Patent and Trademark Office
`
`March 18, 2022
`
`TIDS IS TO CERTIFY THAT A~"NEXED HERETO IS A TRUE COPY FROM
`THE RECORDS OF THIS OFFICE OF:
`
`PATENT NUMBER: ./.,84./.,893
`ISSUE DATE: July .J, 1989
`
`By Authority of the
`Under Secretary of Commerce for Intellectual Property
`and Director of the Cnitecl States Patent and Trademark Office
`
`Certifying Officer
`
`Miltenyi Ex. 1005 Page 1
`
`

`

`United States Patent [19J
`H onsik et al.
`
`[111 Patent Number:
`[45) Date of P atent:
`
`4,844,893
`Jul. 4, 1989
`
`[54) EX VIVO EFFECTOR CELL ACTIVATION
`FOR 'f AR GET CELL .KILLING
`
`{75]
`
`[nventors: Cyril J. Honsik; Ralph A. Reisfeld,
`both of La Jolla, Calif.
`
`[73] Assignee: Scripps Clinic and Research
`Foundation, La Jolla, Calif.
`
`[211 Appl. No.: 916,173
`[22} Filed:
`Oct. 7, 1986
`Int. Cl.4 •••••.••••••••••••.•••••.••••••.••• ••••••••• A61K 39/395
`[51]
`[52] U.S. Cl. ..................................... 424/85.8; 424/93;
`435/240.2; 530/387; 530/828; 530/388;
`530/808
`[58) F ie.Id of Search ..................... 435/2, 4, 7, 29, 948;
`935/107, 108; 424/85, 93; 436/512, 519
`
`[56]
`
`Referenu s Cited
`U.S. PA TENT DOCUMENTS
`4,444,878 4/1984 Paulus .
`4,507,391 3/ 1985 Pukel er al ..
`4,675,287 6/1987 Reisfeld ................................. 435/ 68
`
`OTHER PUBLICATIONS
`Honsik, C. J. et al., Proc. Natl. Acad, Sci., U.S.A.,
`83:7893- 7897, (10- 1986).
`Cheresh, D. A. et al., Proc. Natl. Acad. Sci., U.S.A.,
`82:5155-5159, (8-1985).
`Park, M. M. et al., Cellular Immunology, 84:94-103,
`(1984).
`Mule, J. J. et al., Science, 225:1487-1489, (9-1984).
`
`Rosenberg, S. et al., New Engl. J. Medicin.e,
`313(23):1485-1492, (12-1985).
`Perez, P. et al., J. Exper. Med., 163:166-178, ( l-1986).
`Jung, G. er al., Proc. Natl. Acad. Sci., U.S.A.,
`83:4479-4483, (6-1986).
`Staerz, U. D. et al., Nature, 314:628-631, (4-1985).
`Cancer Research, (1984), 44:806-809, Dippold et al.
`Natural Immunity and Biological Response, (1985),
`4:253, Honsik et al.
`Eur. J. Immunol., (1986), 16:263-270, Staerz et al.
`Proc. Nat'l Acad. Sci. U.S.A., (1986), 83:1453-1457,
`Staerz et al.
`Proc. Nat'l Acad. Sci. U.S.A., (1985), 82:1242- 1246,
`Houghton et at.
`Primary Examiner- Margaret Moskowitz
`Assistant Examiner-Jeff. P. Kushan
`Attorney, Agent, or Firm- Dressler, Goldsmith, Shore,
`Sutker & Milnamow, Ltd.
`ABSTRACT
`[57]
`A method and composition for killing target cells is
`disclosed. The method utilizes ex vivo IL-2 activation
`of leucocyte effector cells and arming the activated
`leucocyte effectors with monoclonal antibodies whose
`Fe portions bind to the IL-2-activated effectors and
`whose paratopic portions immunoreact with an epitope
`expressed on the surfaces of the target cells. The com(cid:173)
`position contains a cytolytic amount of the armed, IL-2-
`activated effector cells clispersed in an aqueous physio(cid:173)
`logically tolerable diluent medium.
`
`20 Claims, 4 Drawing Sheets
`
`Miltenyi Ex. 1005 Page 2
`
`

`

`U.S. Patent
`
`Jul. 4, 1989
`
`Sheet 1 of 4
`
`4,844,893
`
`TARGET
`
`EFFECTOR
`
`COVA LENT BINDING
`
`FIG. I A
`
`T-Fc RECEPTOR BINDING
`
`FIG. I 8
`
`0
`
`'
`
`I/
`
`\\=v
`(C)'\
`---
`
`_ LYSIS_,,
`/
`
`E-Fc RECEPTOR BINDING
`
`FIG. ID
`
`--..: ___ __
`
`/
`'-..._ LYSIS _,,..,,
`
`Miltenyi Ex. 1005 Page 3
`
`

