throbber
8224035
`
`UNITED STATES DEPARTMENT OF COJV[M:ERCE
`
`United Stntcs Pntcnt nnd Trndcmnrk 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: 61/421,470
`FILING DATE: December 09, 2010
`
`Certified by
`
`Under Secretary of Commerce
`for Intellectual Property
`and Director of the United States
`Patent and Trademark Office
`
`Miltenyi Ex. 1020 Page 1
`
`

`

`046483-6001-Pl-US (600640)
`
`TITLE OF THE INVENTION
`Compositions and Methods for Treatment of Chronic Lymphocytic Leukemia
`
`5
`
`STATEMENT REGARDING FEDERALL YSPONSORED RESEARCH OR
`DEVELOPMENT
`This invention was made with U.S. government support under Grant Nos.
`1RO1CA105216, RO1AI057838 and ROl 1113482 awarded by the National Institutes of
`Health (NIH). The government has certain rights in the invention.
`
`10
`
`15
`
`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,
`20 Molecular Therapy, published online Febrnary 23, 2010, pages 1-9; and, Till et al., 2008,
`Blood, 112:2261-2271.
`, Thus, there is an urgent need in the art for compositions and methods for
`treatment of CLL, The present invention addresses this need.
`
`25
`
`30
`
`1
`
`Miltenyi Ex. 1020 Page 2
`
`

`

`DETAILED DESCRIPTION
`The present invention provides compositions and methods for treatment of CLL.
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`Compositions
`As exemplified elsewhere herein, the present invention includes a vector
`comprising an crCD19 CAR transgene, Preferably, the vector is a retroviral vector. More
`preferably, the vectm· is a self-inactivating lentiviral vector as described elsewhere herein.
`The invention also includes autologous cells that are transfected with the vector of
`the invention. Preferably, the cells are T cells and more preferably, they are autologous T
`cells.
`
`Methods of Treating
`As described in detail elsewhere herein, the invention includes a method of
`treating an patient with CLL, wherein the method comprises administering to the patient
`autologous T cells that have been transfected so as to express a CD 19 CAR antigen.
`Preferably, the cells are administered to the patient by infusion.
`The precise protocols used to treat patients are described elsewhere herein and can
`also be found at clinicaltrials.dot.gov under trial no NCT01029366. Interim results from
`this study, where three patients were treated, establish that successful expansion and
`.transduction of T cells was accomplished in all three patients. The actual manufacturing
`of final product was more difficult in CLL patients than in previous trials, CARs with 4-
`1 BB:z signaling domains have massive expansion in vivo in two of three patients with
`advanced CLL. The cells persisted in blood and migrated to bone marrow for at least
`sixty days in substantial numbers post-infusion. CART cells expanded in vivo compared
`to the infused amount. Anti-tumor effects were observed: patient 1 CR, patient 2 PR,
`patient 3 CRn, And) the treatment was promising in chemotherapy refractory patients.
`
`Definitions
`Unless defined otherwise, all technical and scientific tem1s used herein have the
`same meaning as conunonly understood by one of ordinary skill in the at1 to which this
`invention belongs. Although any methods and materials similar or equivalent to those
`
`2
`
`Miltenyi Ex. 1020 Page 3
`
`

`

`5
`
`15
`
`described herein can be used in the practice or testing of the present invention, the
`preferred methods and materials are described.
`As used herein, each of the following terms has the meaning associated with it in
`this section.
`The articles "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 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%,
`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 methods,
`The term "antibody" as used herein, refers to an immunoglobulin molecule, which
`is able to specifically bind to a specific epitope on an antigen, Antibodies can be intact
`immunoglobulins derived from natural sources or from recombinant sources and can be
`immunoactive portions of intact immunoglobulins. Antibodies are typically tetramers 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 and humanized antibodies.
`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 vittually all proteins or peptides, can
`serve as an antigen. Fmihermore, 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 can encode an ''antigen" as that term is used herein. Furthermore, one skilled in
`tl}e art will understand that an antigen need not be encoded 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 can be arranged in various combinations to elicit the
`desired immune response. Moreover, a skilled artisan will understand that an antigen
`
`20
`
`25
`
`30
`
`3
`
`Miltenyi Ex. 1020 Page 4
`
`

