Methods for Isolating and Proliferating Autologous Cancer AntiGen-Specific CD8+ T Cells

Provided is a method for isolating and proliferating autologous cancer antigen-specific CD8+T cells, and more particularly, a method for selecting an epitope recognized by CD8+ T cells from autologous cancer antigens present in blood of individual cancer patients; and isolating autologous cancer antigen-specific CD8+ T cells by using a peptide of the selected epitope, and a method of massively proliferating CD8+ T cells by using the method. According to the present invention, it is possible to isolate autologous cancer antigen-specific CD8+ T cells by using the peptide of the CD8 T cell epitope of the autologous cancer antigen present in blood of individual cancer patients instead of a heterologous antigen. Therefore, by using T cells recognizing the autologous cancer antigen, it is possible to effectively select and eliminate cancer cells derived from the cancer patient's own cells. Thus, T cells can be applied to treatment and alleviation of cancer diseases without side effects.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This U.S. non-provisional patent application claims priority under 35 U.S.C. §119 of Korean Patent Application No. 10-2014-0029198, filed on Mar. 12, 2014, the entire contents of which are hereby incorporated by reference.

STATEMENT REGARDING SEQUENCE LISTING

The Sequence Listing associated with this application is provided in text form in lieu of a paper copy, and is hereby incorporated by reference into the specification. The name of the text file containing the Sequence Listing is 510461_SequenceListing_asfiled31215.txt. The text file is 3 KB, was created on Mar. 12, 2015, and is being submitted electronically via EFS-Web, concurrent with the filing of the specification.

BACKGROUND OF THE INVENTION

The present invention relates to a method for isolating and proliferating autologous cancer antigen-specific CD8+T cells. More particularly, the present invention relates to a method for selecting an epitope recognized by CD8+ T cells from autologous cancer antigens present in blood of individual cancer patients; and isolating autologous cancer antigen-specific CD8+ T cells by using a peptide of the selected epitope, and to a method for mass proliferating CD8+ T cells by using the method.

Since CD8+ T cells have relatively simple functions than other cells such as dendritic cells, CD4+ T cells, and NK cells, it is less likely to cause unexpected side effects during anticancer immunotherapy. Generally, antigen-specific CD8+ T cells are isolated by using MHC class I/peptide multimer, but the method has a drawback in that due to the high cell death rate caused by cell apoptosis after cell isolation, a long period of culture is required to produce a sufficient amount of antigen-specific CD8+ T cells. Accordingly, needed is a surrogate marker which can isolate antigen-specific CD8+ T cells instead of MHC multimer which stimulates a T cell receptor (TCR). Thus, the present inventors have been studying for a long time about the immune regulatory protein, i.e. 4-1BB (CD137).

It has been known that 4-1BB, which is expressed in T cells activated by an inducible costimulatory molecule, particularly enhances a CD8+ T cell activity and as well as increases expression of anti-apoptotic molecules such as Bcl-2, Bcl-XL, and Bfl-1 so that activation-induced cell death (AICD) is inhibited. The characteristic of 4-1BB simulation are suitable for cancer treatment. Thus, based on the characteristic, a therapeutic effect of an anti-4-1BB mAb on a cancer is validated by using an animal model. In the previous study, the present inventors have established a method for isolating and proliferating antigen-specific CD8+ T cells by using the anti-4-1BB antibody on the basis of 4-1BB expression of activated CD8+ T cells in an antigen-specific manner (see Korean registered patent No. 10-0882445). However, in vitro and in vivo half life of an antibody are long, and the total result is shown as combination of a signal transduction result through an Fc receptor and a signal transduction effect through a target protein recognized by the antibody. In addition, in many cases, there are various antibodies for the same antigen, and the antibodies show effects having little differences from each other. To overcome this limitation, it has been developed a method for successfully isolating and proliferating antigen-specific CD8+ T cells by using the pentamer, COMP-4-1BBL protein (see Korean registered patent No. 10-1103603).

Those two patents relate to techniques for isolating/mass-culturing CD8 T cells specific for a viral antigen (e.g., EBV/LMP2A, CMV/pp65) which is a heterologous antigen, and the techniques are relatively easy to implement because the in vivo ratio of those cells are high. However, since most of cancer cells are formed by cells which compose our bodies, it is necessary to selectively isolate and mass culture CD8 T cells which recognize an autologous cancer antigen (self-tumor Ag), wherein the autologous cancer antigen is a protein to form the body and overexpressed in cancer cells while present in a low ratio in normal cells.

SUMMARY OF THE INVENTION

Thus, an object of the present invention is to provide a method for isolating and proliferating autologous cancer antigen-specific CD8+ T cells which makes it possible to selectively isolate and mass culture autologous cancer antigen-specific CD8+ T cells within 31 days, wherein, the autologous cancer antigen is present in the body in an extremely low ratio.

To achieve the object, the present invention provides a method for isolating autologous cancer antigen-specific CD8+ T cells, the method including: a) selecting a CD8+ T cell epitope of the autologous cancer antigen present in blood of a cancer patient; b) culturing a peripheral blood mononuclear cell (PBMC) isolated form blood of the cancer patient in a medium together with a peptide of the epitope and IL-2; c) inducing 4-1BB expression in the cultured cells by adding the peptide same as in step b); and d) culturing cells in which 4-1BB expression is induced on a culture plate coated with an anti-4-IBB antibody, and removing unattached cells.

In the isolation method of the present invention, the autologous cancer antigen in step a) may be selected from the group consisting of hTERT, WT1, NY-ESO1 and MAGE-A3.

In the isolation method of the present invention, the epitope in step b) may be a peptide formed by an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 15.

In the isolation method of the present invention, the induction of expression in step c) may be performed for 12 to 36 hours with culturing.

In the isolation method of the present invention, the culture in step d) may be performed for 1 to 20 minutes.

Further, the present invention provides a method for mass culturing autologous cancer antigen-specific CD8+ T cells, the method including: suspending autologous cancer antigen-specific CD8+ T cells isolated by the method and allogenic PMBCs irradiated with radiation in a medium including IL-2, an anti-CD3 antibody, and autoplasma; and then injecting the suspension into a culture bag; and additionally injecting the medium and culturing.

In the mass culture method of the present invention, the PBMC may be isolated from a healthy doner.

In the mass culture method of the present invention, the culture may be performed for 4 to 15 days.

According to the present invention, it is possible to isolate autologous cancer antigen-specific CD8+ T cells by using the peptide of the CD8 T cell epitope of the autologous cancer antigen present in blood of individual cancer patients instead of the heterologous antigen. Therefore, by using the T cell, which is isolated by the method of the present invention and recognizes the autologous cancer antigen present in an extremely low ratio in a healthy person, it is possible to effectively select and eliminate cancer cells derived from the cancer patient's own cells.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 illustrates a process of selectively isolating and mass culturing autologous cancer cell-specific CD8+ T cells according to the present invention.

FIG. 2 is a flow chart showing a process of epitope screening according to the present invention.

FIG. 3 shows an hTERT epitope screening result using PBMCs obtained from a healthy doner.

FIG. 4 shows a WT1 epitope screening result using PBMCs obtained from the healthy doner.

FIGS. 5 to 7 show hTERT epitope screening results using PBMCs respectively obtained from patients with gastric cancer, lung cancer and pancreatic cancer.

FIGS. 8 and 9 show WT1 epitope screening results using PBMCs respectively obtained from patients with glioblastoma and lung cancers.

FIGS. 10 and 11 show NY-ESO1 epitope screening results using PBMCs respectively obtained from patients with ovarian cancer and sarcoma.

FIGS. 12 and 13 show MAGE-A3 epitope screening results using PBMCs respectively obtained from patients with sarcoma and lung cancers.

FIG. 14 illustrates a pilot production process of an hTERT T cell therapeutic agent.

FIG. 15 illustrates a pilot production process of a WT1 T cell therapeutic agent.

FIG. 16 illustrates a pilot production process of an NY-ESO1 T cell therapeutic agent.

FIG. 17 illustrates a pilot production process of an MAGE-A3 T cell therapeutic agent.

DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS

Since cancer cells are derived from cells which form our body, it is necessary to selectively isolate and mass culture CD8+ T cells specific for an autologous cancer antigen (self-tumor Ag), which is overexpressed in cancer cells, to selectively eliminate cancer cells. However, a T cell, which recognizes the autologous cancer antigen, is present in an extremely low ratio in a healthy person with an activity which is inhibited by immune tolerance. Accordingly, it has not been yet developed a standardized process of selectively isolating and mass culturing autologous cancer antigen-specific CD8+ T cells from blood of a cancer patient. Thus, the present inventors have been developed a standardized technique of a process of selectively isolating and mass culturing CD8+ T cell within 31 days by using an anti-4-1BB antibody, wherein the CD8+ T cell is present in the body in an extremely low ratio and specific for an autologous cancer antigen such as hTERT, WT1, NY-ESO1, and MAGE-A3.

Therefore, the present invention provides a method for isolating autologous cancer antigen-specific CD8+ T cells. Specifically, the method for isolating autologous cancer antigen-specific CD8+ T cells of the present invention includes: a) selecting a CD8+ T cell epitope of the autologous cancer antigen present in blood of a cancer patient; b) culturing a peripheral blood mononuclear cell (PBMC) isolated from blood of the cancer patient in a medium together with a peptide of the epitope and IL-2; c) inducing 4-1BB expression in the cultured cells by adding the peptide same as in step (b); and d) culturing cells, in which 4-1BB expression is induced, on a culture plate coated with an anti-4-IBB antibody, and removing unattached cells.

