METHOD FOR PREPARING GENETICALLY-MODIFIED T CELLS WHICH EXPRESS CHIMERIC ANTIGEN RECEPTOR

Provided is a method for preparing genetically-modified T cells expressing chimeric antigen receptor, comprising: (i) a step of preparing non-proliferative cells holding a viral peptide antigen, which are obtained by stimulating a group of cells comprising T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability; (ii) a step of obtaining genetically-modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced using a transposon method; (iii) a step of mixing the non-proliferative cells prepared by step (i) with the genetically-modified T cells obtained by step (ii), and co-culturing the mixed cells; and (iv) a step of collecting the cells after culture.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
TECHNICAL FIELD

The present invention relates to a method for preparing genetically-modified T cells which express chimeric antigen receptor, and uses therefor. The present application claims priority based on Japanese Patent Application No. 2015-200458 filed on Oct. 8, 2015 and the content of the patent application is hereby incorporated by reference herein in its entirety.

BACKGROUND ART

Gene-modified T-cell therapy (CAR therapy) using a chimeric antigen receptor (hereinafter may be referred to as “CAR”) find more and more clinical application. A CAR typically has a structure composed of a single chain variable region of an antibody as the extracellular domain, to which linked are a transmembrane region, CD3ξ, and an intracellular domain of a molecule which transmits costimulatory signals. The CAR-T cells are activated by binding to the antigen according to specificity of the antibody, and injures the target cells (for example, cancer cells). CAR therapy has advantages such as relatively easy cell preparation, high cytotoxic activity, and sustainable effect, and thus is expected as a new treatment means for refractory subjects and subjects having resistance to conventional therapy. In the actual clinical trials carried out in Europe and the United States, the CAR for the CD19 antigen expressed on the cell surface was gene-introduced into the peripheral blood T-cells collected from patients with chemotherapy-resistant acute lymphoblastic leukemia, cultured, and infused; satisfactory results with a remission rate of 80 to 90% was reported (Grupp S A et al., N Engl J Med, 368(16): 1509-18. 2013; Maude S L et al., N Engl J Med, 371(16): 1507-17.2014; Lee D W et al., Lancet. 2015 Feb. 7; 385(9967): 517-28). In the United States, CAR therapy has been attracting attention as one of the most promising therapies for refractory cancer.

In prior art, the cells used in CAR therapy (CAR-T cells) are prepared using viral vectors. However, the commonly used retroviruses have problem of safety, because the frequency of insertion mutation to proto-oncogene is high (leukemia frequently occurs in gene therapy using hematopoietic stem cells). In addition, in the prior art method, cell lines and fetal bovine serum are used for culturing, so that there is a fear about long-term safety particularly in pediatric patients. Furthermore, the use of a viral vector requires special equipment for cell culturing, which highly increases the treatment cost and causes problem of cost efficiency (Non-Patent Literature 4).

CITATION LIST Non Patent Literature

  • [NPL 1] Grupp S A, Kalos M, Barrett D, Aplenc R, Porter D L, Rheingold S R, Teachey D T, Chew A, Hauck B, Wright J F, Milone M C, Levine B L, June C H. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med, 368(16):1509-18. 2013
  • [NPL 2] Maude S L, Frey N, Shaw P A, Aplenc R, Barrett D M, Bunin N J, Chew A, Gonzalez V E, Zheng Z, Lacey S F, Mahnke Y D, Melenhorst J J, Rheingold S R, Shen A, Teachey D T, Levine B L, June C H, Porter D L, Grupp S A. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med, 371(16):1507-17. 2014
  • [NPL 3] Lee D W, Kochenderfer J N, Stetler-Stevenson M, Cui Y K, Delbrook C, Feldman S A, Fry T J, Orentas R, Sabatino M, Shah N N, Steinberg S M, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg S A, Wayne A S, Mackall C L. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2014
  • [NPL 4] Morgan R A. Faster, cheaper, safer, T-cell engineering. J Immunother, 36(1):1-2. 2013

SUMMARY OF INVENTION Technical Problem

In order to solve the problems in prior art CAR therapy using a viral vector, the use of a transposon method, which is one of genetic modification techniques using a non-viral vector, has been studied. A transposon method allows lasting gene introduction as in the case of a viral vector technique, but has problems that the gene introduction efficiency by a transposon method is lower than a viral vector method, and the cells are damaged by the gene introduction operations (for example, electroporation and its improvement method), and the cell survival rate and the cell proliferation rate decrease. The objects of the present invention are to solve these problems, and contribute to the improvements of clinical application and treatment results of CAR therapy.

Solution to Problem

The inventors eagerly studied to solve the above-described problems. As a result of this, it was found that co-culturing of the T cells after gene introduction operation (genetically-modified T cells) with the separately prepared activated T cells improved the gene introduction efficiency and the survival rate/proliferation rate, and increased the number of the finally obtained CAR-T cells. On the other hand, efficient preparation of virus-specific CAR-T cells was achieved by the strategy of co-culturing of the T cells after gene introduction operation (genetically-modified T cells) with activated T cells holding a viral peptide. The invention described below is based on mainly these results.

[1] A method for preparing genetically-modified T cells expressing chimeric antigen receptor, including the following steps (1) to (4):

(1) a step of preparing non-proliferative cells which are obtained by stimulating a group of cells including T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by a treatment for causing the cells to lose their proliferation capability;

(2) a step of obtaining genetically-modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced using a transposon method;

(3) a step of mixing the non-proliferative cells prepared by step (1) with the genetically-modified T cells obtained by step (2), and co-culturing the mixed cells while stimulating the mixed cells using an anti-CD3 antibody and anti-CD28 antibody; and

(4) a step of collecting the cells after culture.

[2] The preparation method according to [1], wherein a step of culturing the cells after the co-culturing in the presence of a T-cell growth factor is carried out between step (3) and step (4).

[3] The preparation method according to [1] or [2], wherein the period of the co-culturing in step (3) is one day to 14 days.

[4] The preparation method according to any one of [1] to [3], wherein step (3) is carried out in the presence of a T-cell growth factor.

[5] The preparation method according to [4], wherein the T-cell growth factor is IL-15.

[6] The preparation method according to [4], wherein the T-cell growth factor is a combination of IL-15 and IL-7.

[7] A method for preparing genetically-modified T cells expressing chimeric antigen receptor, including the following steps (i) to (iv):

(i) a step of preparing non-proliferative cells holding a viral peptide antigen, which are obtained by stimulating a group of cells including T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability;

(ii) a step of obtaining genetically-modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced using a transposon method;

(iii) a step of mixing the non-proliferative cells prepared by step (i) with the genetically-modified T cells obtained by step (ii), and co-culturing the mixed cells; and

(iv) a step of collecting the cells after culture.

[8] The preparation method according to [7], wherein a step of culturing the cells after the co-culturing in the presence of a T-cell growth factor is carried out between step (iii) and step (iv).

[9] The preparation method according to [7] or [8], wherein the period of the co-culturing in step (iii) is one day to 14 days.

[10] The preparation method according to any one of [7] to [9], wherein step (iii) is carried out in the presence of a T-cell growth factor.

[11] The preparation method according to [10], wherein the T-cell growth factor is IL-15.

[12] The preparation method according to [10], wherein the T-cell growth factor is a combination of IL-15 and IL-7.

[13] The preparation method according to any one of [1] to [12], wherein the group of cells including T-cells is peripheral blood mononuclear cells (PBMCs).

[14] The preparation method according to any one of [1] to [13], wherein the treatment for losing the proliferation capability is irradiation.

[15] The preparation method according to any one of [1] to [14], wherein the transposon method is the PiggyBac transposon method.

[16] The preparation method according to any one of [1] to [15], wherein the target antigen is the CD19, GD2, GMCSF receptor or the IGF receptor.