`

`5
`
`IO
`
`15
`
`20
`
`25
`
`need not be encoded by a "gene" at all. It is readily apparent 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,
`A "coding region" of a gene consists of the nucleotide residues of the coding
`strand of the gene and the nucleotides of the non-coding strand of the gene which are
`homologous with or complementary to, respectively, the coding region of an mRNA
`molecule which is produced by transcription of the gene.
`A "coding region'' of an mRNA molecule also consists of the nucleotide residues
`of the mRN A molecule which are matched with an anti-codon region of a transfer RNA
`molecule during translation of the mRNA molecule or which encode a stop codon. The
`coding region may thus include nucleotide residues corresponding to amino acid residues
`which are not present in the mature protein encoded by the mRNA molecule (e.g., amino
`acid residues in a protein expo1t signal sequence).
`"Encoding" refers to the inherent property 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 defined sequence of
`amino acids and the biological properties resulting therefrom. Thus, a gene encodes a
`protein if transcdption and translation of mRN A corresponding to that gene produces the
`protein in a cell or other biological system. Both the coding strand, the nucleotide
`sequenc~ of which is identical to the mRNA sequence and is usually provided 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 other
`and that encode the same amino acid sequence. Nucleotide sequences that encode
`proteins and RNA may include introns.
`
`4
`
`Miltenyi Ex. 1020 Page 5
`
`

`

`A "disease" is a state of health of an animal, preferably a mammal and more
`preferably, a human, wherein the animal cannot maintain homeostasis, and wherein if the
`disease is not ameliorated then the animal's health continues to deteriorate,
`In contrast, a "disorder" in· an animal is a state of health in which the animal is
`able to maintain homeostasis, but in which the animal's state of health is less favorable
`than it would be in the absence of the disorder. Left untreated, a disorder does not
`necessarily cause a further decrease in the animal's state of health,
`A disease or disorder is "alleviated" if the severity of a symptom of the disease or
`disorder, the frequency with which such a symptom is experienced by a patient, or both,
`is reduced,
`An "effective amount" or "therapeutically effective amount" of a compound is
`that amount of compound which is sufficient to provide a beneficial effect to the subject
`to which the compound is administered. An "effective amount" of a delivery vehicle is
`that amount sufficient to effectively bind or deliver a compound.
`As used herein, the term "fragment," as applied to a nucleic acid, refers to a
`subsequence of a larger nucleic acid, A "fragment" of a nucleic acid can be at least about
`15 nucleotides in length; for example, at least about 50 nucleotides to about 100
`nucleotides; at least about 100 to about 500 nucleotides, at least about 500 to about 1000
`nucleotides, at least about 1000 nucleotides to about 1500 nucleotides; or about 1500
`nucleotides to about 2500 nucleotides; or about 2500 nucleotides (and any integer value
`in between).
`As used herein, the term Hfragment," as applied to a protein or peptide, refers to a
`subsequence of a larger protein or peptide, A "fragment" of a protein or peptide can be at
`least about 20 amino acids in length; for example at least about 50 amino acids in length;
`at least about 100 amino acids in length, at least about 200 amino acids in length, at least
`about 300 amino acids in length, and at least about 400 amino acids in length (and any
`integer value in between).
`The term "inununoglobulin" or "lg", as used herein is defined as a class of
`proteins, which function as antibodies. The five members included in this class of
`proteins are IgA, lgG, IgM, lgD, and IgE. IgA is the primary antibody that is present in
`body secretions, such as saliva, tears, breast milk, gastrointestinal secretions and mucus
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`5
`
`Miltenyi Ex. 1020 Page 6
`
`