In the isolation method of the present invention, the autologous cancer antigen in step a) may be any cancer antigen present in the cancer patient's own body, and a suitable autologous cancer antigen may be selected and used depending on the type of cancer. Preferably, hTERT (GenBank: BAC11010.1), WT1 (GenBank: AAO61088.1), NY-ESO1 (GenBank: CAA05908.1), and MAGE-A3 (NCBI Reference Sequence: NP005353.1), etc. may be used as a typical autologous cancer antigen used in anticancer immunotherapy. The hTERT is an enzyme for synthesizing telomeric DNA at the end of a chromosome and known as a target antigen for various solid cancers including lung cancer, gastric cancer and pancreatic cancer because cancer cells over-activate the enzyme to avoid telomerase-dependent cell death (see Kim N W, et al. Science. 1994; 266:2011-2015). Also, the WT1, which is a gene associated with Wilms tumor, encodes a zinc-finger transcriptional factor which is a protein involved in cell proliferation and differentiation, apoptosis, and development of an organ and kwon as a target antigen of brain and spinal cancer, and lung cancer, etc. (see Call K M, et al., Cell. 1990. 60:509-520; Nakahara Y, et al., Brain Tumor Pathol. 2004. 21:113-6). In addition, the NY-ESO1, one of the proteins belonging to a cancer testis antigen (CTA), has been known to be expressed in various cancer cells including germ cell cancer, sarcoma, and breast cancer; however, it has not been well known about functions of NY-ESO1 in those cells (see Gnjatic S, et al., Adv Cancer Res. 2006; 95:1-30). The MAGE-A3 is a protein belonging to melanoma-associated antigen family. Although any function of the MAGE-A3 in normal cells has not found, it has been known that MAGE-A3 is overexpressed in various cancer cells including lung cancer, sarcoma, and melanoma so that MAGE-A3 is assessed as a target antigen suitable for immunotherapy of a cancer (see Decoster L, et al., Ann Oncol. 2012 June; 23(6):1387-93).

In the isolation method of the present invention, the epitope in step b) may be a peptide formed by an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 15.

In the isolation method of the present invention, the medium in step b) may be a medium including autoplasma, and the culture in step b) may be performed for 12 to 16 days.

In the isolation method of the present invention, the induction of expression in step c) may be performed for 12 to 36 hours with culturing, and the culture in step d) may be performed for 1 to 20 minutes.

Further, the present invention provides a method for mass culturing autologous cancer antigen-specific CD8+ T cells, the method including: suspending autologous cancer antigen-specific CD8+ T cells isolated by the isolation method described above, and allogenic PMBCs irradiated with radiation in a medium including IL-2, an anti-CD3 antibody, and autoplasma, and then injecting the suspension into a culture bag; and additionally injecting the medium and culturing.

In the mass culturing method of the present invention, the PBMCs may be isolated from a normal doner, and the culture may be performed for 4 to 15 days. In particular, during the culture, the medium may be additionally injected on day 4, 7, 9, 11 and 14 of culture.

Hereinafter, the method for isolating and proliferating autologous cancer antigen-specific CD8+ T cells of the present invention will be described in stepwise.

(1) Epitope Screening (Pre-Selection Test)

According to the present invention, autologous cancer antigen-specific CD8 T cells are selectively isolated and proliferated by using a peptide. Since epitopes of an autologous cancer antigen recognized by a CD8 T cell are different depending on HLA-A types and status of individual patients, a CD8 T cell epitope of the autologous cancer antigen present in blood of individual patients is selected through epitope screening so that 3-4 types of peptides for preparing a T cell therapeutic agent are selected.

(2) Proliferation of Autologous Cancer Antigen-Specific CD8 T Cells

Since autologous cancer antigen-specific CD8 T cells are present in blood in 0.1% or less, 3-4 types of peptides for preparation and IL-2 are added to PBMCs isolated from blood, and the mixture is culture for 14 days to induce proliferation of CD8 T cells specific for a peptide derived from the autologous cancer antigen. On day 14 of culture, whole cells are collected and reactivated with the same peptides for 24 hours to induce peptide-specific CD8 T cells to simultaneously express 4-1BB.

(3) Selective Isolation of Autologous Cancer Antigen-Specific CD8 T Cells

Cells reactivated with the peptide are seeded to a culture plate coated with an anti-4-IBB antibody and cultured for 10 minutes to allow CD8 T cells expressing 4-1BB to be attached. Then, unattached cells are entirely removed through washing. Thereafter, IL-2-containing medium is added and culture is then performed for two days to proliferate the isolated T cells and also to allow the T cells to be detached from the culture plate.

(4) Mass Culture of Autologous Cancer Antigen-Specific CD8 T Cells

In a 1 L culture bag, 5×105 of isolated CD8 T cells, 1×108 of irradiated allogenic PBMC cells, 1000 U/ml of IL-2, and 40 ng of an anti-CD3 mAb are mixed, and the medium is periodically added for 14 days to culture cells to a degree of about 109 cells/L such that the cells are proliferated to a level sufficient to be administered to a cancer patient.

Hereinafter, the present invention will be described in more detail with reference to the following examples. However, these examples are only provided to more specifically describe the present invention, and the scope of the invention is not limited thereto.

EXPERIMENTAL EXAMPLE Epitope Screening Process

CD 8 T cell epitopes of an autologous cancer antigen were selected through algorithm. To evaluate a type of CD8 T cell epitope with which T cells present in blood of a cancer patient react, peripheral blood mononuclear cells (PBMCs) were isolated from blood of the cancer patient, washed, and suspended in CTL medium (RPMI1640 medium+4 mM L-glutamine+12.5 mM HEPES+50 μM 2-mercaptoethanol+3% autoplasma) to become the concentration of 1×106 cell/ml. Then, 1 ml of the suspension was aliquoted in a 14 ml round tube. Peptides for each epitope selected by the analysis with algorithm were added to each tube in the concentration of 1 μg/ml. Thereafter, culture in a CO2 incubator was started. Two days after culture, 1 ml of CTL medium including 50 U/ml IL-2 was added to each tube. On day 7, 9, 11, and 13 of culture, 1 ml of the medium was removed and CTL medium including 50 U/ml IL-2 was added. On day 14 of culture, RPMI1640 medium was added to each tube, and the tube was centrifuged for 5 minutes at 1400 rpm. Then, cells were washed three times. Washed cells were suspended in 1 ml CTL medium, and the same peptide was added in the concentration of 5 μg/ml. Thereafter, the resultant was cultured. After 24 hours, cells in each tube were collected and stained with an anti-CD8-PE-Cy5 and anti-4-1BB-PE antibodies for flow cytometry. Then, by analyzing a ratio of CD8 T cell expressing 4-1BB, a type of peptide which CD8 T cell reacted with and was thus activated has been determined. FIG. 2 is a flow chart showing an epitope screening process according to the present invention.

The anti-CD8-PE-Cy5 and anti-4-1BB-PE used in the experiment were purchased from eBioscience (San Diego, Calif., USA). RPMI1640, L-glutamine, HEPES, and 2-mercaptoethanol were purchased from Invitrogen (San Diego, Calif., USA).

Example 1 Selection of Autologous Cancer Antigen and CD8 T Cell Epitope

Based on journals (Scanlan M J, et al., Immunol Rev. 2002 October 188:22-32; Ramakrishnan S, et al., Cancer research. 1998. 58:622-625; Nakahara Y, et al., Brain Tumor Pathol. 2004. 21(3):113-6) evaluating which type of cancer antigen is suitable for immunotherapy of a cancer depending on the type of cancer, an autologous cancer antigen, which is suitable for immunotherapy of frequently occurring cancer in Korean and hard-to treat cancers (e.g., gastric cancer, lung cancer and pancreatic cancer), is selected. hTERT (GenBank: BAC11010.1), WT1 (GenBank: AAO61088.1), NY-ESO1 (GenBank: CAA05908.1), and MAGE-A3 (NCBI Reference Sequence: NP005353.1) are typical autologous cancer antigens used in anticancer immunotherapy in various ways, and types of cancers to which those four cancer antigens are applicable are selected and summarized in Table 1 below.

TABLE 1 Target antigen Patients EBV EBNA1, LMP1, LMP2 EBV-related Tumors Gastric Cancer Nasopharyngeal Carcinoma Hodgkin's lymphoma Non Hodgkin's lymphoma hTERT Lung cancer, Gastric cancer, Pancreatic cancer, melanoma, and other solid cancers WT-1 Glioblastoma, lung cancers Leukemia NY-ESO-1 Ovarian cancer, Sarcoma MAGE-3 Sarcoma, Lung cancer, Melanoma

An amino acid sequence of the selected autologous cancer antigen was analyzed through algorithm to determine an amino acid sequence which is expected as a CD8 T cell epitope (CTLPred: http://www.imtech.res.in/raghava/ctlpred/, NetCTL: http://www.cbs.dtu.dk/services/NetCTL/, SYFPEITHI: http://www.syfpeithi.de/), and a peptide of the selected epitope was chemically synthesized (Peptron Inc; www.peptron.com) and used in epitope screening. CD8 T cell epitopes selected from each autologous cancer antigen are shown in Tables 2 to 5 below.

