throbber
8224035
`
`U'.'IITED STATES DEPARTMENT OF COMMERCE
`
`United Stntcs J>n(c,nt nnd Trndemnrk Office
`
`March 11, 2022
`
`THIS IS TO CERTIFY THAT ANNEXED IS A TRUE COPY FROM THE
`RECORDS OF THIS OFFICE OF THE FILE WRAPPER AND CONTENTS
`OF:
`
`APPLICATION NUMBER: 61/502,649
`FILING DATE: June 29, 2011
`
`Certified by
`
`Under Secretary of Commerce
`for Intellectual Property
`and Oirector of the United States
`Patent and l'rademark Office
`
`Miltenyi Ex. 1019 Page 1
`
`

`

`PTO/SB/16
`0MB 0651-0032
`U.S. Patent and Trademark Office; U.S. DEPARTMENT OF COMMERCE
`PROVISIONAL APPLICATION FOR PATENT COVER SHEET
`This is a request for filing a PROVISIONAL APPLICATION FOR PATENT under 37 CPR l.53(c).
`Electronicall Filed
`
`Given Name (first and middle rif anyl)
`Carl
`
`JUNE
`
`INVENTOR(S)
`
`Family Name or Surname
`
`Residence
`(City and either State or ForeiW1 Countty)
`Merion Station, Pennsylvania
`
`□ Additional inventors are being named 011 the _ _ separately numbered sheets attached hereto
`TITLE OF THE INVENTION (500 characters max)
`COMPOSITIONS AND METHODS FOR TREATMENT OF CHRONIC
`L YMPHOCYTIC LEUKEMIA
`CORRESPONDENCE ADDRESS
`
`Direct all correspondence to:
`
`II Customer Number
`
`10872
`
`I
`
`I
`OR
`0 Firm or Individual Name Riverside Law LLP
`Address
`300 Four Falls Corporate Center, Suite 710
`Address
`300 Conshohocken State Road
`I ZIP I 19428
`West Conshohocken I State
`City
`PA
`I Telephone
`I 215-268-3871
`I Fax
`us
`Country
`215-268-3888
`ENCLOSED APPLICATION PARTS (check all that aooly)
`II Specification Number of Pages 71
`D CD(s), Number
`
`D Other (specify):
`
`II Drawing(s) Numbero/Sheets 15
`□ Application Data Sheet. See 37 CFR 1.76
`METHOD OF PAYMENT OF FILING FEES FOR THIS PROVISIONAL APPLICATION FOR PATENT
`[8] Applicant claims small entity status. See 37 CFR 1.27.
`D A check of money order is enclosed to cover the filing fees
`D Payment by credit card.
`[8J The Commissioner is hereby authorized to charge filing fees or
`credit any overpayment to Deposit Account No. 50-5366
`
`Filing Fee Amount ($110.00):
`
`I $110.00
`
`I
`
`The invention was made by an agency of the United States Government or under a contract with an agency of the United States
`Government.
`□ No.
`t contract number arc: This invention was made with
`[8:lycs, the name of the U.S. Government agency and the Governme
`government support under K24 CA 11787901 and RO 1 CA120409 awarded by the National Institutes of Health
`(NIH).
`
`0n
`
`1K:~~---- }- /-+~-,-----
`
`Signature
`Typed Name: ~-Bennis L. Hna's, Ph.D., J.D.
`
`Date: b ZLl I I I
`
`I
`
`Registration No. 65,315
`Docket No. 46483-6001-P2-US.600881
`Telephone No. 215.268.3888
`USE ONLY FOR FILING A PROVISIONAL APPLICATION FOR PATENT
`
`Miltenyi Ex. 1019 Page 2
`
`