`

`secretions of the respiratory and genitourinary tracts. IgG is the most common
`circulating antibody. IgM is the main immunoglobulin produced in the primary immune
`response in most mammals. It is the most efficient immunoglobulin in agglutination,
`complement fixation, and other antibody responses, and is important in defense against
`bacteria and viruses. IgD is the immunoglobulin that has no known antibody function,
`but may serve as an antigen receptor. IgE is the immunoglobulin that mediates
`immediate hypersensitivity by causing release of mediators from mast cells and basophils
`upon exposure to allergen.
`"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
`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 form, or can exist in a non-native environment such as, for
`example, a host cell.
`"Naturally occurring" as used herein describes a composition that can be found in
`nature as distinct from being mtificially produced. For example, a nucleotide sequence .
`present in an organism, which can be isolated from a source in nature and which has not
`been intentionally modified by a person in the laboratory, is naturally occurring.
`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 nucleotide sequence that
`encodes a protein or an RNA may also include introns to the extent that the nucleotide
`sequence encoding the protein may in some version contain an intrnn(s).
`The term "polynucleotide" as used herein is defined as a chain of nucleotides.
`Furthermore, nucleic acids are polymers of nucleotides. Thus, nucleic acids and
`polynucleotides as used herein are interchangeable. One skilled in the art has the general
`knowledge that nucleic acids are polynucleotides, which can be hydrolyzed into the
`monomeric "nucleotides." The monomeric nucleotides can be hydrolyzed into
`nucleosides. As used herein polynucleotides include, but are not limited to, all nucleic
`acid sequences which are obtained by any means available in the art, including, without
`limitation, recombinant means, i.e., the cloning of nucleic acid sequences from a
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`6
`
`Miltenyi Ex. 1020 Page 7
`
`

`

`5
`
`10
`
`15
`
`20
`
`25
`
`recombinant library or a cell genome, using ordinary cloning technology and PCRTri\ and
`the like, and by synthetic means.
`As used herein, the terms "peptide," "polypeptide," and "protein" are used
`interchangeably, and refer to a compound comprised of amino acid residues covalently
`linked by peptide bonds. A protein or peptide must contain at least two amino acids, and
`no limitation is placed on the maximum number of amino acids that can comprise a
`protein's or peptide's sequence. Polypeptides include any peptide or protein comprising
`two or more amino acids joined to each other by peptide bonds. As used herein, the term
`refers to both short chains, which also commonly are referred to in the art as peptides,
`oligopeptides and oligomers, for example, and to longer chains, which generally are
`referred to in the art as proteins, of which there are many types. "Polypeptides" include,
`for example, biologically active fragments, substantially homologous polypeptides,
`oligopeptides, homodimers, heterodimers, variants of polypeptides, modified
`polypeptides, derivatives, analogs, fusion proteins, among others. The polypeptides
`include natural peptides, recombinant peptides, synthetic peptides, or a combination
`thereof.
`The terms "patientt "subject," "individualt and the like are used interchangeably
`herein, and refer to any animal, or cells thereof whether in vitro or in situ, amenable to
`the methods described herein. Preferably, the patient, subject or individual is a mammal,
`and more preferable, a human.
`The term "treatmenf' as used within the context of the present invention is meant
`to include therapeutic treatment as well as prophylactic, or suppressive measures for the
`disease or disorder. Thus, for example, the term treatment includes the administration of
`an agent prior to or following the onset of a disease or disorder thereby preventing or
`removing all signs of the disease or disorder. As another example, administration of the
`agent after clinical manifestation of the disease to combat the symptoms of the disease
`comprises "treatment" of the disease.
`A "therapeutic" treatment is a treatment administered to a subject who exhibits
`signs of pathology, for the purpose of diminishing or eliminating those signs.
`
`7
`
`Miltenyi Ex. 1020 Page 8
`
`