TABLE 2 Amino acid sequence of hTERT CTL epitopes hTERT-1 AAFRALVAQCL hTERT-2 CLKELVARV hTERT-3 LAFGFALL hTERT-4 VGDDVLVH hTERT-5 FVLVAPSCA hTERT-6 GAATQARP hTERT-7 SGTRHSH hTERT-8 KEQLRPSFLLSSLRPSL hTERT-9 PLFLELL hTERT-10 AAVTPAA hTERT-11 QSIGIRQ hTERT-12 IVNMDYV hTERT-13 RPGLLGASV hTERT-14 TLTDLQP hTERT-15 LLCSLCYG hTERT-16 LVRGVPEYGCVVNLR hTERT-17 YSSYARTSIRASL hTERT-18 IYKILLLQAY hTERT-19 LGAKGAA hTERT-20 YVPLLGSL hTERT-21 QTQLSRKLP hTERT-22 ALEAAANPAL hTERT-23 ILAKFLHWL hTERT-24 RLVDDFLLV hTERT-25 EARPALLTSRLRFIPK hTERT-26 RLFFYRKSV hTERT-27 YLFFYRKSV hTERT-28 DLQVNSLQTV hTERT-29 YLQVNSLQTV hTERT-30 GLLGASVLGL hTERT-31 ALLTSRLRFI hTERT-32 RLTSRVKAL hTERT-33 TYVPLLGSL hTERT-34 CYGDMENKL hTERT-35 AYQVCGPP hTERT-36 VYGFVRACL hTERT-37 VYAETKHFL hTERT-38 DYVVGARTF

TABLE 3 Amino acid sequence of WT1 CTL epitopes HLA-A type Amino acid sequence HLA-A*02 WT1-1 ALLPAVPSL HLA-A*02 WT1-2 DLNALLPAV HLA-A*02 WT1-3 SLGEQQYSV HLA-A*02 WT1-4 RMFPNAPYL HLA-A*02 WT1-5 GVFRGIQDV HLA-A*02 WT1-6 CMTWNQMNL HLA-A*02 WT1-7 SGQFTGTAGA HLA-A*02 WT1-8 VLDFAPPGA HLA-A*24 WT1-9 APGCNKRYF HLA-A*24 WT1-10 QYRIHTHGVF HLA-A*24 WT1-11 AFTVHFSGQF HLA-A*24 WT1-12 RWPSCQKKF HLA-A*24 WT1-13 RVPGVAPTL HLA-A*24 WT1-14 DFKDCERRF HLA-A*24 WT1-15 RTPYSSDNL HLA-A*24 WT1-16 TSEKPFSCR HLA-A*24 WT1-17 FSRSDQLKR HLA-A*24 WT1-18 LSHLQMHSR

TABLE 4 Amino acid sequence of NY-ESO-1 CTL epitopes HLA-A type Amino acid sequence HLA-A*02 NY-1 SLAQDAPPL HLA-A*02 NY-2 SISSCLQQL HLA-A*02 NY-3 LLMWITQCFL HLA-A*02 NY-4 RLLEFYLAM HLA-A*02 NY-5 DAPPLPVPGV HLA-A*02 NY-6 TVSGNILTI HLA-A*02 NY-7 QLQLSISSCL HLA-A*02 NY-8 GVLLKEFTV HLA-A*02 NY-9 ILTIRLTAA HLA-A*02 NY-10 SLLMWITQC HLA-A*24 NY-11 EFTVSGNIL HLA-A*24 NY-12 SGLNGCCR HLA-A*24 NY-13 SSCLQQLSL HLA-A*24 NY-14 FATPMEAEL HLA-A*24 NY-15 ITQCFLPVF HLA-A*24 NY-16 LTAADHRQL HLA-A*24 NY-17 YLAMPFATPM HLA-A*24 NY-18 ATPMEAELAR HLA-A*24 NY-19 ASGPGGGAPR HLA-A*24 NY-20 PVPGVLLKEF

TABLE 5 Amino acid sequence of MAGE-A3 CTL epitopes HLA-A type Amino acid sequence HLA-A*02 M3-1 ALSRKVAEL HLA-A*02 M3-2 LLIIVLAII HLA-A*02 M3-3 GLLIIVLAI HLA-A*02 M3-4 KIWEELSVL HLA-A*02 M3-5 ILGDPKKLL HLA-A*02 M3-6 TLVEVTLGEV HLA-A*02 M3-7 ALVETSYVKV HLA-A*02 M3-8 AALSRKVAEL HLA-A*02 M3-9 LVFGIELMEV HLA-A*02 M3-10 SLPTTMNYPL HLA-A*24 M3-11 SYPPLHEWVL HLA-A*24 M3-12 YIFATCLGL HLA-A*24 M3-13 VFEGREDSIL HLA-A*24 M3-14 EGLEARGEAL HLA-A*24 M3-15 TFPDLESEF HLA-A*24 M3-16 EFLWGPRAL HLA-A*24 M3-17 VAELVHFLL HLA-A*24 M3-18 IFSKASSSL HLA-A*24 M3-19 KVLHHMVKI HLA-A*24 M3-20 VDPIGHLYI HLA-A*24 M3-21 IMPKAGLLI HLA-A*24 M3-22 SILGDPKKL HLA-A*24 M3-23 VKISGGPHI HLA-A*24 M3-24 LGLSYDGLL

Example 2 Epitope Screening on Clinical Cancer Patient

To evaluate whether CD8 T cell epitopes of autologous cancer antigens, i.e. hTERT, WT1, NY-ESO1, and MAGE-A3, selected in Example 1 substantially induce proliferation of CD8 T cells present in blood of a clinical cancer patient, epitope screening as depicted in FIG. 2. was performed. hTERT epitope screening was performed on gastric cancer, lung cancer and pancreatic cancer, as a main subject matter. Also, WT1 epitope screening was performed on brain and spinal cancer and lung cancer; NY-ESO1 epitope screening was performed on ovarian cancer and sarcoma; and MAGE-A3 epitope screening was performed on sarcoma and lung cancer as a main subject matter.

FIG. 3 shows an hTERT epitope screening result using PBMCs obtained from a healthy doner.

FIG. 4 shows a WT1 epitope screening result using PBMCs obtained from the healthy doner.

As shown in FIGS. 3 and 4, CD8 T cell epitopes of hTERT and WT1 did not induce T cell response by PBMCs derived from blood of the healthy doner. Thus, it has been found that the selected epitope of the present invention cannot be recognized by T cells of the healthy doner.

FIGS. 5 to 7 show hTERT epitope screening results using PBMCs respectively obtained from patients with gastric cancer, lung cancer and pancreatic cancer.

FIGS. 8 and 9 show WT1 epitope screening results using PBMCs respectively obtained from patients with glioblastoma and lung cancers.

FIGS. 10 and 11 show NY-ESO1 epitope screening results using PBMCs respectively obtained from patients with ovarian cancer and sarcoma.

FIGS. 12 and 13 show MAGE-A3 epitope screening results using PBMCs respectively obtained from patients with sarcoma and lung cancers.

As shown in FIGS. 5 to 13, as a result of investigating reactivity of CD8 T cells on hTERT, WT1, NY-ESO1, and MAGEA3 by performing epitope screening on PBMCs isolated from blood of the clinical cancer patient, it has been found that in contrast with the result in the health doner, high degree of T cell response was exhibited to the selected autologous cancer antigens. Reactivity on each autologous cancer antigen was then evaluated by repeatedly performing epitope screening on gastric cancer, lung cancer, sarcoma, and ovarian cancer.

In addition, to objectively analyze the epitope screening result, a scoring system was made as shown in Table 6 below.

TABLE 6 The ratio of The ratio of CD8T cells Score 4-1BB-CD8·T cells (compare with average ratio of CD8·T) 0  0-3% ≦1.0 fold 1  0-3% ≧1.0 fold 2 4-10% ≧1.0 fold 3 11-15%  ≧1.0 fold 4 16-20%  ≧1.0 fold 5 ≧20% ≧1.0 fold Although CD8·T cells express the 4-1BB on their surface, in case that the percentage of CD8·T cells is lower than average ratio one point will be deducted from final scores.

According to the criteria in Table 6 above, the epitope screening results were analyzed, and the analyzed results were shown in Tables 7 to 15 below.

Table 7 below shows the result of analyzing hTERT epitope screening using PBMCs obtained from a patient with gastric cancer.