[17] The preparation method according to any one of [1] to [16], wherein the non-proliferative cells and the genetically-modified T cells are derived from an identical individual.

[18] Genetically-modified T cells expressing chimeric antigen receptor obtained by the preparation method according to any one of [1] to [17].

[19] A cell preparation including the genetically-modified T cells according to [18] in a therapeutically effective amount.

[20] A cancer therapy including a step of administering a therapeutically effective amount of the genetically-modified T cells according to [18] to a cancer patient.

BRIEF DESCRIPTION OF DRAWINGS

FIG. 1 depicts preparation and culturing of CAR-T cells by a prior art method (culturing method 1).

FIG. 2 depicts preparation and culturing of CAR-T cells by a novel method (culturing method 2).

FIG. 3 depicts preparation and culturing of CAR-T cells by a novel method (culturing method 3).

FIG. 4 depicts the structure of the pIRII-CAR.CD19.28z vector (SEQ ID NO. 1), composed of the CD19CAR gene held between the 5′ inverted repeat sequence (5′IR) and the 3′ inverted repeat sequence (3′IR). CD19CAR includes a reader sequence (SEQ ID NO. 2), a light chain variable region (VL) (SEQ ID NO. 3), a heavy chain variable region (VH) (SEQ ID NO. 4), an Fc region (CH2, CH3) (SEQ ID NO. 5), a transmembrane region of CD28, an intracellular domain (SEQ ID NO. 6), and CD3ζ (SEQ ID NO. 7).

FIG. 5 depicts the structure of the pCMV-pigBac vector (SEQ ID NO. 8), composed of the piggyback transposase gene placed under control of the CMV immediate early promoter.

FIG. 6 depicts the number of the CAR-T cells obtained by each culturing method (average±standard error).

FIG. 7 depicts the gene introduction rate by each culturing method (average±standard error).

FIG. 8 depicts the number of the CAR-T cells obtained by each culturing method (average±standard error). N=9. Significant differences are found between the novel methods (culturing methods 2 and 3) and the prior art method (culturing method 1). Additionally, culturing method 3 brought a significantly higher number of CAR-T cells than culturing method 2.

FIG. 9 depicts the gene introduction rate by each culturing method (average±standard error). N=9. Significant differences are found between the novel methods (culturing methods 2 and 3) and the prior art method (culturing method 1). Additionally, culturing method 3 had a significantly higher gene introduction rate than culturing method 2.

FIG. 10 depicts the structure of the pIRII-CAR.CD19_optimized vector (SEQ ID NO. 9), which is an optimized vector for CAR gene introduction. In comparison with the structure of the pIRII-CAR.CD19.28z vector, the Fc regions (CH2 and CH3) are deleted.

FIG. 11 depicts the gene introduction rate when the pIRII-CAR.CD19_optimized vector was used. The result of flow cytometry (left) and the proportion of the CAR expression cell in the CD3-positive T cells (right) are represented. N=9.

FIG. 12 depicts the result of cytotoxic activity test. The results of flow cytometry (top) and tumor cell residual rate (bottom) are given. Three kinds of CD19-positive leukemia cell lines (KOPN30bi, SK-9, and TCC-Y/sr) and CAR-T cells were co-cultured, and the cytotoxic activity was evaluated. N=6.

FIG. 13 depicts the result of antitumor activity test. After administering CAR-T cells to tumor-bearing mice, the proliferation of tumor cells was monitored with an in vivo imaging system.

DESCRIPTION OF EMBODIMENTS

The present invention relates to a method for preparing genetically-modified T cells (CAR-T cells) expressing chimeric antigen receptor. The CAR-T cells obtained by the preparation method of the present invention may be used in a CAR therapy. The present invention provides generally two preparation methods, more specifically, a method including co-culturing with activated T cells (for convenience of explanation, the method may be referred to as “the first preparation method”), and a method including co-culturing with activated T cells holding a viral peptide (for convenience of explanation, the method may be referred to as “the second preparation method”). Unless otherwise specified, the cells (for example, T cells) in the present description are human cells.

1. Method Including Co-Culturing with Activated T Cells

In this preparation method (the first preparation method), the following steps (1) to (4) are carried out:

(1) a step of preparing non-proliferative cells which are obtained by stimulating a group of cells including T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by a treatment for causing the cells to lose their proliferation capability;

(2) a step of obtaining genetically-modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced using a transposon method;

(3) a step of mixing the non-proliferative cells prepared by step (1) with the genetically-modified T cells obtained by step (2), and co-culturing the mixed cells while stimulating the mixed cells using an anti-CD3 antibody and anti-CD28 antibody; and

(4) a step of collecting the cells after culture.

Step (1) is a step of obtaining non-proliferative cells used for the protection of the T cells after gene introduction operation (the genetically-modified T cells used in step (2)); firstly, a group of cells including T cells is stimulated with an anti-CD3 antibody and an anti-CD28 antibody. Through this treatment, activated T cells are obtained. As the “a group of cells including T cells”, preferably, PBMCs (peripheral blood mononuclear cells) collected from the peripheral blood are used. The “group of cells including T cells” herein may be, for example, the PBMCs which have been purified to increase the T-cell content, or mononuclear cells collected from the peripheral blood by pheresis.

For example, the T cells in a group of cells can be stimulated with an anti-CD3 antibody and an anti-CD28 antibody by culturing them in a culture vessel (for example, culture dish) coated with an anti-CD3 antibody and an anti-CD28 antibody on the culturing surface for three hours to three days, preferably six hours to two days, and more preferably from 12 hours to one day. The anti-CD3 antibody (for example, the trade name CD3pure antibody provided by Miltenyi Biotec may be used) and the anti-CD28 antibody (for example, the trade name CD28pure antibody provided by Miltenyi Biotec may be used) are commercially available and are easily available. The stimulation in step (1) may be carried out using the magnetic beads coated with an anti-CD3 antibody and an anti-CD28 antibody (for example, Dynabeads T-Activator CD3/CD28 provided by VERITAS). The anti-CD3 antibody is preferably “OKT3” clone.

The cells stimulated with an anti-CD3 antibody and an anti-CD28 antibody are subjected to a treatment for causing them to lose their proliferation capability, preferably after culturing in the presence of a T-cell growth factor. This culturing increases the activity of the cells after the stimulation treatment. The period of the culture is, for example, from one day to 10 days, preferably from two days to seven days, and more preferably from three days to four days. If the culture period is too short, sufficient activation cannot be obtained, and if the culture period is too long, attenuation of the costimulatory molecule may occur. The cells after culture may be once subjected to cryopreservation. In this case, the cells are unfrozen before use, stimulated with the anti-CD3 antibody and the CD28 antibody again (the conditions are the same as described above), and then subjected to “the treatment for losing the proliferation capability”.

Through the “treatment for losing the proliferation capability”, the activated T cells having no proliferation capability (non-proliferative cells) are obtained. The treatment for losing the proliferation capability is typically irradiation, but may use a drug. The irradiation is carried out by, for example, using a T-ray, at an intensity of 25 Gy to 50 Gy, for 15 to 30 minutes.

