throbber
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY(PCT)
`
`(19) World Intellectual Property Organization
`International Bureau
`
`(43) International Publication Date
`3 October 2002 (03.10.2002)
`
`
`
`(10) International Publication Number
`WO 02/077029 A2
`
`(51) International Patent Classification’:
`
`(US). FORMAN,Stephen [US/US]; 2580 Oak Knoll Av-
`enue, San Marino, CA 91108 (US). RAUBITSCHEK,An-
`drew [US/US]; 1691 El Molino, San Marino, CA 91108
`(21) International Application Number:=PCT/US01/42997
`(US).
`
`C07K 14/705
`
`(22) International Filing Date:
`7 November 2001 (07.11.2001)
`
`(25) Filing Language:
`
`(26) Publication Language:
`
`English
`
`English
`
`(30) Priority Data:
`60/246,117
`
`7 November 2000 (07.11.2000)
`
`US
`
`(74) Agents: FIGG, E., Anthonyet al.; Rothwell, Figg, Ernst
`& Manbeck, P.C., Suite 701-E, 555 13th Street, N.W, Wash-
`ington, DC 20004 (US).
`
`(81) Designated States (national): AU, CA, JP, US.
`
`(84) Designated States (regional): European patent (AT, BE,
`CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IT, LU, MC,
`NL,PT, SE, TR).
`
`(71) Applicant: CITY OF HOPE [US/US]; 1500 East Duarte
`Road, Duarte, CA 91010-3000 (US).
`
`without international search report and to be republished
`upon receipt of that report
`
`Published:
`
`(72) Inventors; and
`(75) Inventors/Applicants (for US only): JENSEN, Michael,
`C. [US/US]; 2305 Woodlyn Road, Pasadena, CA 91104
`
`For two-letter codes and other abbreviations, refer to the "Guid-
`ance Notes on Codes andAbbreviations" appearing at the begin-
`ning ofeach regular issue ofthe PCT Gazette.
`
`(54) Title: CD19-SPECIFIC REDIRECTED IMMUNE CELLS
`
`oGh pAn
`
`On COlE1
`
` Pacli751)
`
`
`
`~ Span
`CMV Promoter
`
`
`A
`
`WO02/077029A2
`
`Intron A
`
`CD19R scFvFc:Zeta
`
`chimeric T cell receptor by electroportion using naked DNA encodingthe receptor.
`
`engineered,
`Genetically
`(57) Abstract:
`redirected
`immune
`cells
`CD19-specific
`expressing a cell surface protein having an
`extracellular domain comprising a receptor
`which is specific for CD19, an intracellular
`signaling domain,
`and a transmembrane
`domain.
`Use of such cells for cellular
`
`immunotherapy of CD19* malignancies
`and for abrogating any untoward B cell
`function.
`In one embodiment, the immune
`cell is a T cell and the cell surface protein is
`a single chain svFvFc:€
`receptor where scFc
`designates the Vy and V, chains of a single
`chain monoclonal antibody to CD19, Fe
`represents at least part of a constant region
`of an IgG, and €
`represents the intracellular
`signaling domain of the zeta chain of human
`CD3.
`The extracellular domain scFvFc
`
`and the intracellular domain ¢ are linked
`by a transmembrane domain such as the
`transmembrane domain of CD4. A method
`
`of making a redirected T cell expressing a
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO 02/077029
`
`PCT/US01/42997
`
`CD19-SPECIFIC REDIRECTED IMMUNE CELLS
`
`Cross-reference to Related Application:
`
`[0001]
`
`This application claims priority to Provisional Application Serial No.
`
`60/246,117, filed November 7, 2000, the disclosure of which is incorporated by reference.
`
`BACKGROUND OF THE INVENTION
`
`[0002]
`
`This invention relates to the field of genetically engineered, redirected
`
`immunecells andto the field of cellular immunotherapy of B-cell malignancies, B-cell
`
`lymphoproliferative syndromes and B-cell mediated autoimmunediseases.