TABLE 7 # Peptide Patients 11 12 13 14 15 16 17 18 19 10 11 12 13 14 15 16 17 18 19 20 21 1 KYH 2 1 2 1 2 1 0 2 2 1 6 2 0 2 1 1 1 1 1 2 1 2 SKP 1 1 1 2 1 0 1 1 1 1 1 1 1 1 1 1 1 1 6 1 1 3 KOS 3 2 2 2 1 2 2 2 2 2 2 2 1 2 2 2 2 3 2 2 2 4 JHT 1 0 1 1 1 1 2 1 1 0 1 1 1 1 1 0 1 1 1 1 0 5 AHS 4 1 0 1 0 0 0 0 3 0 0 2 0 4 1 3 3 2 2 1 2 6 POH 2 1 2 1 2 1 1 3 1 1 1 2 1 1 1 0 2 2 3 2 1 7 LHK 4 2 2 4 2 3 2 2 4 4 2 2 3 1 4 4 3 4 4 3 2 8 LOK 4 3 4 3 3 3 2 2 3 3 4 3 4 3 3 3 3 3 3 4 4 9 LMY 4 0 1 1 1 3 1 1 2 1 1 1 2 1 3 3 2 0 2 2 1 10 KYI 3 1 3 2 1 4 4 1 4 0 1 1 1 2 0 4 0 2 2 2 0 11 LJY 2 1 1 1 1 2 1 1 2 1 1 1 1 2 1 1 1 0 2 1 1 12 JOH 2 1 1 3 3 1 1 1 1 2 2 2 3 2 3 1 0 3 2 0 1 13 OYK 2 3 1 3 0 0 3 2 2 2 4 1 3 1 0 1 1 1 3 2 0 14 HSY 2 1 3 3 3 2 3 2 2 3 2 2 3 2 2 1 3 3 2 3 1 15 CHS 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 4 5 5 5 5 16 KWK 0 0 0 3 1 0 1 1 2 2 0 3 0 3 2 3 0 0 0 0 2 17 KTG 2 1 1 3 2 2 2 2 1 3 3 3 3 3 1 1 2 0 3 3 1 18 YSM 1 2 2 1 1 1 1 1 1 3 3 1 3 3 3 1 1 1 1 3 3 19 KIS 4 5 4 5 5 5 5 4 5 5 5 5 5 5 4 4 4 4 2 5 5 20 JHK 2 0 0 0 0 1 5 0 0 0 0 0 0 0 0 0 0 0 3 0 0 21 PBO 1 4 1 1 1 3 2 5 2 1 3 2 1 1 1 1 1 3 1 2 2 22 MSY 3 1 1 1 3 2 3 3 3 3 2 0 5 1 1 1 3 0 3 4 2 23 KDH 5 4 2 2 1 1 2 1 5 1 3 3 1 1 5 3 2 2 1 1 1 24 PGS 2 5 1 3 0 3 0 0 3 3 2 0 0 4 3 1 0 1 1 1 3 25 KJO 2 0 2 0 1 0 2 0 1 2 0 0 2 1 2 0 0 1 0 3 2 26 BUS 0 4 2 0 3 0 0 0 3 2 0 0 2 5 1 0 0 0 2 0 5 27 OYL 0 3 0 0 0 0 0 0 0 3 0 0 0 0 0 0 3 0 0 0 0 28 YBS 3 1 0 0 0 4 0 0 0 3 2 3 0 0 3 3 0 0 0 2 0 29 OJI 1 4 0 0 0 3 3 0 1 0 0 0 2 0 2 0 1 3 1 0 1 30 IJI 3 3 4 4 3 1 3 3 4 3 0 4 2 3 1 3 1 4 1 1 4 31 YSJ 1 1 1 1 0 1 1 2 1 2 0 1 0 1 1 1 1 1 1 1 1 32 LKS 0 2 0 0 0 3 0 0 4 0 0 0 0 0 0 1 0 2 1 1 1 33 NKO 1 1 2 0 1 2 2 0 2 0 2 2 1 0 1 1 1 0 2 1 0 34 LMS 1 0 2 0 1 0 1 2 2 2 0 0 0 0 0 1 0 0 0 1 3 35 YOK 3 1 3 1 1 2 1 1 3 1 1 3 2 1 2 3 3 3 2 1 2 36 AKJ 2 2 2 2 2 2 3 4 2 5 2 2 2 2 3 2 2 2 2 2 2 37 PIM 2 1 4 1 1 1 3 2 4 3 2 2 1 3 1 3 1 1 2 3 1 38 KYS 2 2 2 4 2 2 2 1 1 2 2 2 3 3 4 1 1 1 2 3 2 39 JJM 2 3 3 3 3 5 2 3 3 4 2 3 3 2 3 3 3 2 2 3 2 40 NMO 4 3 4 5 3 2 2 3 4 5 2 4 3 5 4 4 3 4 2 4 2 41 SSS 2 1 2 1 1 2 3 3 3 3 2 3 2 2 2 1 1 2 3 3 3 42 YKJ 0 2 0 0 2 2 4 0 2 5 2 3 1 1 4 2 2 4 1 4 2 43 BJK 1 0 0 3 0 5 0 1 2 1 0 0 0 0 0 1 1 0 0 0 0 44 IYS 3 1 2 0 1 1 0 1 0 0 0 0 0 0 1 0 0 1 0 1 1 45 JJN 2 2 3 3 3 2 3 2 2 1 3 3 3 3 2 3 3 2 3 3 2 46 HYJ 1 5 5 4 4 4 5 1 4 5 4 5 4 5 5 4 1 3 2 5 4 47 NKS 2 0 0 0 0 0 0 0 0 1 3 2 0 3 0 0 0 0 0 0 0 48 YOK 0 0 0 0 1 1 2 0 0 2 2 2 0 0 0 1 0 1 0 1 0 49 MKS 0 0 2 0 0 3 2 3 4 1 1 0 0 1 0 0 0 3 0 2 0 50 PJS 1 0 1 1 0 1 1 1 0 0 0 0 0 0 0 1 1 0 1 0 0 # Peptide Patients 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 1 KYH 1 2 1 1 0 1 6 2 1 1 1 1 1 2 1 1 2 2 SKP 1 1 1 1 1 1 2 0 0 0 1 0 1 0 0 0 0 3 KOS 2 3 2 0 1 2 1 1 2 2 1 0 0 0 0 1 1 4 JHT 0 1 1 0 1 0 0 0 0 0 1 1 1 0 0 0 1 5 AHS 2 3 1 2 3 2 3 0 0 1 1 0 0 1 0 0 1 6 POH 2 1 1 2 1 0 1 1 2 1 2 2 1 0 1 0 2 7 LHK 3 2 2 4 2 3 1 3 3 2 1 0 2 3 1 1 0 8 LOK 3 3 3 2 3 3 3 4 2 2 4 3 3 2 3 3 4 9 LMY 1 2 4 1 0 1 3 1 0 0 1 2 2 2 1 1 0 10 KYI 0 2 2 1 3 4 2 3 3 2 1 1 3 1 0 1 2 11 LJY 1 1 1 1 1 1 0 0 1 2 2 2 1 1 0 2 1 12 JOH 1 1 1 1 2 3 3 2 1 1 1 0 0 1 1 0 1 13 OYK 0 4 1 1 3 3 0 1 2 2 2 1 0 1 1 0 1 14 HSY 3 2 1 2 1 2 3 3 2 1 2 3 1 2 0 2 2 15 CHS 4 4 5 3 4 4 4 3 4 4 2 4 2 3 3 3 4 16 KWK 1 1 0 1 2 2 1 3 3 1 0 2 1 1 1 1 1 17 KTG 1 2 2 2 2 1 1 2 1 1 0 1 3 1 2 1 1 18 YSM 3 1 1 1 1 1 1 1 1 1 1 1 0 0 1 1 1 19 KIS 4 4 5 4 5 5 5 4 5 5 3 4 4 4 5 4 4 20 JHK 0 0 5 0 0 0 3 0 0 0 0 1 1 0 0 0 0 21 PBO 2 3 2 1 0 3 2 2 1 3 2 1 0 0 1 2 2 22 MSY 1 0 1 1 3 4 2 2 1 1 0 1 2 1 0 2 1 23 KDH 1 4 4 2 2 1 3 2 1 1 1 2 4 1 2 2 1 24 PGS 3 0 0 2 1 1 1 1 2 2 4 2 1 1 1 2 2 25 KJO 1 3 1 2 1 2 2 0 1 2 2 2 1 1 2 2 1 26 BUS 3 0 0 0 0 0 0 2 1 0 4 2 2 1 2 1 1 27 OYL 0 0 4 0 0 0 0 0 0 0 0 0 3 0 0 0 0 28 YBS 3 1 0 0 0 1 0 0 0 1 0 0 1 2 3 0 0 29 OJI 1 0 0 0 1 1 1 0 0 1 0 1 2 2 1 0 1 30 IJI 4 1 3 1 2 0 0 1 2 1 2 0 3 0 0 1 2 31 YSJ 1 1 1 0 1 1 2 1 0 0 1 1 1 1 0 1 1 32 LKS 2 3 3 0 1 2 2 0 4 2 3 1 1 0 1 1 2 33 NKO 2 1 1 1 1 0 0 1 1 1 1 1 2 0 1 0 1 34 LMS 0 1 1 1 1 0 0 0 1 1 3 1 2 1 0 1 2 35 YOK 1 2 3 0 1 1 2 0 1 2 3 3 1 2 2 0 1 36 AKJ 2 2 2 1 1 1 1 2 1 0 0 1 2 1 2 1 1 37 PIM 3 1 1 2 2 1 3 4 1 0 0 1 2 3 2 3 1 38 KYS 3 3 3 1 0 1 2 2 1 4 3 3 2 1 3 1 1 39 JJM 2 2 1 2 2 1 1 1 2 3 3 2 3 2 2 1 2 40 NMO 4 3 3 2 1 1 3 3 4 2 1 1 1 2 2 1 2 41 SSS 2 2 3 2 1 1 2 2 3 2 1 1 3 3 2 1 1 42 YKJ 3 3 3 1 1 2 2 1 0 1 0 3 1 1 0 1 1 43 BJK 1 5 0 1 0 0 1 2 3 2 3 1 0 1 1 1 0 44 IYS 0 1 1 0 0 1 1 0 1 1 1 1 1 1 0 0 1 45 JJN 3 2 2 2 3 3 1 1 0 1 2 2 3 1 1 2 3 46 HYJ 4 0 4 3 3 4 4 4 1 1 2 3 2 3 2 1 3 47 NKS 0 2 0 2 0 0 0 1 0 0 0 0 0 0 0 0 0 48 YOK 2 0 0 0 0 0 0 0 2 0 0 2 2 1 0 1 0 49 MKS 1 0 3 1 0 2 1 0 0 5 0 0 2 1 2 0 1 50 PJS 0 0 0 0 0 0 2 0 1 1 0 0 0 0 0 0 1

Table 8 below shows the result of analyzing hTERT epitope screening using PBMCs obtained from a patient with lung cancer.