In the step (2), genetically-modified T cells in which a target antigen-specific chimeric antigen receptor gene has been introduced are obtained. Namely, in the present invention, a CAR gene is introduced to T cells, thereby CAR-T cells are obtained. The gene introduction is preferably carried out by a transposon method. The transposon method is one of the non-viral gene introduction methods. Transposon is the generic name of short gene sequences causing a gene transposition conserved during the process of evolution. A pair of a gene enzyme (transposase) and its specific recognition sequence causes gene transposition. The transposon method may be, for example, the piggyBac transposon method. The piggyBac transposon method uses the transposon isolated from insects (Fraser M J et al., Insect Mol Biol. 1996 May; 5(2): 141-51; Wilson M H et al., Mol Ther. 2007 January; 15(1): 139-45), and allows highly efficient integration into mammal chromosomes. The piggyBac transposon method is actually used for the introduction of the CAR gene (for example, see Nakazawa Y, et al., J Immunother 32: 826-836, 2009; Nakazawa Y et al., J Immunother 6: 3-10, 2013). The transposon method applicable to the present invention is not limited to that using piggyBac, and may use a method using transposon, for example, Sleeping Beauty (Ivics Z, Hackett P B, Plasterk R H, Izsvak Z (1997) Cell 91: 501-510), Frog Prince (Miskey C, Izsvak Z, Plasterk R H, Ivics Z (2003) Nucleic Acids Res 31: 6873-6881), Tol1 (Koga A, Inagaki H, Bessho Y, Hori H. Mol Gen Genet. 1995 Dec. 10; 249 (4): 400-5; Koga A, Shimada A, Kuroki T, Hori H, Kusumi J, Kyono-Hamaguchi Y, Hamaguchi S. J Hum Genet. 2007; 52(7): 628-35. Epub 2007 Jun. 7), Tol2 (Koga A, Hori H, Sakaizumi M (2002) March Biotechnol 4: 6-11; Johnson Ha mL et M R, Yergeau D A, Kuliyev E, Takeda M, Taira M, Kawakami K, Mead P E (2006) Genesis 44: 438-445; Choo B G, Kondrichin I, Parinov S, Emelyanov A, Go W, Toh W C, and Korzh V (2006) BMC Dev Biol 6: 5).

The introduction operation by the transposon method may be carried out by an ordinary method with reference to past literatures (for example, for the piggyBac transposon method, see Nakazawa Y, et al., J Immunother 32: 826-836, 2009, Saha S, Nakazawa Y, Huye L E, Doherty J E, Galvan D L, Rooney C M, Wilson M H. J Vis Exp. 2012 Nov. 5; (69): e423). In a preferred embodiment of the present invention, the piggyBac transposon method is used. Typically, in the piggyBac transposon method, a vector including the gene coding piggyBac transposase (transposase plasmid) and a vector having a structure wherein the gene coding a target protein (CAR gene) is sandwiched between piggyBac inverted repeat sequences (transposon plasmid) are prepared, and these vectors are introduced (transfected) to the target cell. The transfection may use various methods such as electroporation, nucleofection, lipofection, or calcium phosphate method.

Examples of cell into which the CAR gene is introduced (a target cell) include CD4-positive CD8-negative T-cells, CD4-negative CD8-positive T-cells, T-cells prepared from iPS cells, αβ-T-cells, and γδ-T-cells. Various cell populations may be used, as long as they contain the above-described T cells or precursor cells. PBMCs (peripheral blood mononuclear cells) collected from the peripheral blood is one of the preferred target cells. More specifically, in a preferred embodiment, gene introduction operation is carried out on the PBMCs. The PBMCs may be prepared by an ordinary method. The method for preparing the PBMCs may refer to, for example, Saha S, Nakazawa Y, Huye L E, Doherty J E, Galvan D L, Rooney C M, Wilson M H. J Vis Exp. 2012 Nov. 5; (69): e4235.

The cells after the gene introduction operation is subjected to the co-culturing in step (3). Before step (3), the cells after the gene introduction operation may be cultured in the presence of a T-cell growth factor (for example, IL-15 or IL-7).

The CAR gene codes the chimeric antigen receptor (CAR) recognizing a specific target antigen. The CAR is a structural body including an extracellular domain specific to the target, a transmembrane domain, and an intracellular signal domain for the effector function of immunocytes. These domains are explained below.

(a) Extracellular Domain

The extracellular domain specifically binds to the target. For example, the extracellular domain contains the scFv fragment of the anti-target monoclonal antibody. Examples of the monoclonal antibody used herein include rodent antibodies (e.g., mouse, rat, and rabbit antibodies), human antibodies, and humanized antibodies. The humanized monoclonal antibody is prepared by making the structure of the monoclonal antibody of any animal species (for example, mice or rats) analogous to the structure of the human antibody, and includes the human type chimera antibody, which is prepared by substituting only the constant region of an antibody with that of the human antibody, and the human type CDR-grafted antibody, which is prepared by substituting the parts excluding the CDR (complementary determining region) in the constant and variable regions with those of the human antibody (P. T. Johons et al., Nature 321, 522 (1986)). For the purpose of increasing the antigen binding activity of human type CDR-grafted antibodies, already developed are the improvement techniques for the method for choosing a human antibody framework (FR) having high homology for mouse antibodies, the method for preparing humanized antibodies having high homology, and the method for transplanting a mouse CDR in a human antibody, followed by substitution of amino acids in the FR region (e.g., U.S. Pat. Nos. 5,585,089, 5,693,761, 5,693,762, 6,180,370, European Patent Application No. 451216, European Patent Application No. 682040, and Japanese Patent No. 2828340), which can be used for the preparation of humanized antibodies.

The scFv fragment is a structural body wherein the light chain variable region (VL) and heavy chain variable region (VH) of immunoglobulin are linked through a linker, and retains binding ability for the antigen. The linker may be, for example, a peptide linker. The peptide linker is composed of a peptide made by linear linking of amino acids. Typical examples of the peptide linker are the linkers composed of glycine and serine (GGS and GS linkers). The amino acids composing the GGS and GS linkers, glycine and serine, are small in their sizes, and thus hardly form higher-order structures. The length of the linker is not particularly limited. For example, a linker having 5 to 25 amino acid residues may be used. The number of the amino acid residue composing the linker is preferably from 8 to 25, and more preferably from 15 to 20.

The target used herein is typically an antigen which shows specific expression in tumor cells. The “specific expression” means significant or remarkable expression in comparison with the cells other than tumor, and will not intend to confine to those showing no expression in the cells other than tumor. Examples of the target antigen include the CD19 antigen, CD20 antigen, GD2 antigen, CD22 antigen, CD30 antigen, CD33 antigen, CD44 variant 7/8 antigen, CEA antigen, Her2/neu antigen, MUC1 antigen, MUC4 antigen, MUC6 antigen, IL-13 receptor-alpha 2, immunoglobulin light chain, PSMA antigen, and VEGF receptor 2.

The GM-CSF (granulocyte macrophage colony-stimulating factor) receptor expressed in leukemic stem cells/precursor cells may be used as the target. In this case, GM-CSF, which is a ligand of the GM-CSF receptor, is used as the extracellular domain composing CAR. The leukemia stem cells, leukemia precursor cells, leukemia cells and others of bone marrow tumor are used as the targets of the CAR-T cells, whereby the cells applicable to prevention and treatment of myeloproliferative tumor, myelodysplastic/myeloproliferative tumor (CMML, JMML, CML, MDS/MPN-UC), myelodysplastic syndrome, acute myelogenous leukemia, and the like are prepared.

(b) Transmembrane Domain

The transmembrane domain intervenes between the extracellular domain and intracellular signal domain. Examples of the transmembrane domain used herein include CD28, CD3ε, CD8α, CD3, CD4, and 4-1BB. Alternatively, a transmembrane domain composed of an artificially constructed polypeptide may be used.

(c) Intracellular Signal Domain

The intracellular signal domain transmits the signals necessary for exertion of the effector function of immunocytes. More specifically, when the extracellular domain binds with the target antigen, an intracellular signal domain capable of transmitting the signals necessary for activation of immunocytes are used. The intracellular signal domain includes the domain for transmitting the signals through the TCR complex (for convenience, referred to as “the first domain”), and the domain for transmitting the costimulatory signals (for convenience, referred to as “the second domain”). As the first domain, CD3ξ or other intracellular domains such as FcεRIγ may be used. The use of CD3ξ is preferred. As the second domain, the intracellular domain of a costimulatory molecule is used. Examples of the costimulatory molecule include CD28, 4-1BB (CD137), CD2, CD4, CD5, CD134, OX-40, and ICOS. The use of the intracellular domain of CD28 or 4-1BB is preferred.