`

`As used herein, "treating a disease or disorder" means reducing the frequency
`with which a symptom of the disease or disorder is experienced by a patient. Disease and
`disorder are used interchangeably herein.
`"Variant" as the term is used herein, is a nucleic acid sequence or a peptide
`sequence that differs in sequence from a reference nucleic acid sequence or peptide
`sequence respectively, but retains essential propet1ies of the reference molecule.
`Changes in the sequence of a nucleic acid variant may not alter the amino acid sequence
`of a peptide encoded by the reference nucleic acid, or may result in amino acid
`substitutions, additions, deletions, fusions and truncations. Changes in the sequence of
`peptide variants are typically limited or conservative, so that the sequences of the
`reference peptide and the variant are closely similar overall and, in many regions,
`identical. A variant and reference peptide can differ in amino acid sequence by one or
`more substitutions, additions, deletions in any combination. A variant of a nucleic acid
`or peptide can be a naturally occurring such as an allelic variant, or can be a variant that
`is not known to occur naturally. Non-naturally occurring variants of nucleic acids and
`peptides may be made by mutagenesis techniques or by direct synthesis.
`For sequence comparison, typically one sequence acts as a reference sequence, to
`which test sequences may be compared. When using a sequence comparison algorithm,
`test and reference sequences are input into a computer, subsequent coordinates are
`designated, if necessary, and sequence algorithm program parameters are designated. The
`sequence comparison algorithm then calculates the percentage sequence identity for the
`test sequence(s) relative to the reference sequence, based on the designated program
`parameters. ·
`As used herein, "vaccination" is intended for prophylactic or therapeutical
`vaccination.
`Ranges: throughout this disclosure, various aspects of the invention can be
`presented in a range format. It should be understood that the description in range format
`is merely for convenience and brevity and should not be construed as an inflexible
`limitation on the scope of the invention. Accordingly, the description of a range should be
`considered to have specifically disclosed all the possible subranges as well as individual
`numerical values within that range. For example, description of a range such as from 1 to
`
`5
`
`IO
`
`15
`
`20
`
`25
`
`30
`
`8
`
`Miltenyi Ex. 1020 Page 9
`
`

`

`6 should be considered to have specifically disclosed subranges such as from 1 to 3, from
`1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual
`numbers within that range, for example, 1, 2 1 2.7 1 3 1 4, 5, 5.3, and 6. This applies
`regardless of the breadth of the range.
`
`EXPERIMENT AL EXAMPLES
`The invention is further described in detail by reference to the following
`experimental examples. These examples are provided for purposes of illustration only,
`and are not intended to be limiting unless otherwise specified. Thus, the invention should
`in no way be construed as being limited to the following examples, but rather, should be
`construed to encompass any and all variations which become evident as a result of the
`teaching provided herein.
`Without fmiher description, it is believed that one of ordinary skill in the
`ai1 can, using the preceding description and the following illustrative examples, make and
`utilize the compounds of the present invention and practice the claimed methods. The
`following working examples therefore, specifically point out the preferred embodiments
`of the present invention, and are not to be construed as limiting in any way the remainder
`of the disclosure.
`
`5
`
`10
`
`15
`
`9
`
`Miltenyi Ex. 1020 Page 10
`
`

`

`Example I: Chimeric Receptors Containing CD 13 7 Signal Transduction Domains
`
`Mediate Enhanced Survival of T Cells and Increased Anti"Leukemic Efficacy In Vivo
`
`10
`
`Miltenyi Ex. 1020 Page 11
`
`

`

`Abstract
`
`Persistence of T cells engineered with chimeric antigen receptors (CARs) has been a
`
`major barrier to use of these cells for molecularly targeted adoptive immunotherapy. To
`
`address this issue, we created a series of CARs that contain the TCR-s signal
`
`transduction domain with the CD28 and/or CD137 (4-lBB) intracellular domains in
`
`tandem. After short-term expansion, primary human T cells were subjected to lentiviral
`
`gene transfer, resulting in large numbers of cells with >85% CAR expression. In an
`
`immunodeficient mouse xenograft model of primary human pre-B-cell acute
`
`lymphoblastic leukemia, human T cells expressing anti-CDI9 CARs containing CD137
`
`exhibited the greatest anti-leukemic efficacy and prolonged (>6 months) survival in vivo,
`
`and were significantly more effective than cells expressing CARs containing TCR-l;
`
`alone or CD28-s signaling receptors, We uncovered a previously unrecognized, antigen(cid:173)
`
`independent effect of CARs expressing the .CD 13 7 cytoplasmic domain that likely _
`
`contributes to the enh_anced antileukemic efficacy and survival in tumor bearing mice.
`
`Furthermore, our studies revealed significant discrepancies between in vitro and in vivo
`
`surrogate measures of CAR efficacy. Together these results suggest that incorporation of
`
`the CD 137 signaling domain in CARs should improve the persistence of CAR.s in the
`
`tumor microenvironment and hence maximize their anti tumor activity.
`
`Miltenyi Ex. 1020 Page 12
`
`