TABLE 8 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 1 BGO 0 0 0 0 0 1 1 1 0 0 2 1 2 1 2 2 1 0 0 1 0 0 0 0 0 2 LCS 1 0 0 0 0 0 0 1 0 0 1 1 0 0 0 0 4 0 1 0 0 0 1 0 0 3 KKH 3 2 4 1 4 1 1 1 1 0 1 1 1 1 1 1 1 2 2 2 1 1 1 1 1 4 NSD 1 2 4 1 2 3 2 2 1 2 2 2 2 2 2 3 2 2 1 1 2 1 2 1 1 5 CHS 3 3 3 3 2 3 3 0 3 3 2 2 3 3 3 1 0 3 3 3 2 3 0 4 3 6 KCH 1 0 2 2 0 0 0 0 1 2 0 0 1 0 0 0 0 0 0 0 1 0 1 0 0 7 CMR 0 1 1 3 0 0 1 2 0 1 2 0 1 0 0 1 2 2 1 2 1 3 1 1 4 8 HJS 0 0 1 0 2 2 2 1 2 0 0 1 0 0 0 0 0 2 0 0 0 0 0 0 0 9 PBS 3 0 0 1 1 1 3 2 3 1 0 0 2 1 3 4 2 3 1 2 4 1 3 1 1 10 KSH 5 3 2 1 1 3 2 2 4 3 1 1 3 1 1 5 4 1 3 1 2 3 0 3 3 11 LIS 1 1 1 0 0 0 0 1 0 0 0 0 0 0 1 1 1 0 0 0 1 1 1 0 1 12 LKS 1 1 3 2 3 2 1 1 2 2 0 1 1 2 1 5 0 2 2 3 3 1 3 4 2 13 LYS 2 1 2 2 2 2 2 2 0 2 1 0 0 0 0 2 2 1 1 2 1 0 0 1 2 14 SMS 0 0 1 0 1 0 1 2 0 5 0 1 1 0 2 3 2 2 0 0 2 0 2 0 1 15 KHS 0 0 0 0 1 1 1 1 1 0 1 1 0 0 1 1 1 1 1 1 0 1 0 1 1 16 LKT 0 1 2 1 0 0 2 0 0 2 2 2 0 2 2 1 0 0 1 1 0 0 0 0 0 17 EHR 1 0 2 3 1 2 3 0 1 4 0 1 0 0 1 0 0 3 4 1 1 2 2 1 5 18 KYH 4 3 4 4 4 4 4 2 3 5 1 5 1 4 3 2 2 3 3 1 1 1 1 3 2 19 CEH 1 2 0 0 0 0 0 0 2 1 4 0 0 0 0 0 0 0 0 0 1 0 0 2 2 20 KSJ 0 0 1 0 1 0 2 2 1 1 2 2 1 0 1 0 0 2 1 1 0 0 0 0 0

Table 9 below shows the result of analyzing hTERT epitope screening using PBMCs obtained from a patient with pancreatic cancer.

TABLE 9 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 1 KEH 2 4 4 5 4 4 4 5 5 5 4 5 3 5 5 4 4 4 3 4 3 2 4 4 2 2 LSS 1 2 1 1 1 1 2 0 0 1 2 1 1 1 1 0 2 1 1 1 1 2 1 1 1 3 KKS 1 1 0 0 1 1 0 1 0 5 1 5 0 0 0 2 1 0 1 4 0 1 3 1 1 4 KBN 1 1 1 1 1 1 2 1 1 1 1 1 2 2 0 2 1 1 1 1 1 1 2 1 1 5 JKK 4 1 1 4 2 1 0 1 5 4 0 1 0 4 3 1 2 1 2 2 0 2 0 2 2 6 HIS 1 1 1 1 1 1 1 1 1 2 2 1 2 2 5 1 1 0 0 0 3 1 1 0 0 7 PSN 1 1 1 0 1 1 0 0 0 0 0 0 1 1 0 0 0 1 0 0 1 1 0 0 1 8 LJO 1 1 1 1 0 0 0 1 1 1 1 2 1 1 3 2 4 0 0 1 2 0 0 0 1 9 SSH 1 2 4 2 2 3 1 1 0 5 1 1 1 3 1 3 2 2 3 2 2 2 1 1 0 10 KBM 0 0 1 2 1 1 2 3 2 1 1 1 0 2 2 0 2 1 1 1 0 1 1 2 2 11 JTJ 3 1 1 0 1 1 2 1 2 3 0 1 1 0 0 3 3 0 1 1 0 1 0 4 2 12 KKH 0 0 1 0 0 1 0 0 0 0 0 0 0 2 0 0 2 2 1 0 0 0 0 0 2 13 PBH 5 1 1 0 0 1 0 1 1 3 1 0 0 0 0 2 1 1 0 2 1 0 0 1 0 14 LSB 1 1 3 3 0 1 5 0 2 4 1 0 3 1 1 3 1 2 4 2 1 1 4 4 2 15 KMY 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 16 KCJ 2 1 0 1 1 1 1 1 1 1 1 1 0 3 0 2 2 0 2 0 1 2 0 0 0 17 CSH 2 1 3 4 4 4 3 2 2 2 1 1 3 3 3 4 3 2 2 2 2 2 1 2 1 18 CH 4 4 5 5 4 5 4 4 5 5 1 4 4 5 4 2 3 3 3 4 4 4 3 4 4 19 HKS 1 0 1 1 1 0 1 1 1 0 1 0 1 0 0 1 1 0 1 0 1 0 1 1 1 20 YSC 1 1 0 2 0 0 0 0 1 3 0 0 1 0 0 1 0 1 0 1 1 1 1 1 1

Table 10 below shows the result of analyzing WT1 epitope screening using PBMCs obtained from a patient with glioblastoma.

TABLE 10 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 1 KSK 5 5 5 2 2 5 5 5 2 2 3 0 3 3 2 5 4 5 5 2 PJY 3 1 1 2 2 1 0 2 0 1 0 0 1 3 1 3 2 2 2 1 3 JJY 3 1 1 1 1 1 1 1 5 3 2 2 2 2 2 2 2 2 3 1 4 OJH 3 3 2 2 2 2 1 4 3 1 2 2 3 1 1 3 3 2 5 3 5 KYA 0 0 1 1 0 1 1 1 3 1 0 0 0 0 1 0 0 1 1 0 6 YSB 0 0 0 2 1 0 0 0 0 5 5 0 3 0 1 0 0 0 0 1 7 KKJ 1 1 2 1 1 2 1 1 1 1 1 2 2 2 1 2 2 1 2 1 8 NKH 2 3 1 2 3 2 2 3 2 2 1 1 1 3 3 4 4 2 2 3 9 AKM 0 0 0 0 1 1 0 2 1 0 0 1 1 0 1 1 0 1 1 0 10 HIS 1 1 0 1 1 1 0 1 1 1 1 0 0 1 0 0 0 0 0 1 11 CHY 4 3 4 3 2 3 2 4 5 4 3 2 2 3 2 3 2 3 3 3 12 JYS 3 4 2 2 4 5 2 4 4 4 2 3 2 3 3 4 4 2 3 3 13 HIS 2 1 1 1 1 0 1 1 1 2 0 0 1 0 1 0 0 1 0 2 14 ABS 2 2 2 1 4 1 1 4 3 3 4 1 1 1 2 4 2 2 2 2 15 LYJ 3 2 2 2 2 0 1 2 1 2 3 1 1 3 0 1 1 1 3 0 16 SBS 1 2 0 0 1 1 0 1 2 1 0 0 0 2 2 1 2 2 1 1 17 PJB 2 2 2 1 4 2 1 1 1 1 1 1 1 1 1 3 1 1 1 1 18 JJS 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 19 LYH 1 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 1 1 20 SSO 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

Table 11 below shows the result of analyzing WT1 epitope screening using PBMCs obtained from a patient with lung cancer.

TABLE 11 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 1 BGO 2 1 0 0 1 0 2 2 3 1 1 1 0 0 2 4 1 1 1 1 2 LCS 1 0 0 1 0 0 0 0 0 1 1 0 1 0 0 0 0 1 0 0 3 KKH 0 1 1 2 2 0 2 2 4 5 2 1 1 1 1 1 3 1 2 1 4 NSD 2 1 2 1 2 1 1 2 2 1 2 3 3 3 3 2 3 1 1 1 5 CHS 3 4 3 3 3 3 3 3 0 2 2 1 4 4 3 5 3 3 4 3 6 KCH 0 0 1 1 2 2 1 1 1 1 1 0 0 0 0 0 0 1 0 2 7 CMR 2 0 2 2 1 3 0 0 0 1 0 0 3 0 0 0 1 0 0 0 8 HJS 0 0 0 1 1 2 1 1 1 4 0 0 0 1 0 1 0 0 0 3 9 PBS 0 2 0 2 0 0 0 2 0 0 2 3 3 1 3 2 3 1 1 3 10 KSH 3 3 3 3 3 1 3 3 4 3 2 2 1 1 2 3 3 3 3 3 11 LJS 1 1 1 1 0 0 1 1 1 0 1 1 0 0 0 0 0 1 1 1 12 LKS 2 2 2 1 3 3 2 1 2 0 2 2 2 4 4 1 0 4 2 0 13 LYS 0 3 2 0 1 1 3 0 1 1 1 1 0 0 0 1 0 0 1 1 14 SMS 2 1 1 2 1 1 1 1 4 0 1 0 1 1 1 1 1 1 1 1 15 KHS 1 1 1 0 0 1 0 1 1 1 1 1 1 1 0 1 1 1 1 1 16 LKT 2 0 0 0 0 1 0 1 0 1 0 0 2 1 1 0 0 5 1 0 17 EHR 2 1 0 3 2 1 1 1 3 1 4 2 1 2 1 1 3 0 3 2 18 KYH 4 3 3 3 3 4 3 3 3 3 1 1 1 2 3 2 2 1 1 3 19 CEH 0 1 2 0 0 0 0 0 2 0 0 0 0 0 0 1 0 0 0 2 20 KSJ 2 1 1 2 1 0 3 1 1 5 3 0 0 1 2 1 1 0 1 1

Table 12 below shows the result of analyzing NY-ESO1 epitope screening using PBMCs obtained from a patient with ovarian cancer.