`

`Attorney Docket No. 046483-600 l-P2-US .60088 I
`
`TITLE OF THE INVENTION
`
`COMPOSITIONS AND METHODS FOR TREATMENT OF CHRONIC
`
`L YMPHOCYTIC LEUKEMIA
`
`STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
`
`DEVELOPMENT
`
`This invention was made with government support under K24
`CAI 1787901 and ROI CA120409 awarded by the National Institutes of Health (NIH).
`The government has certain rights in the invention.
`
`BACKGROUND OF THE INVENTION
`
`The large majority of patients having B-cell malignancies, including
`chronic lymphocytic leukemia (CLL), will die from their disease. One approach to
`treating these patients is to genetically modify T cells to target antigens expressed on
`tumor cells through the expression of chimeric antigen receptors (CARs). CARs are
`antigen receptors that are designed to recognize cell surface antigens in a human
`leukocyte antigen-independent manner. Attempts in using genetically modified cells
`expressing CARs to treat these types of patients have met with very limited success. See
`for example, Brentjens et al., 2010, Molecular Therapy, 18:4, 666-668; Morgan et al.,
`2010, Molecular Therapy, published online Febrnary 23, 2010, pages 1-9; and, Till et al.,
`2008, Blood, 112:2261-2271.
`
`In most cancers, tumor-specific antigens are not yet well defined, but in B
`cell malignancies, CD 19 is an attractive tumor target. Expression of CD 19 is restricted to
`normal and malignant B cells (Uckun, et al. Blood, 1988, 71: 13-29), so that CD 19 is a
`widely accepted target to safely test CARs. While CARs can trigger T-cell activation in a
`manner similar to an endogenous T-cell receptor, a major impediment to the clinical
`application of this technology to date has been limited in vivo expansion of CAR+ T
`cells, rapid disappearance of the cells after infusion, and disappointing clinical activity
`(Jena, et al., Blood, 2010, I 16: 1035-1044; Uckun, et al. Blood, 1988, 71: 13-29).
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`1
`
`Miltenyi Ex. 1019 Page 3
`
`

`

`Thus, there is an urgent need in the aii for compositions and methods for
`
`treatment of CLL. The present invention addresses this need.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`5
`
`The following detailed description of preferred embodiments of the
`
`invention will be better understood when read in conjunction with the appended
`
`drawings. For the purpose of illustrating the invention, there are shown in the drawings
`
`embodiments which are presently preferred. It should be understood, however, that the
`
`invention is not limited to the precise arrangements and instrumentalities of the
`
`10
`
`embodiments shown in the drawings.
`
`Figure 1, comprising Figures IA through IC, depicts a schematic
`
`representation of the gene-transfer vector and transgene, gene modified T cell
`
`manufacturing and clinical protocol design. Figure 1 A depicts the lentiviral vectors and
`
`transgene that show the major functional elements. A vesicular stomatitis vims protein G
`
`15
`
`pseudotyped clinical grade lentiviral vector ( designated pELPs 19BBz) directing
`
`expression of anti-CD19 scFv derived from FMC63 murine monoclonal antibody, human
`
`CD8a hinge and transmembrane domain, and human 4-lBB and CD3zeta signaling
`
`domains was produced. Constitutive expression of the trans gene was directed by
`
`inclusion of an EF-la (elongation factor-la promoter); LTR, long terminal repeat; RRE,
`
`20
`
`rev response element. ( cPPT) and the central termination sequence (CTS). Figure is not
`
`to scale. Figure 1B depicts T cell manufacturing. Autologous cells were obtained via an
`
`apheresis, and T cells were enriched by mononuclear cell elutriation, washed and residual
`
`leukemia cells depleted by addition of anti-CD3/CD28 coated paramagnetic beads for
`
`positive selection and activation ofT cells. Lentiviral vector was added at the time of
`
`25
`
`cell activation and was washed out on day 3 post culture initiation. Cells were expanded
`
`on a rocking platform device (WAVE Bioreactor System) for 8-12 days. On the final day
`
`of culture the beads were removed by passage over a magnetic field and the CART 19 T
`
`cells harvested and cryopreserved in infusible medium. Figure 1 C depicts the clinical
`
`protocol design. Patients were given lymphodepleting chemotherapy as described,
`
`30
`
`followed by CART19 infusion #1 by i.v. gravity flow drip over a period of 15-20
`
`minutes. The infusion was given using a split dose approach over 3 days (10%, 30%,
`
`2
`
`Miltenyi Ex. 1019 Page 4
`
`