`

`U.S. Patent
`
`Jul. 4, 1989
`
`Sheet 2 of 4
`
`4,844,893
`
`FIG. 2
`
`u
`~
`u
`a.,
`C2. 40
`"' ~ 0
`
`20
`
`0 - - - - - - - - . - - - - - - - - - - -
`lDO
`25
`50
`Effector to Target Ratio
`
`Miltenyi Ex. 1005 Page 4
`
`

`

`U.S. Patent
`
`Jul. 4, 1989
`
`Sheet 3 of 4
`
`4,844,893
`
`FIG. 3
`
`100
`
`BO
`
`60
`
`V,
`
`·;:;;
`>-
`_,J
`
`L.l -u
`
`(U
`0..
`
`en 40
`~
`
`20
`
`D
`
`Normal #1
`
`Normal #2
`
`Patient #1
`
`Patient #2
`
`Patient #3
`
`Patient#4
`
`Miltenyi Ex. 1005 Page 5
`
`

`

`U.S. Patent
`
`Jul. 4, 1989
`
`Sheet 4 of 4
`
`4,844,893
`
`FIG. 4
`
`rll·2 AUGMENTATION OF AOC C
`
`100
`
`90
`
`80
`
`70
`
`·:;; 60
`.....
`>(cid:173)
`~ - 50
`u ..,
`~ 40
`
`C.
`
`30
`
`20
`
`10
`
`100:l
`
`50:1
`Effector: Target
`
`25: 1
`
`Miltenyi Ex. 1005 Page 6
`
`