`

`With the advent of efficient gene transfer technologies, such as murine retroviral
`
`and HIV-derived lentiviral vectors, it has become feasible to confer novel antigenic
`
`specificity to T cells by transfer of chimeric antigen receptors (CARs) with stable, long(cid:173)
`
`tenn expression. This technology has been used to generate T cells specific for HIV and
`
`several human tumor antigens, and some of these engineered T cells have been tested in
`
`Phase I/II studies in humans demonstrating the feasibility and relative safety of this
`
`approach [1-3]. One study has demonstrated anti-tumor activity in patients with
`
`neuroblastoma given a single CAR infusion [4].
`
`CARs combine the antigen recognition domain of antibody ~th the intracellular
`
`· domain of the TCR-t;, chain or FcyRI protein into a single chimeric protein that are
`
`capable of triggering T-cell activation in a manner very similar to that of the endogenous
`
`TCR [5, 6]. Several studies de1nonstrate that the addition of co-stimulatory domains,
`
`particularly the intracellular domain of CD28 can significantly augment the ability of
`
`these receptors to stimulate cytokine secretion and enhance antitumor•efficacy in pre(cid:173)
`
`clinical animal models using both solid tumors and leukemia that lack the expression of
`
`the CD28 receptor ligands CD80 and CD86 [7-9]. Inclusion of domains from receptors
`
`such as the TNF receptor family members, CD134(OX-40) and CDI37 (4-lBB) into
`
`CARs has also been shown to augment CAR-mediated T-cell responses [10,11]. Gene
`
`transfer approaches using these engineered CARs may therefore provide significant
`
`improvements over current adoptive immunotherapy strategies that must rely upon the
`
`endogenous TCR specificities, for which significant issues of TCR repertoire limitation
`
`and impaired tumor MHC class I expression may exist.
`
`Miltenyi Ex. 1020 Page 13
`
`

`

`In this study, we have addressed the issue of limited in vivo persistence of CARs
`
`by defining the relative contributions ofTCR-s, CD137 and CD28 signaling domains in
`
`mice engrafted with hematopoietic malignancies. We chose the human CD 19 antigen as
`
`our initial target for several reasons: 1) CD19 displays a pattern of expression that is
`
`highly restricted to B-cells and B-cell progenitor cells [12], 2) CD 19 does not appear to
`
`be expressed by hematopoietic stem cells permitting the targeting of the B-cell lineage
`
`without affecting other hematopoietic lineages (13], and 3) CD 19 is widely expressed by
`
`malignant cells that are derived from the B cell lineage including most lymphomas and
`
`lymphocytic leukemias (14]. After optimizing the generation of CARs with an efficient
`
`T-cell culture process, in vitro studies indicate that inc01poration of either CD28 or 4-
`
`IBB signaling domains enhances activity over TCR--s, confirming previous studies. In
`
`contrast, compared to CARs that contain CD28, our in vivo studies indicate that CARs
`
`containing CD 137 have superior antileukemic efficacy and improved persistence in a
`
`primary human acute lymphoblastic leukemia xenograft model. Furthermore, we also
`
`find that CARs expressing CD137 signaling domains can provide significant activity that
`
`appears to be antigen independent and may contribute to the efficacy of CARs in vivo.
`
`Results
`
`E/ficie11t generation CAR. T cells using artificial bead-based APCs and lentivira/ gene .
`
`transfer
`
`Lentiviral vectors can transfer genes with into activated CD4+ and CDS+ human
`
`T cells with high efficiency but_ expression of the vector-encoded transgene depends on
`
`the internal promoter that drives its transcription. Therefore, successful CAR expression
`
`Miltenyi Ex. 1020 Page 14
`
`