`

`60%) beginning 1 to 5 days after completion of chemotherapy. Endpoint assays were
`
`conducted on study week 4. At the conclusion of active monitoring, subjects were
`
`transferred to a destination protocol for long term follow up as per FDA guidance.
`
`Figure 2, comprising Figures 2A through 2F, is a series of images
`
`5
`
`demonstrating sustained in vivo expansion and persistence in blood and marrow of
`
`CART19 cells. DNA isolated from whole blood as depicted in Figure 2A through 2C or
`
`marrow as depicted in Figure 2D through 2F, samples obtained from UPN 01 as depicted
`
`in Figure 2A and 2D, UPN 02 as depicted in Figure 2B and 2E and VPN 03 as depicted
`
`in Figure 2C and 2F was subjected in bulk to Q-PCR analysis using a qualified assay to
`
`10
`
`detect and quantify CART19 sequences, Each data point represents the average of
`
`triplicate measurements on 100-200 ng genomic DNA, with maximal % CV less than
`
`1.56%, Pass/fail parameters for the assay included pre-established ranges for slope and
`
`efficiency of amplification, and amplification of a reference sample. The lower limit of
`
`quantification for the assay established by the standard curve range was 2 copies
`
`15
`
`transgene/microgram genomic DNA; sample values below that number are considered
`estimates and presented if at least 2/3 replicates generated a Ct value with % CV for the
`
`values 15%. CART19 cells were infused at day 0, 1, and 2 for VPN 01 and UPN 03, and
`
`days 0, 1, 2 and 11 for UPN 02.
`
`Figure 3, comprising Figures 3A through 3D, is a series of images
`
`20
`
`demonstrating serum and bone marrow cytokines before and after CART cell infusion;
`
`longitudinal measurements of changes in serum cytokines, chemokines and cytokine
`
`receptors in VPN O 1 as depicted in Figure 3A, UPN 02 as depicted in Figure 3B and UPN
`
`03 as depicted in Figure 3C, on the indicated day after CART19 cell infusion and serial
`
`assessments of the same analytes in the bone marrow from VPN 03 as depicted in Figure
`
`25
`
`3D. Samples were subjected to bulk to multiplex analysis using Luminex bead array
`
`technology and pre-assembled and validated multiplex kits. Analytes with a >=3 fold
`
`change are indicated, and plotted as relative change from baseline as depicted in Figure
`
`3A through 3C or as absolute values as depicted in Figure 3D. Absolute values for each
`
`analyte at each time-point were derived from a recombinant protein-based standard curve
`
`30
`
`over a 3-fold 8-point dilution series, with upper and lower limits of quantification
`
`(ULOQ, LLOQ) determined by the 80-120% observed/expected cutoff values for the
`
`3
`
`Miltenyi Ex. 1019 Page 5
`
`

`

`standard curves. Each sample was evaluated in duplicate with average values calculated
`
`and% CV in most cases less than 10%. To accommodate consolidated data presentation
`in the context of the wide range for the absolute values, data are presented as fold-change
`over the baseline value for each analyte. In cases where baseline values were not
`
`5
`
`detectable, half of the lowest standard curve value was used as the baseline value.
`Standard curve ranges for analytes and baseline (day 0) values (listed in parentheses
`
`sequentially for UPNOI, 02 and 03), all in pg/ml: ILl-Ra: 35.5-29,3 I 8 (689, 301, 287);
`IL-6: 2.7-4,572 (7, IO. I, 8.7); IFN-y: 11.2-23,972 (2.8, ND, 4.2 ); CXCLIO: 2.1-5,319
`
`(481,115,287); MIP-lp: 3.3-7,233 (99.7, 371,174); MCP-1: 4.8-3,600 (403,560,828);
`CXCL9: 48.2-3,700 (1,412, 126, 177); IL2-Ra: 13.4-34,210 (4,319, 9,477,610); IL-8:
`
`10
`
`2.4-5,278 (15.3, 14.5, 14.6); IL-10: 6.7-13,874 (8.5, 5.4, 0.7); MIP-la: 7.1-13,778 (57.6,
`
`57.3, 48.1).
`
`Figure 4, comprising Figures 4A through 4D, is a series of images
`
`depicting prolonged surface CART19 expression and establishment of functional
`
`15 memory CARs in vivo. Figure 4A depicts detection of CAR-expressing CD3+
`
`lymphocytes and absence of B cells in periphery and marrow. Freshly processed
`
`peripheral blood or marrow mononuclear cells obtained from UPN 03 at day 169 post(cid:173)
`
`CART19 cell infusion were evaluated by tlow-cytometry for surface expression of
`
`CARI 9 (top) or presence of B cells (bottom); as a control, PBMC obtained from a
`
`20
`
`healthy donor ND365 were stained. The gating strategy for the CD3+ and B cell
`
`populations is presented in Figure 9. To evaluate CAR19 expression in CD3+
`lymphocytes, samples were co-stained with antibodies to CD14-PE-Cy7 and CD16-PE(cid:173)
`
`Cy7 (dump channel) and CD3-FITC, positively gated on CD3+, and evaluated for
`
`CAR19 expression in the CDS+ and CDS-lymphocyte compartments by co-staining with
`
`25
`
`CD8a-PE and the anti-CARI 9 idiotype antibody conjugated to Alexa-64 7. Data in plots
`
`are gated on the dump channel-negative/CD3-positive cell population. To evaluate the
`
`presence of B cells, samples were co-stained with antibodies to CD14-APC and CD3-
`
`FITC (dump channels) and evaluated for the presence of B cells in the dump channel(cid:173)
`
`negative fraction by co-staining with antibodies to CD20-PE and CD19-PE-Cy-7. In all
`
`30
`
`cases, negative gate quadrants were established on no-stain controls as depicted in
`
`Figures 4B and 4C. T cell immunophenotyping of CD4+ (panel b) and CDS+ (panel c) T
`
`4
`
`Miltenyi Ex. 1019 Page 6
`
`

