Conjugates of WT 1-derived peptides and compositions comprising the same

文档序号:1131751 发布日期:2020-10-02 浏览:19次 中文

阅读说明:本技术 Wt1衍生肽的缀合物和包含其的组合物 (Conjugates of WT 1-derived peptides and compositions comprising the same ) 是由 坂仁志 高梨洋辅 于 2018-12-26 设计创作,主要内容包括:本公开包括式(1)的化合物或其药学上可接受的盐,<Image he="115" wi="246" file="DEST_PATH_IMAGE002.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>其中癌抗原肽A是由7至30个氨基酸残基组成并含有至少一个半胱氨酸残基的MHC I类限制性肽,其中所述癌抗原肽A的半胱氨酸残基经由二硫键与R<Sup>1</Sup>结合;且R<Sup>1</Sup>是氢、式(2)的基团、式(3)的基团或癌抗原肽D,其中式(2)的基团是<Image he="97" wi="350" file="DEST_PATH_IMAGE004.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>其中X<Sup>a</Sup>和Y<Sup>a</Sup>独立地代表单键或由1至4个氨基酸残基组成的二价肽基团,条件是X<Sup>a</Sup>和Y<Sup>a</Sup>中氨基酸残基数量之和为0至4的整数,且癌抗原肽B是由9至30个氨基酸残基组成的MHC II类限制性肽,其中癌抗原肽B的N端氨基酸的氨基与式(2)中的Y<Sup>a</Sup>结合,并且癌抗原肽B的C端氨基酸的羰基与式(2)中的羟基结合,并且式(1)与式(2)经由二硫键结合,式(3)的基团是<Image he="91" wi="366" file="DEST_PATH_IMAGE006.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>其中X<Sup>b</Sup>和Y<Sup>b</Sup>独立地代表单键或由1至4个氨基酸残基组成的二价肽基团,条件是X<Sup>b</Sup>和Y<Sup>b</Sup>中氨基酸残基数量之和为0至4的整数,且癌抗原肽C是由9至30个氨基酸残基组成的MHC II类限制性肽,其中癌抗原肽C的C端氨基酸的羰基与式(3)中的X<Sup>b</Sup>结合,并且癌抗原肽C的N端氨基酸的氨基与式(3)中的氢原子结合,并且式(1)与式(3)经由二硫键结合,且癌抗原肽D是由9至30个氨基酸残基组成并含有至少一个半胱氨酸的MHC II类限制性肽,其中癌抗原肽D的半胱氨酸残基经由二硫键与R<Sup>1</Sup>结合。(The present disclosure includes compounds of formula (1) or a pharmaceutically acceptable salt thereof, wherein the cancer antigen peptide A is an MHC class I restricted peptide consisting of 7 to 30 amino acid residues and containing at least one cysteine residue, wherein the cysteine residue of the cancer antigen peptide A is linked to R via a disulfide bond 1 Combining; and R is 1 Is hydrogen, a group of formula (2), a group of formula (3) or a cancer antigen peptide D, wherein the group of formula (2) is Wherein X a And Y a Independently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that X a And Y a The sum of the number of amino acid residues is an integer of 0 to 4, and the cancer antigen peptide B is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues, wherein the amino group of the N-terminal amino acid of the cancer antigen peptide B is the same as that of Y in the formula (2) a And the carbonyl group of the C-terminal amino acid of the cancer antigen peptide B is bonded to the hydroxyl group in the formula (2), and the formula (1) and the formula (2) are bonded via a disulfide bond, the group of the formula (3) is Wherein X b And Y b Independently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that X b And Y b The sum of the number of amino acid residues is an integer of 0 to 4, and the cancer antigen peptide C is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues, wherein the carbonyl group of the C-terminal amino acid of the cancer antigen peptide C is bonded to X in the formula (3) b And an amino group of the N-terminal amino acid of the cancer antigen peptide C is bonded to a hydrogen atom in the formula (3), and the formula (1) and the formula (3) are bonded via a disulfide bond, and the cancer antigen peptide D is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues and containing at least one cysteine, wherein the cysteine residue of the cancer antigen peptide D is bonded to R via a disulfide bond 1 And (4) combining.)

1. A compound of formula (1) or a pharmaceutically acceptable salt thereof,

wherein the cancer antigen peptide A is an MHC class I restricted WT1 peptide consisting of 7 to 30 amino acid residues and containing at least one cysteine residue, wherein the cysteine residue of the cancer antigen peptide A is linked to R via a disulfide bond1Combining; and is

R1Is a group of formula (2), a group of formula (3) or a cancer antigen peptide D,

wherein the radical of formula (2) is

Wherein XaAnd YaIndependently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XaAnd YaThe sum of the number of amino acid residues is an integer of 0 to 4, and

the cancer antigen peptide B is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues, wherein the amino group of the N-terminal amino acid of the cancer antigen peptide B is identical to Y in the formula (2)aAnd the carbonyl group of the C-terminal amino acid of the cancer antigen peptide B is bound to the hydroxyl group in formula (2), and the formulas (1) and (2) are bound via a disulfide bond,

the radical of formula (3) is

Wherein XbAnd YbIndependently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XbAnd YbThe sum of the number of amino acid residues is an integer of 0 to 4, and

the cancer antigen peptide C is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues, wherein the carbonyl group of the C-terminal amino acid of the cancer antigen peptide C is bonded to X in the formula (3)bAnd an amino group of the N-terminal amino acid of the cancer antigen peptide C is bonded to a hydrogen atom in the formula (3), and the formula (1) and the formula (3) are bonded via a disulfide bond, and

the cancer antigen peptide D is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues and containing at least one cysteine, wherein the cysteine residue of the cancer antigen peptide D is linked to R via a disulfide bond1And (4) combining.

2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide a is an MHC class I-restricted WT1 peptide consisting of 7 to 12 amino acid residues.

3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide a is a peptide comprising the amino acid sequence:

Figure 181237DEST_PATH_IMAGE004

or a peptide comprising an amino acid sequence that differs from the amino acid sequence of SEQ ID NO. 3 by deletion, substitution, or addition of one to three amino acid residues and having the ability to induce CTLs.

4. The compound of claim 3, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide A is a peptide comprising an amino acid sequence selected from the group consisting of:

Figure 220826DEST_PATH_IMAGE005

5. the compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein R1Is a radical of formula (2).

6. The compound of claim 5 or a pharmaceutically acceptable salt thereof, wherein XaAnd YaIs a single bond.

7. The compound of claim 5 or 6, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide B is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

8. The compound of any one of claims 5-7, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide B is a peptide consisting of an amino acid sequence selected from the group consisting of:

or a peptide consisting of an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS: 217-248 by deletion, substitution or addition of one to three amino acid residues and having the ability to induce helper T cells.

9. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein R1Is a radical of formula (3).

10. The compound of claim 9, or a pharmaceutically acceptable salt thereof, wherein XbAnd YbIs a single bond.

11. The compound of claim 9 or 10, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide C is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

12. The compound of any one of claims 9-11, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide C is a peptide consisting of an amino acid sequence selected from the group consisting of:

Figure 25708DEST_PATH_IMAGE010

or a peptide consisting of an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS: 217-248 by deletion, substitution or addition of one to three amino acid residues and having the ability to induce helper T cells.

13. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein R1Is cancer antigen peptide D.

14. The compound of claim 13, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide D is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

15. The compound of claim 13 or 14, or a pharmaceutically acceptable salt thereof, wherein the cancer antigen peptide D is a peptide consisting of an amino acid sequence containing at least one cysteine residue selected from the group consisting of:

Figure 880620DEST_PATH_IMAGE011

Figure 229734DEST_PATH_IMAGE012

or a peptide which is different from the amino acid sequence selected from the group consisting of SEQ ID NOS: 217-248 by deletion, substitution or addition of one to three amino acid residues and which has an amino acid sequence containing at least one cysteine residue and has an ability to induce helper T cells.

16. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (4):

Figure 994647DEST_PATH_IMAGE013

wherein C-C in said formula means that the C residues are linked together by a disulfide bond.

17. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (5):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond.

18. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (6):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond.

19. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (7):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond.

20. A composition comprising a compound of any one of claims 1-19, or a pharmaceutically acceptable salt thereof, and at least one cancer antigen peptide E, or a pharmaceutically acceptable salt thereof, wherein the at least one cancer antigen peptide E is an MHC class I-restricted WT1 peptide consisting of 7 to 30 amino acid residues.

21. The composition of claim 20, wherein the at least one cancer antigen peptide E is an MHC class I-restricted WT1 peptide consisting of 7 to 12 amino acid residues.

22. The composition of claim 20 or 21, wherein said at least one cancer antigen peptide E is a peptide other than cancer antigen peptide a.

23. The composition of any one of claims 20-22, wherein said at least one cancer antigen peptide E comprises a peptide consisting of an amino acid sequence selected from the group consisting of:

or a peptide consisting of an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 3, 5, 6 and 7 by deletion, substitution or addition of one to three amino acid residues and having the ability to induce CTLs.

24. A composition comprising at least one compound selected from the group consisting of:

a compound of formula (4):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond,

a compound of formula (5):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond,

a compound of formula (6):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond, and

a compound of formula (7):

wherein C-C in said formula means that the C residues are linked together by a disulfide bond,

or a pharmaceutically acceptable salt thereof, and

at least one peptide consisting of an amino acid sequence selected from the group consisting of:

or a pharmaceutically acceptable salt thereof.

25. A pharmaceutical composition comprising a compound of any one of claims 1-19 or a pharmaceutically acceptable salt thereof or a composition of any one of claims 20-24, and a pharmaceutically acceptable carrier.

26. The pharmaceutical composition of claim 25, wherein the pharmaceutical composition is for use as a composition for treating a cancer associated with WT1 gene expression or an elevated level of WT1 gene expression.

27. The pharmaceutical composition of claim 25 or 26, wherein the pharmaceutical composition is used as a composition for inducing CTLs in cellular immunotherapy of cancer.

28. The pharmaceutical composition of any one of claims 25-27, wherein the pharmaceutical composition is used as a cancer vaccine.

29. The pharmaceutical composition of any one of claims 26-28, wherein the cancer is a blood cancer selected from leukemia, myelodysplastic syndrome, multiple myeloma, and malignant lymphoma, or a solid cancer selected from gastric cancer, colorectal cancer, lung cancer, breast cancer, germ cell cancer, liver cancer, skin cancer, bladder cancer, prostate cancer, uterine cancer, cervical cancer, ovarian cancer, and brain tumor.

30. A method of treating or preventing cancer, comprising administering to a WT1 positive subject in need thereof a therapeutically or prophylactically effective amount of a compound of any one of claims 1-19, or a pharmaceutically acceptable salt thereof, a composition of any one of claims 20-24, or a pharmaceutical composition of any one of claims 25-29.

Technical Field

This application claims the benefit of japanese patent application No. 2017-251568, which is incorporated herein by reference in its entirety.

The present disclosure relates to cancer immunotherapy and includes conjugates containing cancer antigen peptides from cancer antigen protein WT1 and compositions comprising the conjugates.

Background

The WT1 gene has been isolated as a response gene to Wilms' tumor, which is kidney cancer in children. Leukemia and some solid cancers are known to be associated with high expression of WT 1. The WT1 protein is one of cancer antigen proteins that have attracted much attention for use in cancer vaccines (non-patent document 1).

Cellular immunity, especially cytotoxic T cells (cytotoxic T lymphocytes, hereinafter also referred to as CTLs), plays an important role in eliminating cancer cells in vivo. After being recognized by precursor T cells of an antigen peptide having 8 to 13 amino acid residues (i.e., cancer antigen peptide) derived from a cancer antigen protein, CTLs that attack cancer cells are derived from the precursor T cells by differentiation and proliferation of the precursor T cells, and are complexed with MHC class I molecules. As for the WT1 protein, the following cancer antigen peptides (patent document 1) or modified peptides thereof (patent document 2) have been reported, which bind to and are presented by MHC class I:

WT1126-134peptide: RMFPNAPYL (Arg-Met-Phe-Pro-Asn-Ala-Pro-Tyr-Leu) (SEQ ID NO:2),

WT1235-243peptide: CMTWNQMNL (Cys-Met-Thr-Trp-Asn-Gln-Met-Asn-Leu) (SEQ ID NO:3), and

WT1235-243(2M → Y) peptide: CYTWNQMNL (Cys-Tyr-Thr-Trp-Asn-Gln-Met-Asn-Leu) (SEQ ID NO: 4).

In cancer immunotherapy, the activation of helper T cells is also important for enhancing the function of other T cells such as CTLs (non-patent documents 2 and 3). It is generally believed that degradation of antigenic proteins in lysosomes within cells produces a peptide fragment consisting of approximately 10 to 25 amino acid residues, and a portion of the peptide fragment binds to MHC class II molecules and is presented to the TCR-CD3 complex on helper T cells to activate the cell. With respect to the WT1 protein, the following cancer antigen peptides, which bind to MHC class II and are presented by MHC class II, have been reported (patent documents 3 and 4):

WT135-52peptide: WAPVLDFAPPGASAYGSL (Trp-Ala-Pro-Val-Leu-Asp-Phe-Ala-Pro-ro-Gly-Ala-Ser-Ala-Tyr-Gly-Ser-Leu) (SEQ ID NO: 237)

WT1330-349Peptide: PGCNKRYFKLSHLQMHSRKHTG (Pro-Gly-Cys-Asn-Lys-Arg-Tyr-Phe-Lys-Leu-Ser-His-Leu-Gln-Met-His-Ser-Arg-Lys-His-Thr-Gly) (SEQ ID NO: 233)

In order to induce CTLs more efficiently, a cocktail vaccine comprising two different peptides, an MHC class II restricted peptide and an MHC class I restricted peptide, has been widely used (non-patent documents 4 to 7). Also reported are cocktail vaccines comprising cancer antigen peptide derived from WT1 protein (non-patent documents 8 and 9). However, since the cocktail vaccine contains an antigenic peptide composed of different amino acids and thus may exhibit different physical properties, it is generally difficult to prepare an optimal preparation that effectively induces CTLs corresponding to the peptide.

Long-chain peptide vaccines are also thought to be effective in inducing CTLs, but may be difficult to produce as proteins. Also, since the long-chain peptide vaccine is composed of antigen peptides to be presented on MHC class I and class II molecules, respectively, which are bound to each other via a spacer peptide, it is difficult to control and predict the sites at which they are cleaved by intracellular enzymes.

Another alternative that has been reported to effectively induce CTLs is MHC class I-restricted peptides from WT1 protein via disulfide bonds or conjugates of MHC class I-restricted peptides with MHC class II-restricted peptides (patent document 5). The conjugate of patent document 5 is characterized in that the MHC class I-restricted peptide is provided by the function of ERAP1 (endoplasmic reticulum aminopeptidase 1), that ERAP1 is one of trimming enzymes reported to be able to cleave cancer antigen peptide precursors (non-patent documents 10 to 12), and that the conjugate must contain at least one MHC class I-restricted peptide to which a cysteine residue is added. Therefore, in the development of a WT1 conjugate vaccine capable of inducing CTLs efficiently while having good physicochemical properties, further improvement is desired in view of easy manufacture and simple manufacturing control and wide application of the vaccine.