The linking form of the first and second domains is not particularly limited, and preferably the second domain is disposed on the transmembrane domain side, because it is known that co-stimulation was strongly transmitted when CD3ξ was linked distally in a past case. The same or different kinds of plural intracellular domains may be linked in tandem to form the first domain. The same holds true for the second domain.

The first and second domains may be directly linked, or a linker may intervene between them. The linker may be, for example, a peptide linker. The peptide linker is composed of peptides which are linear chains of amino acids. The structure and characteristics of the peptide linker are as described above. However, the linker used herein may be composed solely of glycine. The length of the linker is not particularly limited. For example, a linker composed of 2 to 15 amino acid residues may be used.

(d) Other Elements

A leader sequence (signal peptide) is used to promote CAR secretion. For example, the leader sequence of the GM-CSF receptor may be used. In addition, the structure is preferably composed of an extracellular domain and a transmembrane domain linked together through a spacer domain. More specifically, the CAR according to a preferred embodiment contains a spacer domain between the extracellular domain and transmembrane domain. The spacer domain is used for promoting linking between the CAR and target antigen. For example, the Fc fragment of a human IgG (for example, human IgG1 or human IgG4) m ay be used as the spacer domain. Alternatively, a part of the extracellular domain of CD28 or a part of the extracellular domain of CD8a may be used as the spacer domain. A spacer domain may be placed between the transmembrane domain and intracellular signal domain.

There are some reports on the experiments and clinical studies using CARs (for example, Rossig C, et al. Mol Ther 10: 5-18, 2004; Dotti G, et al. Hu m Gene Ther 20: 1229-1239, 2009; Ngo M C, et al. Hum Mol Genet 20 (R1): R93-99, 2011; Ahmed N, et al. Mol Ther 17: 1779-1787, 2009; Pule M A, et al. Nat Med 14: 1264-1270, 2008; Louis C U, et al. Blood 118: 6050-6056, 2011; Kochenderfer J N, et al. Blood 116: 4099-4102, 2010; Kochenderfer J N, et al. Blood 119: 2709-2720, 2012; Porter D L, et al. N Engl J Med 365: 725-733, 2011; Kalos M, et al. Sci Transl Med 3: 95ra73, 2011; Brentjens R J, et al. Blood 1 18: 4817-4828, 2011; and Brentjens R J, et al. Sci Transl Med 5: 177 ra38, 2013), and the CARs in the present invention may be constructed with reference to these reports.

In the transposon plasmid, a poly-A additional signal sequence is disposed down stream of the CAR gene. Transcription is terminated by the use of the poly-A additional signal sequence. The poly-A additional signal sequence may be, for example, the poly-A additional sequence of SV40, or the poly-A additional sequence of a bovine-derived growth hormone gene.

The transposon plasmid may include, for example, a gene for detection (for example, a reporter gene, cell or tissue-specific gene, or selectable marker gene), an enhancer sequence, and a WRPE sequence. The gene for detection is used for the judgement of success/failure or efficiency of the introduction the expression cassette, detection of CAR expression or judgement of the expression efficiency, and selection and collection of the cells having expressed the CAR gene. On the other hand, the enhancer sequence is used for improving the expression efficiency. Examples of the gene for detection include the neo gene im parting resistance against neomycin, the npt gene (Herrera Estrella, EMBO J. 2 (1983), 987-995) and npt II gene (Messing & Vierra. Gene 1 9: 259-268(1982)) imparting resistance against kanamycin, the hph gene imparting resistance against hygromycin (Blochinger & Diggl mann, Mol Cell Bio 4: 2929-2931), and the dhfr gene imparting resistance against Methotrexate (Bourouis et al., EMBO J. 2 (7)) (the aforementioned are marker genes); genes of fluorescence proteins such as the luciferase gene (Giacomin, P1. Sci. 116 (1996), 59 to 72; Scikantha, J. Bact. 178 (1996), 121), the β-glucuronidase (GUS) gene, GFP (Gerdes, FEBS Lett. 389 (1996), 44-47), and their variants (EGFP and d2EGFP) (the aforementioned are reporter genes); and the epidermal growth factor receptor (EGFR) gene deficient in the intracellular domain. The gene for detection is linked to the CAR gene through, for example, a bicistronic control sequence (for example, internal ribo some entry site (IRES)) and a sequence coding a self cleavage peptide. Examples of the self cleavage peptide include, but not limited to, the 2A peptide (T2 A) derived from Thosea asigna virus. Known examples of the self cleavage peptide include the 2A peptide (F2A) derived from the Foo-and-mouse disease virus (FMDV), the 2A peptide (E2A) derived from equine rhinitis A virus (ERAV), and the 2A peptide (P2A) derived from porcine teschovirus (PTV-1).

In step (3), the non-proliferative cells prepared in step (1) and the genetically-modified T cells obtained in step (2) are mixed, and the mixed cells are co-cultured while stimulating the mixed cells using an anti-CD3 antibody and an anti-CD28 antibody. As a result of this, stimulation through the costimulatory molecules by the non-proliferative cells and stimulation by an anti-CD3 antibody and an anti-CD28 antibody are added, whereby the genetically-modified T cells are activated, and their survival and proliferation are promoted.

The ratio between the number of the non-proliferative cells used for co-culturing and the number of the genetically-modified T cells (the number of the non-proliferative cells/the number of genetically-modified T cells) is not particularly limited, and, for example, from 0.025 to 0.5.

In order to improve the survival rate/proliferation rate of the cells, it is preferred to use a culture solution containing a T-cell growth factor in the activation treatment. The T-cell growth factor is preferably IL-15. Preferably, a culture solution containing IL-15 and IL-7 is used. The concentration of IL-15 is, for example, from 5 ng/mL to 10 ng/mL. The concentration of IL-7 is, for example, from 5 ng/mL to 10 ng/mL. The T-cell growth factor such as IL-15 or IL-7 may be prepared according to a common procedure. Alternatively, a commercial product may be used. Although the use of animal T-cell growth factors other than human ones will not be excluded, the T-cell growth factor used herein is usually derived from human (may be a recombinant). The growth factors such as human IL-15 and human IL-7 are readily available (for example, provided by Miltenyi Biotec, R&D systems).

A culture medium containing blood serum (for example, human blood serum or fetal bovine serum) may be used, but the use of a serum-free medium allows the preparation of cells having advantages of high safety in clinical application, and little difference in the culture efficiency among blood serum lots. Specific examples of the serum-free medium for T cells include TexMACS™ (Miltenyi Biotec) and AIM V (registered trademark) (Thermo Fisher Scientific). When a blood serum is used, the blood serum is preferably a self-blood serum, or a blood serum collected from the individual who is the origin of the genetically-modified T cells obtained in step (2) (typically, the patient to receive administration of the chimeric antigen receptor genetically-modified T cells obtained by the preparation method of the present invention). The basal culture medium is the one suitable for culture of T cells, and specific examples include the above-listed TexMACS™ and AIM V (registered trademark). Other culture conditions may be common ones, as long as they are suitable for the survival and proliferation of T cells. For example, the culture is carried out in a CO2 incubator adjusted at 37° C. (CO2 concentration: 5%). The stimulation by an anti-CD3 antibody and an anti-CD28 antibody is the same as that in step (1), so the explanations thereof are omitted.

The period of the co-culturing in step (3) is, for example from one day to 10 days, preferably from one day to seven days, and more preferably two days to four days. If the culture period is too short, sufficient effect cannot be obtained, and if the culture period is too long, the activity (vital force) of the cells may decrease.

In step (4) following step (3), the cells after culture are collected. The collection operation may use an ordinary method. For example, the collection is carried out by pipetting or centrifugation treatment.