`

`cell subsets. Frozen peripheral blood samples from UPN 03 obtained by apheresis at day
`
`56 and 169 post T cell infusion were rested overnight in culture medium with no added
`
`factors, washed, and subjected to multi-parametric immunophenotyping for expression of
`
`markers of T cell memory, activation, and exhaustion. The gating strategy, as depicted in
`
`5
`
`Figure 8, involved an initial gating on dump channel (CD14, CD16, Live/Dead Aqua)(cid:173)
`
`negative and CD3-positive cells, followed by positive gates on CD4+ and CDS+ cells.
`
`Gates and quadrants were established using FMO controls (CAR, CD45RA, PD-1, CD25,
`
`CD127, CCR?) or by gating on positive cell populations (CD3, CD4, CDS) and clearly
`
`delineated subsets (CD27, CD28, CD57); data were displayed after bi-exponential
`
`10
`
`transformation for objective visualization of events. Figure 4D depicts functional
`
`competence of persisting CAR cells. Frozen peripheral blood samples from UPN 03
`
`obtained by apheresis at day 56 and 169 post T cell infusion were rested overnight in
`
`culture medium with no added factors, washed, and evaluated directly ex-vivo for the
`
`ability to recognize CD 19-expressing target cells using CD 107 de granulation assays.
`
`15
`
`Following a two-hour incubation in the presence of anti-CD2S, anti-CD49d, and CD 107-
`
`FITC, cell mixtures were harvested, washed, and subjected to multi-parametric flow
`
`cytometric analysis to evaluate the ability of CART19 cells to de-granulate in response to
`
`CD 19-expressing targets. The gating strategy involved an initial gate on dump channels
`
`(CD14-PE-Cy7, CD16-PE-Cy7, Live/Dead Aqua)-negative and CD3-PE-positive cells,
`
`20
`
`followed by gating on CDS-PE-Texas Red-positive cells; presented data is for the CDS+
`
`gated population. In all cases, negative gate quadrants were established on no-stain
`
`controls.
`
`Figure 5, comprising Figures 5A through 5C, is series of images depicting
`
`the results of experiments evaluating clinical responses after infusion ofCART19 cells.
`
`25
`
`Figure 5A depicts that UPN 02 was treated with two cycles ofrituximab and
`
`bendamustine with minimal response (RIB, arrow). CART19 T cells were infused
`
`beginning 4 days after bendamustine only (B, arrow). The rituximab and bendamustine(cid:173)
`
`resistant leukemia was rapidly cleared from blood, as indicated by a decrease in the
`
`absolute lymphocyte count (ALC) from 60,600/itl to 200/µl within 18 days of the
`
`30
`
`infusion. Corticosteroid treatment was staiied on day 18 post infusion due to malaise and
`
`non-infectious febrile syndrome. The reference line ( dotted) indicates upper limit of
`
`5
`
`Miltenyi Ex. 1019 Page 7
`
`