CITATION LIST

Patent document

Patent document 1: WO 00/06602

Patent document 2: WO 02/079253

Patent document 3: WO2007/047764

Patent document 4: WO 2010/123065

Patent document 5: WO2014/157692

Non-patent document

Non-patent document 1: clin Cancer Res, 2009; 15(17); 5323-37

Non-patent document 2: cancer Res, 2002, 62(22), 6438-

Non-patent document 3: j Immunotherer, 2001; 24(3); 195-204-

Non-patent document 4: cancer Journal, 2011, 17(5), 343-

Non-patent document 5: cancer Sci, 2013, 104(1), 15-21

Non-patent document 6: adv Immunol. 2012; 114; 51-76

Non-patent document 7: recemt Pat Inflex Allergy Drug Discov. 2015, 9(1), 38-45

Non-patent document 8: blood, 2010, 116(2), 171-9

Non-patent document 9: cancer Immunol Immunother, 2010, 59(10), 1467-79

Non-patent document 10: proceedings of the National Academy of Sciences of United states of America 2005, 102(47), 17107-

Non-patent document 11: the Journal of Immunology, 2009; 183; 5526-

Non-patent document 12: the Journal of Immunology, 2010; 184; 4725-.

Summary of The Invention

Technical problem

The object of the present disclosure is to provide a conjugate of MHC class I restricted peptide and MHC class ii restricted peptide each from WT1 protein capable of effectively inducing CTLs, and a composition comprising the conjugate, which can be used as a cocktail vaccine.

The inventors have made intensive studies to overcome the above-mentioned difficulties, and found that a cysteine residue in an MHC class I-restricted peptide containing at least one cysteine residue can bind to a cysteine residue in or added to an MHC class II-restricted peptide via a disulfide bond. Subsequently, the inventors provided a conjugate which is capable of inducing CTLs efficiently and has favorable physicochemical stability while being easily produced and performing simple production control because it does not require addition of a cysteine residue to an MHC class I-restricted peptide. In addition, the conjugates can be used widely. Furthermore, the inventors have found that a cocktail vaccine comprising the conjugate and an MHC class I restricted peptide monomer induces CTLs more efficiently. Thus, the inventors have completed the present invention.

Solution to the problem

(1) First aspect

1. A compound of formula (1):

Figure 662365DEST_PATH_IMAGE001

wherein the cancer antigen peptide A is an MHC class I restricted peptide consisting of 7 to 30 amino acid residues and containing at least one cysteine residue, wherein the cysteine residue of the cancer antigen peptide A is linked to R via a disulfide bond1Combining; and is

R1Is hydrogen, a group of formula (2), a group of formula (3) or a cancer antigen peptide D,

wherein the radical of formula (2) is

Wherein XaAnd YaIndependently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XaAnd YaThe sum of the number of amino acid residues is an integer of 0 to 4, and

cancer antigen peptide B is MHC class II consisting of 9 to 30 amino acid residuesA restricted peptide in which the amino group of the N-terminal amino acid of the cancer antigen peptide B is identical to Y in the formula (2)aAnd the carbonyl group of the C-terminal amino acid of the cancer antigen peptide B is bound to the hydroxyl group in formula (2), and the formulas (1) and (2) are bound via a disulfide bond,

the radical of formula (3) is

Wherein XbAnd YbIndependently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XbAnd YbThe sum of the number of amino acid residues is an integer of 0 to 4, and

the cancer antigen peptide C is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues, wherein the carbonyl group of the C-terminal amino acid of the cancer antigen peptide C is bonded to X in the formula (3)bAnd an amino group of the N-terminal amino acid of the cancer antigen peptide C is bonded to a hydrogen atom in the formula (3), and the formula (1) and the formula (3) are bonded via a disulfide bond, and

the cancer antigen peptide D is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues and containing at least one cysteine, wherein the cysteine residue of the cancer antigen peptide D is linked to R via a disulfide bond1Combining;

or a pharmaceutically acceptable salt thereof.

2. A compound of item 1 or a pharmaceutically acceptable salt thereof, wherein the cancer antigen peptide A is a peptide consisting of 7 to 15 amino acid residues and is HLA-A, HLA-B or HLA-Cw-restricted.

3. A compound of item 1 or 2 or a pharmaceutically acceptable salt thereof, wherein the cancer antigen peptide A consists of 7 to 15 amino acid residues and is selected from HLA-A1, HLA-A2, HLA-A3, HLA-A11, HLA-A24, HLA-A28, HLA-A29, HLA-A31, HLA-A33, HLA-A34, HLA-A68.1, HLA-A1101, HLA-A0201, HLA-A0205, HLA-A3101, HLA-A3302, HLA-B7, HLA-B8, HLA-B13, HLA-B14, HLA-B35, HLA-B40, HLA-B60, HLA-B61, HLA-B62, HLA-B2702, HLA-B2705, HLA-B1, HLA-B3801, HLA-B3701, HLA-B3901, HLA-B3902, HLA-B3903, HLA-B4402, HLA-B4403, HLA-B4402, HLA-B, At least one HLA-typing-restricted peptide selected from the group consisting of HLA-B5201, HLA-B5801, HLA-Cw2, HLA-Cw3, HLA-Cw6, HLA-Cw7, HLA-Cw8, HLA-Cw16, HLA-Cw0301, HLA-Cw0401, HLA-Cw0602 and HLA-Cw 0702.

4. A compound of any one of items 1 to 3, wherein the cancer antigen peptide A is an MHC class I restricted peptide derived from a cancer antigen protein selected from the group consisting of WT1, MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A10, MAGE-A12, BAGE, DAM-6, DAM-10, GAGE-1, GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7B, GAGE-8, NA88-A, NY-ESO-1, NY-ESO-1a, MART-1/Melan-A, MC1R, Gp100, PSA, PSM, tyrosinase, proteolytic enzyme 3, TRP-1, TRP-2, ART-4, CAMEL, CEA, Cyp-B, and the like, or a pharmaceutically acceptable salt thereof, Her2/neu, VEGFR, hTERT, hTRT, iCE, MUC1, MUC2, PRAME, P15, RU1, RU2, SART-1, SART-2, SART-3, AFP, beta-catenin, caspase-8, CDK-4, ELF2, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, myosin, RAGE, SART-2, TRP-2, 707-AP, Survivin (Survivin), Livin, and SYT-SSX.

5. The compound of any one of items 1 to 4, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide A is an MHC class I restricted WT1 peptide consisting of 7 to 12 amino acid residues.

6. The compound of any one of items 1 to 5, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide A is an MHC class I restricted WT1 peptide consisting of 8 to 10 amino acid residues.

7. The compound of any one of items 1 to 6, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide A is an MHC class I restricted WT1 peptide consisting of 9 amino acid residues.

8. A compound of item 7, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide a is a peptide comprising the amino acid sequence:

CMTWNQMNL (SEQ ID NO: 3)

or a peptide comprising an amino acid sequence different from the amino acid sequence of SEQ ID NO. 3 by changing one or several amino acid residues and having the ability to induce CTL.

9. A compound of item 8, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide a is a peptide comprising an amino acid sequence selected from the group consisting of:

CMTWNQMNL (SEQ ID NO:3) and

CYTWNQMNL (SEQ ID NO: 4)。

10. a compound of any one of items 1-9, or a pharmaceutically acceptable salt thereof, wherein R1Is a radical of formula (2).

11. A compound of any one of items 1 to 10, or a pharmaceutically acceptable salt thereof, wherein XaIs a divalent peptide radical consisting of two amino acid residues, and YaIs a single bond; xaAnd YaIndependently is a divalent peptide group consisting of one amino acid residue; xaIs a single bond, and YaIs a divalent peptide group consisting of two amino acid residues; xaIs a divalent peptide radical consisting of one amino acid residue, and YaIs a single bond; xaIs a single bond, and YaIs a divalent peptide group consisting of one amino acid residue; or XaAnd YaIs a single bond.

12. A compound of item 11 or a pharmaceutically acceptable salt thereof, wherein XaIs a single bond, and YaIs a single bond or a divalent peptide group consisting of one amino acid residue.

13. A compound of item 11 or a pharmaceutically acceptable salt thereof, wherein XaIs a single bond or a divalent peptide group consisting of one amino acid residue, and YaIs a single bond.

14. A compound of any one of items 11-13, or a pharmaceutically acceptable salt thereof, wherein XaAnd YaIs a single bond.

15. A compound of any one of items 1-14, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide B is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues.

16. The compound of any one of clauses 15, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide B is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

17. A compound of any one of items 1-16, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide B is a peptide comprising an amino acid sequence selected from the group consisting of:

Figure 687599DEST_PATH_IMAGE005

or a peptide comprising an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS 217-248 by changing one or several amino acid residues and having the ability to induce helper T cells.

18. The peptide of any one of items 1 to 17, wherein the MHC class II molecule is selected from DRB1 × 0101, DRB1 × 0405, DRB1 × 0802, DRB1 × 0803, DRB1 × 0901, DRB1 × 1201, DRB1 × 1403, DRB1 × 1501, DSB1 × 1502, DPB1 × 0201, DPB1 × 0202, DPB1 × 0402, DPB1 × 0501, DPB1 × 0901, DQB1 × 0301, DQB1 × 0302, DQB1 × 0401, DQB1 × 0501, DQB1 × 0601, DQB1 × 0602, and DRB 010 5.

19. The peptide of item 18, wherein the MHC class II molecule is selected from the group consisting of DRB1 x 0101, DRB1 x 0405, DSB1 x 1502, DPB1 x 0201, DPB1 x 0202, and DQB1 x 0601.

20. A compound of any one of items 1-9, or a pharmaceutically acceptable salt thereof, wherein R1Is a radical of formula (3).

21. A compound of any one of items 1-9 and 20, or a pharmaceutically acceptable salt thereof, wherein XbIs a divalent peptide radical consisting of two amino acid residues, and YbIs a single bond; xbAnd YbIndependently is a divalent peptide group consisting of one amino acid residue; xbIs a single bond, and YbIs a divalent peptide group consisting of two amino acid residues; xbIs a divalent peptide radical consisting of one amino acid residue, and YbIs a single bond; xbIs a single bond, and YbIs a divalent peptide group consisting of one amino acid residue; or XbAnd YbIs a single bond.

22. A compound of item 21 or a pharmaceutically acceptable salt thereof, wherein XbIs a single bondAnd Y isbIs a single bond or a divalent peptide group consisting of one amino acid residue.

23. A compound of item 21 or a pharmaceutically acceptable salt thereof, wherein XbIs a single bond or a divalent peptide group consisting of one amino acid residue, and YbIs a single bond.

24. A compound of item 21 or a pharmaceutically acceptable salt thereof, wherein XbAnd YbIs a single bond.

25. A compound of any one of items 1-9 and 20-24, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide C is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues.

26. The compound of any one of items 1-9 and 20-25, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide C is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

27. A compound of any one of items 1-9 and 20-26, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide C is a peptide comprising an amino acid sequence selected from the group consisting of:

Figure 187815DEST_PATH_IMAGE007

or a peptide comprising an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS 217-248 by changing one or several amino acid residues and having the ability to induce helper T cells.

28. The peptide of any one of items 1-9 and 20-27, wherein the MHC class II molecule is selected from DRB1 0101, DRB1 0405, DRB1 0802, DRB1 0803, DRB1 0901, DRB1 1201, DRB1 1403, DRB1 1501, DSB1, DPB1 0201, DPB1 0202, DPB1 0402, DPB1 0501, DPB1 0901, DQB1 0301, dq 1 0302, DQB1 0601, DQB1 0501, DQB1 0601, DQB1 and DRB 5.

29. The peptide of item 28, wherein the MHC class II molecule is selected from the group consisting of DRB1 x 0101, DRB1 x 0405, DSB1 x 1502, DPB1 x 0201, DPB1 x 0202, and DQB1 x 0601.

30. A compound of any one of items 1-9, or a pharmaceutically acceptable salt thereof, wherein R1Is cancer antigen peptide D.

31. A compound of any one of items 1-9 and 30, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide D is an MHC class II restricted WT1 peptide consisting of 9 to 30 amino acid residues.

32. The compound of item 31 or a pharmaceutically acceptable salt thereof, wherein the cancer antigen peptide D is an MHC class II restricted WT1 peptide consisting of 10 to 25 amino acid residues.

33. The compound of item 32, or a pharmaceutically acceptable salt thereof, wherein cancer antigen peptide D is a peptide comprising an amino acid sequence containing at least one cysteine residue selected from the group consisting of:

Figure 380561DEST_PATH_IMAGE009

or a peptide which differs from the amino acid sequence selected from the group consisting of SEQ ID NOS 217-248 by changing one or several amino acid residues and which has an amino acid sequence of at least one cysteine residue and has an ability to induce helper T cells.

34. The peptide of any one of items 1-10 and 30-33, wherein the MHC class II molecule is selected from DRB1 0101, DRB1 0405, DRB1 0802, DRB1 0803, DRB1 0901, DRB1 1201, DRB1 1403, DRB1 1501, DSB1, DPB1 0201, DPB1 0202, DPB1 0402, DPB1 0501, DPB1 0901, DQB1 0301, dq 1 0302, DQB1 0101, DQB1 0501, DQB1 0601, DQB1 and DRB 5.

35. The peptide of item 34, wherein the MHC class II molecule is selected from the group consisting of DRB1 x 0101, DRB1 x 0405, DSB1 x 1502, DPB1 x 0201, DPB1 x 0202, and DQB1 x 0601.

36. A compound of item 1 or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (4):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond.

37. A compound of item 1 or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (5):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond.

38. A compound of item 1 or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (6):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond.

39. A compound of item 1 or a pharmaceutically acceptable salt thereof, wherein the compound of formula (1) is a compound of formula (7):

Figure 729929DEST_PATH_IMAGE013

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond.

40. A composition comprising a compound of any one of items 1-39, or a pharmaceutically acceptable salt thereof, and at least one cancer antigen peptide E, or a pharmaceutically acceptable salt thereof, wherein the at least one cancer antigen peptide E is an MHC class I-restricted WT1 peptide consisting of 7 to 30 amino acid residues.

41. The composition of item 41 or 42, wherein the at least one cancer antigen peptide E consists of 7 to 15 amino acid residues and is selected from the group consisting of HLA-A1, HLA-A2, HLA-A3, HLA-A11, HLA-A24, HLA-A28, HLA-A29, HLA-A31, HLA-A33, HLA-A34, HLA-A68.1, HLA-A5201101, HLA-A0201, HLA-A0205, HLA-A3101, HLA-A3302, HLA-B7, HLA-B8, HLA-B13, HLA-B14, HLA-B35, HLA-B40, HLA-B60, HLA-B61, HLA-B62, HLA-B2702, HLA-B2705, HLA-B1, HLA-B3801, HLA-B370B 3901, HLA-B3902, HLA-B5103, HLA-B5101, HLA-B4401, HLA-B4402, HLA-B-3, HLA-, HLA-B5801, HLA-Cw2, HLA-Cw3, HLA-Cw6, HLA-Cw7, HLA-Cw8or, HLA-Cw16, HLA-Cw0301, HLA-Cw0401, HLA-Cw0602 and HLA-Cw 0702.