In a preferred embodiment, the cells after the co-culturing are cultured in the presence of a T-cell growth factor between step (3) and step (4). This step allows efficient cell expansion, and also improves the survival rate of cells.

The T-cell growth factor may be, for example, IL-15 or IL-7. Preferably, in the same manner as in step (3), the cells are cultured on a medium containing IL-15 and IL-7. The culture period is, for example, from one day to 21 days, preferably from five days to 18 days, and more preferably from 10 days to 14 days. If the culture period is too short, the number of cells will not increase sufficiently, and if the culture period is too long, the activity (vital force) of the cells may decrease, and the cells may be exhausted or fatigued. The cells may be subcultured during culturing. Additionally, the culture medium is replaced as necessary during culturing. For example, about ⅓ to ⅔ of the culture solution is replaced with a new culture medium once every three days.

2. Method Including Co-Culturing with T Cells Holding Viral Peptide

Another embodiment (second preparation method) of the present invention relates to a method for preparing a virus specific chimeric antigen receptor genetically-modified T cells (hereinafter referred to as “virus-specific CAR-T cells”). The virus-specific CAR-T cells have important advantages in clinical application, such as their use in autotransplantation improves internal persistence by stimulation from a viral T cell receptor, and their use in allogeneic transplantation further allows the preparation of CAR-T from a transplanted donor owing to the reduction of allogeneic immunity reaction (GVHD), and creates possibility of drug formulation of CAR-T cells from a third party donor. Actually, there is a report that virus-specific CAR-T cells survive longer in the body (Pule M A, et al. Nat Med. 2008 November; 14 (11): 1264-70). In addition, the report of a third party-derived EBV-specific CTL clinical study (Annual Review Blood 2015, published in January 2015, Chugai-Igakusha) supports high level of safety of virus-specific cytotoxic T cells (CTLs).

The preparation method of this embodiment includes the following steps (i) to (iv). The items not referred herein (for example, the method for preparing a group of cells including T cells, basic operation of stimulation by an anti-CD3 antibody and an anti-CD28 antibody, the method of the treatment for losing proliferation capability, the operation of gene introduction by a transposon method, basic operation of co-culturing, and the method for collecting cells) are the same as those in the above-described first preparation method, so that repetitive description thereof will be omitted, and corresponding explanation is incorporated.

(i) A step of preparing non-proliferative cells holding a viral peptide antigen, which are obtained by stimulating a group of cells including T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability

(ii) A step of obtaining genetically-modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced using a transposon method

(iii) A step of mixing the non-proliferative cells prepared by step (i) with the genetically-modified T cells obtained by step (ii), and co-culturing the mixed cells

(iv) A step of collecting the cells after culture.

In step (i), firstly, the group of cells including T cells are stimulated with an anti-CD3 antibody and an anti-CD28 antibody, thereby obtaining activated T cells. Thereafter, the cells are subjected to culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability. As a result of this, non-proliferative “activated T cells holding a viral peptide antigen on the cell surface” (hereinafter referred to as “viral peptide-holding non-proliferative cells”) are obtained. The order of culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability is not particularly limited. Accordingly, the proliferation capability may be lost after culturing in the presence of the viral peptide antigen, or the cells may be cultured in the presence of the viral peptide antigen after they were caused to lose their proliferation capability. Preferably, the former order is adopted in the expectation that the intake of the viral peptide antigen would be better than before the loss of proliferation capability. In order to culture the cells in the presence of the viral peptide antigen, for example, a culture medium containing the viral peptide antigen is used. Alternatively, the viral peptide antigen may be added to the culture medium during culturing. The addition concentration of the viral peptide antigen is, for example, from 0.5 μg/mL to 1 μg/mL. The culture period is, for example, from 10 minutes to 5 hours, and preferably from 20 minutes to 3 hours. The “viral peptide antigen” in the present description means an epitope peptide or a long peptide containing an epitope which can induce cytotoxic T cells (CTLs) specific to a specific virus. Examples of the viral peptide antigen include, but not limited to, antigen peptides of adenovirus (AdV) (for example, see WO 2007015540 A1), antigen peptides of cytomegalovirus (CMV) (for example, see Japanese Unexamined Patent Application Publication No. 2002-255997, Japanese Unexamined Patent Application Publication No. 2004-242599, and Japanese Unexamined Patent Application Publication No. 2012-87126), and antigen peptides of Epstein-Barr virus (EBV) (for example, see WO 2007049737 A1, Japanese Patent Application No. 2011-177487, and Japanese Unexamined Patent Application Publication No. 2006-188513). The viral peptide antigen can be prepared by a common procedure (for example, a solution-phase synthesis method or a solid-phase synthesis method) based on the sequence information. Some viral peptide antigens are commercially available (for example, provided by Medical & Biological Laboratories Co., Ltd., Takara Bio, Inc., and Miltenyi Biotec).

One antigen peptide may be used, but usually two or more antigen peptides (an antigen peptide mixture) are used. For example, an AdV antigen peptide mixture, a CMV antigen peptide mixture, or an EBV antigen peptide mixture, or a combination of two or more of these antigen peptide mixtures (for example, a mixture of the AdV antigen peptide mixture, CMV antigen peptide mixture, and EBV antigen peptide mixture) is used. The combination of two or more antigen peptides allows obtainment of plural activated T cells having different targets (antigen peptides), which would increase the subjects (patients) to whom the CAR-T cells obtained by the preparation method of the present invention is effective (the improvement of cover rate). When determining which virus the antigen peptide to be used is derived from, the use of the CAR-T cells obtained by the preparation method of the present invention, specifically the disease and the disease state of the patient to be treated may be considered. For example, when the treatment of recurrent leukemia after transplantation of hematopoietic stem cells is aimed at, it is suggested that the antigen peptide mixture of EBV virus be used alone or in combination with other viral antigen peptide mixture. The AdV antigen peptide mixture, CMV antigen peptide mixture, and EBV antigen peptide mixture are commercially available (for example, PepTivator (registered trademark) AdV5 Hexon, PepTivator (registered trademark) CMV pp65, PepTivator (registered trademark) EBV EBNA-1, PepTivator (registered trademark), and EBV BZLF1 provided by Miltenyi Biotec, and PepMix™ Collection HCMV, PepMix™ EBV (EBNA1) provided JPT Peptide Technologies, and the like) are easily available.

Step (ii) is the same as step (2) in the above-described first preparation method of the present invention. Through this step, the genetically-modified T cells (CAR-T cells) are obtained.

In step (iii), the non-proliferative cells (viral peptide-holding non-proliferative cells) prepared in step (i) and the genetically-modified T cells obtained in step (ii) are mixed, and the mixed cells are co-cultured. As a result of this, stimulation through the costimulatory molecules by the non-proliferative cells and stimulation by the viral antigen peptide are added, whereby the virus antigen specific genetically-modified T cells are activated, and their survival and proliferation are promoted.

The ratio between the number of the non-proliferative cells used for co-culturing and the number of the genetically-modified T cells (the number of non-proliferative cells/the number of genetically-modified T cells) is not particularly limited, and, for example, from 0.025 to 0.5.

In this step, in principle, stimulation by an anti-CD3 antibody and an anti-CD28 antibody is not applied for the purposes of, for example, selectively proliferating the virus-specific CAR-T cells, or preventing exhaustion and fatigue of the T cells by avoiding strong stimulation. On the other hand, in order to increase the survival rate/proliferation rate of the cells, it is preferred that a culture solution containing a T-cell growth factor be used during the co-culturing. The T-cell growth factor is preferably IL-15. Preferably, a culture solution containing IL-15 and IL-7 are used. The addition amount of IL-15 is, for example, from 5 ng/mL to 10 ng/mL. In the same manner, the addition amount of IL-7 is, for example, from 5 ng/mL to 10 ng/mL. The conditions not referred herein (for example, possibility of the use of the blood serum, the basal culture medium, and the incubation temperature) are the same as those in step (3) of the above-described first preparation method of the present invention.