`

`normal for ALC. Figure 5B depicts the results of example experiments staining
`
`sequential bone marrow biopsy or clot specimens from patient UPN 01 and 03 for CD20.
`
`Pretreatment infiltration with leukemia present in both patients was absent on post
`
`treatment specimens accompanied by normalization of cellularity and trilineage
`
`5
`
`hematopoiesis. UPN 0 1 has not had any CLL cells detected as assessed by flow
`
`cytometry, cytogenetics and fluorescence in~situ hybridization or normal B cells detected
`
`by flow cytometry in bone marrow or blood. UPN 03 had 5% residual normal CDS(cid:173)
`
`negative B cells confirmed by flow cytometry on d +23, which also showed them to be
`
`polyclonal; no normal B cells were detected at day + 176. Figure SC depicts the results of
`
`10
`
`experiments using sequential CT imaging to assess the rapid resolution of chemotherapy(cid:173)
`
`resistant generalized lymphadenopathy. Bilateral axillary masses resolved by 83 (UPN
`
`01) and 31 (UPN 03) days post infusion, as indicated by arrows and circle.
`
`Figure 6, comprising Figures 6A through 6C, is a series of images
`
`depicting absolute lymphocyte counts and total CART19+ cells in circulation for UPN
`
`15
`
`01, 02, 03. The total number oflymphocytes (Total normal and CLL cells) vs. Total
`
`CART19+ cells in circulation is plotted for all 3 subjects using the absolute lymphocyte
`
`count from CBC values, and assuming a 5.0 L volume of blood, The total number of
`
`CART19 cells in circulation was calculated by using the tandem CBC values with
`
`absolute lymphocyte counts and the Q-PCR marking values as depicted in Figure 2,
`converting copies/µg DNA to average % marking as described elsewhere herein. The Q(cid:173)
`
`20
`
`PCR % marking was found to correlate closely ( <2 fold variation) with the flow
`
`cytometric characterization of the infusion products (see Methods) and with data from
`
`samples where concomitant flow cytometry data was available to directly enumerate
`
`CART19 cells by staining.
`
`25
`
`Figure 7, comprising Figures 7 A through 7D is a series of images
`
`depicting experiments involving the direct ex vivo detection of CART19-positive cells in
`
`UPN-01 PBMC 71 days post-T cell infusion. UPN-01 PBMC collected either fresh post(cid:173)
`
`apheresis on day71 day post infusion, or frozen at the time of apheresis for manufacture
`
`of the T cell product(baseline) and viably thawed prior to the staining, were subjected to
`
`30
`
`flow-cytometric analysis to detect the presence of CAR Tl 9 cells that express the CARI 9
`
`moiety on the surface. To evaluate the expression of CAR19 in lymphocytes, samples
`
`6
`
`Miltenyi Ex. 1019 Page 8
`
`

`

`were co-stained with CD3-PE and the anti-CAR19 idiotype antibody conjugated to
`
`Alexa-647, or co-stained with CD3-PE alone (FMO for CAR19). Figure 7A depicts that
`
`an initial lymphocyte gate was established based on forward and side scatter (FSC vs
`SSC), followed by gating on CD3+ cells. Figure 7B depicts CD3+ lymphocyte gate;
`
`5
`
`Figure 7C depicts CAR idiotype stain; Figure 7D depicts CAR idiotype FMO. The
`
`CAR19-positive gate was established on the CAR19 FMO samples.
`
`Figure 8, comprising Figures 8A through 8C, is a series of images
`
`depicting the gating strategy to identify CART19 expression by using polychromatic
`
`flow cytometry in UPN 03 blood specimens. The gating strategy for Figure 8C is shown
`
`10
`
`for the UPN 03 Day 56 sample and is representative of the strategy used on the UPN 03
`
`Day 169 sample. Figure 8A depicts primary gate: Dump (CD14, CD16, LIVE/dead
`
`Aqua) negative, CD3-positive. Figure 8B depicts secondary gates: CD4-positive, CD8-
`
`positive. Figure 8C depicts tertiary gates: CARI 9-positive and CARI 9-negative,
`
`established on CAR FMO samples (right-most panels).
`
`15
`
`Figure 9 depicts the gating strategy to directly identify CART19
`
`expression and B cells in blood and marrow specimens. The gating strategy for Figure
`
`4A, which shows detection of CAR-expressing CD3+ lymphocytes and absence of B
`
`cells in periphery and marrow: Leftplot: Cell gate; Upper panel: positive gate for CD3+
`cells, Lower panel: negative gate (CD14-negative, CD3-negative) for B cells. NC365,
`
`20
`
`peripheral blood control cells from a healthy donor
`
`Figure 10 is an image summarizing the patient demographics and
`
`response.
`
`DETAILED DESCRIPTION
`
`25
`
`The present invention relates generally to the treatment of a patient having
`
`Chronic Lymphocytic Leukemia (CLL) or at risk of having CLL using lymphocyte
`
`infusion. Preferably, autologous lymphocyte infusion is used in the treatment.
`
`Autologous PBMCs are collected from a patient in need of treatment and T cells are
`
`activated and expanded using the methods described herein and known in the art and then
`
`30
`
`infused back into the patient.
`
`7
`
`Miltenyi Ex. 1019 Page 9
`
`