42. The composition of clause 40 or 41, wherein the at least one cancer antigen peptide E is an MHC class I-restricted WT1 peptide consisting of 7 to 12 amino acid residues.

43. The composition of item 42, wherein the at least one cancer antigen peptide E is an MHC class I-restricted WT1 peptide consisting of 8 to 10 amino acid residues.

44. The composition of item 43, wherein the at least one cancer antigen peptide E is an MHC class I restricted WT1 peptide consisting of 9 amino acid residues.

45. The composition of any one of items 40-44, wherein the at least one cancer antigen peptide E is a peptide other than cancer antigen peptide A.

46. The compound of any one of items 40-45, or a pharmaceutically acceptable salt thereof, wherein the at least one cancer antigen peptide E comprises a peptide comprising an amino acid sequence selected from the group consisting of:

or a peptide comprising an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 3 and 5 to 7 by changing one or several amino acid residues and having the ability to induce CTLs.

47. A composition comprising at least one compound selected from the group consisting of

A compound of formula (4):

Figure 37950DEST_PATH_IMAGE015

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond,

a compound of formula (5):

Figure 142434DEST_PATH_IMAGE016

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond,

a compound of formula (6):

Figure 291887DEST_PATH_IMAGE017

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond, and

a compound of formula (7):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond,

or a pharmaceutically acceptable salt thereof, and

at least one peptide consisting of an amino acid sequence selected from the group consisting of:

Figure 933138DEST_PATH_IMAGE019

or a pharmaceutically acceptable salt thereof.

48. A pharmaceutical composition comprising a compound of any one of items 1-39 or a pharmaceutically acceptable salt thereof or a composition of any one of claims 40 to 47, and a pharmaceutically acceptable carrier.

49. The pharmaceutical composition of item 48, wherein the compound of any one of items 1 to 39 or a pharmaceutically acceptable salt thereof is used in combination with at least one cancer antigen peptide E, wherein the at least one cancer antigen peptide E is an MHC class I restricted peptide consisting of 7 to 30 amino acid residues.

50. The pharmaceutical composition of item 49, wherein the compound or pharmaceutically acceptable salt thereof is administered concurrently with at least one cancer antigen peptide E.

51. The pharmaceutical composition of item 49, wherein the compound or pharmaceutically acceptable salt thereof is administered prior to the administration of the at least one cancer antigen peptide E.

52. The pharmaceutical composition of item 49, wherein the compound or pharmaceutically acceptable salt thereof is administered after the administration of the at least one cancer antigen peptide E.

53. The pharmaceutical composition of any one of items 48 to 52, wherein the pharmaceutical composition is for use as a composition for treating a cancer associated with WT1 gene expression or an elevated level of WT1 gene expression.

54. The pharmaceutical composition of any one of items 48 to 52, wherein the pharmaceutical composition is used as a composition for inducing CTLs in cellular immunotherapy of cancer.

55. The pharmaceutical composition of any one of items 48-52, wherein the pharmaceutical composition is for use as a cancer vaccine.

56. The pharmaceutical composition of any one of items 53-55, wherein the cancer is a blood cancer or a solid cancer.

57. The pharmaceutical composition of item 56, wherein the cancer is a blood cancer selected from the group consisting of leukemia, myelodysplastic syndrome, multiple myeloma, and malignant lymphoma.

58. The pharmaceutical composition of item 56, wherein the cancer is a solid cancer selected from the group consisting of gastric cancer, colorectal cancer, lung cancer, breast cancer, germ cell cancer, liver cancer, skin cancer, bladder cancer, prostate cancer, uterine cancer, cervical cancer, ovarian cancer, and brain tumor.

59. Use of a compound of any one of items 1-39, or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-49, or a pharmaceutical composition of any one of items 48-58 for the manufacture of a cancer vaccine.

60. A method of treating or preventing cancer, comprising administering to a WT1 positive subject in need thereof a therapeutically or prophylactically effective amount of a compound of any one of items 1-39, or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-47, or a pharmaceutical composition of any one of items 48-58.

61. A method of obtaining an MHC class I restricted peptide and an MHC class II restricted peptide comprising reacting a compound of any one of items 1-39, or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-47, or a pharmaceutical composition of any one of items 48-58 with ERAP 1.

62. A method of synthesizing a compound of formula (1), comprising forming a disulfide bond between a cysteine residue of cancer antigen peptide a as described in any one of items 1 to 19, 36 and 37 and a cysteine residue linked to the N-terminus of cancer antigen peptide B as described in any one of the items.

63. A method of synthesizing a compound of formula (1), comprising forming a disulfide bond between a cysteine residue of cancer antigen peptide a as described in any one of items 1 to 9, 20 to 29, 38 and 39 and a cysteine residue attached to the C-terminus of cancer antigen peptide C as described in any one of the items.

64. A method of synthesizing a compound of formula (1), comprising forming a disulfide bond between a cysteine residue of cancer antigen peptide a as described in any one of items 1 to 9 and 30 to 35 and a cysteine residue of cancer antigen peptide D as described in any one of the items.

65. An MHC class I-restricted peptide consisting of 7 to 30 amino acid residues for use in combination with a compound of any one of items 1-39 or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-47, or a pharmaceutical composition of any one of items 48-58.

66. A compound of any one of items 1-39 or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-47, or a pharmaceutical composition of any one of items 48-58, for use in combination with at least one MHC class I-restricted peptide consisting of 7 to 30 amino acid residues.

67. A kit for treating cancer, wherein the kit comprises at least one MHC class I restricted peptide consisting of 7 to 30 amino acid residues, and a compound of any one of items 1-39 or a pharmaceutically acceptable salt thereof, a composition of any one of items 40-47, or a pharmaceutical composition of any one of items 48-58.

Effects of the invention

The present disclosure provides compounds of formula (1) and compositions comprising the compounds, which are useful for treating or preventing cancer.

Brief description of the drawings

FIG. 1 shows the results of testing the in vivo CTL-inducing ability in HLA-A2402 transgenic mice in the IFN γ ELISPOT assay of Experimental example 1 using the peptide of SEQ ID NO. 4 synthesized in reference example 2 and the compound of formula (5) synthesized in example 1.

FIG. 2 shows the results of testing the ability to induce IFN γ -producing cells in vivo in HLA-A0201 transgenic mice in an IFN γ ELISPOT assay of test example 2 using the peptide of SEQ ID NO:2 synthesized in reference example 1 and the compound of formula (5) synthesized in example 1.

Description of the embodiments

Embodiments of the present application are described in detail below.

As used herein, "amino acid residue" refers to a single amino acid unit among the amino acids that make up a peptide or protein molecule. The "amino acid residue" may be a natural or non-natural α -amino acid residue, β -amino acid residue, γ -amino acid residue or-amino acid residue, more particularly a natural α -amino acid residue, ornithine residue, homoserine residue, homocysteine residue, β -alanine residue, γ -aminobutyric acid or-aminopentanoic acid. When the "amino acid residue" is optically active, it includes L-form and D-form, preferably L-form.

For the description of "amino acid residues", abbreviations thereof may be used. The following is a list of abbreviations:

ala or A: alanine residue

a: d-alanine residue

Arg or R: arginine residue

Asn or N: asparagine residue

Asp or D: aspartic acid residue

Cys or C: cysteine residue

Gln or Q: glutamine residue

Glu or E: glutamic acid residue

Gly or G: glycine residue

His or H: histidine residue

Ile or I: isoleucine residues

Leu or L: leucine residue

Lys or K: lysine residue

Met or M: methionine residue

Phe or F: phenylalanine residue

Pro or P: proline residue

Ser or S: serine residue

Thr or T: threonine residues

Trp or W: tryptophan residue

Tyr or Y: tyrosine residue

Val or V: valine residue

Abu: 2-aminobutyric acid residue (also referred to as alpha-aminobutyric acid residue)

Orn: ornithine residue

Cit: citrulline residue

Cha: cyclohexylalanine residue

Ahx: 2-aminocaproic acid residue.

The amino acid sequence of a "peptide" is described herein according to the general description such that its N-terminal amino acid residue is located on the left side and its C-terminal amino acid residue is located on the right side. Unless otherwise stated, in the "peptide", the amino group of the N-terminal amino acid is bonded to a hydrogen atom (to become a free amino group), and the carbonyl group of the C-terminal amino acid is bonded to a hydroxyl group. A divalent peptide group refers to a peptide group that is capable of binding to other chemical moieties via the N-terminal amino group and via the C-terminal carbonyl group. Unless otherwise specified, in the peptide corresponding to the partial structure of the compound of formula (1), for example, the compound of any one of formulas (4) to (7), the amino group of the N-terminal amino acid is bonded to a hydrogen atom, and the carbonyl group of the C-terminal amino acid is bonded to a hydroxyl group.

"cancer antigen peptide A" is an MHC class I restricted peptide consisting of 7 to 30 amino acid residues and contains at least one cysteine residue. In the formula (1), the cysteine residue of the cancer antigen peptide A is linked to R via a disulfide bond1And (4) combining.

In the formula (1), "R1"is hydrogen, a group of formula (2), a group of formula (3) or a cancer antigen peptide D. Preferably, R1Is a radical of formula (2), a radical of formula (3) or a carcinostatic agentThe propeptide D is more preferably a group of formula (2) or a group of formula (3).

When R is1When it is a group of formula (2), the compound of formula (1) is a compound of formula (1-1):

wherein Xa、YaAnd cancer antigen peptide B has the same meaning as defined above for formula (2).

“Xa"and" Ya"independently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XaAnd YaThe sum of the number of amino acid residues is an integer from 0 to 4. For example, when XaAnd YaWhen the sum of the number of amino acid residues is an integer of 0, XaAnd YaMust be a single bond; and when XaAnd YaWhen the sum of the number of amino acid residues is an integer of 4, XaAnd YaEach may be a divalent peptide radical consisting of two amino acid residues, or XaMay be a divalent peptide group consisting of three amino acid residues, and YaMay be a divalent peptide group consisting of one amino acid residue, or XaMay be a divalent peptide group consisting of four amino acid residues, and YaMay be a single bond.

XaAnd YaThe sum of the number of amino acid residues is preferably an integer from 0 to 2, more preferably an integer from 0 to 1, or most preferably zero. That is, most preferably, XaAnd YaAre all single bonds.

When X is presentaAnd YaWhen the sum of the number of amino acid residues is an integer of 2, XaMay be a divalent peptide group consisting of two amino acid residues, and YaMay be a single bond; xa and Ya may independently be a bivalent peptide group consisting of one amino acid residue; or Xa may be a single bond and Ya is a divalent peptide group consisting of two amino acid residues.

When X is presentaAnd YaAmmonia in ammoniaWhen the sum of the number of amino acid residues is an integer of 1, XaMay be a divalent peptide group consisting of one amino acid residue, and YaMay be a single bond; or XaMay be a single bond, and YaMay be a divalent peptide group consisting of one amino acid residue. In a preferred embodiment, XaIs a single bond, and YaIs a residue of alanine, leucine or methionine; or XaIs a residue of alanine, leucine or methionine, and YaIs a single bond.

"cancer antigen peptide B" is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues. In the formula (2) (or the formula (1-1)), the amino group of the N-terminal amino acid of the cancer antigen peptide B and Y in the formula (2)aAnd the carbonyl group of the C-terminal amino acid of cancer antigen peptide B is bound to the hydroxyl group in formula (2).

When R is1When it is a group of formula (3), the compound of formula (1) is a compound of formula (1-2):

wherein Xb、YbAnd cancer antigen peptide C has the same meaning as defined above for formula (3).

“Xb"and" Yb"independently represents a single bond or a divalent peptide group consisting of 1 to 4 amino acid residues, with the proviso that XbAnd YbThe sum of the number of amino acid residues is an integer from 0 to 4. For example, when XbAnd YbWhen the sum of the number of amino acid residues is an integer of 0, XbAnd YbAll must be single bonds; and when XbAnd YbWhen the sum of the number of amino acid residues is an integer of 4, XbAnd YbEach may be a divalent peptide radical consisting of two amino acid residues, or XbMay be a divalent peptide group consisting of three amino acid residues, and YbMay be a divalent peptide group consisting of one amino acid residue, or XbMay be a divalent peptide group consisting of four amino acid residues, and YbMay be a single bond.

XbAnd YbThe sum of the number of amino acid residues is preferably an integer from 0 to 2, more preferably an integer from 0 to 1, or most preferably zero. That is, most preferably, XbAnd YbAre all single bonds.

For example, when XbAnd YbWhen the sum of the number of amino acid residues is an integer of 2, XbMay be a divalent peptide group consisting of two amino acid residues, and YbMay be a single bond, XbAnd YbEach of which may independently be a divalent peptide group consisting of one amino acid residue, or XbMay be a single bond, and YbMay be a divalent peptide group consisting of two amino acid residues.

When X is presentbAnd YbWhen the sum of the number of amino acid residues is an integer of 1, XbMay be a divalent peptide group consisting of one amino acid residue, and YbMay be a single bond, or XbMay be a single bond, and YbMay be a divalent peptide group consisting of one amino acid residue. In a preferred embodiment, XbIs a single bond, and YbIs a residue of alanine, leucine or methionine, or XbIs a residue of alanine, leucine or methionine, and YbIs a single bond.

"cancer antigen peptide C" is an MHC class II restricted peptide consisting of 9 to 30 amino acid residues. In formula (3) (or formula (1-2)), the carbonyl group of the C-terminal amino acid of the cancer antigen peptide C is bound to Yb in formula (3), and the amino group of the N-terminal amino acid of the cancer antigen peptide C is bound to the hydrogen atom in formula (3).

When R is1When it is a peptide D, the compound of formula (1) is a compound of formula (1-3):

wherein cancer antigen peptide D has the same meaning as defined above.

"cancer antigen peptide D" is a peptide consisting of 9 to 30 amino groupsAn MHC class II restricted peptide consisting of acid residues and containing at least one cysteine. When R is1In the case of the cancer antigen peptide D, the cysteine residue of the cancer antigen peptide D is bonded to R via a disulfide bond1And (4) combining.

The cancer antigen peptide D contains at least one cysteine residue in its amino acid sequence. The number of cysteine residues is preferably 1 to 3, more preferably 1 to 2, or most preferably 1.

The term "WT 1 peptide", which is synonymous with "partial peptide of WT1 protein", as used herein, refers to a peptide consisting of consecutive amino acid residues of the amino acid sequence of the human WT1 protein of SEQ ID NO: 1, or an allosteric peptide thereof having the ability to induce CTL or helper T cells.