`

`1
`
`4,844,893
`
`EX VIVO EFFECTOR CELL ACITV ATION FOR
`TARGET CELL KILLING
`
`The present invention was made with support of the 5
`Government of the United States, and the Government
`of the United States has certain rights in the invention.
`
`DESCRIPTION
`TECHNICAL FIELD
`The present invention relates to a cytotoxicity
`method, and particularly to a method for killing target
`cells such as tumor cells with antibody-directed, ex
`vivo-activated effector leucocytes.
`
`BACKGROUND OF THE INVENTION
`Neuroectodermal tumors are highly malignant, and
`include neuroblastomas, small cell cancinoma of the
`lung, gliomas, neuroblastomas and melanomas. Of the
`neuroectodermal tumors, neuroblastomas occur during 20
`infancy and early childhood. Except for Wilms' tumor,
`they are the most common retroperitoneal tumors in
`children. Neuroblastomas arise most commonly in the
`adrenal medulla, but they may also develop in other
`sympathetic ganglia within the thorax or abdomen. 25
`These tumors metastasize early with widespread in(cid:173)
`volvement of lymph nodes, liver, bone, lung and mar(cid:173)
`row. The prognosis is often good when the tumor is
`diagnosed prior to obvious metastasis, but with metasta(cid:173)
`sis, prognosis is poor despite the extensive use of radical 30
`surgery, deep X-ray therapy, and chemotherapeutic
`agents.
`Several antigenic determinants have recently been
`detected on neuroblastoma cells with monoclonal anti(cid:173)
`bodies (Mabs). See Seeger, Ann. Intern. Med., 91, 873 35
`(1982); Wikstrand et al., Cancer Res., 42, 267 (1982);
`Wikstrand et al., J. N euroimmunlogy, 3, 43 (I 982); Ei(cid:173)
`senbarth et al., Proc. Natl. Acad. Sci. USA, 76, 4913
`(1979); Liao et al.1 Eur. J. Immunol., 11, 450 (1981);
`Seeger et al., Cancer Res., 4, 2714 (1981); Kennett et al., 40
`Advances in N euroblastama Research, p . 209, Raven
`Press, N.Y. (Evans ed.) (1980); Seeger et al., J.
`Immwzol., 128, 983 (1982); Kemshead et a)., Pediatr.
`Res., 15, 1282 (1981). Gangliosides GD3 and GD2 are
`among the antigenic determinants detected on neuro- 45
`blastoma cells.
`Gangliosides (sialic acid-bearing glycolipids) are thus
`rapidly being characterized as tumor markers that are
`relevant
`target antigens for monoclonal antibody
`(Mab)-roediated immunotberapy [Dippold et al. ( 1983) 50
`Cancer Res. 44;806-810; Houghten et al. (1985) Proc.
`Natl. Acad. Sci. USA 82:1242- 1246; Hellstrom et al.
`(1985) Proc. Natl. Acad. Scl, USA 82:1499-1502; Che(cid:173)
`resh et al. (1985) Proc. Natl Acad. Sci USA
`82:5155-5159; Honsik et al. (1985) Natural Immunity 55
`and Biological Response 4:253; and Steplewski et al.
`(1985) Proc. Natl. Acad. Sci. USA 82:8653- 8657).
`The disialoganglioside G D 3, is expressed preferen(cid:173)
`tially on human mel.anorna cells [Dippold et al. (1980)
`Proc. Natl. Acad. Sci, USA 77:6114-6118; Cheresh et al. 60
`(1984) Proc. Natl. Acad. Sci., USA 81:5767-5771], and is
`an effective target in vitro for both complement(cid:173)
`medfated tumor cytolysis and antibody-dependent cel(cid:173)
`lular cytoxicity (ADCC) by Mabs of the l gG3 subclass
`[Hellstrom et al. (1985) Proc. Natl. Acad. Sci. , USA 65
`82:1499-1502; Cheresh et al. (1985) Proc. Natl Acad.
`Sci .• USA 82:5155-5159; and Honsik. et al. (1985) Natu-
`ral Immunity and Biological Response 4;253]. Mono-
`
`2
`clonal antibodies of the IgG3 class directed against
`GD3 have been reported to effectively suppress the
`establishment of human melanoma tumors in the xeno-