`

`In some instances, the T cells are genetically modified prior to
`
`administering to a patient in need thereof, Preferably, the cell can be genetically
`
`modified to stably express an antibody binding domain on its surface, conferring novel
`
`antigen specificity that is MHC independent. Chimeric antigen receptors (CAR) are an
`
`5
`
`application of this approach that combines an antigen recognition domain of a specific
`
`antibody with an intracellular domain of the CD3-zeta chain or FcyRI protein into a
`
`single chimeric protein.
`
`In one embodiment, the invention includes autologous cells that are
`
`transfected with a vector comprising an aCD 19 CAR transgene. Preferably, the vector is
`
`10
`
`a retroviral vector. More preferably, the vector is a self-inactivating lentiviral vector as
`
`described elsewhere herein. For example, chime1ic antigen receptors (CAR) are an
`
`application of this approach that combines an antigen recognition domain of a specific
`
`antibody with an intracellular domain of the CD3-zeta chain or FcyRI protein into a
`
`single chimeric protein.
`
`15
`
`In one embodiment, the CAR T cells further express a 4-1 BB signaling
`
`domain. For example, the CART cells of the invention can be generated by introducing
`
`a lentiviral vector comprising aCD19, CD8a hinge and transmembrane domain, and
`
`human 4-lBB and CD3zeta signaling domains into the cells. CAR-modified T cells of
`
`the invention are able to replicate in vivo resulting in long-term persistence that can lead
`
`20
`
`to sustained tumor control.
`
`In various embodiments, the T cells administered to the patient, or their
`
`progeny, persist in the patient for at least four months, five months, six months, seven
`
`months, eight months, nine months, ten months, eleven months, twelve months, thirteen
`
`months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen
`
`25 months, nineteen months, twenty months, twenty-one months, twenty-two months,
`
`twenty-three months, two years, three years, four years, or five years after administration
`
`of the T cell to the patient.
`
`In yet another embodiment, the invention relates generally to the treatment
`
`of a patient at risk of developing CLL. For example, treating a malignancy or an
`
`30
`
`autoimmune disease in which chemotherapy and/or immunotherapy in a patient results in
`
`8
`
`Miltenyi Ex. 1019 Page 10
`
`