`
`The publications and other materials used herein to illuminate the background ofthe
`
`invention or provide additionaldetails respecting the practice are incorporated by
`
`reference.
`
`[0003]
`
`Approximately half of all hematopoietic stem cell transplantation (HSC)
`
`procedures performed inthe United States are for the treatment of hematologic
`
`malignancy [1]. Theinitial obstacles for successful HSC transplantation were in large part
`
`due to inadequate treatment modalities for ameliorating regimen-related toxicities and for
`
`controlling opportunistic infections and graft-versus-host disease (GVHD)[2-5]. As
`
`supportive care measures have improved overthe last decade, post-transplant disease
`
`relapse has emerged as the major impediment to improving the outcomeofthis patient
`population [6-10]. The inability of maximally intensive preparative regimens combined
`with immunologic graft-versus-tumorreactivity to eradicate minimal residual disease is
`
`the mechanism oftreatment failure in allogeneic transplantation while, in the autologous
`
`setting, tumor contamination of the stem cell graft can also contribute to post-transplant
`
`relapse [11]. Targeting minimalresidual disease early after transplantation is one strategy
`to consolidate the tumor cytoreduction achieved with myeloablative preparative regimens
`
`and purge, in vivo, malignant cells transferred with autologous stem cell grafts. The utility
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`of therapeutic modalities for targeting minimal residual disease shortly following stem
`
`cell rescue is dependent on both a limited spectrum of toxicity and the susceptibility of
`
`residual tumorcells to the modality's antitumor effector mechanism(s). The successful
`elimination of persistent minimalresidual disease should not only have a major impact on
`the outcomeoftransplantation for hematologic malignancy utilizing current
`
`myeloablative preparative regimens but may also provide opportunities to decrease the
`
`intensity of these regimens andtheir attendanttoxicities.
`
`[0004]
`
`The prognosis for patients with bcr-abl positive Acute Lymphoblastic
`
`Leukemia (ALL) treated with chemotherapy is poor and allogeneic transplantation has
`
`offered a curative option for many patients when an appropriate donor was available. For
`example at the City of Hope, 76 patients with bcr-abl positive ALL were treated with
`allogeneic Bone Marrow Transplantation (BMT) from a HLA matched donor. Of these
`patients, 26 were in first remission, 35 were transplantedafter first remission. The two
`year probability of disease free survival was 68% with a 10% relapserate in those patients
`transplanted in first remission whereas for those patients transplantedafter first remission,
`the disease-free survival and relapse rate were 36% and 38%,respectively [12]. Post-
`transplant Polymerase Chain Reaction (PCR) screening of blood and marrow for ber-abl
`transcript is under evaluation as a molecular screening tool for identifying early those
`transplant recipients at high risk for later developmentof overt relapse [13,14]. Patients
`
`for whom detectable p190 transcript was detected following BMThad a 6.7 higher
`
`incidence of overt relapse than PCR negative patients. The median time from the
`
`development ofa positive signal to morphologic relapse was 80-90 days in these patients.
`The identification of patients in the earliest phases of post-transplant relapse affords the
`
`opportunity for making therapeutic interventions when tumor burden is low and
`potentially most amenableto salvage therapy.
`[0005]
`Recent advancesin the field of immunology have elucidated many of the
`
`molecular underpinnings of immune system regulation and have provided novel
`
`opportunities for therapeutic immune system manipulation, including tumor
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`immunotherapy. Evidence supporting the potential of immune-mediated eradication of
`
`residual tumorcells following allogeneic transplantation can be inferred by comparing the
`
`disparate relapse rates between recipients of syngeneic and non-T cell depleted matched
`
`sibling transplants. Patients with chronic myelogenous leukemia in chronic phase (CML-
`CP), acute myelogenousleukemia in first complete remission (1* CR), and acute
`
`lymphoblastic leukemia in 1 CR who received a marrow transplant from a syngeneic
`
`donorhad an actuarial probability of relapse at 3 years of 45%, 49%, and 41%,
`
`respectively, whereas the rates for recipients of a non-T depleted marrow transplant from
`
`an HLA identical sibling for the same diseases were 12%, 20%, and 24%, respectively
`
`[15-17]. The reduction of relapse rates following allogeneic bone marrow transplantation
`
`has been most significant in patients who develop acute and/or chronic GVHD.Currently,
`efforts are focused on developing strategies to selectively augmentthe graft-versus-
`leukemia (GVL) responsein order to reduce post-transplant relapse rates without the
`
`attendant toxicities of augmented GVHD.