The viral peptide-holding non-proliferative cells may be added during step (iii). Alternatively, the co-cultured cells are collected, mixed with the viral peptide-holding non-proliferative cells, and then co-culturing is carried out again. These operations may be repeated twice or more times. In this manner, the improvement of the induction rate of the virus-specific CAR-T cells and the increase of the number of the virus-specific CAR-T cells are expected by carrying out plural times of the stimulation or activation using the viral peptide-holding non-proliferative cells. The viral peptide-holding non-proliferative cells used herein are prepared anew, or a portion of the preserved cells which have been prepared in step (i).

In step (iii), the period of co-culturing is, for example, from one day to 21 days, preferably from five days to 18 days, and more preferably from 10 days to 14 days. If the culture period is too short, sufficient effect cannot be obtained, and if the culture period is too long, the activity (vital force) of the cells may decrease, and the cells may be exhausted or fatigued.

Before the co-culturing with the viral peptide-holding non-proliferative cells, the genetically-modified T cells obtained in step (ii) may be co-cultured with viral peptide-holding non-proliferative PBMCs (peripheral blood mononuclear cells). In this embodiment, the cells obtained by co-culturing the genetically-modified T cells obtained in step (ii) with the viral peptide-holding non-proliferative PBMCs, and the viral peptide-holding non-proliferative cells prepared in step (i) are mixed, and the mixture is co-cultured. The viral peptide-holding non-proliferative PBMCs herein can be prepared by subjecting PBMCs to culturing in the presence of a viral peptide antigen and a treatment for causing them to lose their proliferation capability. Specifically, for example, PBMC isolated from the peripheral blood are irradiated, and then cultured in the presence of a viral peptide antigen, thus obtaining viral peptide-holding non-proliferative PBMCs. The number of blood collection for carrying out the present invention can be reduced by preparing viral peptide-holding non-proliferative PBMCs using a portion of the PBMCs isolated from the peripheral blood obtained by one time of blood collection, and preparing genetically-modified T cells from another portion, which will bring a markedly big advantage in clinical application. In particular, when the viral peptide-holding non-proliferative cells (the cells used for the second step co-culturing) are prepared by carrying out step (i) using the remaining PBMCs, the three kinds of necessary cells, more specifically, the genetically-modified T cells, viral peptide-holding non-proliferative PBMCs used for co-culturing with these cells, and the viral peptide-holding non-proliferative cells used for the second step co-culturing can be prepared by one time of blood collection, which markedly reduces the burden imposed on the patient in the treatment using the CAR-T cells obtained in the present invention.

In step (iv) following step (iii), the cells after culture are collected. In the same manner as in the above-described first preparation method of the present invention, the step of culturing the co-cultured cells in the presence of the T-cell growth factor (expanded culturing) may be carried out between step (iii) and step (iv). For this cell expansion, viral peptide-holding non-proliferative cells may be added, or viral peptide-holding non-proliferative cells may be added during the cell expansion.

3. Genetically-Modified T Cells which Express Chimeric Antigen Receptor and and Uses Thereof

A further aspect of the present invention relates to the genetically-modified T cells which express chimeric antigen receptor gene obtained in the preparation method of the present invention (hereinafter referred to as “CAR-T cells of the present invention”) and uses thereof. The CAR-T cells of the present invention can be used for treatment, prevention, or improvement of various diseases (hereinafter referred to as “target diseases”) to which the CAR therapy is likely effective. Representative examples of the target disease include, but not limited to, cancer. Examples of the target disease include various B-cell lymphoma (follicular lymphoma, diffuse large B-cell lymphoma, mantle cell lymphoma, MALT lymphoma, intravascular B-cell lymphoma, and CD20-positive Hodgkin's lymphoma), myeloproliferative tumor, myelodysplastic/myeloproliferative tumor (CMML, JMML, CML, MDS/MPN-UC), myelodysplastic syndromes, acute myelocytic leukemia, neuroblastoma, brain tumor, Ewing's sarcoma, osteosarcoma, retinoblastoma, small cell lung cancer, melanoma, ovarian cancer, rhabdomyosarcoma, kidney cancer, pancreas cancer, malignant mesothelioma, and prostate cancer. “Treatment” include alleviation (moderation) of symptoms or associated symptoms characteristic to the target diseases, inhibition or retard of deterioration of symptoms. “Prevention” means prevention or retard of development/expression of diseases (disorders) or their symptoms, or decrease of the risk of development/expression. On the other hand, “improvement” means alleviation (moderation), change for the better, amelioration, or healing (containing partial healing).

The CAR-T cells of the present invention may be prepared in the form of a cell preparation. The cell preparation of the present invention contains the CAR-T cells of the present invention in a therapeutically effective amount. For example, 104 to 1010 cells are contained for one administration (one dose). The cell preparation may contain dimethylsulfoxide (DMSO) or serum albumin for the purpose of cell protection, antibiotics for the purpose of preventing bacterial contamination, and various components for (for example, vitamins, cytokine, growth factors, and steroids) for the purpose of cell activation, proliferation, or inductive differentiation.

The administration route of the CAR-T cells or cell preparation of the present invention is not particularly limited. For example, they are administered by intravenous injection, intraarterial injection, intraportal injection, intradermal injection, hypodermic injection, intramuscular injection, or intraperitoneal injection. Local administration may be used in place of systemic administration. Examples of the local administration include direct injection into the target tissues, body parts, and organs. The administration schedule may be made according to the sex, age, body weight, and pathology of the subject (patient). A single dose or continuous or periodical multiple doses may be used.

EXAMPLES <Study of Preparation Efficiency and Gene Introduction Efficiency of CAR-T Cells>

The CAR therapy using a transposon method is advantageous especially in terms of safety in comparison with the case using a viral vector. On the other hand, there are problems that the gene introduction efficiency is low, the cells tend to be damaged by the operation during gene introduction (for example, electroporation), and the number of cells to be obtained is small. In order to solve these problems, the following study was carried out.

1. Material

(1) Antibody

Anti-CD3 antibody (Miltenyi Biotec)

Anti-CD28 antibody (Miltenyi Biotec)

(2) Culture Medium

TexMACS (Miltenyi Biotec)

(3) Cytokine

Recombinant human IL-7 (Miltenyi Biotec)

Recombinant human IL-15 (Miltenyi Biotec)

(4) Viral Peptide Mix

PepTivator (registered trademark) CMV pp65-premium grade, human (Miltenyi Biotec)

PepTivator (registered trademark) AdV5 Hexon-premium grade, human (Miltenyi Biotec)

PepTivator (registered trademark) EBV EBNA-1-premium grade, human (Miltenyi Biotec)

PepTivator (registered trademark) EBV BZLF1-premium grade, human (Miltenyi Biotec)

(5) Plasmid

pIRII-CD19CARvector (expressing CAR)

pCMV-piggyBacvector (expressing piggyBac transposase)

(6) Cell culture vessel

24-well uncoated tissue culture plate (Falcon)

24-well tissue culture plate (Falcon)

G-Rex10 (Wilson Wolf)

2. Method

(1) Preparation of Activated T Cells

(1-1) Preparation of Anti-CD3 Antibody/Anti-CD28 Antibody Coated (Immunized) Plate

An anti-CD3 antibody and an anti-CD28 antibody were diluted with PBS to 1 mg/mL, and added to a 24-well uncoated plate at a rate of 0.5 mL/well. The plate is allowed to stand in an incubator at 37° C. for 2 to 4 hours. The PBS that diluted the antibodies is aspirated, and each well is washed once with 1 mL of PBS.