`transplant nude mouse mode [Hellstrom et al. (] 985)
`Proc. Natl, Acad. Sci.. USA 82:1499-1502; and Cheresh
`et al. (1985) Proc. Natl, Acad, Sci., USA 82:5155-5159).
`Murine mononuclear splenocytes "armed" whh anti(cid:173)
`GD3 Mabs were recently reported to eradicate well
`established and progressively growing human mela-
`10 noma tumors in nude mice [Honsik et al. (1985) Natural
`Immunity and Biological Response 4:253). Additionally,
`Houghten et al. (1985) Proc. N atl. Acad. Sci., USA
`82:1242- 1246, using Mab R24 (IgG3) directed to GD3
`(discussed in U.S. Pat. No. 4,507,391), observed major
`15 tumor regressions in 3 of 11 melanoma patients treated
`with that antibody in a Phase I clinical trial. It was also
`reported in Dippo}d et al., Cancer Res .. 44, 806 (1984),
`that Mab R24 could kill GD3-containing human mela-
`noma cells in vitro after prolonged exposure (greater
`than 24 hours) to the antibody suggesting an additional,
`as yet undefined, mechanism of tumor cell killing.
`T aken together, these findings demonstrate that the
`potential therapeutic efficacy of anti-GD3 Mabs war(cid:173)
`rants further study of ganglioside as immunotherapeutic
`targets.
`The fact that the GD2 antigen was shown to be heav(cid:173)
`ily expressed on most excised melanoma and SCCL
`tumors, as well as on numerous tumor cell lines, Pu.lee!
`et al., J. Exp. Med., 155, 1133 (1982) and yet is virtually
`absent from most normal tissues, suggests that it might
`be a good target antigen for in vivo specific immuno-
`therapy, and tumor imaging.
`In a recent report by Kipps et al., J. Exp. Med., 161,
`1 ( 1985), using isotype switch variants of a Mab directed
`to an epitope on Class I human histocompatibility anti(cid:173)
`gens, an IgG2a isotype variant was shown to be. more
`effective in directing ADCC than the corresponding
`lgG I or an lgG2b variant. Recent work from our own
`laboratory, Schulz et al.
`( 1985) J. Exp. Med.
`161:1315- 1325, also showed specific cytolysis. That
`work, using Mab 9.2.27, an lgG2a monoclonal antibody
`that 1mmunoreacts with a chondritin sulfate proteogly(cid:173)
`can that is preferentially expressed on human melanoma
`cells, illustrated that human melanoma tumors, estab(cid:173)
`lished and progressively growing in nude mice, could
`be eradicated by simultaneous injection of that Mab
`along with a relatively large dose of mononuclear
`s plenocytes. Neither the splenocytes nor the antibodies
`alone achieved significant tumor regression.
`Park et al., Cellular Immunol., 84, 94 (1984), reported
`that a monoclonal antibody of IgG2b isotype could
`sensitize K.562 human erythroleukernia cells to ADCC-
`mediated lysis. In that case, it was reported that the
`Mab accelerated killing of the target cells by large gran(cid:173)
`ular lymphocytes known to be enriched in natural killer
`(NK) cells.
`T aken together, the results of these studies indicate
`that monoclonal antibodies may not only be useful rea(cid:173)
`gents for the immunotherapy of cancer, but also that
`different Mabs can induce tumor killing by several dif-
`ferent or even a combination of effector mechanisms.
`Some additional recent reports suggest that mouse
`monoclonal antibodies are relatively well tolerated in
`humans and pose minimal risks and few, If any, side
`effects, Oldham et al., J. C/i11. Oncol, 2, 1235 (1984). In
`using murine Mab 9.2.27, discussed before, to treat mel(cid:173)
`anoma patients, that antibody was shown by the above
`
`Miltenyi Ex. 1005 Page 7
`
`