`
`[0006]
`
`Studies in animal models have established that donor MHC-restricted CD8*
`
`and CD4* a/f* T cells specific for minor histocompatibility antigens encoded by
`
`polymorphic genes that differ between the donor andrecipient are the principle mediators
`
`of acute GVHD and GVL [18-21]. Recently, patients with CML in chronic phase who
`
`relapse after allogeneic BMT havebeenidentified as a patient population for whom the
`
`infusion of donor lymphocytes (DLI) successfully promotes a GVL effect [22,23].
`
`Complete response rates of approximately 75% are achieved with DLIcell doses in the
`range of 0.25-12.3x10° mononuclear cells/kg [24]. Although the antitumoractivity of
`
`donor lymphocyte infusion underscores the potential of cellular immunotherapy for CML,
`
`the clinical benefit of DLI has not been generalizable to all forms of hematologic
`
`malignancy. Relapsed ALL is muchless responsive to DLI with a reported CR rate of
`
`less than 20%; when tumor responses are observed, they are typically associated with
`
`significant GVHD morbidity and mortality [25]. In order to increase the therapeutic ratio
`
`of DLI, genetic modification of donor lymphocytes to express a suicide gene is being
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`.
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`evaluated as a strategy to permit the in vivo ablation of donor lymphocytes should toxicity
`
`from GVHDwarrant this maneuver [26,27]. Alternately, efforts are underwayto identify
`
`genes encoding minor histocompatibility antigens (mHA’s) with restricted hematopoietic
`expression thatelicit donor antigen-specific T cell responses. The isolation, ex vivo
`expansion, andre-infusion of donor-derived clones specific for these mHA’s hasthe
`potential of selectively augmenting GVL following allogeneic bone marrow
`
`transplantation [28-30].
`[0007]
`Non-transformed B-cells and malignant B-cells express an array ofcell-
`surface moleculesthat define their lineage commitment and stage of maturation. These
`
`were identified initially by murine monoclonal antibodies and more recently by molecular
`genetic techniques. Expression of several of these cell-surface molecules is highly
`restricted to B-cells and their malignant counterparts. CD20 is a clinically useful cell-
`
`surface target for B-cell lymphoma immunotherapy with anti-CD20 monoclonal
`antibodies. This 33-kDaprotein hasstructural features consistent with its ability to
`
`function as a calcium ion channel and is expressed on normal pre-B and mature B cells,
`
`but not hematopoietic stem cells nor plasmacells [31-33]. CD20 does not modulate nor
`does it shed from the cell surface [34]. Jn vitro studies have demonstrated that CD20
`
`crosslinking by anti-CD20 monoclonalantibodies can trigger apoptosis of lymphomacells
`[35,36]. -Clinical trials evaluating the antitumoractivity of chimeric anti-CD20 antibody
`IDEC-C2B8 (Rituximab) in patients with relapsed follicular lymphoma have documented
`tumor responsesin nearly half the patients treated, although the clinical effect is usually
`transient [37-40]. Despite the prolonged ablation of normal CD20* B-cells, patients
`receiving Rituximab have not manifested complications attributable to B-cell
`lymphopenia [41]. Radioimmunotherapy with *I-conjugated and °°Y-conjugated anti-
`CD20antibodies also has shown promisingclinical activity in patients with
`
`relapsed/refractory high-grade Non-Hodgkins Lymphomabut hematopoietic toxicities
`from radiation have been significant, often requiring stem cell support [42].