TABLE 12 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 1 CEH 0 0 0 1 0 0 2 1 2 0 2 2 0 1 1 0 0 2 1 0 2 LBS 1 1 0 1 1 0 0 1 1 0 0 1 0 1 0 0 1 2 0 0 3 KSY 1 0 3 2 3 1 1 2 0 2 1 2 1 1 0 1 1 0 0 1 4 KMS 1 1 1 2 2 3 4 2 4 1 5 1 1 3 1 2 4 3 1 1 5 HSY 0 0 0 0 0 0 0 0 2 2 0 0 1 0 0 0 0 1 0 3 6 KHJ 1 2 3 1 1 1 2 1 1 3 3 1 2 2 3 1 1 1 1 2 7 LKS 1 0 5 1 1 1 1 1 1 5 3 2 1 1 4 1 1 1 5 1 8 KHM 1 1 5 3 0 1 2 0 2 1 1 0 0 2 0 1 1 1 0 3 9 JMJ 2 1 1 2 0 0 0 1 1 1 1 0 1 0 0 0 0 1 1 0 10 YSG 1 0 1 1 1 0 0 0 1 1 0 1 1 1 1 0 0 1 1 0 11 KMH 1 0 2 2 0 2 0 2 1 2 0 2 1 1 1 2 0 2 2 2 12 JYS 3 3 2 3 2 4 2 2 3 1 3 2 2 1 1 4 2 2 2 2 13 PJS 0 0 1 0 1 0 2 1 2 1 1 0 0 1 1 0 3 0 0 0 14 SSS 0 2 1 2 2 3 1 0 0 0 0 0 0 2 0 0 1 1 0 0 15 HYH 0 2 0 2 0 1 2 0 2 2 0 0 0 0 1 1 0 1 2 0 16 KHS 3 3 3 3 5 5 4 4 3 3 5 5 2 2 4 3 3 2 4 4 17 KYS 0 1 5 3 1 0 1 2 0 2 1 2 0 2 0 4 0 1 2 2 18 YEK 1 1 2 0 0 3 0 5 2 1 1 2 3 4 0 2 0 4 0 5 19 LYJ 1 1 2 2 1 2 2 2 1 2 1 0 1 1 0 1 0 2 1 1 20 KKS 0 1 0 0 0 1 0 0 2 3 1 0 0 0 1 2 2 2 1 3 21 HHS 0 0 1 0 0 2 2 0 1 0 0 0 0 1 2 1 1 1 0 0 22 LKS 1 2 4 2 0 1 2 1 1 1 2 0 2 2 0 1 2 1 0 1 23 PSD 0 0 0 1 0 1 1 0 0 0 0 0 0 0 0 0 0 0 0 0 24 SES 3 1 0 0 0 0 1 2 2 1 2 0 1 0 0 0 2 0 0 0 25 JHY 2 1 2 1 1 2 2 1 2 2 2 2 1 1 2 2 2 1 1 1 26 KKM 1 0 1 3 3 4 0 1 3 2 3 1 1 2 2 1 3 3 4 2 27 KYJ 2 2 2 3 2 2 1 2 1 2 2 2 1 1 0 1 1 2 0 0 28 KGH 5 5 5 4 4 5 5 4 5 4 4 5 4 4 4 5 4 5 5 4 29 KSK 1 1 4 1 3 1 1 1 2 3 3 1 2 2 2 1 1 2 0 1 30 SJH 5 2 1 3 2 5 4 4 3 0 4 1 3 2 1 4 2 2 5 2

Table 13 below shows the result of analyzing WT1 epitope screening using PBMCs obtained from a patient with sarcoma.

TABLE 13 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 1 L S 1 3 5 1 1 1 1 2 1 1 1 1 2 1 1 2 2 2 1 1 2 SJV 3 3 3 4 5 1 0 3 1 1 1 1 3 2 1 3 4 4 1 1 3 AVS 1 1 2 2 1 1 2 1 2 4 2 2 1 1 2 1 2 3 1 1 4 KDS 1 1 1 1 1 1 1 1 1 1 1 1 1 1 2 1 1 3 1 1 5 LSM 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 2 0 6 CSK 0 2 1 1 1 0 0 0 0 1 0 0 0 1 1 1 2 1 0 2 7 JJS 2 1 0 2 4 3 1 2 2 1 3 0 4 2 2 0 2 1 1 3 8 PVS 1 1 3 2 0 0 2 1 1 1 0 1 1 1 1 1 0 1 0 0 9 LYS 1 1 5 1 1 1 1 1 1 2 4 4 1 0 2 3 1 1 1 0 10 KYS 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 11 VIH 0 0 0 0 0 0 0 0 0 0 0 0 3 0 0 0 0 0 0 0 12 KVC 1 1 0 1 1 0 0 0 0 3 0 1 0 0 0 1 1 1 1 0 13 LSH 1 2 3 1 0 0 1 1 1 0 0 0 0 0 1 2 1 0 0 1 14 ESJ 0 1 1 1 1 0 0 0 0 0 0 0 1 0 0 0 0 0 1 0 15 HSH 0 0 2 0 0 0 0 1 0 0 0 0 0 0 1 0 0 0 0 0 16 PCI 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 17 KJS 2 2 5 2 2 0 1 1 1 3 0 0 1 0 2 2 1 1 1 1 18 JBK 1 1 1 0 1 0 1 0 1 1 1 1 1 1 1 1 0 1 0 1 19 CYR 2 0 2 1 1 2 0 1 1 3 2 1 0 1 0 5 2 3 0 0 20 KNY 0 1 0 1 1 2 1 0 2 0 1 1 0 0 1 1 0 2 2 1 21 KKC 4 3 2 3 3 3 5 5 5 4 3 2 5 3 5 2 5 4 3 2 22 KMS 1 0 5 5 3 5 2 2 1 1 3 3 0 0 3 0 0 3 2 1 23 KMJ 1 1 5 1 2 1 1 1 0 3 0 1 0 0 1 1 0 1 1 0 24 LMJ 0 0 2 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 25 PHS 0 0 2 4 2 5 1 3 1 3 1 1 1 1 5 2 2 1 1 4 26 VSH 2 1 2 2 1 1 1 1 3 5 2 2 0 1 1 2 1 2 1 1 27 HSW 0 1 5 1 2 1 3 0 3 2 1 2 0 2 0 0 1 1 0 1 28 SSB 1 0 1 2 2 2 2 2 1 1 1 2 1 2 1 1 2 1 2 2 29 JSH 0 0 0 0 0 0 1 1 1 1 0 0 0 0 1 1 0 0 2 1 30 RCH 2 3 2 2 1 2 3 1 3 1 3 1 2 4 1 2 1 1 1 3 indicates data missing or illegible when filed

Table 14 below shows the result of analyzing MAGE-A3 epitope screening using PBMCs obtained from a patient with sarcoma.

TABLE 14 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 1 LVS 1 2 1 0 1 1 1 1 2 1 1 0 1 1 1 1 0 2 2 2 2 1 1 1 2 SJY 3 1 0 2 3 3 0 2 1 1 2 5 1 4 3 5 0 0 1 1 4 4 1 1 3 AVS 1 2 3 2 1 1 1 3 2 2 1 0 3 2 2 2 3 4 2 1 2 3 2 1 4 KDS 0 1 1 1 1 2 2 2 1 0 1 0 4 1 2 3 1 0 1 1 0 1 2 5 5 ESM 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 6 CSK 2 0 1 0 0 0 0 0 1 1 0 0 0 1 0 1 0 0 1 1 1 0 1 1 7 JJS 1 0 2 1 0 3 3 3 4 3 0 1 1 1 2 2 0 1 0 2 0 0 1 1 8 PYS 1 1 0 1 1 1 1 0 1 2 1 0 1 0 0 0 0 0 2 1 2 1 0 0 9 LYS 3 1 4 0 1 2 0 1 1 1 1 1 0 0 3 0 1 1 4 1 0 0 1 1 10 KYS 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 11 YIH 0 1 2 0 0 0 0 0 1 0 2 0 0 0 0 0 0 0 0 0 0 0 0 0 12 KYC 2 1 0 0 0 1 0 1 0 0 0 0 0 0 0 0 0 2 0 0 2 0 0 0 13 LSH 0 0 0 1 0 1 0 0 1 0 1 3 0 2 0 0 1 1 1 1 2 0 0 0 14 ESJ 0 0 0 1 0 1 0 0 1 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 15 HBH 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 16 PCI 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 17 KJS 2 2 2 1 1 0 0 1 2 2 1 3 0 2 2 0 1 1 1 1 0 2 1 0 18 JBK 1 0 0 1 0 0 0 0 1 0 1 1 0 1 0 0 1 1 1 0 1 1 1 0 19 CYR 0 1 1 0 2 0 1 1 1 0 5 0 5 1 2 2 0 0 3 0 2 0 2 1 20 KNY 1 1 1 2 2 2 2 1 1 1 2 0 1 1 1 3 0 0 2 1 1 1 0 3 21 KKC 2 2 5 4 3 4 4 3 4 3 1 3 3 4 3 2 4 4 4 5 2 3 3 3 22 KMS 1 1 2 1 0 2 1 3 1 1 0 2 2 0 1 1 1 1 2 1 2 4 5 3 23 KJM 1 0 1 1 1 1 0 0 0 1 0 1 0 0 1 0 0 0 1 0 1 1 1 1 24 LMJ 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 25 PHS 1 1 2 1 0 1 4 3 0 4 0 1 0 2 1 0 0 1 1 2 1 2 4 2 26 YSH 0 1 1 2 1 3 1 1 2 0 3 3 1 1 2 1 1 1 3 0 0 1 3 3 27 HSW 0 1 1 1 1 1 3 1 0 4 2 0 2 1 0 0 1 0 0 3 2 4 0 1 28 SSB 2 1 2 2 2 2 1 2 0 1 2 0 3 0 2 1 0 2 0 2 0 0 0 3 29 JSH 1 0 0 0 0 0 1 1 0 0 1 0 1 0 1 2 0 0 0 0 0 0 0 0 30 RCH 1 1 1 3 2 1 1 3 1 3 3 1 2 1 1 1 2 1 3 2 2 1 2 1