The term "MHC class I restricted" refers to the ability of a peptide to bind to a Major Histocompatibility Complex (MHC) class I molecule and induce CTLs.

Human MHC is called Human Leukocyte Antigen (HLA). HLA molecules corresponding to MHC class I molecules include HLA-A, B, Cw, F and G subtypes. HLA-A, HLA-B or HLA-Cw restriction is preferred as MHC class I restriction of the "MHC class I restricted" peptide.

Allelic polymorphisms are known for individual subtypes of HLC. For HLA-A, 27 or more types of polymorphisms including HLA-A1, HLA-A0201, and HLA-A24 are known. For HLA-B, 59 or more types of polymorphisms including HLA-B7, HLA-B40, and HLA-B4403 are known. For HLA-Cw, 10 or more types of polymorphisms are known including HLA-Cw0301, HLA-Cw0401, and HLA-Cw 0602. Among such polymorphisms, HLA-A0201 and HLA-A24 are preferred.

The term "MHC class I restricted peptide" as used herein refers to a peptide capable of binding to an MHC class I molecule so as to be presented in a complex form and inducing CTLs from precursor T cells that recognize the complex in vitro and/or in vivo (in other words, a peptide having the ability to induce CTLs). The "MHC class I-restricted WT1 peptide" is a WT1 peptide, which is an "MHC class I-restricted peptide". The "MHC class I-restricted peptide" and "MHC class I-restricted WT1 peptide" are generally referred to herein as a "killer peptide" and "WT 1 killer peptide", respectively. The "MHC class I-restricted peptide" may consist of any number of sequences of any type of amino acid, as long as it functions as the "MHC class I-restricted peptide" defined above. However, the longer the peptide chain, the more easily it is degraded by proteolytic enzymes. Also, too small a peptide may not be successfully captured in the peptide binding groove of MHC class I molecules. The "MHC class I restricted peptide" typically consists of 7 to 30 amino acid residues, preferably 7 to 15 amino acid residues, more preferably 8 to 12 amino acid residues, still more preferably 8 to 11 amino acid residues or most preferably 8or 9 amino acid residues.

An "MHC class I-restricted peptide" consisting of 7 to 12 amino acid residues, preferably 9 amino acid residues, may also be referred to as an "MHC class I-restricted epitope". The term "MHC class I-restricted epitope" refers to a peptide corresponding to the actual peptide that is complexed with and presented by an MHC class I molecule. That is, the term "MHC class I-restricted peptide" includes peptides that provide an "MHC class I-restricted epitope" in vitro or in vivo by a process such as digestion with a proteasome and/or a protease and/or cleavage (also referred to as trimming) by ERAP1 to an appropriate peptide length.

An "MHC class I-restricted epitope" can be derived from an "MHC class I-restricted peptide" by degradation with a proteasome and/or a protease and subsequent trimming (cleavage) by ERAP1, wherein the C-terminal amino acid residue and the N-terminal amino acid residue of the "MHC class I-restricted epitope" can be determined by the action of the proteasome/protease and ERAP1, respectively. Thus, the "MHC class I-restricted peptide" may be a peptide consisting of 7 to 30 amino acid residues, wherein 0 to 23 amino acid residues are linked via its C-terminal carbonyl group to an "MHC class I-restricted epitope" consisting of 7 to 12 residues.

The term "peptide comprising an amino acid sequence" as used herein refers to a peptide having a given amino acid sequence, which may optionally have additional amino acid residue sequences attached to the N-terminal and/or C-terminal amino acids of the given sequence, as is commonly understood.

The term "allosteric peptide" as used herein refers to a peptide consisting of an amino acid sequence that differs from the amino acid sequence of the original peptide by the alteration of one or several amino acid residues. In the allosteric peptides, one or several amino acid residues, such as 1 to 9 amino acid residues, preferably 1 to 5, 1 to 4 or 1 to 3 amino acid residues, more preferably 1 to 2 amino acid residues or most preferably one amino acid residue, are deleted from, substituted to and/or added to the amino acid sequence of the original peptide. The number of amino acids deleted from, substituted for, and/or added to the amino acid sequence of the original peptide may preferably be 1 to 5, 1 to 4, or 1 to 3, more preferably 1 to 2, or most preferably 1. Amino acid substitutions for the altered peptide can be made with any type of amino acid at any position of the amino acid residue in the original sequence. Conservative amino acid substitutions are preferred. For example, Asp may be substituted for Glu; replacement of Phe with Tyr; substitution of Ile for Leu; replacement of Ala with Ser; or His by Arg. Amino acid additions or deletions may preferably be made at the N-terminus or C-terminus of the peptide. However, amino acid addition or deletion may be performed internally. Amino acid additions (or insertions) or substitutions can be made with any of the amino acids encoded by the twenty genes or even any unnatural amino acid.

The killer peptides consisting of an altered amino acid sequence as used herein are also referred to as "allosteric killer peptides". In allosteric killer peptides, amino acid substitutions may be made, in particular at amino acid position 1 (N-terminus), 2, 3 or C-terminus, for example at position 1 (N-terminus), 2, 3 or 9 (C-terminus) in peptides consisting of nine amino acid residues. When the allosteric killer peptide has an added (or inserted) amino acid residue, the number of added amino acids is preferably 1 or 2, or more preferably one. The amino acid addition is preferably performed to the N-terminus or C-terminus, and more preferably to the C-terminus. When the killer peptide is changed by amino acid deletion, the number of the deleted amino acids is preferably 1 or 2, or more preferably one. The amino acid deletion is preferably performed at the N-terminus or C-terminus, and more preferably at the C-terminus.

Each HLA subtype is polymorphic. Peptides that can be complexed with polymorphic sequences of HLA antigens have a specific pattern of amino acid sequences (i.e., binding motifs) in order to bind to the polymorphic sequences of HLA antigens. Amino acid substitutions may be made at any of the amino acid residues that make up such binding motifs. For example, HLA-A24 binding peptides consisting of 8 to 11 amino acid residues are known to have Tyr, Phe, Met or Trp at position 2 and Phe, Leu, Ile, Trp or Met at the C-terminus (J. Immunol.,152, p. 3913, 1994; J. Immunol., 155, p. 4307, 1994; Immunogenetics, 41, p. 178, 1995). Thus, for example, a peptide consisting of nine amino acid residues may be altered by amino acid substitutions to have Tyr, Phe, Met or Trp at position 2 and/or Phe, Leu, Ile, Trp or Met at position 9 (C-terminus) to obtain an allosteric peptide that can be used as an allosteric killer peptide. Likewise, HLA-a0201 binding peptides consisting of 8 to 11 amino acid residues are known to have Leu or Met at position 2 and Val or Leu at the C-terminus. Thus, for example, a peptide consisting of nine amino acid residues may be altered by amino acid substitutions to have Leu or Met at position 2 and/or Val or Leu at position 9 (C-terminus) to obtain an allosteric peptide that can be used as an allosteric killer peptide.

Examples of "MHC class I restricted WT1 peptides" include the peptides shown in tables 1-44. In the table, the column "position" shows the position in the amino acid sequence of human WT1 of SEQ ID NO. 1 corresponding to each peptide.

Figure 632881DEST_PATH_IMAGE023

Figure 508783DEST_PATH_IMAGE028

Figure 886213DEST_PATH_IMAGE031

Figure 594385DEST_PATH_IMAGE034

Figure 82610DEST_PATH_IMAGE035

Figure 861342DEST_PATH_IMAGE036

Figure 603776DEST_PATH_IMAGE037

The "MHC class I restricted WT1 peptide" may preferably be a peptide comprising an amino acid sequence selected from the group consisting of:

Figure 869148DEST_PATH_IMAGE038

or a peptide comprising an amino acid sequence which differs from an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 3, 5, 6 and 7 by changing one or several amino acid residues. Peptides consisting of amino acid sequences selected from the group consisting of SEQ ID NOS: 2, 3, 5, 6 and 7 are also preferred.

Examples of allosteric killer peptides include the following peptides:

as an allosteric killer peptide of PMFPNAPYL (SEQ ID NO:2),

as

Figure 912245DEST_PATH_IMAGE040

The allosteric killer peptide of (a),

Figure 26963DEST_PATH_IMAGE041

(wherein Xaa is Ser or Ala) or

(wherein Xaa isOr

AsThe allosteric killer peptide of (a),

as

Figure 395335DEST_PATH_IMAGE047

The allosteric killer peptide of (a),

or

And

as

Figure 860241DEST_PATH_IMAGE051

The allosteric killer peptide of (a),

the term "MHC class II restricted" refers to the ability of a peptide to bind to MHC class II molecules and induce helper T cells.

HLA's corresponding to MHC class II-molecules have subtypes including HLA-DR, DQ and DP subtypes. HLA-DR, HLA-DQ or HLA-DP restriction is preferred as MHC class II restriction of the "MHC class II restricted" peptide. More preferred is a restriction of a subtype selected from the group consisting of: DRB1 0101, DRB1 0405, DRB1 0802, DRB1 0803, DRB1 0901, DRB1 1201, DRB1 1403, DRB1 1501, DRB1 1502, DPB1 0201, DPB1 0202, DPB1 0402, DPB1 0501, DPB1 0901, DQB1 0401, DQB1 0302, DQB1 0601, DQB1 0501, DQB1 0601, DQB1 0502 and DRB5 0102. Restriction of a subtype selected from DRB1 × 0101, DRB1 × 0405, DRB1 × 1502, DPB1 × 0201, DPB1 × 0202 and DQB1 × 0601 is most preferred.

The term "MHC class II restricted peptide" as used herein refers to a peptide capable of binding to an MHC class II molecule and inducing helper T cells in vitro and/or in vivo (in other words, a peptide having the ability to induce helper T cells). The "MHC class II restricted WT1 peptide" is a WT1 peptide, which is an "MHC class II restricted peptide". The "MHC class II restricted peptide" and "MHC class II restricted WT1 peptide" are generally referred to herein as the "helper peptide" and "WT 1 helper peptide", respectively. The "MHC class II restricted WT1 peptide" may consist of any number of sequences of any type of amino acid, as long as it functions as the "MHC class II restricted WT1 peptide" defined above. However, the longer the peptide chain, the more easily it is degraded by proteolytic enzymes. Also, too small a peptide may not be successfully captured in the peptide binding groove of MHC class II molecules. The "MHC class II restricted WT1 peptide" typically consists of 9 to 30 amino acid residues, preferably 10 to 25 amino acid residues, more preferably 12 to 24 amino acid residues, or even more preferably 15 or 22 amino acid residues.

Examples of "MHC class II restricted WT1 peptides" include peptides comprising an amino acid sequence selected from the group consisting of:

Figure 948950DEST_PATH_IMAGE053

Figure 648660DEST_PATH_IMAGE054

or a peptide comprising an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS 217-241 by changing one or several amino acid residues and having the ability to induce helper T cells.

As submitted in PCT application No. PCT/JP2017/042760, the inventors have found that some WT1 helper peptides generally have the motif: KLSHL from the amino acid sequence of WT1 protein. The motif KLSHL corresponds to the amino acid sequence at positions 336-340 of SEQ ID NO: 1. Thus, in one embodiment, an "MHC class II restricted WT1 peptide" is a partial peptide of the WT1 protein consisting of an amino acid sequence of 9 to 30 amino acid residues including the motif KLSHL as a part thereof, or a peptide consisting of an amino acid sequence different from the amino acid sequence of the partial peptide by changing one or several amino acid residues and having the ability to induce helper T cells.

In a further embodiment, the "MHC class II restricted WT1 peptide" is a partial peptide of the WT1 protein consisting of an amino acid sequence comprising 10 to 25 amino acid residues, 12 to 24 amino acid residues, or 15 to 22 amino acid residues of which KLSHL is a part, or a peptide consisting of an amino acid sequence different from that of the partial peptide by changing one or several amino acid residues and having the ability to induce helper T cells.

The term "peptide comprising an amino acid sequence" as defined above refers to a peptide having a given amino acid sequence, which may optionally have additional amino acid residue sequences attached to the N-terminal and/or C-terminal amino acids of the given sequence, as is commonly understood. Thus, when the partial peptide of the WT1 protein consists of an amino acid sequence including the motif KLSHL as a part of 9 to 30 amino acid residues, 10 to 25 amino acid residues, 12 to 24 amino acid residues, or 15 to 22 amino acid residues, the partial peptide of the WT1 protein is a peptide corresponding to the amino acid sequence portion of SEQ ID NO: 1 consisting of the motif KLSHL and a sequence directly linked to the essential number of amino acid residues on the N-terminus and/or C-terminus of the motif. In particular, such partial peptides correspond to an amino acid sequence consisting of 9 to 30 amino acids, 10 to 25 amino acids, 12 to 24 amino acids, or 15 to 22 amino acids in succession, which comprises the motif KLSHL from SEQ ID NO. 272, which corresponds to the sequence of amino acids 311-365 of SEQ ID NO. 1.

The amino acid sequence at position 311-365 of SEQ ID NO: 1 (corresponding to the amino acid sequence of SEQ ID NO: 272) is as follows:

examples of "MHC class II restricted WT1 peptide" comprising the motif KLSHL as a part thereof include peptides comprising an amino acid sequence selected from the group consisting of:

Figure 208624DEST_PATH_IMAGE057

or a peptide comprising an amino acid sequence different from the amino acid sequence selected from the group consisting of SEQ ID NOS 221-236 and 242-271 by changing one or several amino acid residues and having the ability to induce helper T cells.

Examples of "MHC class II restricted WT1 peptide comprising at least one cysteine residue" include peptides comprising an amino acid sequence comprising at least one cysteine residue selected from the group consisting of:

Figure 842846DEST_PATH_IMAGE060

or a peptide comprising an amino acid sequence containing at least one cysteine residue which differs from the amino acid sequence selected from the group consisting of SEQ ID NOS 217-271 by changing one or several amino acid residues and having the ability to induce helper T cells.

Preferred examples of "MHC class II restricted WT1 peptide comprising at least one cysteine residue" include peptides comprising an amino acid sequence comprising at least one cysteine residue selected from the group consisting of:

Figure 696008DEST_PATH_IMAGE061

the MHC class II restricted peptide may be complexed with an MHC class II molecule in any one of the HLA-DR, HLA-DQ or HLA-DP subclasses. In a preferred embodiment, the MHC class II restricted peptide induces helper T cells by binding to an MHC class II molecule selected from the group consisting of DRB1 0101, DRB1 0405, DRB1 0802, DRB1 0803, DRB1 0901, DRB1 1201, DRB1 1403, DRB1 1501, DRB1 1502, DPB1 0201, DPB1 0202, DPB 580402, DPB1 0501, DPB1 0901, DQB1 0301, DQB1 0302, DQB 360401, DQB1 0501, DQB 11 and DRB 010 5. More preferably, the MHC class II restricted peptide induces helper T cells by binding to an MHC class II molecule selected from the group consisting of DRB1 x 0101, DRB1 x 0405, DRB1 x 1403, DRB1 x 1502, DPB1 x 0201, DPB1 x 0202, DPB1 x 0901, DQB1 x 0301, DQB1 x 0601 and DRB5 x 0102. Most preferably, the MHC class II restricted peptide induces helper T cells by binding to an MHC class II molecule selected from DRB1 x 0101, DRB1 x 0405, DRB1 x 1502, DPB1 x 0201, DPB1 x 0202 and DQB1 x 0601.