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`[0008]
`
`Unlike CD20, CD19 is expressed on all human B-cells beginning from the
`
`initial commitmentof stem cells to the B lineage andpersisting until terminal
`
`differentiation into plasmacells [43]. CD19 is a type I transmembrane protein that
`
`associates with the complement 2 (CD21), TAPA-1, and Leu13 antigens forming a B-cell
`
`signal transduction complex. This complex participates in the regulation of B-cell
`proliferation [44]. Although CD19 does not shed from thecell surface, it does internalize
`[45]. Accordingly, targeting CD19 with monoclonal antibodies conjugated with toxin
`molecules is currently being investigated as a strategy to specifically deliver cytotoxic
`
`agents to the intracellular compartment of malignant B-cells [46-48]. Anti-CD19
`antibody conjugatedto blocked ricin and poke-weedantiviral protein (PAP) dramatically
`
`increase specificity and potency of leukemiacell killing both in ex vivo bone marrow
`purging procedures and when administered to NOD-SCIDanimals inoculated with CD19*
`leukemiacells [49]. Jn vitro leukemia progenitor cell assays have provided evidence that
`
`the small percentage of leukemic blasts with the capacity for self-renewal express CD19
`
`on their cell surface. This conclusion was derived from the observations that leukemic
`
`progenitor activity is observed exclusively in fresh marrow samples sorted for CD19
`positive cells and is not observed in the CD19 negative cell population [50]. Additionally
`B43-PAPtreatmentof relapsed leukemic marrow specimensablates progenitorcell
`
`activity while a PAP conjugated antibody with an irrelevant specificity had no such
`
`activity [51]. Systemic administration of the CD19-specific immunotoxin B43-PAP is
`
`currently undergoing investigation in phase I/II clinicaltrials in patients with high risk
`
`pre-B ALL[52].
`
`[0009]
`
`Despite the antitumoractivity of monoclonal anti-CD20 and anti-CD19
`
`antibody therapy observed in clinicaltrials, the high rate of relapse in these patients
`
`underscores the limited capacity of current antibody-based immunotherapyto eliminateall
`
`tumorcells [53]. In contrast, the adoptive transfer of tumor-specific T cells can result in
`complete tumoreradication in animal models and a limited numberofclinical settings
`
`[54,55]. Theability of transferred T cells to directly recognize and lyse tumortargets,
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`produce cytokines that recruit and activate antigen non-specific antitumor effector cells,
`migrate into tumor masses, and proliferate following tumor recognition all contributeto
`
`the immunologic clearance of tumorby T cells [56]. Expression-cloning technologies
`have recently permitted the genetic identification of a growing number of genes expressed
`by human tumors to which T cell responses have beenisolated [57,58]. To date leukemia
`and lymphoma-specific antigens have not been identified that are both broadly expressed
`by malignant B-cells and elicit T cell responses. Consequently, preclinical and clinical
`investigation has focused on combining antibody targeting of tumors with T cell effector
`
`mechanisms by constructing bispecific antibodies consisting of CD20 or CD19 binding
`
`sites and a bindingsite for a cell-surface CD3 complex epitope. Such bispecific
`antibodies can co-localize leukemia and lymphomatargets with activated T cells resulting
`
`in target cell lysis in vitro [59-61]. The in vivo antitumoractivity of such bispecific
`antibodies has been limited, however, both in animal models as well as in clinical practice
`
`[62]. The discrepancy betweenin vitro activity and in vivo effect likely reflects the
`inherent limitations in antibody immunotherapy compoundedbythe obstacles associated
`
`with engaging T cells and tumor cells via a soluble linker in a mannerthat yields a
`
`persistent and functional cellular immuneresponse [63].