`

`4,844,893
`
`4
`3
`pressed the Thy-1.1 alloantigen were killed by coating
`workers to localize specifically to the tumor site with
`little if any adverse side effects, but provided no appar-
`the target cells with the bispecific hybrid antibody and
`subsequent admixture of the coated target cells with
`ent clinical improvement of the disease over the period
`various dilutions of cytotoxic T lymphocytes bearing
`of time of their study in stage four patients with large
`5 the antigen recognized by the other paratopic portion of
`tutnor burdens.
`the hybrid. Potential target cells that did not bear the
`Another form of anti-tumor immunotherapy involves
`Thy-1.1 antigen were not killed. Related studies by that
`the exposure of mononuclear lymphocytes to the lym-
`group can be found in Staerz and Bevan ( 1986) Proc.
`phokine interleukin-2 (IL-2) to generate lymphokine
`Natl. Acad. Sci. USA, 83:1453-1457; Staerz. and Bevan
`activated killer (LAK) cells [Yron et al. (1980) J.
`Immunol 125:238-245; Lotze et al. ( 1981) Cancer Res. JO (1986) Eur. J. Jmmunol., 16:263-170; and Staerz and
`Bevan (1985) Eur. J. Immunol., 15:1172-1177.
`41:4420--4425; Odmm et al. (1982) J. Exp. Med.
`155:1823-1841; Grimm et al. (1983) J. Exp. Med.
`Still further, Perez et al. (1986) J. &p. Med.
`158:1356-1361].
`163:166-178 described a bispecific heteroconjugate
`For e~ample, Rosenberg ~t al. reported that the use of
`antibody that reportedly targets tIL-2-activated periph-
`~ecombmant ~1:,-2 (rl~•2),stlmulat~d human LAK ceJls 15 era! blood mononuclear cells (PBMCs) to tissue. Thus,
`m ph_ase _I cltmcal tr1~ls res~ted m mar~ed tumor _re-
`one portion of that heteroconjugate binds through its
`gress1on m several pattents with progressively growmg
`paratope [Fab fragment of Mab OKT3 (ATCC CRL
`neoplasms [Rosenberg et al: (1985) _New Engl. J. Med.
`8001)] to the activated PBMCs, while the other para-
`313:1485-1492]. In those_ clinical tnals, large numbers
`tope of the heteroconjugate (Fab fragment of a target-
`(I0'.0-1011) of human penpheral blood Jeuko~ytes were 20 binding Mab) binds to the target tissue.
`subJ~ted to doses of rIL-2 (1000 (!/1.~~ lO c~ls/ml)
`In their study, Perez et al. depleted the lymphocyte
`ex vtvo ~or up to 9~ hours befor~ being mJect~d ~!rave-
`effector cell population ofmonocytes as well as Leu-11-
`nously (i._v.) back. ~to ~e P~tt_ent~. These mdivtduals
`positive (Leu 11 +)cells.They reported that their IL-2-
`then received addition~ t.v. tnJ:ctmos of rIL-2 {up t_o
`activated effectors exhibited the TS antigen (were TS+)
`
`:oo,~ ~/kg body weight) dunng the course of theu- 25" and that elimination of T8+cells using Mab OKT8
`~~TCC CRL 8014) and complem~nt_eliminated cyiox-
`r~lthonugh some dramatic tumor regressions were
`ictty. Removal of T 4+ cells by a Similar strat~gy [Mab
`observed in several patients with extensive neoplasms,
`the treatment also produced a number of relatively O~!~ (A TCC CRL 8002) plus complement] m~re~sed
`severe clinical problems. Among those clinical prob- 30 lys1s m the presence and absence of IL-2 achvatmn.
`!ems were fluid retention, pulmonary edema and occa-
`Those ~uthor~ a~so reported t~e loss of abo~t one-~alf o(
`sional respiratory distress requiring intubation. Those
`the lyuc act1v1ty when_ ac~1vated, hybnd antibody-
`problems were a result of the direct toxicity effects of
`coated effectors were maintained free of IL-2 between 8
`rIL-2 infusions.
`and 24 hours.
`In a still more recent report, Rosenberg et al. (1986) 35 Bispecific heterobifunctional _ monoclon~l para topic
`Science 233:1318-1321, reported use of a three-part mo-
`~olecules 3?d methods of their preparation are also
`dality consisting of (a) an immunosuppressing drug,
`disclosed m U.S.
`Pat. No.
`4,444, 8_78
`and
`PC1;'/US82/01766 (W~ 83/02285), whose d1sclo~ures
`cyclophosphamide, along with (b) rIL-2-expanded tu-
`mor-infiltrating lymphocytes ('TIL) obtained from re-
`are t~corp?rated herem by reference. The .te~hmques
`sected t\llllors and (c) a relatively low dose of rIL-2 to 40 described m U.S. Pat. No. 4,350,626 for linking Pab
`successfully cure twelve mice with MC-38 colon adeno-
`portions of anti-tumor antibodies to tbe ricin A subunit
`carcinoma of advanced hepatic metastases, and to cure
`call: als<? b~ utilized for !~king F~b po_rtions of_ desired
`up to 50 percent of mice with advanced pulmonary
`antibodies m the preparation of bJSpectfic hybnds.
`metastases. The rIL-2 was reportedly administered sys-
`BRIEF SUMMARY OF THE INVENTION
`temically at 25,000 units three times per day for four 45
`days. Those tumors reportedly djd not respond to LAK
`therapy.
`Several groups have reported the use of bispecific
`heteroconjugate (hybrid) antibodies for mediating tar(cid:173)
`get cell lysis. For example, Jung et al. (1986) Proc. Narl. so
`Acad. Sci. USA 83:4479-4483 reported effective killing
`of human melanoma target cells that were coated with
`a bispecific hybrid antibody composed of one paratopic
`molecule that immunoreacted with an antigen on the
`target cell and a second paratopic molecule (OKT3, 55
`ATCC CRL 8001) that immunoreacted with human T
`cells. Human T cells previously stimulated by contact
`with OKT3 were used as the effector cells.
`Bevan, Staerz and co-workers have published several
`papers dealing with the use ofbispecific hybrid antibod. 60
`ies to kill target cells in murine systems. For example,
`Staerz et al. (1985) Nature 314:628-631 reported use of
`a hybrid antibody one of whose paratopic portions
`immunoreacted with an allotypic epitope on the T cell
`receptor of about 25 percelit of peripheral T lympho- 65
`cytes and the other of whose paratopic portions im(cid:173)
`munoreacted with the Thy-1.l alloantigen. That report
`showed that target S. AKR lymphoma cells that ex-
`
`The present invention contemplates a method for
`killing target cells and a composition useful in the
`method. The method utilizes effector cells that are acti(cid:173)
`vated ex vivo, freed from the activating agent, armed
`with antibodies that immunoreact with the target cells
`and thereafter co-incubated with target cells.
`Thus, one aspect of the invention contemplates a
`method of specifically killing target cells and comprises
`the steps of:
`{a) Activating ex vivo a culture of leucocytes such as
`peripheral blood mononuclear cells (PBMCs) or periph(cid:173)
`eral blood lymphocytes (PBLs) that contain lgGl,
`lgG2a, lgG2b, or lgG3 antibody Fe receptors with an
`amount of interleukin-2 (IL-2) sufficient to enhance the
`natural killer activity of those cells and form IL-2-
`activitated effector cells.
`(b) The IL-2-activated effectors are then separated
`from toxic amounts of IL-2, and the IL·2•freed cells are
`collected.
`(c) The Fe receptors of those IL-2-activated effector
`cells are then bound with monoclonal antibodies (Mabs)
`of class IgGl, lgG2a, IgG2b, or tgG3 Fe portions, to
`form armed, IL-2-activated effector cells. The para-
`
`Miltenyi Ex. 1005 Page 8
`
`