Helper peptides consisting of altered amino acid sequences are also referred to herein as "allosteric helper peptides". If a partial peptide of WT1 protein, which is useful as a WT1 helper peptide, consisting of an amino acid sequence comprising 9 to 30 amino acid residues of the sequence KLSHL is altered, the allosteric peptide preferably has a modification in an amino acid residue other than the sequence KLSHL. The "MHC class II restricted WT1 peptide" as "cancer antigen peptide B" may have an additional sequence of amino acid residues preferably linked to its C-terminus. The "MHC class II restricted WT1 peptide" as the "cancer antigen peptide C" may have an additional sequence of amino acid residues preferably linked to the N-terminus thereof.

Amino acid substitutions may be made at any of the amino acid residues that constitute the binding motif for HLA antigens. When the helper peptide consisting of an amino acid sequence comprising nine amino acid residues comprising the binding motif for HLA-DRB1 x 0405 is altered by amino acid substitutions, the substitutions may preferably be made at positions 1 (N-terminus), 4, 6 and/or 9 (C-terminus). In a preferred embodiment, the helper peptide consisting of a sequence comprising nine amino acid residues of the binding motif to HLA-DRB1 x 0405 may be altered by amino acid substitutions to have amino acid residues selected from:

phenylalanine, tryptophan, valine, isoleucine, leucine or methionine at position 1 (N-terminus);

valine, isoleucine, methionine, aspartic acid and glutamic acid at position 4;

asparagine, serine, threonine, glutamine, lysine, and aspartic acid at position 6; and/or

Aspartic acid, glutamic acid and glutamine at position 9 (C-terminus).

Peptides may have modifications in amino acid residues in their sequence. The modification can be carried out by conventional methods, for example by esterification, alkylation, halogenation, phosphorylation, sulfonation or amidation of functional groups in the amino acid residue. Amino acid modifications in a peptide may also be the addition of any moiety to the N-and/or C-terminus of the peptide. Peptides can be modified by the addition of such moieties so that the solubility of the peptide can be modulated, the peptide is stabilized against, for example, proteolytic degradation, the peptide is directed to a particular tissue or organ, or the capture of the peptide by antigen presenting cells is improved.

In the peptide, the amino group of the N-terminal amino acid thereof or the carboxyl group of the C-terminal amino acid thereof may be modified. The amino group may be modified, for example, by the addition of one to three groups selected from C1-6Alkyl, phenyl, cycloalkyl or acyl radicals such as C1-6Alkanoyl, phenyl-C1-6Alkanoyl radical, C5-7-cycloalkylcarbonyl, C1-6-alkylsulfonyl, phenylsulfonyl, C2-6-alkoxy-carbonyl, phenyl-alkoxycarbonyl, C5-7-cycloalkoxy-carbonyl or phenoxycarbonyl. The carboxyl group of the C-terminal amino acid can be converted, for example, into an ester, such as C1-6Alkyl esters, phenyl-C0-6Alkyl esters or C5-7Cycloalkyl esters, or conversion to amides, e.g. unsubstituted amides, mono-or di-C1-6Alkyl amides, mono-or di-phenyl-C0-6-alkylamides or disubstituted amides wherein the two substituents together with the nitrogen to which they are attached form a5 to 7 membered azacycloalkane.

In peptides, the bonds between amino acid residues may be peptide bonds or other types of bonds, such as carbon-carbon bonds, carbon-nitrogen bonds or carbon-sulfur bonds. The peptides described herein may comprise one or more D-amino acid residues.

The above description of modifications in peptides is merely illustrative, and variations thereof are possible to those skilled in the art. Such modified peptides may be prepared, tested or used by one of ordinary skill in the art.

Examples of the compound of formula (1) include compounds of formula (4):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond;

a compound of formula (5):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond;

a compound of formula (6):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond; and

a compound of formula (7):

wherein C-C as shown in the formula means that the C residues are linked together by a disulfide bond.

The ability of the peptide to induce CTL or helper T cells can be confirmed by conventional methods. CTL induction can be confirmed, for example, by counting CTLs by the HLA tetramer method (int. J. Cancer:100, 565-. The induction of CTL by the HLA-A24-restricted peptide can be confirmed by using an HLA-A24 mouse model as described in WO 02/47474 or int. J. Cancer:100, 565-570 (2002). Induction of helper T cells can be demonstrated, for example, by Cancer immunol. 51: 271 (2002) or the methods described in the examples section herein.

The peptides or compounds described herein, or intermediate peptides used in their Synthesis, may be synthesized according to the methods described in the examples section herein, or by using conventional techniques of Peptide Synthesis, such as those described in Peptide Synthesis, Interscience, New York, 1966; the Proteins, volume 2, academic press inc., New York, 1976; peptide Synthesis, Maruzen co., ltd., 1975; basicsand Experiment of Peptide Synthesis, Maruzen co., ltd., 1985; or Development of pharmaceutical Product subset 14, Peptide Synthesis, Hirokawa Shoten, 1991. Examples of such techniques include solid phase synthesis by Fmoc method or Boc method, or liquid phase synthesis by sequential condensation of Boc-amino acid or Z-amino acid in liquid phase (wherein Fmoc refers to 9-fluorenylmethoxycarbonyl, Boc refers to t-butoxycarbonyl, and Z refers to benzyloxycarbonyl). The peptide can be obtained by genetic techniques using a nucleotide sequence encoding the peptide according to a conventionally known method as described, for example, in Molecular Cloning, T. Maniatis et al, CSH Laboratory (1983), DNA Cloning, DM. Glover, IRL PRESS (1985).

In the synthesis of compounds of formula (1), functional groups on intermediate compounds, such as amino, carboxyl or thiol groups, may be protected with appropriate protecting groups, or deprotected as necessary by conventional techniques. For information on such protecting Groups, or methods of protection or deprotection, reference may be made to "Protective Groups in Organic Synthesis 2 nd edition (John Wiley & Sons, Inc.; 1990)". As the protecting group for a mercapto group, an acetamidomethyl group or a trityl group can be used.

When the compound of formula (1) includes a disulfide bond, a linkage is formed between two different cysteine-containing peptide components in the compound, or between a cysteine-containing peptide component and a cysteine residue in the compound. Such disulfide bonds can be formed, for example, by methods described in the following documents: peptide Synthesis, Interscience, New York, 1966; the Proteins, volume 2, Academic Press Inc., New York, 1976; peptesynthesis, Maruzen co., ltd., 1975; basics and experience of peptidesynthesis, Maruzen co., ltd., 1985; or Development of Pharmaceutical product sequential 14, Peptide Synthesis, Hirokawa Shoten, 1991.

Specifically, to prepare a compound having a disulfide bond (disulfide) from a peptide having one cysteine residue, the peptide may be subjected to a deprotection reaction to remove any protecting groups on functional groups including a thiol group on the cysteine residue, and then treated under oxidizing conditions in an inert solvent to form a disulfide bond. Disulfides can also be prepared from two different intermediates having a thiol group by treating them in a suitable solvent under oxidative conditions. The oxidation conditions for disulfide bond formation are known in the art of peptide synthesis. For example, known methods of iodine oxidation, air oxidation under basic conditions, or oxidation by an oxidizing agent under basic or acidic conditions may be used for the formation of disulfide bonds. As the oxidizing agent, iodine, dimethyl sulfoxide (DMSO), or potassium ferricyanide may be used. As reaction solvent, water, acetic acid, methanol, chloroform, DMF or DMSO or a mixture thereof may be used. Such oxidation conditions generally result in products in the form of mixtures of symmetric and asymmetric disulfides. The desired asymmetric disulfide can be recovered or purified by appropriate chromatography or recrystallization. Intermediates having an activated thiol group, such as a thiol group conjugated with an Npys group (3-nitro-2-pyridinesulfophenyl) may be used. To form a disulfide bond at a given thiol group on an intermediate, the group may be activated with 2,2' -dithiobis (5-nitropyridine) prior to coupling to another intermediate (Tetrahedron letters, Vol. 37, No. 9, p. 1347-.

The methods described above can be used to make disulfides from peptides having more than one cysteine residue. In this case, however, a mixture of different disulfides with disulfide bonds between different cysteine residues may be formed. In order to selectively prepare products dimerized via disulfide bonds between specific positions of monomers, different protecting groups may be used in combination to protect functional groups on cysteine residues. Examples of combinations of such protecting groups include a combination of MeBzl (methylbenzyl) and Acm (acetamidomethyl); trt (trityl) and Acm; npys (3-nitro-2-pyridylthio) and Acm; and S-Bu-t (S-t-butyl) and Acm. For example, when a peptide protected with a combination of MeBzl and Acm is used for selective disulfide bond formation, all MeBzl protecting groups and Acm protecting groups on functional groups on amino acid residues other than certain cysteine residues can be removed in a first step. Subsequently, by treating the peptide monomers in solution under air oxidation conditions, disulfide bonds can be formed between the selectively deprotected cysteine residues of the monomers. Subsequently, a further disulfide bond can be formed on the newly deprotected cysteine residue by removing the remaining Acm protecting group and treating with iodine under oxidative conditions.

The synthesized peptide, compound or intermediate may be purified by any purification method known in the art or in the field of peptide chemistry. Examples of such purification techniques include various types of chromatography (e.g., silica gel column chromatography, ion exchange column chromatography, gel filtration or reverse phase chromatography) and recrystallization from solvents (e.g., alcohols such as methanol, ethanol or 2-propanol, ethers such as diethyl ether, esters such as ethyl acetate, aromatic hydrocarbons such as benzene or toluene, ketones such as acetone, hydrocarbons such as hexane, aprotic solvents such as dimethylformamide or acetonitrile, water, or mixtures thereof). For further usable purification methods, reference may be made, for example, to volume 1 of Jikken KagakuKouza (the chemical Society of Japan ed., Maruzen). Methods for purification of disulfides are also described in the following documents: peptide Synthesis, Interscience, New York, 1966; the proteins, volume 2, Academic Press Inc., New York, 1976; peptide synthesis, Maruzen co., ltd., 1975; basics and experience of peptide synthesis, maruzen co., ltd., 1985; or Development of Pharmaceutical Product sequential volume 14, Peptide Synthesis, Hirokawa Shoten, 1991. Purification by HPLC is preferred.

The compounds of formula (1) may have one or more asymmetric centers. Such compounds can be prepared from starting materials (amino acids) having the corresponding asymmetric centers. Further, by including an optical resolution step in the synthesis process, the compound of formula (1) can be obtained with high optical purity. For example, according to the diastereomer method for optical resolution, the compound of formula (1) or the intermediate may be treated with an optically active acid (for example, a monocarboxylic acid such as mandelic acid, N-benzyloxyalanine or lactic acid, a dicarboxylic acid such as tartaric acid, o-diisopropylidene tartaric acid or malic acid, or a sulfonic acid such as camphorsulfonic acid or bromocamphorsulfonic acid) in an inert solvent (for example, an alcohol such as methanol, ethanol or 2-propanol, an ether such as diethyl ether, an ester such as ethyl acetate, an aromatic hydrocarbon such as toluene, an aprotic solvent such as acetonitrile, or a mixture thereof) to form a salt. For optical resolution of the compound of formula (1) or an intermediate having an acidic functional group (e.g., carboxyl group), a salt thereof can be formed with an optically active amine (e.g., an organic amine such as α -phenylethylamine, kinin, quinidine, cinchonidine, cinchonine, or strychnine).

The salts may be formed at temperatures from room temperature to the boiling point of the solvent used. In order to obtain the product in a highly optically pure form, it may be desirable to raise the temperature over a period of time to near the boiling point of the solvent. The salt formed is crystallized and subsequently filtered, optionally cooled to increase the yield. The optically active acid or amine for salt formation may be used in an amount of about 0.5 to about 2.0 equivalents, preferably about 1 equivalent, relative to the amount of the compound to be optically resolved. The crystalline product may optionally be further purified by recrystallization from an inert solvent (e.g., an alcohol such as methanol, ethanol or 2-propanol, an ether such as diethyl ether, an ester such as ethyl acetate, a hydrocarbon solvent such as toluene, an aprotic solvent such as acetonitrile, or mixtures thereof). The product recovered in salt form can optionally be converted to the free base or acid by treatment with an acid or base.

The term "pharmaceutically acceptable salts" as used herein includes both acid addition salts and base addition salts. The acid addition salt may be an inorganic acid salt such as a hydrochloride, hydrobromide, sulfate, hydroiodide, nitrate or phosphate, or an organic acid salt such as a citrate, oxalate, acetate, formate, propionate, benzoate, trifluoroacetate, maleate, tartrate, methanesulfonate, benzenesulfonate or p-toluenesulfonate. The base addition salts may be salts with inorganic bases such as sodium, potassium, calcium, magnesium or ammonium salts, salts with organic bases such as triethylammonium, triethanolammonium, pyridinium or diisopropylammonium salts. The "pharmaceutically acceptable salts" also include salts with basic or acidic amino acids, such as arginine, aspartic acid or glutamic acid. The term "peptide" or "compound" as used herein includes peptides or compounds in the form of pharmaceutically acceptable salts, unless the context requires otherwise.

The present disclosure includes, as only one step, a hydrate or solvate, such as an ethanol solvate, of a compound or peptide of formula (1) described herein, or a pharmaceutically acceptable salt thereof. The present disclosure also includes any stereoisomer, such as diastereomer or enantiomer, as well as any crystalline form of the compound or peptide described herein.

The compounds of formula (1) or pharmaceutically acceptable salts thereof described herein are useful for treating or preventing (including preventing recurrence of) cancer with WT1 gene expression or cancer with increased levels of WT1 gene expression. For example, the compound of formula (1) or a pharmaceutically acceptable salt thereof may be an active ingredient of a pharmaceutical composition (e.g., cancer vaccine), or a composition for inducing CTLs in cellular immunotherapy of cancer.

The compound of formula (1) described herein or a pharmaceutically acceptable salt thereof can be used for treating or preventing (including preventing recurrence of) "cancer accompanied by WT1 gene expression" or "cancer accompanied by increased WT1 gene expression level".

Examples of such cancers include blood cancers such as leukemia, myelodysplastic syndrome, multiple myeloma, and malignant lymphoma, as well as solid cancers such as gastric cancer, colorectal cancer, lung cancer, breast cancer, germ cell cancer, liver cancer, skin cancer, bladder cancer, prostate cancer, uterine cancer, cervical cancer, ovarian cancer, glioblastoma multiforme, malignant melanoma, non-small cell lung cancer, renal cell carcinoma, or brain tumors.