`
`[00010]
`
`The safety of adoptively transferring antigen-specific CTL clones in
`
`humanswasoriginally examined in bone marrow transplant patients who received donor-
`
`derived CMV-specific T cells [56]. Previous studies have demonstrated that the
`
`reconstitution of endogenous CMV-specific T cell responses following allogeneic bone
`
`marrow transplantation (BMT)correlates with protection from the development of severe
`
`CMVdisease [64]. In an effort to reconstitute deficient CMV immunity following BMT,
`
`CD8* CMV-specific CTL clones were generated from CMV seropositive HLA-matched
`
`sibling donors, expanded, and infused into sibling BMTrecipients at risk for developing
`CMVdisease. Fourteen patients were treated with four weekly escalating doses of these
`CMV-specific CTL clones to a maximum cell dose of 10° cells/m? without any attendant
`toxicity [65]. Peripheral blood samples obtained from recipients of adoptively transferred
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`T cell clones were evaluated for in vivo persistence of transferred cells. The recoverable
`CMV-specific CTL activity increased after each successive infusion of CTL clones, and
`persisted at least 12 weeks after the last infusion. However, long term persistence of
`CD8*clones without a concurrent CD4* helper response was not observed. Nopatients
`developed CMV viremia or disease. These results demonstrate that ex-vivo expanded
`CMV-specific CTL clones can be safely transferred to BMTrecipients and can persist in
`vivo as functional effector cells that may provide protection from the development of
`
`CMVdisease.
`
`A complication of bone marrow transplantation, particularly when marrow
`[00011]
`is depleted of T cells, is the development of EBV-associated lymphoproliferative disease
`[66]. This rapidly progressiveproliferation ofEBV-transformed B-cells mimics
`immunoblastic lymphomaand is a consequence of deficient EBV-specific T cell
`immunity in individuals harboring latent virus or immunologically naive individuals
`receiving a virus inoculum with their marrow graft. Clinicaltrials by Rooneyetal. have
`demonstrated that adoptively transferred ex-vivo expanded donor-derived EBV-specific T
`cell lines can protect patients at high risk for developmentof this complication as well as
`mediate the eradication of clinically evident EBV-transformed B cells [54]. No
`significant toxicities were observed in the forty-one children treated with cell doses in the
`range of 4x10’ to 1.2x10° cells/m’.
`[00012]
`Genetic modification of T cells used in clinical trials has been utilized to
`
`markcells for in vivo tracking and to endow T cells with novel functional properties.
`
`Retroviral vectors have been used most extensively for this purpose duetotheir relatively
`
`high transduction efficiency and lowin vitro toxicity to T cells [67]. These vectors,
`however, are time consuming and expensive to prepare as clinical grade material and
`
`must be meticulously screened for the absence of replication competent viral mutants
`
`[68]. Rooneyet al. transduced EBV-reactive T cell lines with the NeoR geneto facilitate
`assessmentofcell persistence in vivo by PCR specific for this marker gene [69]. Riddell
`et al. have conducted a PhaseI trial to augment HIV-specific immunity in HIV
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`seropositive individuals by adoptive transfer using HIV-specific CD8* CTL clones[70].
`
`These clones were transduced with the retroviral vector tgLS'HyTK which directs the
`
`synthesis of a bifunctional fusion protein incorporating hygromycin phosphotransferase
`
`and herpes virus thymidine kinase (HSV-TK)permitting in vitro selection with
`
`hygromycin and potential in vivo ablation of transferred cells with gancyclovir. Six HIV
`
`infected patients were treated with a series of four escalating cell dose infusions without
`
`toxicities, with a maximum cell dose of 5x10 cells/m? [70].
`
`[00013]
`
`Asan alternate to viral gene therapy vectors, Nabel et al. used plasmid
`
`DNAencoding an expression cassette for an anti-HIV gene in a PhaseI clinicaltrial.
`
`Plasmid DNA wasintroduced into T cells by particle bombardment with a gene gun [71].
`Genetically modified T cells were expandedand infused back into HIV-infected study
`subjects. Although this study demonstrated the feasibility of using a non-viral genetic
`
`modification strategy for primary human T cells, one limitation of this approach is the
`
`episomal propagation of the plasmid vector in T cells. Unlike chromosomally integrated
`
`transferred DNA, episomal propagation of plasmid DNAcarriesthe risk of loss of
`
`transferred genetic material with cell replication and of repetitive random chromosomal
`
`integration events.