(1-2) Culturing on Anti-CD3 Antibody/Anti-CD28 Antibody Coated Plate

Day 0: The PBMC isolated from the peripheral blood is diluted to 5×105/mL with TexMACS containing 5 ng/mL of IL-15, and dispensed to the anti-CD3 antibody/anti-CD28 antibody coated plate at 2 mL to each well.

Day 1: The cells are transferred to a 24-well tissue culture plate. Half the amount of the culture solution is replaced, and IL-15 is added to 5 ng/mL.

Day 4: IL-15 is added to 5 ng/mL.

Day 7: The cells are collected, dispensed, and cryopreserved.

(1-3) Re-Stimulation of Activated T Cells

Day 0: Cryopreserved cells are unfrozen, washed twice, diluted to 1×106/mL with TexMACS™ containing IL15 at 5 ng/mL, and dispensed to an anti-CD3 antibody/anti-CD28 antibody coated plate at 2 Ml to each well.

Day 3: The cells are collected, and used for CAR-T culturing.

(2) Preparation and Culturing of CAR-T by Prior Art Method (See Culturing Method 1 and FIG. 1)

Day 0: Mononuclear cells are isolated from the peripheral blood, and counted. To 1×107 mononuclear cells, pIRII-CAR.CD19.28z vector (FIG. 4) and pCMV-pigBac vector (FIG. 5) are added in amounts of 5 μg, and the gene is introduced by electroporation (nucleofection) using 4D nucleofector (Lonza). Thereafter, the cells are floated in TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15, and culturing is started on a 24-well plate in an incubator at 37° C.

Day 1: The cells are stimulated with the anti-CD3 antibody/anti-CD28 antibody coated plate.

Day 4: The cells are transferred to G-Rex10, and cultured with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 7: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 10: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 14: Culturing is finished.

(3) Preparation and Culturing of CAR-T by Activated T Cells Addition Method (See Culturing Method 2 and FIG. 2)

Day 0: Mononuclear cells are isolated from the peripheral blood. To 1×107 mononuclear cells, pIRII-CAR.CD19.28z vector (FIG. 4) and pCMV-pigBac vector (FIG. 5) are added in amounts of 5 μg, and the gene is introduced by electroporation (nucleofection). The gene-introduced cells and 5×105 irradiated activated T cells are mixed, the mixture is floated in TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15, and culturing is started on a 24-well plate in an incubator at 37° C.

Day 1: The cells are stimulated with the anti-CD3 antibody/anti-CD28 antibody coated plate.

Day 4: The cells are transferred to G-Rex10, and cultured with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 7: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 10: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 14: Culturing is finished.

(4) Preparation and Culturing of CAR-T by Viral Peptide-Added Activated T Cells Addition Method (See Culturing Method 3 and FIG. 3)

Day 0: 5×105 activated T cells after irradiation are mixed with the viral peptides (50 ng each of PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1, and PepTivator EBV BZLF1), and incubated at 37° C. for 30 minutes. To 1×107 mononuclear cells, pIRII-CAR.CD19.28z vector (FIG. 4) and pCMV-pigBac vector (FIG. 5) are added in amounts of 5 μg, and the gene is introduced by electroporation (nucleofection). The gene-introduced cells and the irradiated viral peptide-added activated T cells are mixed, the mixture is floated in TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15, and culturing is started on a 24-well plate in an incubator at 37° C.

Day 2 to Day 5: As necessary, half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 7: The cells are collected and counted. 2×106 viral peptide-added activated T cells papered in the same manner as described above and the collected cells are floated in 30 mL of a culture solution containing IL-15 (5 ng/mL) and IL-7 (10 ng/mL), and culturing is started with G-Rex10.

Day 10: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 14: Culturing is finished.

3. Result

After completion of culturing (Day 14), the number of proliferated cells, the number of CAR-T cells, and the gene introduction efficiency (CAR-T cells/all living cells) were determined for each culturing method, and the results were compared between culturing methods 1 to 3. Measuring of the number of proliferated cells used a hemocytometer, and the number of CAR-T cells and the gene introduction efficiency were calculated from the result of flow cytometry analysis.

The number of CAR-T cells were 1.18×106 (±0.509×106) in culturing method 1, 7.13×106 (±3.25×106) in culturing method 2, and 1.09×107 (±2.98×106) in culturing method 3 (FIG. 6). The gene introduction efficiency was 3.22% (±1.42) in culturing method 1, 10.3% (±4.21) in culturing method 2, and 26.9% (±2.79) in culturing method 3 (FIG. 7). The values in the parentheses are standard errors.

The number of the donors of the peripheral blood used for the preparation of CAR-T cells (N=9) was increased, and further studied. As a result of this, the number of CAR-T cells was 2.5×106 (±0.72×106) in culturing method 1, 7.6×106 (±2.6×106) in culturing method 2, and 17.2×106 (±5.8×106) in culturing method 3 (FIG. 8). The gene introduction efficiency was 4.6% (±1.1) in culturing method 1, 10.7% (±2.7) in culturing method 2, and 33.0% (±3.4) in culturing method 3 (FIG. 9). The values in the parentheses are standard errors.

On the other hand, as a result of optimization of the plasmid (FIG. 10), in culturing method 3, the gene introduction efficiency of about 50% on average was achieved (FIG. 11).

As described above, in the novel culturing methods (culturing methods 2 and 3), in comparison with the prior art method (culturing method 1), the number of the CAR-T cells increased, and the gene introduction efficiency also increased. In culturing method 2, the increase of the number of CAR-T cells and the enhancement of the gene introduction efficiency were likely caused by the cell stimulation by the expression of costimulatory molecules (protective action on gene-introduced cells damaged during electroporation), cytokine stimulation by culturing micro-environment, and others. In culturing method 3, the increase of the number of CAR-T cells and the enhancement of the gene introduction efficiency were likely caused by the cell stimulation by the expression of costimulatory molecules, cytokine stimulation by culturing micro-environment, relatively moderate cell stimulation from the virus-specific T cells receptor, and others.

It has been found that the novel two culturing methods, more specifically, the novel culturing method (culturing method 2) using activated T cells and the novel culturing method (culturing method 3) using viral peptide-added activated T cells can solve the problems with CAR therapy using a transposon method. It is expected that the use of these culturing methods will further promote and expand the clinical application of CAR therapy. Culturing method 3 may cause the decrease of alloreactivity (the anticipated effect of virus-specific CTL), which can allow the possibility of the use of the CAR-T cells derived from the third party, and increase the internal persistency due to stimulation of the viral antigen receptor by internal viruses, and thus is expected to further promote safety and enhance the therapeutic effect.

<Improvement of Culturing Method 3>

1. Method

Day 0: Mononuclear cells (PBMCs) are isolated from the peripheral blood. A portion (1×106 PBMCs) is irradiated, mixed with the viral peptides (50 ng each of PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1, and PepTivator EBV BZLF1), and incubated at 37° C. for 30 minutes. To 1×107 PBMCs, pIRII-CAR.CD19-optimized vector (FIG. 10) and pCMV-pigBac vector (FIG. 5) are added in amounts of 5 μg, and the gene is introduced by electroporation (nucleofection). The gene-introduced cells and the irradiated viral peptide-added PBMCs are mixed, the mixture is floated on TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15, and culturing is started on a 24-well plate in an incubator at 37° C. The activated T cells are prepared from the remaining PBMCs according to the above-described method (1).

Day 2 to Day 5: As necessary, half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 7: The cells are collected and counted. On the other hand, 2×106 activated T cells prepared by the above-described method are irradiated, and then the viral peptides (each 50 ng of PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1, and PepTivator EBV BZLF1) are added, and incubated at 37° C. for 30 minutes. The 2×106 viral peptide-added activated T cells thus obtained and the collected cells are floated in 30 mL of a culture solution containing IL-15 (5 ng/mL) and IL-7 (10 ng/mL), and culturing is started with G-Rex10.