`

`significant immunosuppression in the patient that raises the risk of the patient of
`
`developing CLL.
`
`Definitions
`
`5
`
`Unless defined otherwise, all technical and scientific terms used herein
`
`have the same meaning as commonly understood by one of ordinary skill in the art to
`
`which the invention pertains. Although any methods and materials similar or equivalent
`
`to those described herein can be used in the practice for testing of the present invention,
`
`the preferred materials and methods are described herein. In describing and claiming the
`
`10
`
`present invention, the following terminology will be used.
`
`It is also to be understood that the terminology used herein is for the
`
`purpose of describing particular embodiments only, and is not intended to be limiting.
`
`The at1icles "a" and "an" are used herein to refer to one or to more than
`
`one (i.e., to at least one) of the grammatical object of the article. By way of example, "an
`
`15
`
`element" means one element or more than one element.
`
`"About" as used herein when referring to a measurable value such as an
`
`amount, a temporal duration, and the like, is meant to encompass variations of ±20% or
`
`±10%, more preferably ±5%, even more preferably ±1 %, and still more preferably ±0.1 %
`
`from the specified value, as such variations are appropriate to perform the disclosed
`
`20 methods.
`
`The term "antibody," as used herein, refers to an immunoglobulin
`
`molecule which specifically binds with an antigen. Antibodies can be intact
`
`immunoglobulins derived from natural sources or from recombinant sources and can be
`
`immunoreactive portions of intact immunoglobulins. Antibodies are typically tetramers
`
`25
`
`of immunoglobulin molecules. The antibodies in the present invention may exist in a
`
`variety of forms including, for example, polyclonal antibodies, monoclonal antibodies,
`
`Fv, Fab and F(ab)2, as well as single chain antibodies (scFv) and humanized antibodies
`(Harlow et al., 1999, In: Using Antibodies: A Laboratory Manual, Cold Spring Harbor
`
`Laboratory Press, NY; Harlow et al., 1989, In: Antibodies: A Laboratory Manual, Cold
`
`30
`
`Spring Harbor, New York; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-
`
`5883; Bird et al., 1988, Science 242:423-426).
`
`9
`
`Miltenyi Ex. 1019 Page 11
`
`

`

`The term "antigen" or "Ag" as used herein is defined as a molecule that
`
`provokes an immune response. This immune response may involve either antibody
`
`production, or the activation of specific immunologically-competent cells, or both. The
`
`skilled artisan will understand that any macromolecule, including virtually all proteins or
`
`5
`
`peptides, can serve as an antigen. Furthermore, antigens can be derived from
`
`recombinant or genomic DNA. A skilled artisan will understand that any DNA, which
`
`comprises a nucleotide sequences or a partial nucleotide sequence encoding a protein that
`
`elicits an immune response therefore encodes an "antigen" as that term is used herein.
`
`Furthermore, one skilled in the art will understand that an antigen need not be encoded
`
`10
`
`solely by a full length nucleotide sequence of a gene. It is readily apparent that the
`
`present invention includes, but is not limited to, the use of partial nucleotide sequences of
`
`more than one gene and that these nucleotide sequences are arranged in various
`
`combinations to elicit the desired immune response. Moreover, a skilled artisan will
`
`understand that an antigen need not be encoded by a "gene" at all. It is readily apparent
`
`15
`
`that an antigen can be generated synthesized or can be derived from a biological sample.
`
`Such a biological sample can include, but is not limited to a tissue sample, a tumor
`
`sample, a cell or a biological fluid.
`
`The term "anti-tumor effect" as used herein, refers to a biological effect
`
`which can be manifested by a decrease in tumor volume, a decrease in the number of
`
`20
`
`tumor cells, a decrease in the number of metastases, an increase in life expectancy, or
`
`amelioration of various physiological symptoms associated with the cancerous condition.
`
`An "anti-tumor effect" can also be manifested by the ability of the peptides,
`
`polynucleotides, cells and antibodies of the invention in prevention of the occurrence of
`
`tumor in the first place.
`
`25
`
`As used herein, the term "autologous" is meant to refer to any material
`
`derived from the same individual to which it is later to be re-introduced into the
`
`individual.
`
`species.
`
`"Allogeneic" refers to a graft derived from a different animal of the same
`
`30
`
`"Xenogeneic" refers to a graft derived from an animal of a different
`
`species.
`
`10
`
`Miltenyi Ex. 1019 Page 12
`
`