`
`[00014]
`
`Chimeric antigen receptors engineered to consist of an extracellular single
`
`chain antibody (scFvFc) fused to the intracellular signaling domain of the T cell antigen
`
`receptor complex zeta chain (€) have the ability, when expressed in T cells, to redirect
`antigen recognition based on the monoclonal antibody’s specificity [72]. The design of
`
`scFvFc:¢ receptors with target specificities for tumorcell-surface epitopesis a
`
`conceptually attractive strategy to generate antitumor immuneeffector cells for adoptive
`
`therapy as it does not rely on pre-existing anti-tumor immunity. These receptors are
`
`“universal” in that they bind antigen in a MHC independentfashion, thus, one receptor
`
`construct can be used to treat a population of patients with antigen positive tumors.
`
`Several constructs for targeting human tumors have been described in the literature
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`including receptors with specificities for Her2/Neu, CEA, ERRB-2, CD44v6,and
`
`epitopes selectively expressed on renal cell carcinoma [73-77]. These epitopesall share
`
`the common characteristic of being cell-surface moieties accessible to scFv binding by the
`
`chimeric T cell receptor. Jn vitro studies have demonstrated that both CD4* and CD8* T
`
`cell effector functions canbe triggered via these receptors. Moreover, animal models
`
`have demonstrated the capacity of adoptively transferred scFvFc:¢ expressing T cells to
`
`eradicate established tumors [78]. The function of primary human T cells expressing
`
`tumor-specific scFvFc:C receptors have been evaluated in vitro; these cells specifically
`
`lyse tumortargets and secrete an array of pro-inflammatory cytokines including IL-2,
`
`TNF, IFN-y, and GM-CSF [79]. Phase I pilot adoptive therapy studies are underway
`
`utilizing autologous scFvFc:C-expressing T cells specific for HIV gp120 in HIV infected
`
`individuals and autologous scFvFc:C-expressing T cells with specificity for TAG-72
`expressed on a variety of adenocarcinomasincluding breast and colorectal
`
`adenocarcinoma.
`
`[00015]
`
`Investigators at City of Hope have engineered a CD20-specific scFvFc:¢
`
`receptor constructfor the purposeof targeting CD20+ B-cell malignancy [80]. Preclinical
`
`laboratory studies have demonstrated thefeasibility of isolating and expanding from
`
`healthy individuals and lymphoma patients CD8+ CTL clonesthat contain a single copy
`
`of unrearranged chromosomally integrated vector DNA and express the CD20-specific
`
`scFvFc:z6 receptor [81]. To accomplish this, purified linear plasmid DNA containing the
`
`chimeric receptor sequence underthetranscriptional control of the CMV immediate/early
`
`promoter and the NeoR gene under the transcriptional control of the SV40 early promoter
`was introduced into activated human peripheral blood mononuclear cells by exposure of
`
`cells and DNAto a brief electrical current, a procedure called electroporation [82].
`
`Utilizing selection, cloning, and expansion methods currently employed in FDA-approved
`
`clinical trials at the FHCRC, gene modified CD8+ CTL clones with CD20-specific
`
`cytolytic activity have been generated from each of six healthy volunteers in 15 separate
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`electroporation procedures [81]. These clones when co-cultured with a panel of human
`
`CD20+ lymphomacell lines proliferate, specifically lyse target cells, and are stimulated to
`
`produce cytokines.
`[00016]
`| It is desired to develop additional redirected immunecells and, in a
`preferred embodiment, redirectedTcells, for treating B-cell malignancies and B-cell
`
`mediated autoimmunedisease.