`

`4,844,893
`
`5
`6
`FIG. 1 is a schematic representation of various strate-
`topic portions of useful Mabs bind to (imnmnoreact
`with) an antigen expressed on the surface of target cells.
`gies of target cell lysis mediated by antibodies (the Y-
`(d) A cytotoxic amount of the armed, IL-2-activated
`shaped entity) and an effector cells. In strategy A, a
`effector cells is contacted with target cells.
`monoclonal antibody that immunoreacts with an epi-
`(e) That contact is maintained for a time period suffi- 5 tope on the effector cell such as the T cell receptor is
`cient to kill the target cells. This method is useful both
`depicted as being covalently linked to the surface of the
`in vitro and in vivo. The contact and maintenance steps
`target ceU. Strategy B illustrates a monoclonal antibody
`are carried out out in the substantial absence of exo-
`that immunoreacts with an effector cell epitope com-
`geneously supplied IL-2.
`plexed with the target cell via the Fe receptor of the
`Another embodiment of the above method contem- 10 target cell (T-Fc binding). Strategy C illustrates the use
`plates contacting the target cells with a cytolytic
`of a heterobifunctional hybrid monoclonal antibody
`amount of bispecific hybrid monoclonal paratopic mo!-
`that immunoreacts with an epitope on the target cell
`ecules that contain a first anti-T3 paratopic portion that
`and with an epitope on the effector cell; the two por-
`immunoreacts with the T3 antigen expressed on the
`tions of the hybrid antibody are illustrated by wider and
`surfaces of T cells and a second paratopic portion that 15 narrower lines. Aspects of strategy C can be urilized
`immunoreacts with a second epitope expressed on the
`herein. Strategy D illustrates the use of a monoclonal
`surface of the target cells. That second target cell epi-
`antibody that immunoreacts with an epitope on the
`tape is different from the antigen bound by the above,
`target molecule and is complexed to the effector cell via
`tirst-named Mabs, and the immunoreaction of those
`the Fe receptor of the effector cell (E-Fc binding).
`first-named Mabs does not interfere with the immunore- 20 Strategy Dis the principal strategy utilized in the inven-
`action of the second para topic portion of the bispecific
`tion discussed herein. This figure is adapted from Staerz
`hybrid molecules, or vice versa.
`et al., "Targeting for T-Lymphocyte-Mediated Lysis by
`The bispecific hybrid molecules can be contacted
`Hybrid Antibodies" in Cyrolytic Lymphocytes and Com-
`plement, E. Podack, ed., CRC Press, Boca Raron, Fla.
`with the target cells before, substantually simulta-
`neously, or after the contact with the armed, IL-2- 25 (in press).
`activated effector cells. Preferably, that contact is sub-
`FIG. 2 is a graph illustrating augmentation of anti-
`stantially simultaneous with contact by the armed, IL-2·
`body-dependent cellular cytotoxicity (ADCC) induced
`activated effectors. Most preferably, the bispecific hy-
`by short-term ex vivo activation of human peripheral
`brid molecules are coated on the surfaces of anti-T3-
`blood mononuclear cells (PBMCs) with recombinant
`activated T cells that express the T3 antigen prior to 30 interleukin-2 (rlL-2) on 51Cr-labeled M21 human mela-
`noma cells in vitro. Human PBMCs at 106 cells per
`contacting the target cells, and those hybrid-coated or
`-armed anti-T3-activated T cells are present along with
`milliliter (cells/ml) were activated by admixture with
`the armed, IL-2-activated effector cells.
`250 units/milliliter (U/ml) of rlL-2 at 37 degrees C. for
`Another aspect of this invention contemplates a tar-
`different time periods. The PBMCs were thereafter
`get cell killing composition. That composition com- 35 washed substantially free of toxic amounts of rlL-2 and
`prises an aqueous physiologically tolerable diluent me-
`admixed with Mab 11C64 and target M2 l cells at the
`dium that contains dispersed therein armed, IL-2-
`effector to target cell ratios (E:T) shown on the ab-
`activated effector cells from a leucocyte population in
`scissa. The otdinate is in units of percent specific lysis of
`an amount effective to kill target cells (a cytolytic
`the target cells. Quadruplicate determinations were
`amount). The IL-2-activated effector cells are armed 40 made for each point with standard deviations of less
`with monoclonal antibodies of class lgG1, IgG2a,
`than about 10 percenl The ex vivo activation times
`lgG2b, or IgG3 that are bound to the Fe receptors of
`utilized were as follows: No rIL-2 (O); 15 minutes of
`the activated cells, and whose paratopic portions im-
`rIL-2 activation (
`); 30 minutes of rlL-2 activation
`munoreact with an antigen expressed on the surfaces of
`( ); I hour of activation (A); 2 hours of activation ( );
`45 and 4 hours of activation (0).
`the target cells.
`The composition can also contain a cytotoxic amount
`FIG. 3 is a bar graph showing cytolysis by ADCC of
`51Cr-labeled M21 human melanoma cells challenged
`of bispecific hybrid monoclonal paratopic molecules
`that contain first anti-T3 paratopic portions that im-
`with PBMCs from four melanoma patients and two
`munoreact with the T3 antigen expressed on the sur-
`normal, asymptomatic persons used as controls, using a
`faces of T cells and second paratopic portions that im- 50 4-hour assay time. Patients 1, 2 and 4 were in remission
`munoreact with a second epitope expressed on the sur-
`whereas patient 3 had advanced disease and a large
`face of the target cells, as discussed before. Preferably,
`tumor burden. An effector to target cell ratio of 50: I
`the bispecific molecules are present in the composition
`was utilized with the following effector co-incubations
`coated on the surfaces of anti•T3-activited T cells as
`prior to effector-target admixture: 500 U/ ml rlL-2
`armed, anti-T3-activated T cells.
`55 (closed bars): 25 micrograms per milliliter (ug/rnl) Mab
`The present invention has several benefits and ad van-
`11 C64 (hatched bars); and 500 U /ml rIL-2 plus 25
`tages. Saliem among those benefits and advantages is
`ug/ml Mab 11C64 (open bars).
`that a leucocyte population can be activated and armed
`FIG. 4 is another set of bar graphs that illustrate
`ex vivo and then utilized to kill target cells in the ab,
`augmentation of ADCC by IL-2-activated human
`seoce of exogeneously supplied IL-2, thereby avoiding 60 PBMCs. Here, PBMCs from a normal, asymptomatic
`the toxicity problems associated with administration of
`huUlan donor were admixed with target human 51Cr-
`exogenously supplied IL-2 to a recipient animal.
`labeled M21 melanoma cells at different effector to
`Still further benefits and advantages of the invention
`target cell ratios, as shown. The P13MC treatments
`will be apparent from the description of the invention
`during the 4-hour assay illustrated included: admixture
`that follows.
`65 with 250 U/ml of rIL-2 (horizontallY.-hatched bars);
`admixture with Mab 11C64 at 25 micrograms per milli(cid:173)
`liter (ug/ ml) {diagonally-hatched bars); and co-incuba(cid:173)
`tion with 250 U/ml of rIL-2 plus 25 ug/ml Mab 11C64
`
`BRIEF DESCRIPTION OF THE ORA WINGS
`In the Figures forming a portion of this disclosure:
`
`Miltenyi Ex. 1005 Page 9
`
`