Day 10: Half the amount of the culture solution is replaced with TexMACS™ containing 10 ng/mL of IL-7 and 5 ng/mL of IL15.

Day 14: Culturing is finished.

2. Result

After completion of culturing (day 14), the number of CAR-T cells and the gene introduction efficiency (CAR-T cells/all living cells) were determined. The number of CAR-T cells and the gene introduction efficiency were 2.81×107 and 55.1% in the first experiment, 1.03×107 and 52.0% in the second experiment, and 9.22×106 and 42.3% in the third experiment. These high gene introduction efficiencies were achieved. This culturing method allows obtainment of CAR-T cells by one time of blood collection, and has an advantage that the burden on patients is reduced.

<Evaluation of Activity of CAR-T Cells>

The cytotoxic activity and antitumor activity of the CAR-T cells prepared by the novel culturing method were evaluated by the following methods.

1. Experiment of Co-Culturing with CD19-Positive Leukemia Cell Line

CAR-T cells were prepared from the peripheral blood of six normal subjects using culturing method 3. CAR-T cells (1×105) and a CD19-positive leukemia cell line (5×105) were co-cultured for seven days in the 10% fetal bovine serum-containing RPMI1640 culture medium (effector cell:target cell=1:5). The control used the activated T cells without gene introduction (T cells prepared by culturing method 3, except for the gene introduction operation) in place of CAR-T cells. The cells were collected after co-culturing for seven days, stained with an anti-CD19 antibody and an anti-CD3 antibody, and then the number of cells was counted by counting beads and a flowcytometer. The number of residual tumor cells was calculated by the following calculation formula, and the cytotoxic activity of the CAR-T cells was evaluated. The above experiment was carried out using three kinds of CD19-positive leukemia cell lines (KOPN30bi, SK-9, and TCC-Y/sr).


Standardized tumor cell residual rate (%)=100×(the number of residual tumor cells in the well co-cultured with T cells−the number of tumor cells in the well not co-cultured with T cells)/the number of tumor cells in the well not co-cultured with T cells

The result is given in FIG. 12. When co-cultured with CAR-T cells, the residual rate of KOPN30bi was 0.2% (the control was 89%), the residual rate of SK-9 was 0.03% (the control was 87%), and TCC-Y/sr was 0.03% (the control was 86%), indicating that the CAR-T cells had strong cytotoxic activity.

2. Evaluation by Tumor-Bearing Mouse

The CAR-T cells (1×107) prepared by culturing method 3 were injected into the NSG mice (five animals in each test section), to which the CD19-positive cell strain (luciferase gene-introduced Daudi, 1×106) had been injected three days before (D−3) from their tail vein, from their tail vein (DO). In the control (NT), activated T cells with no introduced gene (the T cells prepared by culturing method 3 except for the gene introduction operation) were injected. As necessary, luciferin was intraperitoneally administered, and the mouse was photographed using an in vivo imaging system.

As given in FIG. 13, proliferation of the tumor cells was not observed in the mice treated with the proliferated CAR-T cells, in contrast to the mice treated with the activated T cells with no gene introduction (control (NT)) in which the tumor cells proliferated. More specifically, the CAR-T cells strongly suppressed proliferation of tumor in the tumor-bearing mice.

3. Summary

As described above, it was confirmed that the CAR-T cells prepared by the novel culturing method exhibits high activity by the in vitro and in vivo experiments. More specifically, it was indicated that the novel culturing method is highly effective as a means for efficiently preparing CAR-T cells having high therapeutic effect.

INDUSTRIAL APPLICABILITY

The present invention increases the CAR gene introduction efficiency and the cell proliferation rate in the preparation of CAR-T cells using a transposon method. More specifically, the present invention increases practicability of the highly safe method for preparing CAR-T cells using a transposon method, thereby promoting clinical application of CAR therapy, and contributing to the increase of the therapeutic effect of CAR therapy.

The present invention is not limited only to the description of the above embodiments. A variety of modifications which are within the scopes of the following claims and which are achieved easily by a person skilled in the art are included in the present invention.

SEQUENCE LISTING

Claims

1-6. (canceled)

7. A method for preparing genetically-modified T cells expressing chimeric antigen receptor, comprising the following steps (i-a) or (i-b), and (ii) to (iv): or

(i-a) a step of preparing non-proliferative cells holding a viral peptide antigen, which are obtained by stimulating a group of cells comprising T cells using an anti-CD3 antibody and an anti-CD28 antibody followed by culturing in the presence of the viral peptide antigen and a treatment for causing the cells to lose their proliferation capability;
(i-b) a step of preparing viral peptide-holding non-proliferative peripheral blood mononuclear cells, which are prepared by culturing peripheral blood mononuclear cells in the presence of the viral peptide antigen, and a treatment for causing the cells to lose their proliferation capability; and
(ii) a step of introducing a target antigen-specific chimeric antigen receptor gene into T cells using a transposon method and thereby obtaining the genetically-modified T cells;
(iii) a step of mixing the non-proliferative cells prepared by step (i-a) or the viral peptide-holding non-proliferative peripheral blood mononuclear cells prepared by step (i-b), with the genetically-modified T cells obtained by step (ii), and co-culturing the mixed cells; and
(iv) a step of collecting the cells after culture;
wherein the step (i-a) or (i-b) is conducted in advance of step (ii).

8. The preparation method according to claim 7, wherein, after the cells are co-cultured, a step of culturing the co-cultured cells in the presence of a T-cell growth factor is carried out between step (iii) and step (iv).

9. The preparation method according to claim 7, wherein the period of the co-culturing in step (iii) is one day to 14 days.

10. The preparation method according to claim 7, wherein step (iii) is carried out in the presence of a T-cell growth factor.

11. The preparation method according to claim 10, wherein the T-cell growth factor is IL-15.

12. The preparation method according to claim 10, wherein the T-cell growth factor is a combination of IL-15 and IL-7.

13. The preparation method according to claim 1, wherein the group of cells comprising T cells is peripheral blood mononuclear cells (PBMCs).

14. The preparation method according to claim 1, wherein the treatment for losing the proliferation capability is irradiation.

15. The preparation method according to claim 1, wherein the transposon method comprises preparing a vector including the gene coding transposase and a vector having a structure wherein the gene coding a target protein is sandwiched between inverted repeat sequences, and introducing these vectors to the target cell.

16. The preparation method according to claim 1, wherein the target antigen is the CD19, GD2, GMCSF receptor or the IGF receptor.

17. The preparation method according to claim 1, wherein the non-proliferative cells or the viral peptide-holding non-proliferative peripheral blood mononuclear cells, and the genetically-modified T cells are derived from an identical individual.

18-20. (canceled)

21. The preparation method according to claim 1, which comprises step (i-a).

22. The preparation method according to claim 1, which comprises step (i-b).

Patent History
Publication number: 20230040878
Type: Application
Filed: Oct 19, 2022
Publication Date: Feb 9, 2023
Applicant: NATIONAL UNIVERSITY CORPORATION TOKAI NATIONAL HIGHER EDUCATION AND RESEARCH SYSTEM (Nagoya-shi)
Inventors: Nobuhiro Nishio (Nagoya-shi), Yozo Nakazawa (Matsumoto City), Miyuki Tanaka (Matsumoto City), Daisuke Morita (Matsumoto City), Yoshiyuki Takahashi (Nagoya-shi)
Application Number: 17/969,071
Classifications
International Classification: A61K 35/17 (20150101); C07K 14/705 (20060101); C12N 15/09 (20060101); C12N 5/0783 (20100101); C07K 14/725 (20060101); A61P 35/00 (20060101); C12N 5/16 (20060101); C07K 19/00 (20060101); C12N 5/10 (20060101); C07K 14/075 (20060101); A61K 39/00 (20060101);