`
`SUMMARYOF THE INVENTION
`
`[00017]
`
`In one aspect, the present invention provides genetically engineered T cells
`
`which express and bear onthe cell surface membrane a CD19-specific chimeric T cell
`
`receptor(referred to herein as “CD19R”) having an intracellular signaling domain, a
`
`transmembrane domain (TM) and a CD19-specific extracellular domain (also referred to
`
`herein as “CD19-specific T cells”). The present invention also provides the CD19-
`
`specific chimeric T cell receptors, DNA constructs encoding the receptors, and plasmid
`
`expression vectors containing the constructs in proper orientation for expression.
`
`[00018]
`
`In a second aspect, the present invention provides a methodoftreating a
`
`CD19* malignancy in a mammal which comprises administering CD19-specific T cells to
`the mammalin a therapeutically effective amount. In one embodiment, CD8* CD19-
`specific T cells are administered, preferably with CD4* CD19-specific T cells. Ina
`
`second embodiment, CD4* CD19-specific T cells are administered to a mammal,
`
`preferably with CD8* cytotoxic lymphocytes which do not express the CD19-specific
`chimeric receptor of the invention, optionally in combination with CD8* CD19-specific
`
`redirected T cells.
`
`In another aspect, the present invention provides a methodof abrogating
`[00019]
`any untoward B cell function in a mammal which comprises administering to the mammal
`CD19-specific redirected T cells in a therapeutically effective amount. These untoward B
`
`cell functions can include B-cell mediated autoimmunedisease (e.g., lupus or rheumatoid
`
`arthritis) as well as any unwanted specific immune responseto a given antigen.
`
`10
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`[00020]
`
`In another aspect, the present invention provides a method of making and
`
`expanding the CD19-specific redirected T cells which comprises transfecting T cells with
`
`an expression vector containing a DNA construct encoding the CD19-specific chimeric
`receptor, then stimulating the cells with CD19" cells, recombinant CD19,or an antibody
`
`to the receptor to cause the cells to proliferate. In one embodiment, the redirected T cells
`
`are prepared by electroporation. In a second embodiment,the redirected T cells are
`
`prepared by using viral vectors.
`
`[00021]
`
`In another aspect, the present invention provides a method oftargeting
`
`Natural Killer (NK) cells which express and bear onthe cell surface membrane a CD19-
`
`specific chimeric immunereceptor having an intracellular signaling domain, a
`
`transmembrane domain (TM)and a CD19-specific extracellular domain.
`- [00022]
`In another aspect, the present invention provides a method oftargeting
`macrophagecells which express and bear onthe cell surface membrane a CD19-specific
`
`chimeric immunereceptor having an intracellular signaling domain, a transmembrane
`
`domain (TM) and a CD19-specific extracellular domain.
`
`[00023]
`
`In another aspect, the present invention provides a method oftargeting
`
`neutrophils cells which express and bear on the cell surface membrane a CD19-specific
`
`chimeric immunereceptor having an intracellular signaling domain, a transmembrane
`
`domain (TM)and a CD19-specific extracellular domain.
`
`[00024]
`
`In another aspect, the present invention provides a method oftargeting
`
`stem cells which express and bear on the cell surface membrane a CD19-specific chimeric
`
`immunereceptor having an intracellular signaling domain, a transmembrane domain
`
`(TM) and a CD19-specific extracellular domain.
`
`[00025]
`
`In another aspect, the invention provides a CD-19-specific chimeric T-cell
`
`receptor comprising an intracellular signalling domain, a transmembrane domain and a
`
`CD19-specific extracellular domain.
`
`[00026]
`
`In one embodiment, the CD19-specific chimeric T cell receptor of the
`
`invention comprises scFvFc:¢, where scFvFc represents the extracellular domain, scFv
`
`11
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`designates the V,, and V, chains ofa single chain monoclonal antibody to CD19, Fe
`represents at least part of a constant region of an IgG,, and ¢ represents the intracellular
`
`signaling domain of the zeta chain of human CD3.
`
`[00027]
`
`In another embodiment, the CD19-specific chimeric T cell receptor of the
`
`invention comprises the scFvFc extracellular domain and the ¢ intracellular domain are
`
`linked by the transmembrane domain of human CD4.