`

`and gene therapies with CAR-expressing T cells rely on the ability ofT cells to maintain
`
`adequate receptor expression over long periods of time, We tested several promoters to
`
`identify the one with the highest stable expression in both primary CD4+ and CD8+ T
`
`cells. Transduction was perfonned at limiting dilution to ensure that the cells have a
`
`single integrated vector per cell (data not shown). While the CMV promoter exhibited
`
`high levels of expression of a GFP transgene early after transduction, expression .
`
`decreased to < 25% of the initial expression after 10 days of culture (Fig. 1 b ). The
`
`distribution of CMV-driven GFP expression was also quite variable compared with the
`
`other promoters tested (Suppl. Fig. S1). In contrast, the EF-la promoter not only
`
`induced the highest level of GFP expression but also optimally maintained it in both CD4
`
`cells and CD8 cells (Fig. 1b). These findings confirmed and extended other studies in
`
`primary human T cells [15]. The EF-la promoter was therefore selected for all future
`
`studies using CARs. By using lentiviral vectors and transductions at an MOI of 5, the
`
`different CARs could be expressed high expression in > 85% primary human T cells
`
`(Fig. le). Western blotting under both reducing and non-reducing conditions
`
`demonstrated that the CARs are present as both covalent dimers and monomers within T
`
`cells (Suppl. Fig. S2). Using the artificial bead-based APC system previously described
`
`by our laboratory [16], > 50-fold expansion of CAR+ T cells could be achieved over the
`
`course of transduction and growth in~ 10 days (Fig. ld).
`
`Fu11ctio11al characterization of anti-CD19 CAR-expressing primary ltuma11 T cells
`
`To enhance the functionality of the immunoreceptor, we introduce the signal
`
`transduction domains of CD28 or CD 13 7 in the TCR-l; containing CAR (Fig. 1 a).
`
`Similar to data reported by other groups [11,17], the introduction of costimulatory
`
`Miltenyi Ex. 1020 Page 15
`
`

`

`domains into CARs does not improve the antigen-specific cytotoxicity triggered by these
`
`receptors (Fig. 2b). Lytic activity of transduced T cells against K562 target cells
`
`expressing CD 19 correlated with the transduction efficiency of the T cells ( data not
`
`shown). CAR-triggered cytotoxicity is antigen-specific with only negligible lysis ofwild(cid:173)
`
`type K562 cells that lack expression of the CD19 antigen (Fig. 2a). CAR+ T cells are
`
`also able to efficiently kill primary pre-BALL cells that express physiologic levels of
`
`CD19 (Fig. 2b). Of note, these primary ALL cells lack expression of endogenous CO80
`
`or CD86 ( data not shown).
`
`Following CAR activation with CD19+ K562 cells, CD4+ T cells expressing
`
`CARs produced abundant quantitiesofIL-2 and IFN-y (Fig. 3) comparable to cells
`
`stimulated via the endogenous TCR and CD28 receptors (data not shown). T cells
`
`expressing CD28 and CD 13 7 domain containing CARs produced greater quantities of IL-
`
`2 when compared with cells expressing the aCD19-<'.; receptor (Fig. 3). The production of
`
`the type 2 cytokines, IL-4 and IL- I 0, by CD4+ T cells was also stimulated by all of the
`
`CARs tested; however, the levels of these cytokines were much.lower, consistent with the
`
`Th I-like phenotype ofT cells generated by anti-CD3 and CD28 stimulatory beads [16). It
`
`was notable that the incorporation of the CD 13 7 domain into CARs decreased the
`
`production of these type 2 cytokines, consistent with previous repo1is of the 4~ lBB
`
`signaling pathway .in natural T cells [18]. All CARs stimulated IFN-y production by
`
`CDS+ T cells. These findings confirm that the addition of co-stimulatory domains into
`
`CARs modulates cytokine secretion in a manner that is dependent on the type of
`
`costimulatory domain [7,S,17,19]. However, it is less well appreciated that the pattern of
`
`cytokine expression is altered by incorporation of different signal transduction domains
`
`Miltenyi Ex. 1020 Page 16
`
`