Table 15 below shows the result of analyzing MAGE-A3 epitope screening using PBMCs obtained from a patient with lung cancer.

TABLE 15 # Peptide Patients 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 1 BGO 2 1 3 1 1 4 1 1 1 1 2 2 2 1 1 1 1 1 2 1 2 3 0 1 2 LCS 0 0 0 0 0 0 1 1 2 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 3 KKH 1 1 1 1 5 1 1 5 5 5 1 1 1 1 1 1 1 1 5 1 5 1 1 1 4 NSD 1 1 2 2 2 1 1 2 2 2 2 1 1 3 0 4 0 2 3 1 1 2 1 1 5 CHS 3 2 3 3 4 4 3 2 3 3 2 3 3 3 3 4 4 3 0 2 1 2 2 1 6 KCH 0 0 0 0 2 1 1 1 0 2 0 1 0 0 2 0 0 0 0 2 0 0 1 1 7 CMR 0 0 2 0 4 0 0 0 0 2 0 0 3 2 2 0 0 0 2 0 0 0 0 2 8 HJS 0 0 2 0 1 0 0 2 1 0 0 2 1 1 1 1 1 2 0 0 0 0 3 2 9 PBS 0 4 2 3 2 1 2 4 1 1 0 4 1 2 2 0 1 2 3 2 0 0 2 10 KSH 2 3 1 1 2 2 2 3 3 3 1 2 1 2 2 2 2 3 4 1 3 2 2 3 11 LJS 1 1 1 1 1 0 1 1 1 0 1 0 1 1 0 1 0 0 1 0 0 0 0 0 12 LKS 3 5 2 0 2 2 1 5 4 1 1 0 4 1 0 2 1 1 2 2 0 2 2 2 13 LYS 1 4 0 1 0 2 1 0 2 0 2 2 0 0 0 2 2 5 0 2 1 1 1 1 14 SMS 1 2 2 2 0 2 1 0 2 0 1 1 2 1 1 0 2 1 0 1 2 1 0 3 15 KHS 0 1 1 1 1 0 1 1 1 0 0 1 1 1 1 1 0 1 1 0 1 0 1 0 16 LKT 1 0 0 0 2 0 2 2 1 1 2 0 0 3 2 2 1 1 0 2 1 1 1 1 17 EHR 3 1 2 1 1 1 1 4 0 0 1 1 3 1 3 2 2 0 0 0 1 5 1 3 18 KVH 1 4 3 2 2 2 3 2 1 3 1 2 3 2 2 2 2 2 1 3 2 3 3 2 19 CEH 0 0 2 2 0 0 0 0 0 0 2 3 0 1 0 0 0 0 2 0 0 0 0 0 20 KSJ 2 1 1 0 0 0 0 2 0 2 3 1 2 1 2 0 0 0 0 3 2 2 3 2

As shown in Tables 7 to 13, when CD8 T cells express 4-1BB with at least score 3 by peptide stimulation, these cells can be effectively isolated by using an anti-4-1BB antibody. For each type of cancer, since the ratio of T cell responsive to the autologous cancer antigen with at least score 3 was in a degree of 40 to 50%, it has been determined that a T cell therapeutic agent can be prepared by using the selected epitopes of the autologous cancer antigen. The selected peptides are as follows: hTERT peptide: CLKELVARV (SEQ ID NO: 1), PLFLELL (SEQ ID NO: 2), AAVTPAA (SEQ ID NO: 3); WT1 peptide: SLGEQQVSV (SEQ ID NO: 4), RMFPNAPVL (SEQ ID NO: 5), CMTWNQMNL (SEQ ID NO: 6), VLDFAPPGA (SEQ ID NO: 7); NY-ESO1 peptide: SISSCLQQL (SEQ ID NO: 8), RLLEFYLAM (SEQ ID NO: 9), GVLLKEFTV (SEQ ID NO: 10), ILTIRLTAA (SEQ ID NO: 11); and MAGE-A3 peptide: LLIIVLAII (SEQ ID NO: 12), KIWEELSVL (SEQ ID NO: 13), LVFGIELMEV (SEQ ID NO: 14), SLPTTMNYPL (SEQ ID NO: 15).

Example 3 Pilot Production of T Cell Therapeutic Agent Specific for Autologous Cancer Antigen

Through epitope screening, 3-4 types of peptides of epitopes for each autologous cancer antigen suitable for preparation of a T cell therapeutic agent were selected, and pilot production of a T cell therapeutic agent specific for hTERT, WT1, NY-ESO1, and MAGE-AC was performed by using the peptides. Production of a T cell therapeutic agent includes three steps, that is, a first proliferation of autologous anticancer T cells, isolation and mass culture.

(1) Proliferation of Autologous Cancer Antigen-Specific CD8 T Cells

50 ml of blood was collected from a cancer patient who has been proven to have one or more of epitope having the score of at least 3, through epitope screening.

1) Isolation of PBMCs from blood of a patient: 7 ml of blood was allowed to slowly flow a 15 ml comical tube filled with 7 ml of ficollhypaque such that the blood was overlaid on supernatant of the ficoll solution. The tube was centrifuged for 20 minutes at 2000 rpm at room temperature, and then only a cell layer, which is located between ficoll and plasma and has white color, was collected, washed, and used as PBMCs.

2) Isolated PBMCs were suspended in CTL medium (RPMI1640 medium+4 mM L-glutamine+12.5 mM HEPES+50 μM 2-mercaptoethanol+3% autoplasma) to become the concentration of 1×106 cells/ml, 3-4 types of peptides selected by epitope screening of the present invention were added such that the concentration of each peptide became 1 μg/ml. 1 ml of the cell suspension was aliquoted in a 14 ml round tube, and cultured in a CO2 incubator.

3) Two days after culture, 1 ml of CTL medium including 50 U/ml IL-2 (Proleukin, Novatis) was added to each tube.

4) On day 7, 9, 11, and 13 of culture, 1 ml of the supernatant medium was removed, and CTL medium including 50 U/ml IL-2 was added.

5) After 14 days of culture, cells in each tube were collected in a 50 ml comical tube. Then, RPMI1640 medium was added, and the tube was then centrifuged for 5 minutes at 1400 rpm to wash cells. The process was repeated twice more.

6) The washed cells were suspended in CTL medium such that the concentration of cells became 2×106 cells/ml. Then, 3-4 types of the same peptides were added in the concentration of 5 μg/ml, respectively, and thereafter the resultant was cultured.

(2) Selection of Autologous Cancer Antigen-Specific CD8 T Cells

1) After 24 hours of culture, PBMCs, which were reactivated for a day, were collected, and washed twice with RPMI1640 medium. Then, PBMCs were suspended in CTL medium such that the concentration of PBMCs became 5×106 cells/ml. Thereafter, 50 U/ml of IL-2 was added.

2) 1 ml of the cells were added to a 6-well or 12-well culture plate which was coated with an anti-4-1BB antibody in the concentration of 50 μg/ml for a day, and then the resultant was cultured for 10 minutes in a CO2 incubator.

After 10 minutes of culture, washing was performed to remove all cells which were not attached to the plate. Then, 2 to 4 ml of CTL medium including 1000 U/ml IL-2 was added to each well, and the resultant was cultured for two days in a CO2 incubator.

(3) Mass Culture of Autologous Cancer Antigen-Specific CD8 T Cells

1) Whole cells, which were isolated with the anti-4-1BB antibody and cultured for two days, were collected, washed with RPMI1640 medium twice and counted.

2) PBMCs were isolated from a healthy doner; suspend such that the concentration of PBMCs became 1×108 cells/ml; and irradiated with radiation of 3000 rad to induce cell death so that the resultant was used as a culture additive capable of providing costimulation which is needed to induce proliferation of T cells.