Other examples of cancers that may be treated or prevented by a compound of formula (1) or a pharmaceutically acceptable salt thereof include bone cancer, pancreatic cancer, head and neck cancer, cutaneous or intraocular malignant melanoma, rectal cancer, cancer of the anal region, cancer of the testis, cancer of the fallopian tubes, cancer of the endometrium, cancer of the cervix, cancer of the vagina, cancer of the vulva, hodgkin's disease, non-hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, chronic or acute leukemia such as acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia or chronic lymphocytic leukemia, solid tumors of childhood, lymphocytic lymphoma, cancer of the kidney or ureter, renal pelvis, Central Nervous System (CNS) tumors, primary CNS lymphoma, tumor angiogenesis, cancer of the brain, cancer of the, Spinal cord tumors, brain stem gliomas, pituitary adenomas, kaposi's sarcoma, epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma, environmentally induced cancers, including asbestos-induced cancers, and combinations thereof.

The compounds of formula (1) or pharmaceutically acceptable salts thereof described herein may be used in combination with at least one different cancer antigen peptide, in particular an MHC class I-restricted WT1 peptide or an MHC class II-restricted WT1 peptide, conjugates thereof, or pharmaceutically acceptable salts thereof. Examples of different cancer antigen peptides or conjugates include peptides or derivatives thereof, or conjugates thereof, described in the following publications: WO 2000/006602, WO 2002/079253, WO 2003/106682, WO 2004/026897, WO2004/063903, WO 2007/063903, WO 2010/123065, WO2014/157692, WO 2005/053618, WO2007/047764, WO 2007/120673, WO 2005/045027, WO 2010037395, WO 2000/018795, WO2002/028414, WO 2003/037060 and WO 2004/100870.

In one embodiment, the compound of formula (1) or a pharmaceutically acceptable salt thereof is used in combination with at least one cancer antigen peptide E or a pharmaceutically acceptable salt thereof, wherein the at least one cancer antigen peptide E is an MHC class I-restricted peptide consisting of 7 to 30 amino acid residues, preferably an MHC class I-restricted WT1 peptide.

The compound of formula (1) or a pharmaceutically acceptable salt thereof, or a combination of the compound of formula (1) or a pharmaceutically acceptable salt thereof and at least one different cancer antigen peptide or a pharmaceutically acceptable salt thereof may be used in combination with at least one different drug (hereinafter referred to as a co-administered drug).

The co-administered drug may be an "immunomodulator". The term "immunomodulator" as used herein refers to any agent that controls the transmission of costimulatory signals generated by antigen-presenting cells during T cell activation by interacting with molecules involved in the transmission of costimulatory signals and present on antigen-presenting cells and/or T cells, as well as any agent that directly or indirectly controls the function of molecules involved in the establishment of immune tolerance (immunosuppression) in the immune system. Since cancer antigen peptides effectively increase tumor-reactive CTLs in tumors, they may be used as agents to be co-administered with immunomodulators, to reduce the necessary dose of immunomodulators or to reduce adverse events caused by immunomodulators. Thus, by using the WT1 antigenic peptide in combination with an immunomodulator, the present disclosure provides a therapy with improved efficacy and safety to a patient.

The "immunomodulator" may be an agent in the form of an antibody, nucleic acid, protein, peptide or small compound, but is not limited thereto. "antibodies" as "immunomodulators" include antibody fragments. Examples of antibody fragments include the heavy and light chain variable regions of an antibody (VH and VL), F (ab ')2, Fab', Fab, Fv, Fd, sdFv and scFV. "protein" as an "immunomodulator" refers to any protein other than an antibody. Examples of "immune modulators" include immune checkpoint inhibitors, co-stimulatory molecule agonists, immune activators, and low-molecular inhibitors.

The "immune checkpoint inhibitor" inhibits immune suppression induced by cancer cells or antigen presenting cells. Examples of immune checkpoint inhibitors include, but are not limited to, agents directed against a molecule selected from the group consisting of: (1) CTLA-4 (e.g., ipilimumab and teximumab); (2) PD-1 (e.g., nivolumab, pembrolizumab, AMP-224, AMP-514(MEDI0680), and pidilizumab (CT-011)); (3) LAG-3 (e.g., IMP-321 and BMS-986016); (4) BTLA; (5) KIR (e.g., IPH 2101); (6) TIM-3; (7) PD-L1 (e.g., Duvaluzumab (MEDI4736), MPDL3280A, BMS-936559, and Avermezumab (MSB 0010718C)); (8) PD-L2; (9) B7-H3 (e.g., MGA-271); (10) B7-H4; (11) HVEM; (12) GAL 9; (13) CD 160; (14) VISTA; (15) BTNL 2; (16) TIGIT; (17) PVR; (18) BTN1a 1; (19) BTN2a 2; (20) BTN3A2 (Nat Rev Drug Discov. 2013; 12: 130-146; Nikkei Medical cancer review 2014; 9; Nat Rev Immunol. 2014; 14: 559-69); and (21) CSF 1-R.

"costimulatory molecule agonists" enhance T cell activation by transmitting accessory signals via costimulatory molecules on T cells and/or antigen presenting cells and attenuate the immunosuppressive effects of cancer cells or antigen presenting cells. Examples of co-stimulatory molecule agonists include, but are not limited to, agents directed against a molecule selected from the group consisting of: (1) 4-1 BB; (2) 4-1 BB-L; (3) OX 40; (4) OX 40-L; (5) GITR; (6) CD 28; (7) CD 40; (8) CD 40-L; (9) ICOS; (10) ICOS-L; (11) LIGHT; and (12) CD 27.

The "immune activator" effectively stimulates killer T cells in lymph nodes by directly or indirectly activating immune cells such as T cells and dendritic cells. Examples of immune activators include, but are not limited to, Toll-like receptor (TLR) agonists, stimulators of interferon gene (STING) agonists, cytokines, and agents against Heat Shock Proteins (HSPs).

Examples of "Toll-like receptor (TLR) agonists" include, but are not limited to, TLR1/2 agonists, TLR2 agonists, TLR3 agonists (e.g., PolyI: C), TLR4 agonists (e.g., S-type lipopolysaccharide, paclitaxel, lipid a, and monophosphoryl lipid a), TLR5 agonists (e.g., flagellin), TLR6/2 agonists (e.g., MALP-2), TLR7 agonists, TLR7/8 agonists (e.g., galdomot, imiquimod, loxoribine, and resiquimod (R)), TLR7/9 agonists (e.g., hydroxychloroquine sulfate), TLR8 agonists (e.g., mototonimod 848 (VTX-2337)), TLR9 agonists (e.g., CpG-ODN), and TLR11 agonists (e.g., arrestins).

Examples of "cytokines" include, but are not limited to, IL-1 α, IL-1 β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, Interferon (INF) - α, INF- β, INF- γ, SCF, GM-CSF, G-CSF, M-CSF, erythropoietin, thrombopoietin, Macrophage Inflammatory Protein (MIP), and Monocyte Chemotactic Protein (MCP).

Examples of "Heat Shock Proteins (HSPs)" include, but are not limited to, HSP70, HSP90, HSP90 α, HSP90 β, HSP105, HSP72, and HSP 40. Agents against heat shock proteins include HSP inhibitors. Examples of inhibitors of HSP90 include, but are not limited to, tanespimycin (17-AAG), lomispide (AUY-922, NVP-AUY 922), apramycin (17-DMAG) hydrochloride, Ganetespib (STA-9090), BIIB021, onaprisib (onalespib) (AT 13387), geldanamycin, NVP-BEP800, SNX-2112 (PF-04928473), PF-4929113 (SNX-5422), KW-2478, XL888, VER155008, VER-50589, CH5138303, VER-49009, NMS-E973, PU-H71, HSP990 (NVP-HSP 990), and KNK 437.

Examples of "low molecular inhibitors" include, but are not limited to, histone deacetylase inhibitors, histone demethylase inhibitors, histone acetyltransferase inhibitors, histone methyltransferase inhibitors, DNA methyltransferase inhibitors, anthracyclines, platinum agents, MAPK inhibitors, beta-catenin inhibitors, STAT3 inhibitors, NF-kB inhibitors, JAK inhibitors, mTOR inhibitors, IDO inhibitors, COX-2 inhibitors, CXCR4 inhibitors, and arginase inhibitors.

Examples of "histone deacetylase inhibitors" include, but are not limited to, vorinostat (SAHA, MK 0683), entinostat (MS-275), panobinostat (LBH 589), trichostatin A (TSA), motinostat (MGCD 0103), BG45, BRD73954, belinostat (PXD 101), romidepsin (FK 228, depsipeptide), 4SC-202, HPOB, LMK-235, CAY10603, Taquinmod, TMP269, nextarat A, JNEColinostat (ACY-1215), RGFP966, RG2833 (FP 109 FP), scriptaid, tubastatin A, practinostat (RGacin) (SB 939), CUDC-101, M344, PCI-34051, darussitast (LAQ), bacstatin A hydrochloride, Abystatin (PCI-2451), RGsciostat (RG-3542), Curstat (RG-3542, Curtatt-2358, hydrastin-055 (RG) and D, and D-1588, and their salts (DRY-1215, RGF) and their hydrochloride, CHR-2485, CHR-3996, DAC-060, FRM-0334 (EVP-0334), MGCD-290, CXD-101 (AZD-9468), CG200745, arginine butyrate, sulforaphane, SHP-141, CUDC-907, YM753 (OBP-801), sodium valproate, apicidin, and CI994 (Tadenland).

Examples of "histone demethylase inhibitors" include, but are not limited to, GSK J4 HCl, OG-L002, JIB-04, IOX1, SP2509, ORY-1001 (RG-6016), GSK J1, ML324, and GSK-LSD 12 HCl.

Examples of "histone acetyltransferase inhibitors" include, but are not limited to, C646, MG149, remodelain, and anacardic acid.

Examples of "histone methyltransferase inhibitors" include, but are not limited to, pinometstat (EPZ 5676), EPZ005678, GSK343, BIX01294, tazemetostat (EPZ 6438), 3-deazaadenine A (DZNeP) HCl, UNC1999, MM-102, SGC0946, entacapone, EPZ015666, UNC0379, EI1, MI-2 (menin-MLL inhibitor), MI-3 (menin-MLL inhibitor), PFI-2, GSK126, EPZ04777, BRD4770, GSK-2816126, and UNC 0631.

Examples of "DNA methyltransferase inhibitors" include, but are not limited to, decitabine, azatadine, RG108, thioguanine, zebularine, SGI-110, CC-486, SGI-1027, lomeguazatine, and procainamide hydrochloride.

An "anthracycline" is inserted between DNA strands to inhibit DNA relaxation. Examples of anthracyclines include, but are not limited to, doxorubicin, liposomal doxorubicin, daunorubicin, pirarubicin, epirubicin, idarubicin, aclarubicin, amrubicin, aloin, and mitoxantrone.

Examples of "platinum agents" include, but are not limited to, cisplatin, carboplatin, miriplatin, nedaplatin, satraplatin (JM-126), oxaliplatin (ELOXATIN), triplatin tetranitrate, and DDS agents thereof.

Examples of "MAPK inhibitors" include, but are not limited to SB203580, dammar (BIRB 796), SB202190 (FHIPI), LY2228820, VX-702, SB239063, pemetrexed (ARRY-614), PH-797804, VX-745, and TAK-715.

Examples of "β -catenin inhibitors" include, but are not limited to, XAV-939, ICG-001, IWR-1-endo, Wnt-C59 (C59), LGK-974, KY02111, IWP-2, IWP-L6, WIKI4, and FH 535.

Examples of "STAT 3 inhibitors" include, but are not limited to, S3I-201, Static, niclosamide, nifuroxazide, napobucasin (BBI 608), cryptotanshinone, HO-3867, WHI-P154, FLLL32, STA-21, WP1066, and SH-4-54.

Examples of "NF-kB inhibitors" include, but are not limited to, QNZ (EVP 4593), sodium 4-aminosalicylate, JSH-23, caffeic acid phenethyl ester, sodium salicylate, andrographolide, and SC 75741.

Examples of "JAK inhibitors" include, but are not limited to, ruxotinib (INCB 018424), tofacitinib (CP-690550) citrate, AZD1480, phenanthrotinib (SAR 302503, TG 101348), AT9283, tyrphostin B42 (AG-490), molotinib (CYT 387), tofacitinib (CP-690550, tasocitinib), WP1066, TG101209, gandoltinib (gandottinib) (LY 2784544), NVP-BSK 8052 HCl, balitinib (LY 3009104, INCB 02850), CEP 960, CEP-33779, parkitinib (SB 1518), WHI-P154, XL019, S-ruxotinib (INCB 018424), ZM 923 HCl, dactinib (VX-39509), ceritinib (PRT 070, PRT 0620), non-rosotinib (FLLL 0624), GLPG 207015, GLJ 366983, non-GLJ-366376, and non-Gl analogs.

Examples of "mTOR inhibitors" include, but are not limited to, sirolimus (rapamycin), deforolimus (AP 23573, MK-8669), everolimus (RAD-001), temsirolimus (CCI-779, NSC 683864), umirolimus (ABT-578), sirolimus A9 (temsirolimus), AZD8055, KU-0063794, voxtalisib (XL 765, SAR 2458409), MHY1485, dacrolimus (BEZ 235, NVP-BEZ 235), PI-103, and torkinib (PP 242).

Examples of "IDO inhibitors" include, but are not limited to, NLG919, INCB024360 analogs, indoimod (NLG-8189), and epratstat (INCB 024360).

Examples of "COX-2 inhibitors" include, but are not limited to, valdecoxib, rofecoxib, carprofen, celecoxib, lumiracoxib, tolfenamic acid, nimesulide, niflumic acid, asanal, lornoxicam, meclofenamate sodium, amfenac sodium hydrate, diclofenac sodium, ketoprofen, ketorolac, naproxen sodium, indomethacin, ibuprofen, aspirin, mefenamic acid, bromfenac sodium, oxaprozin, zatoprofen, and nepafenac.

Examples of "CXCR 4 inhibitors" include, but are not limited to, WZ811, plerixafor (AMD 3100), and plerixafor 8HCl (AMD 31008 HCl).

The co-administered drug may also be one or more drugs selected from the group consisting of "hormonal therapy agents", "immunotherapeutic agents", "biopharmaceuticals", "cell growth factors", "cell growth factor inhibitors", "cell growth factor receptor inhibitors", "radiotherapeutic agents", "adjuvant agents" and "chemotherapeutic agents". For example, one to five drugs, one to three drugs, or one drug selected from the above drug groups may be used in combination with the peptide described herein, or the compound of formula (1), or a pharmaceutically acceptable salt thereof, or a combination thereof.

Examples of "hormone therapy agents" include adrenocortical hormone agents (e.g., steroidal anti-inflammatory drugs, estrogen preparations, progesterone preparations, and androgen preparations), antiestrogens, estrogen control agents, estrogen synthesis inhibitors, antiandrogens, androgen control agents, androgen synthesis inhibitors, LH-RH agonist preparations, LH-RH antagonist preparations, aromatase inhibitors, steroid lactonase inhibitors, contraceptive pills, retinoids, and agents that delay retinoid metabolism.