`

`The term "cancer" as used herein is defined as disease characterized by the
`
`rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or
`
`through the bloodstream and lymphatic system to other parts of the body. Examples of
`
`various cancers include but are not limited to, breast cancer, prostate cancer, ovarian
`
`5
`
`cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer,
`liver cancer, brain cancer, lymphoma, leukemia, lung cancer and the like.
`
`"Encoding" refers to the inherent prope1iy of specific sequences of
`
`nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as
`
`templates for synthesis of other polymers and macromolecules in biological processes
`having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a
`
`10
`
`defined sequence of amino acids and the biological prope1iies resulting therefrom. Thus,
`a gene encodes a protein if transcription and translation of mRNA corresponding to that
`
`gene produces the protein in a cell or other biological system, Both the coding strand, the
`
`nucleotide sequence of which is identical to the mRNA sequence and is usually provided
`
`15
`
`in sequence listings, and the non-coding strand, used as the template for transcription of a
`
`gene or cDNA, can be referred to as encoding the protein or other product of that gene or
`
`cDNA.
`
`Unless otherwise specified, a "nucleotide sequence encoding an amino
`
`acid sequence" includes all nucleotide sequences that are degenerate versions of each
`
`20
`
`other and that encode the same amino acid sequence. Nucleotide sequences that encode
`
`proteins and RNA may include intrans.
`
`"Effective amount" or "therapeutically effective amount" are used
`
`interchangeably herein, and refer to an amount of a compound, formulation, material, or
`
`composition, as described herein effective to achieve a particular biological result. Such
`
`25
`
`results may include, but are not limited to, the inhibition of virus infection as determined
`
`by any means suitable in the art.
`
`As used herein "endogenous" refers to any material from or produced
`
`inside an organism, cell, tissue or system.
`
`As used herein, the term "exogenous" refers to any material introduced
`
`30
`
`from or produced outside an organism, cell, tissue or system.
`
`11
`
`Miltenyi Ex. 1019 Page 13
`
`

`

`The term "expression" as used herein is defined as the transcription and/or
`
`translation of a particular nucleotide sequence driven by its promoter.
`
`"Expression vector" refers to a vector comprising a recombinant
`
`polynucleotide comprising expression control sequences operatively linked to a
`
`5
`
`nucleotide sequence to be expressed. An expression vector comprises sufficient cis(cid:173)
`
`acting elements for expression; other elements for expression can be supplied by the host
`
`cell or in an in vitro expression system. Expression vectors include all those known in
`
`the aii, such as cosmids, plasmids (e.g., naked or contained in liposomes) and virnses
`
`(e.g., lentivirnses, retroviruses, adenovirnses, and adeno-associated viruses) that
`
`10
`
`incorporate the recombinant polynucleotide.
`
`"Homologous" as used herein, refers to the subunit sequence identity
`
`between two polymeric molecules, e.g., between two nucleic acid molecules, such as, two
`
`DNA molecules or two RNA molecules, or between two polypeptide molecules. When a
`
`subunit position in both of the two molecules is occupied by the same monomeric
`
`15
`
`subunit; e.g., if a position in each of two DNA molecules is occupied by adenine, then
`
`they are homologous at that position. The homology between two sequences is a direct
`
`fimction of the number of matching or homologous positions; e.g., if half (e.g., five
`
`positions in a polymer ten subunits in length) of the positions in two sequences are
`
`homologous, the two sequences are 50% homologous; if 90% of the positions (e.g., 9 of
`
`20
`
`10), are matched or homologous, the two sequences are 90% homologous.
`
`As used herein, an "instructional material" includes a publication, a
`
`recording, a diagram, or any other medium of expression which can be used to
`
`communicate the usefulness of the compositions and methods of the invention. The
`
`instrnctional mate1ial of the kit of the invention may, for example, be affixed to a
`
`25
`
`container which contains the nucleic acid, peptide, and/or composition of the invention or
`
`be shipped together with a container which contains the nucleic acid, peptide, and/or
`
`composition. Alternatively, the instructional material may be shipped separately from the
`
`container with the intention that the instructional material and the compound be used
`
`cooperatively by the recipient.
`
`30
`
`"Isolated" means altered or removed from the natural state. For example,
`
`a nucleic acid or a peptide naturally present in a living animal is not "isolated," but the
`
`12
`
`Miltenyi Ex. 1019 Page 14
`
`

`

`5
`
`10
`
`15
`
`20
`
`25
`
`same nucleic acid or peptide partially or completely separated from the coexisting
`materials of its natural state is "isolated." An isolated nucleic acid or protein can exist in
`substantially purified fonn, or can exist in a non-native environment such as, for
`example, a host cell.
`
`In the context of the present invention, the following abbreviations for the
`commonly occurring nucleic acid bases are used. "A" refers to adenosine, "C" refers to
`cytosine, "G" refers to guanosine, "T" refers to thymidine, and "U" refers to uridine.
`Unless otherwise specified, a "nucleotide sequence encoding an amino
`acid sequence" includes all nucleotide sequences that are degenerate versions of each
`other and that encode the same amino acid sequence. The phrase n

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