`
`[00028]
`
`In another embodiment, the CD19-specific chimeric T cell receptor of the
`
`invention comprises amino acids 23-634 of SEQ ID NO:2.
`
`[00029]
`
`In another aspect, the invention provides a plasmid expression vector
`
`containing a DNA construct encoding a chimeric T-cell receptor of the invention in
`
`properorientation for expression.
`
`BRIEF DESCRIPTION OF THE FIGURES
`
`[00030]
`
`Figures 1A-1C show the double-stranded DNA sequence and amino acid
`
`sequence for the CD19:zeta chimeric immunoreceptor of the present invention, SEQ ID
`
`NO.1 and show the source of the DNA segments found in the chimeric immunoreceptor.
`
`[00031]
`
`[00032]
`
`Figure 2 is a schematic representation of the plasmid pwG-CD19R/HyTK.
`
`Figure 3 shows Western blot analyses which demonstrate the expression of
`
`the CD19R/scFvFe:¢ chimeric receptor.
`
`[00033]
`
`Figure 4 is a graphical representation showing the antigen-specific cytolytic
`
`activity of T-cells expressing the CD19R/scFvFe:¢ chimeric receptor.
`
`[00034]
`
`Figure 5 is a graphical representation of the production of interferon-y by T
`
`cells expressing the CD19R/scFvFc:¢ chimeric receptor that are incubatedin the presence
`
`of variouscell lines expressing CD-19.
`
`[00035]
`
`Figure 6 A-E are graphical representations showing the antigen-specific
`
`cytolytic activity of CD19R/scFvFc:¢ chimeric receptor redirected T-cell clones.
`
`12
`
`UPenn Ex. 2028
`
`Miltenyi v. UPenn
`IPR2022-00853
`
`UPenn Ex. 2028
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`WO02/077029
`
`PCT/US01/42997
`
`DETAILED DESCRIPTION OF THE INVENTION
`
`[00036]
`
`The present invention is directed to genetically engineered, redirected T
`
`cells and to their use for cellular immunotherapy of B-cell malignancies, Epstein Barr
`
`Virus-related lymphoproliferative disorders, and B-cell mediated autoimmunediseases.
`[00037]
`In one aspect, the present invention provides genetically engineered T cells
`which express and bear on the cell surface membrane a CD19-specific chimeric T cell
`
`receptor having an intracellular signaling domain, a transmembrane domain and a CD19-
`
`specific extracellular domain (referred to herein as CD19-specific T cells). The
`
`extracellular domain comprises a CD19-specific receptor. Individual T cells of the
`invention may be CD4*/CD8, CD4/CD8*, CD4/CD8’ or CD4*/CD8". The T cells may be
`a mixed population of CD4*/CD8and CD4/CD8*'cells or a population of a single clone.
`CD4*Tcells of the invention produce IL-2 when co-cultured in vitro with CD19"
`lymphomacells. CD8* T cells of the invention lyse CD19* human lymphomatarget cells
`
`whenco-cultured in vitro with the target cells. The invention further provides the CD19-
`
`specific chimeric T cell receptors, DNA constructs encoding the receptors, and plasmid
`
`expression vectors containing the constructs in proper orientation for expression.
`
`[00038}
`
`In a preferred embodiment, CD19-specific redirected T cells express CD19-
`
`specific chimeric receptor scFvFe:¢, where scFv designates the Vy and V, chains of a
`single chain monoclonal antibody to CD19,Fc represents at least part of a constant region
`
`of a human IgG,, and ¢ represents the intracellular signaling domain of the zeta chain of
`
`human CD3. The extracellular domain scFvFc andthe intracellular domain ¢ are linked
`
`by a transmembrane domain such as the transmembrane domain of CD4. In other
`
`embodiments, the human Fc constant region may be provided by other species of antibody
`
`such as IgG, for example.
`
`[00039]
`
`In a specific preferred embodiment, a full length scFvFc:¢ cDNA,
`
`designated SEQ ID NO.1 or “CD19R:zeta,” comprises the

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