Examples of "hormonal therapy agents" include fosfestrol, diethylstilbestrol, fluoxymesterol, clomestranol, methyltestosterone, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, cyproterone acetate, danazol, allylestrenol, gestrinone, mepartricin, raloxifene, oxymetafene, levomeloxifene, tamoxifen citrate, toremifene citrate, idoxifene, contraceptive pills, cyclopentadecylepithioandrostane, dihydrotestolactone, aminoglutethimide, goserelin acetate, ethylamide, leuprolide (leromelide), droloxifene, epitroxitol, ethisterol sulfonate, estramustine, fadrozole hydrochloride, anastrozole, tetrazole, ketoconazole, letrozole, exemestane, clomazole, formestane, flutamide, bicalutamide, nilutamide, dexrazol, dexirectionalamine, dexamethasone, nefirlufilgrazone, dexamethasone, fexofenadone, levansetron, dexamethasone, levansetron, levamisole, levomison, levosalbutamol, and the like, Prednisolone, betamethasone, triamcinolone, abiraterone, liazole, bexarotene and DN 101.

Examples of "immunotherapeutics" include streptolysin, coriolus versicolor polysaccharide, cezopyran, lentinan, ubenimex, Interferon (IFN) -alpha, Interferon (IFN) -beta, Interferon (IFN) -gamma, interleukins, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, lymphotoxins, bcg, corynebacterium parvum, levamisole, polysaccharide K, phenylimidazolepropionic acid, anti-CTLA 4 antibodies, anti-PD-1 antibodies, and TLR agonists (e.g., TLR7 agonists, TLR8 agonists, TLR9 agonists).

Examples of "biopharmaceuticals" include, but are not limited to, interleukin-2 (aldesleukin), interferon-a, interferon- β, interferon- γ, Erythropoietin (EPO), granulocyte colony stimulating factor (filgrastim), granulocyte macrophage colony stimulating factor (samustine), IL13-PE38QQR, Bacillus Calmette-Guerin, levamisole, octreotide, CPG7909, Provenge, GVAX, Myvax, Favld, lenalidomide, trastuzumab, rituximab, gemtuzumab, alemtuzumab, endostatin, ibritumomab, tositumomab, cetuximab, zamumab, ofatumumab, HGS-ETR1, pertuzumab, M200, SGN-30, matuzumab, adalimumab, disumab, temumab, MDX-003, tussib, 060-060, Vitaxin, MDX-101, MDX-010, DPC4 antibodies, NF-1 antibodies, NF-2 antibodies, Rb antibodies, p53 antibodies, WT1 antibodies, BRCA1 antibodies, BRCA2 antibodies, gangliosides (GM 2), Prostate Specific Antigen (PSA), alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), melanoma associated antigens (MART-1, gap100, MAGE 1,3 tyrosine), papillomavirus E6 and E7 fragments, and DDS formulations thereof.

With respect to "cell growth factor", "cell growth factor inhibitor" and "cell growth factor receptor inhibitor", the cell growth factor may be any agent that promotes cell proliferation. For example, the cell growth factor may be a peptide having a molecular weight of no more than 20,000 and which can bind to a receptor and function at low concentrations.

Examples of "cell growth factors" include, but are not limited to, Epidermal Growth Factor (EGF), insulin-like growth factors (IGF (e.g., insulin, IGF-1 and IGF-2)), transforming growth factors (TGF (e.g., TGF-alpha and TGF-beta)), Nerve Growth Factor (NGF), brain-derived neurotrophic factor (BDNF), Vascular Endothelial Growth Factor (VEGF), colony Stimulating Factors (CSF) (e.g., granulocyte colony stimulating factor (G-CSF)), granulocyte macrophage colony stimulating factor (GM-CSF)), platelet-derived growth factor (PDGF), Erythropoietin (EPO), Fibroblast Growth Factors (FGF) (e.g., acidic FGF, basic FGF, keratinocyte growth factor (KGK), and FGF-10)), Hepatocyte Growth Factor (HGF), heregulin, and angiogenin. The term "cell growth factor" is synonymous with the term "growth factor".

Examples of "cell growth factor inhibitors" include, but are not limited to, epidermal growth factor inhibitors (EGF inhibitors), insulin-like growth factor inhibitors (IGF inhibitors), nerve growth factor inhibitors (NGF inhibitors), brain-derived neurotrophic factor inhibitors (BDNF inhibitors), vascular endothelial cell growth factor inhibitors (VEGF inhibitors), colony stimulating factor inhibitors (CSF inhibitors), platelet-derived growth factor inhibitors (PDGF inhibitors), erythropoietin inhibitors (EPO inhibitors), fibroblast growth factor inhibitors (FGF inhibitors), hepatocyte growth factor inhibitors (HGF inhibitors), heregulin inhibitors, and angiogenin inhibitors. The term "cell growth factor inhibitor" is synonymous with the term "growth factor inhibitor".

Examples of "cell growth factor receptor inhibitors" include, but are not limited to, epidermal growth factor receptor inhibitors (EGFR inhibitors), insulin-like growth factor receptor inhibitors (IGFR inhibitors), nerve growth factor receptor inhibitors (NGFR inhibitors), brain-derived neurotrophic factor receptor inhibitors (BDNFR inhibitors), vascular endothelial growth factor receptor inhibitors (VEGFR inhibitors), colony stimulating factor receptor inhibitors (CSFR inhibitors), platelet-derived growth factor receptor inhibitors (PDGFR inhibitors), erythropoietin receptor inhibitors (EPOR inhibitors), fibroblast growth factor receptor inhibitors (FGFR inhibitors), hepatocyte growth factor receptor inhibitors (HGFR inhibitors), heregulin receptor inhibitors, and angiopoietin receptor inhibitors. The term "inhibitor of a cell growth factor receptor" is synonymous with the term "inhibitor of a growth factor receptor".

Examples of "radiotherapeutic agents" include, but are not limited to, radioactive materials and radiosensitizers.

An "adjuvant" is a drug used together with an anticancer agent to suppress side effects or vomiting caused by the anticancer agent. Examples of "adjuvants" include, but are not limited to, aprepitant, ondansetron, lorazepam, dexamethasone, diphenhydramine, ranitidine, cimetidine, ranitidine, famotidine, cimetidine, Procrit, recombinant human erythropoietin, filgrastim, alprenil, leucovorin, and granulocyte-macrophage colony stimulating factor (GM-CSF).

Examples of "chemotherapeutic agents" include, but are not limited to, alkylating agents, platinum agents, antimetabolites, topoisomerase inhibitors, DNA intercalators, antimitotic agents, antitumor antibiotics, plant-derived anticancer agents, epigenomic drugs, immunomodulators, molecularly targeted drugs, angiogenesis inhibitors and other chemotherapeutic agents. Some typical examples of chemotherapeutic agents are listed below.

Examples of "alkylating agents" include, but are not limited to, nitrogen mustard N-oxide hydrochloride, chlorambucil, cyclophosphamide, ifosfamide, thiotepa, carboquone, improsulfate, busulfan, nimustine hydrochloride, dibromomannitol, melphalan, dacarbazine, procarbazine, ranimustine, estramustine sodium phosphate, tritamine, carmustine, lomustine, streptozotocin, bromopropiperazine, epidoxine, hexamethylmelamine, AMOMATIN, dibromospiro ammonium chloride, fotemustine, prednimustine, bendamustine, uracil mustard, semustine, pyrimidotene, ribomustin, temozolomide, busulfan, chloroacetamide, setastine, adolesin, cysteamine nitrosurea, bizelesin, mechlorethamine, uracil mustard, sultaine, gansufamide, gansufam, mechlorethamine, thizine, etc, Triimine quinone, procarbazine, canflunomide (canfosfamide), nitrosourea and DDS formulations thereof.

Examples of "platinum agents" include, but are not limited to, cisplatin, carboplatin, miriplatin, nedaplatin, satraplatin, oxaliplatin, triplatin tetranitrate, and DDS agents thereof.

Examples of "antimetabolites" include, but are not limited to, antifolates, pyrimidine metabolism inhibitors, purine metabolism inhibitors, ribonucleotide reductase inhibitors, and nucleotide analogs.

Examples of "antimetabolites" include, but are not limited to, mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, pemetrexed, eoshitabin, enocitabine, cytarabine octadecylphosphate, ancitabine hydrochloride, 5-FU agents (e.g., fluorouracil, Carzonal, Bennan, Lunachol, Lunapon, tegafur-uracil, tegafur-gimerazine-oxonate (TS-1), UFT, doxifluridine, carmofur, gallocotidine, ethimidin and capecitabine), aminopterin, nelarabine, leucovorin, Tabloid, tetracaine, calcium folinate, calcium levofolinate, drotabine, ethidium, fludarabine, gemcitabine, hydroxyurea, pentostatin, picomole, idoxuridine, mitoguazone, furamectin, timothiazolirtine, trimebutamiodarone, and trimebutine, Floxuridine, leucovorin, hydroxyurea, thioguanine, asparaginase, bortezomib, raltitrexed, clofarabine, enocitabine, sabotaabine, azacytidine, sulfadiazine, sulfamethoxazole, trimethoprim, Liproxstatin-1, D4476, xanthohumol, Epacadostat (INCB 024360), Vidofludumus, P7C3, GMX1778 (GMS 828), NCT-501, SW033291, Ro61-8048 and DDS formulations thereof.

Examples of "topoisomerase inhibitors" include, but are not limited to, doxorubicin, daunorubicin, epirubicin, idarubicin, anthracenedione, mitoxantrone, mitomycin C, bleomycin, dactinomycin, mithramycin, irinotecan, camptothecin, rubitecan, belotecan, etoposide, teniposide, topotecan, amsacrine, and DDS formulations thereof.

Examples of "DNA intercalators" include, but are not limited to, proflavine, doxorubicin (adriamycin), daunomycin, dactinomycin, thalidomide, and DDS formulations thereof.

Examples of "antimitotic agents" include, but are not limited to, paclitaxel derivatives (e.g., DHA paclitaxel, polyglutamic acid paclitaxel, nab-paclitaxel, micellar paclitaxel, 7 α -glucosyloxyacetyl paclitaxel, and BMS-275183), docetaxel, vinorelbine, vincristine, vinblastine, vindesine, vinzolidine, etoposide, teniposide, ixabepilone, larotaxel, otaxel, titaxel, isxepin, colchicine, vinflunine, and DDS formulations thereof.

Examples of "antitumor antibiotics" include, but are not limited to, actinomycin D, actinomycin C, mitomycin C, chromomycin A3, mithramycin A, bleomycin hydrochloride, bleomycin sulfate, pellomycin sulfate, daunomycin hydrochloride, doxorubicin hydrochloride, pirarubicin hydrochloride, epirubicin hydrochloride, amrubicin hydrochloride, neocarzinostat, setastin, mithramycin, sarcomycin, carzinostatin, mitomycin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride, liposomal doxorubicin, and DDS formulations thereof.

Examples of "plant-derived anticancer agents" include, but are not limited to, irinotecan, topotecan (nogitecan), etoposide phosphate, eribulin, sobuzosin, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel injection, docetaxel, DJ-927, vinorelbine, topotecan, and DDS formulations thereof.

Examples of "epigenomic agents" include, but are not limited to, DNA methylation inhibitors, Histone Deacetylase (HDAC) inhibitors, DNA methyltransferase (DNMT) inhibitors, histone deacetylase activators, histone demethylase inhibitors, and methylated nucleotides.

Specific examples of "epigenomic drugs" include, but are not limited to, vorinostat, belinostat, motinostat (MGCD 0103), entinostat (SNDX-275), romidepsin, azacytidine, decitabine, GSK 28795522 Hl, SGC707, ary-1001 (RG-6016), PFI-4, SirReal2, GSK2801, CPI-360, GSK503, AMI-1, CPI-169, and DDS formulations thereof.

Examples of "immunomodulators" include, but are not limited to, thalidomide, lenalidomide, pomalidomide and DDS formulations thereof.

The "molecularly targeted drug" may be a small compound or an antibody. Examples of "molecularly targeted drugs" include, but are not limited to, kinase inhibitors, proteasome inhibitors, monoclonal antibodies, mTOR inhibitors, TNF inhibitors, and T cell inhibitors.

Examples of "kinase inhibitors" include, but are not limited to, tyrosine kinase inhibitors, serine/threonine kinase inhibitors, Raf kinase inhibitors, Cyclin Dependent Kinase (CDK) inhibitors, and mitogen-activated protein kinase (MEK) inhibitors.

Specific examples of "kinase inhibitors" include, but are not limited to, imatinib, gefitinib, erlotinib, afatinib, dasatinib, bosutinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, lapatinib, nilotinib, crizotinib, ceritinib, luotinib, tofacitinib, ibrutinib, sorafenib, verofenib, dabrafenib, palbociclib, trametinib, regorafenib, cedivanib, lestatinib, tivatinib, canertinib, pelitinib, tesivatinib, cedatinib, saidinib, indomositinib, mircotatinib, foronib, cabozantinib, neritinib, lenacitinib, neratinib, volatinib, saratinib (icotinib-62), trovadorib, saratinib, valacitinib, valcanitinib, valtinib, valcani, AEE788, PD0325901, PD153035, TK787, ancatin (BBI 503), E6201, E7050 and DDS formulations thereof.

Examples of "proteasome inhibitors" include, but are not limited to, bortezomib, carfilzomib, and DDS formulations thereof.

Examples of "monoclonal antibodies" include, but are not limited to, anti-CD 22 antibodies, anti-CD 20 antibodies, anti-CD 25 antibodies, anti-CD 30 antibodies, anti-CD 33 antibodies, anti-CD 5 antibodies, anti-CD 52 antibodies, anti-epidermal growth factor receptor antibodies (EGFR antibodies), anti-vascular endothelial cell growth factor antibodies (VEGF antibodies), anti-TNF- α antibodies, anti-IL-1 receptor antibodies, anti-IL-2 receptor antibodies, anti-IL-5 receptor antibodies, anti-IL-6 receptor antibodies, anti-HER 2 antibodies, anti-IgE antibodies, anti-IgG antibodies, anti-RS virus antibodies, anti-CCR 4 antibodies, anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4, CD 152) antibodies, anti-PD-1 antibodies, anti-nuclear factor κ B ligand (RANKL) receptor activator antibodies, anti-c-Met antibodies, and anti-CXCR 4 antibodies.

Specific examples of "monoclonal antibodies" include, but are not limited to, ibritumomab tiuxetan, rituximab, cetuximab, infliximab, basiliximab, brevizumab, bevacizumab, omalizumab, meprilizumab, gemtuzumab, palivizumab, ranibizumab, certolizumab, omelizumab (ocrelizumab), moglicalizumab, eculizumab, pertuzumab, alemtuzumab, eculizumab, alemtuzumab, ecumab, panitumumab, ofatumumab, golimumab, adalimumab, ramucirumab, nivolumab, anakinumab, mumab, ipilimumab, matuzumab, farletuzumab, mor-004, a-b ab 009, DDS, and formulations thereof.