`

`4,844,893
`
`5
`
`8
`7
`duced when an antigen is immunologically bound by an
`(open bars). The percentage of specific lysis shown was
`obtained after subtraction of values for NK Jysis.
`antibody or a molecule containing a paratope. An im(cid:173)
`munoreactant is therefore a specific type of complex
`DETAILED DESCRIPTION OF THE
`formed between molecules.
`INVENTION
`The term "intact antibody" is used herein to distin-
`1. DEFINITIONS
`guish a complete molecule secreted by a cell from
`The term "antibody'' refers to a molecule that Is a
`other, smaller, molecules that also contain the paratope
`necessary for biological activity in an immunoreaction
`member of a family of glycosylated proteins called
`immunoglobulins that can specifically combine with an
`with an antigen.
`antigen. Such an antibody combines with its antigen by lO The paratopic molecules useful in the present inven-
`a specific immunologic binding interaction between the
`tion are monoclonal paratopic molecules. A ··mono-
`antigenic determinant (epitope) of the antigen and the
`clonal antibody" (often referred to herein as a "Mab") is
`antibody combining site (paratope) of the antibody.
`a antibody produced by clones of a hybridoma that
`An "antibody combining site" is that structural por-
`secretes but one kind of antibody molecule, and a mono-
`tion of an antibody molecule comprised of heavy and 15 clonal paratopic molecule is a monoclonal antibody.
`The hybridoma cell is fused from an antibody-produc-
`light chain variable and hypervariable regions that spe-
`cifically binds antigen. Using the nomenclature of
`ing cell and a myeloma or other self-perpetuating cell
`Jerne, Ann. lmmunoL (Inst, Pasteur), 125C, 373 (1974)
`line. Such antibodies were first described by Kohler and
`an antibody combining site is usually referred to herein Milstein, Nature, 256, 495-497 (1975), which descrip-
`20 tion is incorporated herein by reference.
`as a "paratope" •
`1:he word "~tigen'' ~as been used hist?rically to
`The terms "monoclonal paratopic molecule", "para•
`designate ~ entity that. 1s boun_d by an antibody, ~nd
`topic molecule", "monoclonal antibody" and "Mab"
`also to d~1gnate the entity that mduc~s _the produc~on
`are used interchangeably herein to refer to an intact
`of the antibody. More current usage limits the meanmg
`monoclonal antibody.
`of antigen!? that entity.~?und by an antibod_y, wher~ 25 The words "secrete" and "produce" are often used
`interchangeably in the art as to cells from which anti-
`the word. immunogen. is used for the e1:1t1ty _that Ill·
`duc~s ~tlbody productto~. Where_ an ~~ty ~iscussed
`body molecules are obtained. Cells that roduce anti-
`herem IS both 1mmunogemc and antlgemc 1t will gener-
`.
`p
`.
`.
`'
`bodies may, however, not secrete those molecules mto
`all b t
`ti
`d
`f .
`Y e erme an an gen.
`11
`h .
`.
`Th h b .d
`t

`e y rt oma ce s o mterest
`th 30 t ell' environment.
`· d t
`"
`t"
`Tho phraso ,.
`t·
`'b d'