3) To a 50 ml comical tube, were added 5×105 cells of the isolated CD8 T cells and 1×108 cells of irradiated allogenic PBMCs. Then, ALyS505N medium (CELL SCIENCE & TECHNOLOGY INST., INC. (CSTI)) including 1,000 U/ml of IL-2, 40 ng/ml of an anti-CD3 mAb (BD Bioscience) and 3% of autoplasma was added q.s. to 50 ml.

4) 50 ml of cell suspension was injected in a 1 L culture bag and cultured in a CO2 incubator.

5) After 4 days of culture, 50 ml of ALyS505N medium including 1,000 U/ml of IL-2, and 3% of autoplasma was additionally injected to the 1 L culture bag.

6) After 7 days of culture, 100 ml of ALyS505N medium including 1,000 U/ml of IL-2, and 3% of autoplasma was additionally injected to the 1 L culture bag.

7) After 9 days of culture, 300 ml of ALyS505N medium including 1,000 U/ml of IL-2, and 3% of autoplasma was additionally injected to the 1 L culture bag.

8) After 11 days of culture, 500 ml of ALyS505N medium including 1,000 U/ml of IL-2, and 3% of autoplasma was additionally injected to the 1 L culture bag.

9) After 14 days of culture, whole cells in the 1 L culture bag were collected, and washed with injectable physiological saline three times. Then, the cells were suspended in injectable physiological saline including 5% of albumin to fill a complete product of a T cell therapeutic agent.

As above, 3-4 types of peptides having a score of at least 3 which can induce T cell response were selected from a clinical cancer patient trough epitope screening of the present invention. The pilot production of hTERT, WT, NY-ESO1, and MAGE-A3 T cell therapeutic agents was performed by using 50 cc of blood. The results were summarized in FIGS. 6 to 9.

FIG. 14 illustrates a process of pilot production of an hTERT T cell therapeutic agent.

In FIG. 14, PBMCs were isolated from 50 cc of blood of a gastric cancer patient having HLA-A*24 allele. Three types of hTERT peptides, i.e. CLKELVARV (SEQ ID NO: 1), PLFLELL (SEQ ID NO: 2), and AAVTPAA (SEQ ID NO: 3) were added in the concentration of 1 μg/ml for each. Then, the resultant was cultured according to the process described in “(1) proliferation of autologous cancer antigen-specific CD8 T cells” in Example 3 above. After 14 days of culture, whole cells were collected, and T cells, which reacted with the hTERT peptide, and were thus proliferated, were isolated/proliferated according to the process in “(2) selection of autologous cancer antigen-specific CD8 T cells”. The isolated T cells were mass cultured to a level sufficient to be administered to a cancer patient through the process in “(3) mass culture of autologous cancer antigen-specific CD8 T cells”. The cultured final cells were analyzed as particular TCRVb type T cells having low catabiosis and a working function through flow cytometry.

FIG. 15 illustrates a process of pilot production of a WT1 T cell therapeutic agent.

In FIG. 15, PBMCs were isolated from 50 cc of blood of a malignant glioblastoma patient having HLA-A*24 allele. Four types of WT1 peptides, i.e., SLGEQQVSV (SEQ ID NO: 4), RMFPNAPVL (SEQ ID NO: 5), CMTWNQMNL (SEQ ID NO: 6), and VLDFAPPGA (SEQ ID NO: 7) were added in the concentration of 1 μg/ml for each. Then, the resultant was cultured according to the process described in “(1) proliferation of autologous cancer antigen-specific CD8 T cells” in Example 3 above. After 14 days of culture, whole cells were collected, and T cells, which reacted with the WT1 peptide, and were thus proliferated, were isolated/proliferated according to the process in “(2) selection of autologous cancer antigen-specific CD8 T cells”. The isolated T cells were mass cultured to a level sufficient to be administered to a cancer patient through the process in “(3) mass culture of autologous cancer antigen-specific CD8 T cells”. The cultured final cells were analyzed as particular TCRVb type T cells having low catabiosis and a working function through flow cytometry.

FIG. 16 illustrates a process of pilot production of an NY-ESO1 T cell therapeutic agent.

In FIG. 16, PBMCs were isolated from 50 cc of blood of an ovarian cancer patient having HLA-A*02 allele. Four types of NY-ESO1 peptides, i.e. SISSCLQQL (SEQ ID NO: 8), RLLEFYLAM (SEQ ID NO: 9), GVLLKEFTV (SEQ ID NO: 10), and ILTIRLTAA (SEQ ID NO: 11) were added in the concentration of 1 μg/ml for each. Then, the resultant was cultured according to the process described in “(1) proliferation of autologous cancer antigen-specific CD8 T cells” in Example 3 above. After 14 days of culture, whole cells were collected, and T cells, which reacted with the NY-ESO-1 peptide and were thus proliferated, were isolated/proliferated according to the process in “(2) selection of autologous cancer antigen-specific CD8 T cells”. The isolated T cells were mass cultured to a level sufficient to be administered to a cancer patient through the process in “(3) mass culture of autologous cancer antigen specific CD8 T cells”. The cultured final cells were analyzed as a particular TCRVb type T cells having low catabiosis and a working function through flow cytometry.

FIG. 17 illustrates a process of pilot production of an MAGE-A3 T cell therapeutic agent.

In FIG. 17, PBMCs were isolated from 50 cc of blood of a sarcoma patient having HLA-A*02 allele. Four types of MAGE-A3 peptides, i.e. LLIIVLAII (SEQ ID NO: 12), KIWEELSVL (SEQ ID NO: 13), LVFGIELMEV (SEQ ID NO: 14), and SLPTTMNYPL (SEQ ID NO: 15)] were added in the concentration of 1 μg/ml for each. Then, the resultant was cultured according to the process described in “(1) proliferation of autologous cancer antigen-specific CD8 T cells” in Example 3 above. After 14 days of culture, whole cells were collected, and T cells, which reacted with the MAGE-A3 peptide were thus proliferated, were isolated/proliferated according to the process in “(2) selection of autologous cancer antigen-specific CD8 T cells”. The isolated T cells were mass cultured to a level sufficient to be administered to a cancer patient through the process in “(3) mass culture of autologous cancer antigen specific CD8 T cells”. The cultured final cells were analyzed as a particular TCRVb type T cells having low catabiosis and a working function through flow cytometry.

Hereto, the present invention is described referring to preferred examples thereof. A person with ordinary skill in the art to which the present invention pertain would understand that the present invention could be implemented in various aspects different from each other without departing from the essential feature of the present invention. Therefore, the disclosed examples are to be construed as being illustrative, and not restrictive. The scope of the present invention is to be determined by the appended claims, not detailed description above, and all modifications fallen within the equivalent range should be interpreted to be included in the present invention.

Claims

1. A method for isolating autologous cancer antigen-specific CD8+ T cells, the method comprising:

a) selecting a CD8+ T cell epitope of an autologous cancer antigen present in blood of a cancer patient;
b) culturing a peripheral blood mononuclear cell (PBMC) isolated from blood of the cancer patient in a medium together with a peptide of the epitope and IL-2;
c) inducing 4-1BB expression in the cultured cells by adding the peptide same as in step b); and
d) culturing cells in which 4-1BB expression is induced on a culture plate coated with an anti-4-IBB antibody, and then removing unattached cells.

2. The method of claim 1, wherein the autologous cancer antigen in step a) is selected from the group consisting of hTERT (GenBank: BAC11010.1), WT1 (GenBank: AA061088.1), NY-ESO1 (GenBank: CAA05908.1), and MAGE-A3 (NCBI Reference Sequence: NP—005353.1).

3. The method of claim 1, wherein the epitope in step b) is a peptide formed by an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 15.

4. The method of claim 1, wherein the medium in step b) is a medium comprising autoplasma.

5. The method of claim 1, wherein the culture in step b) is performed for 12 to 16 days.

6. The method of claim 1, wherein the induction of expression in step c) is performed for 12 to 36 hours with culturing.

7. The method of claim 1, wherein the culture in step d) is performed for 1 to 20 minutes.

8. A method for mass culturing autologous cancer antigen-specific CD8+ T cells, the method comprising: suspending autologous cancer antigen-specific CD8+ T cells isolated by the method of claim 1, and allogenic PBMC irradiated with radiation in a medium comprising IL-2, an anti-CD3 antibody, and autoplasma, and then injecting the suspension into a culture bag; and additionally injecting the medium and culturing.

9. The method of claim 8, wherein the PBMC is isolated from a healthy doner.

10. The method of claim 8, wherein the culture is performed for 4 to 15 days.

11. The method of claim 10, wherein during the culture, the medium is additionally injected on day 4, 7, 9, 11 and 14 of culture.

Patent History
Publication number: 20150259646
Type: Application
Filed: Mar 12, 2015
Publication Date: Sep 17, 2015
Inventors: Byoung Se Kwon (Gyeonggi-do), Hyun-Guy Kang (Seoul), Kwang-Hui Kim (Gyeonggi-do), Young-Woo Kim (Gyeonggi-do), Young-Ho Kim (Gyeonggi-do), Byung-Kiu Park (Seoul), Sang-Yoon Park (Seoul), Sang-Jae Park (Gyeonggi-do), Hyeon-Seok Eom (Seoul), Ho-Sik Oh (Ulsan), Heon Yoo (Seoul), Don-Gil Lee (Gyeonggi-do), Seung-Hoon Lee (Seoul), Young-Joo Lee (Seoul), Jin-Soo Lee (Gyeonggi-do), Beom-Kyu Choi (Gyeonggi-do)
Application Number: 14/656,355
Classifications
International Classification: C12N 5/0783 (20060101);