`

`into the CARs. These differences may have imp011ant consequences for the functionality
`
`ofT cells engineered to express CARs.
`
`The effects of costimulatory domains on CAR-driven T cell proliferation
`
`The generation of a robust and sustained anti-twnor immune response requires not
`
`only triggering of cytotoxicity and cytokine production but also stimulation of T-cell
`
`proliferation. To assess the relative contribution of different costimulatory domains to
`
`proliferative signals delivered by CARs~ we engineered primary human T cells to express
`
`CARs in conjunction with GFP to permit evaluation of both CAR+ and CAR"T cells in
`
`the same culture. Following T-cell re-stimulation with CDI9+ K562 (K562-CD19 cells))
`
`T cells expressing the a CD 19-28-l; receptor exhibited proliferation comparable to that
`
`obtained with full stimulation of the endogenous TCR complex with K562 cells loaded
`
`with anti-CD3 and CD28 antibodies~ a condition shown previously to support long-term
`
`expansion of primary human T cells (KT32-BBL) [20] (Fig, 4a[v]). The aCD19-28-BB(cid:173)
`
`S triple receptor also stimulated CD19 driven proliferation (Fig. 4a[iv]), but to a lesser
`
`extent that the aCD19-28-l; double costimulatory receptor. No significant proliferation
`
`was observed when these same T cells. were stimulated with wild-type K562 cells lacking
`
`the CD19 antigen (K.562 wt). As previously shown by other investigators [17,19)21)22),
`
`T cells expressing the a CD 19...t;, receptor showed little proliferation upon exposure to the.
`
`surrogate CD 19 antigen (Fig. 4a[ii]), demonstrating the dependence of CAR-driven
`
`proliferation on co-stimulatory signals.
`
`Unexpectedly, T cells containing the aCD 19-BB-s double costimulatocy domain
`
`CAR had significantly increased proliferative capacity during in vitro expansion
`
`Miltenyi Ex. 1020 Page 17
`
`

`

`independently of receptor ligation with the surrogate CD19 antigen (Fig. 4a[iii] and 4b).
`
`This increased proliferation was obse1ved in both CD4+ and CD8+ T cells (data not
`
`shown), and it was associated with a prolonged blast phase after the initial stimulation
`
`and transduction, as revealed by a longer maintenance of an elevated mean cellular
`
`volume (Fig. 4c), a parameter that correlates well with log phase proliferation ofT cells
`
`[16]. These findings suggest that incorporation of the CD137 intracellular domain
`
`mediates antigen-independent activity that is similar to that provided by the natural 4-
`
`lBB receptor in T cells following ligation [23]. As a result of the enhanced proliferation
`
`obse1ved following the initial activation of T cells via aCD3/aCD28-coated beads used
`
`to enhance T-cell transduction [24,25] (Fig 4b), CAR+ T cells expressing the aCD19-
`
`BB-½ receptor had relatively low CAR-driven proliferation (Fig. 4a[iii]).
`
`Evaluating anti-tumor responses of CAR+ human primary T cells in vivo
`
`Other than the antigen-in.dependent proliferation of the 4-lBB containing CAR,
`
`the above in vitro findings, in aggregate, suggested that the aCD19-28-l; CAR would be
`
`the most effective receptor for generating a sustained anti-leukemic T_cell response in
`
`vivo. We evaluated the in vivo efficacy of aCD19 CARs in vivo model of ALL (Fig. 5a)
`
`in which primary human pre-BALL cel

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