`re,ers o
`..
`.. an 1gemc e ermman
`h
`h ·
`e
`•

`1 1
`erem secrete monoc ona anti o 1es mt~ t eir environ-
`actual structural portion of the antigen that is immuno-
`logically bound by an antibody combining site. The
`men~. Ne~ert~eless, such c~lls ~e sometunes ~eferr~d to
`Jeme nomenclature defines an antigenic determinant as
`~erem as an~body-producmg cells'. an~. their antt~~~-
`1es 3:e so~etunes referred_ ~o as. bemg produced m
`an "epitope".
`The term "biologically active" refers at least to the 35 keepmg Wt~. the phrase ~t!lized m the_ art,
`. T~e tei1;I' .supern~tant'_ 1s us~d herein to refer to the
`ability to specifically bind antigen or specific antibody
`m vitro hqu1d med!um 10 which cells are cultured.
`combining site, although other general or effector capa-
`bility may be present as well. Biological activity of a M?noclonal paratop1<: molecules ~roduced by the .hy-
`paratopic molecule containing an antibody combining
`bn~oma cultures. of mter_est herem are secreted mto
`site is evidenced by the immunologic reaction of the 40 their cul~ure medium env_rronment. Therefore the cul-
`ture medium supe~tant 1s one prefe~ed so~ce of ~e
`antibody paratope (combining site) with its epitope
`(antigenic detenninant) upon their admixture in an
`monoclonal paratop1_c molecules and 1s readily obtatn-
`aqueous medium to fonn an immunoreactant, at )east at
`able free from hybndoma cells by well known tech-
`physiological pH values and ionic strengtbs. Preferably,
`niques. El!;emplary of such techniques is low speed cen-
`biological activity occurs under biologicitl conditions· 45 trifugation to sediment cells out of the liquid medium.
`i.e., those conditions wherein a paratope-containing Monoclonal paratopic molecules can alternatively be
`obtained from ascites tumor fluid (ascites fluid) of labo-
`molecule of this invention binds to its epitope within a
`pH value range of about 5 to about 9, at ionic strengths
`ratory animals into which the hybridoma tissue was
`such as that of distilled water to that of about one molar
`introduced. Both methods are described hereinafter.
`sodium chloride, and at temperatures of about 4 degrees 50
`Il. GENERAL DISCUSSION
`C. to about 45 degrees C. The monoclonal paratopic
`As already noted, several strategies have been devel(cid:173)
`molecules useful herein are biologically active.
`oped to obtain cytolysis. Treatments such as systemic
`"ELISA'' refers to an enzyme-linked immunosorbent
`administration of rIL-2 and the sole use of LAK cells
`assay that employs an antigen or antibody bound to a
`produced by activation with rIL-2 have been shown to
`solid phase and an enzyme-antibody or enzyme-antigen 55
`be too non-specific for generalized use. The more re(cid:173)
`conjugate to detect and quantify the amount of antigen
`cently reported results with TIL cells and IL-2 is re(cid:173)
`or antibody present in a sample. A description of the
`ELISA technique is found in Chapter 22 of the 4th
`portedly effective in mice, but still utilizes systemic
`Edition of Basic and Clinical Immunology by D. P. Sites
`administration of rlL-2. Since IL-2 can be toxic, it
`would be beneficial if such systemic uses could be
`et al., published by Lange Medical Publications of Los 60
`avoided.
`Altos, Calif. in 1982 and in U.S. Pat. Nos. 3,654,090;
`3,850,752; and 4,016,043, which are incorporated herein
`As also pointed out previously, the exquisite specific(cid:173)
`by reference.
`ity of monoclonal antibodies has been brought to bear
`"Enzyme" refers to a protein capable of accelerating
`upon cytolysis of pre-specified target cell populations.
`or producing by catalytic action some change in a sub- 65
`However, when used as the only modality, results with
`strate for which it is often specific.
`monoclonals alone have been uneven.
`"Immunoreactant" as used herein refers to the prod(cid:173)
`Four strategies that utilize target-ceU specific Mabs
`uct of an immunological reaction; i.e., that entity pro-
`along with effector cells are iJlustrated schematically in
`
`Miltenyi Ex. 1005 Page 10
`
`

`

`9
`FIG. 1. Strategy A of that Figure illustrates use ofMabs
`whose paratopic portions immunoreact with an epitope
`Ort the effector cell and whose Fe portions are cova(cid:173)
`lently linked to the target cell. Strategy B utilizes Fe
`receptors on the target cell to bind to the antibody 5
`(T-Fc receptor binding), while the paratope binds to the
`effector cell. Strategy C utilizes a hybrid monoclonal
`antibody, a first paratopic portion of which im(cid:173)
`munoreacts with the effector cell whereas the second
`paratopic po

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