Examples of "mTOR inhibitors" include, but are not limited to, everolimus (RAD 001), rapamycin (sirolimus), AZD8055, temsirolimus (CCI-779, NSC 683864), KU-0063794, voxtalisib (XL-765, SAR 24409), MHY1485, dactulisib (BEZ 235), PI-103, torkinib (PP 242), deforolimus (MK-8669), INK-128 (MLN 0128), Torin1, omisib (GSK 2126458, GSK 458), OSI-027, PF-04691502, apitolisib (GDC-0980, RG 7422), GSK 9615, gedatolisib (PF-05212384, PKI-587), WYE-132, PP121, WYE-354, AZD2014, WYE-2, WYE-105797, WYE-51799, BGT-68226, Wye-202, WZ-202, and its formulation.

Examples of "TNF inhibitors" include, but are not limited to, etanercept, lenalidomide (CC-5013), pomalidomide, thalidomide, necrostatin-1, and QNZ (EVP 4593).

Examples of "T cell inhibitors" include, but are not limited to, abacavir.

Examples of "angiogenesis inhibitors" include, but are not limited to, CM101, IFN- α, IL-12, platelet factor-4, suramin, semanib, thrombospondin, VEGFR antagonists, combinations of angiogenesis inhibiting steroids and heparin, cartilage derived angiogenesis inhibitors, matrix metalloproteinase inhibitors, batimastat, marimastat, angiogenesis inhibitors, endostatin, 2-methoxyestradiol, ticagreland, thrombospondin, α V β 3 inhibitors, linodiamide, ADH-1, E7820, and DDS preparations thereof.

Examples of "other chemotherapeutic agents" include, but are not limited to, finasteride, sobuzolne, obatoclax, ethacryloxide, tipifarnib, and lonafarnib.

When the compound of formula (1) or a pharmaceutically acceptable salt thereof described herein is used in combination with at least one cancer antigen peptide or a pharmaceutically acceptable salt thereof and/or a co-administered drug, these active agents may be formulated in separate compositions or incorporated into a single composition. In one embodiment, the compound of formula (1) or a pharmaceutically acceptable salt thereof and the cancer antigen peptide are incorporated into a single composition. In another embodiment, the compound of formula (1) or a pharmaceutically acceptable salt thereof and the cancer antigen peptide are formulated in separate compositions. The composition may comprise one or more compounds of formula (1) or a pharmaceutically acceptable salt thereof and/or one or more cancer antigen peptides. Compositions comprising a compound of formula (1) or a pharmaceutically acceptable salt thereof or a cancer antigen peptide may be provided with dosages and instructions for administration for use of the composition in combination with other active agents. The composition comprising the compound of formula (1) or a pharmaceutically acceptable salt thereof and the composition comprising the cancer antigen peptide may be incorporated into a single kit. Such kits may further comprise instructions for dosage and administration so that the compositions are used in combination, or may be packaged. Where more than one active agent is administered in combination, the agents may be administered according to the same dosing regimen or different dosing regimens.

The compositions of the present disclosure may comprise, as an active agent, a compound of formula (1) described herein, the peptide, or a pharmaceutically acceptable salt thereof, or a combination thereof, and a pharmaceutically acceptable carrier. In addition, the compositions of the present disclosure may further comprise or be administered in combination with a suitable adjuvant to enhance the induction of WT 1-specific CTLs and/or helper T cells by the composition.

By "pharmaceutically acceptable carrier" is meant a carrier that is non-toxic to the cells or mammal exposed to the carrier at the amount or concentration at which it is used. In some embodiments, a pH buffered aqueous solution is used as a pharmaceutically acceptable carrier. Examples of "pharmaceutically acceptable carriers" include buffers (such as phosphate, citrate, lactate, tartrate, trifluoroacetate and other organic acids); antioxidants (such as ascorbic acid); low molecular weight polypeptides (less than about 10 residues); proteins (such as serum albumin, gelatin, or immunoglobulins); hydrophilic polymers (such as polyvinylpyrrolidone); amino acids (such as glycine, glutamine, asparagine, arginine, methionine or lysine); monosaccharides, disaccharides, and other carbohydrates (such as glucose, mannose, or dextrins); chelating agents (such as EDTA); sugar alcohols (such as mannitol, trehalose or sorbitol); stabilizers (such as diethylenetriaminepentaacetic acid); salt-forming counterions (e.g., sodium); solubilizers (e.g., polysorbate 80 and/or nonionic surfactants (e.g., TWEEN, polyethylene glycol (PEG) and PLURONICS)). Slowly metabolized macromolecular materials such as proteins, polypeptides, liposomes, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers or inactive viral particles can also be used as pharmaceutically acceptable carriers. For administration, the compounds or peptides of formula (1) described herein may be formulated in liposome formulations, attached to beads of micron-sized diameter, or associated with liquid carriers.

The adjuvant may be any of the adjuvants described in Clin. Microbiol. Rev., 7: 277-289, 1994. Specifically, the adjuvant may be a microorganism-derived agent, GM-CSF, a cytokine (e.g., interleukin-2, interleukin-7, or interleukin-12), a plant-derived agent, a marine organism-derived agent, a mineral gel (e.g., aluminum hydroxide), lysolecithin, a surfactant (e.g., pluronic polyol), a polyanion, a peptide, or an oil emulsion (emulsion formulation). Examples of the microorganism-derived agents include lipid a, monophosphoryl lipid a which is a derivative of lipid a, killed bacteria (e.g., mycobacterial bacteria such as BCG bacteria), bacteria-derived proteins, polynucleotides, freund's incomplete adjuvant, freund's complete adjuvant, cell wall skeleton components (e.g., BCG-CWS), Trehalose Dimycolate (TDM).

The adjuvant may also be a precipitation adjuvant or an oil adjuvant. The precipitation adjuvant may be a suspension of an inorganic substance adsorbing the peptide. Examples of precipitation adjuvants include sodium hydroxide, aluminum hydroxide (Alum), calcium phosphate, aluminum phosphate, Alum, Pepesu, and carboxyvinyl polymers. The oil adjuvant may be an oil emulsifier capable of emulsifying peptides by forming micelles comprising an aqueous peptide phase encapsulated in a mineral oil film. Examples of oil adjuvants include, but are not limited to, liquid paraffin, lanolin, Freund's adjuvant (Freund's complete adjuvant, Freund's incomplete adjuvant), Montanide, and W/O emulsion (see WO 2006/078059).

The compositions of the present disclosure may be provided as dosage forms for oral administration or parenteral administration. Examples of dosage forms for parenteral administration include injectable preparations, external preparations, suppositories, inhalable preparations or nasal preparations. In a preferred embodiment, the compositions of the present disclosure are provided as injectable formulations.

Injectable formulations may be in the form of solutions, suspensions or emulsions comprising one or more active agents dissolved, dispersed or emulsified in the liquid used for injection, or may be provided as solid formulations comprising the active agent dissolved or dispersed in the liquid used for injection prior to use. The liquid for injection may comprise distilled water, physiological saline, vegetable oil, propylene glycol, polyethylene glycol, alcohol (e.g., ethanol), or a combination thereof for injection. The injectable formulation may additionally comprise stabilizers, solubilizing aids (such as glutamic acid, aspartic acid or polysorbate 80), dispersants, emulsifiers, analgesics, buffers, preservatives or other suitable additives. In order to provide an injectable preparation in the form of a sterile preparation, it may be sterilized in the final step of its production, or aseptically produced throughout its production. Formulations for injection may be presented as a sterile solid preparation, e.g., a lyophilized preparation, which may be reconstituted in sterile water for injection or other suitable sterile liquid prior to use.

The external preparation may be in the form of an ointment, gel, cream, plaster, patch, liniment, spray, inhalant, aerosol, eye drop or nasal drop, which may be prepared according to a conventionally known preparation method, and may contain one or more active agents.

Ointments may be prepared according to generally known methods of preparation, for example by mixing one or more active agents into an ointment base by grinding or melting. For preparing the ointment, any conventionally used ointment base may be used, which may comprise higher fatty acids or fatty acid esters (such as adipic acid, myristic acid, palmitic acid, stearic acid or oleic acid, or esters thereof), waxes (such as beeswax, spermaceti or ozokerite), surfactants (such as polyoxyethylene alkyl ether phosphate), higher alcohols (such as cetyl alcohol, stearyl alcohol or cetostearyl alcohol), silicone oils (such as dimethylpolysiloxane), hydrocarbons (such as hydrophilic petrolatum, white petrolatum, purified lanolin or liquid paraffin), glycols (such as ethylene glycol, diethylene glycol, propylene glycol, polyethylene glycol or polyethylene glycol (macrogol)), vegetable oils (such as castor oil, olive oil, sesame oil or turpentine), animal oils (such as mink oil, egg yolk oil, squalane or squalene), water, absorption enhancers, skin protectants or combinations thereof. The ointment may additionally contain humectants, preservatives, stabilizers, antioxidants, fragrances or other suitable additives.

The pharmaceutical compositions in gel form may be prepared according to conventionally known methods of preparation, for example by melt mixing one or more active agents into a gel matrix. For preparing the gel, any conventionally used pharmaceutical gel base may be used, which may comprise a lower alcohol (such as ethanol or isopropanol), a gelling agent (such as carboxymethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose or ethyl cellulose), a neutralizing agent (such as triethanolamine or diisopropanolamine), a surfactant (such as polyoxyethylene glycol monostearate), a gum, water, an absorption enhancer, a skin protectant, or a combination thereof. The gel may additionally comprise preservatives, antioxidants, fragrances or any other suitable additives.

Pharmaceutical compositions in the form of creams may be prepared according to conventionally known manufacturing methods, for example by mixing one or more active agents into a pharmaceutical cream base by melting or emulsifying. For the preparation of a cream, any conventionally used pharmaceutical cream base may be used, which may contain higher fatty acid esters, lower alcohols, hydrocarbons, polyols (such as propylene glycol or 1, 3-butylene glycol), higher alcohols (such as 2-hexyldecanol or cetyl alcohol), emulsifiers (such as polyoxyethylene alkyl ethers or fatty acid esters), water, absorption enhancers, skin protectants, or combinations thereof. A cream may additionally comprise preservatives, antioxidants, fragrances or any other suitable additives.

Pharmaceutical compositions in the form of plasters may be prepared according to conventionally known manufacturing methods, for example by mixing one or more active agents into a plaster matrix by melting and applying the mixture to a support. For the preparation of plasters, any conventionally used pharmaceutical plaster base may be used, which may comprise thickeners (such as polyacrylic acid, polyvinylpyrrolidone, gum arabic, starch, gelatin or methylcellulose), humectants (such as urea, glycerol or propylene glycol), fillers (such as kaolin, zinc oxide, talc, calcium or magnesium), water, solubilising aids, viscosity-increasing agents, skin-protecting agents or combinations thereof. The plaster may additionally contain preservatives, antioxidants, fragrances or any other suitable additives.

Pharmaceutical compositions in the form of a patch can be prepared according to conventionally known manufacturing methods, for example by mixing one or more active agents into a patch matrix by melting and applying the mixture to a support. For preparing the patch, any conventionally used drug patch base may be used, which may comprise polymers, oils or fats, higher fatty acids, viscosity increasing agents, skin protecting agents, or combinations thereof. The patch may additionally contain preservatives, antioxidants, fragrances or any other suitable additives.

Pharmaceutical compositions in the form of liniments may be prepared according to conventionally known methods of preparation, for example by dissolving, dispersing or emulsifying one or more active agents in a vehicle which may comprise water, an alcohol (such as ethanol or polyethylene glycol), a higher fatty acid, glycerol, a soap, an emulsifier, a dispersant or a combination thereof. The liniment may additionally contain preservatives, antioxidants, fragrances or any other suitable additives.

Pharmaceutical compositions in the form of sprays or inhalants may comprise the active agent and optionally a stabilizing agent, such as sodium bisulfite, or a tonicity agent or buffer, such as sodium chloride, sodium citrate or citric acid, in a vehicle.

Pharmaceutical compositions for inhalation dosage forms may be in the form of an aerosol, inhalable powder or inhalable liquid, or may be provided as a liquid concentrate that is dissolved or dispersed in water or any other suitable vehicle to form an inhalable formulation prior to use. The formulation for inhalation may be prepared according to conventionally known preparation methods. The inhalable liquids may optionally contain preservatives (such as benzalkonium chloride or parabens), colorants, buffers (such as sodium phosphate or acetate), tonicity agents (such as sodium chloride or concentrated glycerin), thickeners (such as carboxyvinyl polymers), absorption enhancers, or other suitable additives. The inhalable powder may optionally contain a lubricant (such as stearic acid or a salt thereof), a binder (such as starch or dextrin), a filler (such as lactose or cellulose), a colorant, a preservative (such as benzalkonium chloride or a paraben), an absorption enhancer, or other suitable additives. For the administration of inhalable liquids, use is generally made of a spraying device (such as a nebulizer or atomizer). The inhalable powder may be dispensed by a powder inhalation device.

Pharmaceutical compositions in the form of sprays may comprise the active agent and optionally a stabilising agent (such as sodium bisulphite), or a tonicity agent or buffer (such as sodium chloride, sodium citrate or citric acid) in a vehicle. Sprays can be prepared according to the preparation methods described, for example, in US 2,868,691 or US 3,095,355.

The pharmaceutical composition may be prepared in other parenteral dosage forms. For example, one or more active agents may be formulated as a rectal suppository or vaginal suppository by conventionally known methods of preparation.

In one embodiment, a composition comprising a compound or peptide of formula (1), or a pharmaceutically acceptable salt thereof, or a combination thereof, described herein comprises one or more pharmaceutically acceptable carriers selected from trehalose, mannitol, methionine, citric acid, lactic acid, tartaric acid, acetic acid, trifluoroacetic acid, and a pH adjusting agent.

The compound or peptide of formula (1), or a pharmaceutically acceptable salt thereof, or a co-administered drug described herein may be administered to a subject by a suitable method depending on the disease to be treated, the condition of the subject, the target administration site, or other factors. For example, parenteral administration, preferably intravenous, intramuscular, intradermal, or subcutaneous administration by injection or infusion, may be employed. The compounds or peptides described herein may be administered in lymphocyte therapy or DC (dendritic cell) therapy. When the immunomodulator is co-administered, the immunomodulator may be administered transdermally or transmucosally by intranasal, buccal, vaginal, rectal or sublingual administration.

The dosage frequency or dosage interval may be appropriately selected depending on the disease to be treated, the condition of the subject, the route of administration, or other factors. Application is generally repeated, preferably every few days or months.

The compound or peptide of formula (1) or a pharmaceutically acceptable salt thereof, or co-administered drug, if any, described herein may be administered to a subject in an appropriate amount depending on the disease to be treated, the condition of the subject, the route of administration, or other factors. The compound or peptide or pharmaceutically acceptable salt thereof may be administered in an amount of 0.0001 mg to 1000 mg, preferably 0.001 mg to 1000 mg, more preferably 0.1 mg to 10 mg at a time. The co-administered drug may be administered in an amount appropriately selected based on the known clinical dose of the drug. For example, an immunomodulator as a co-administered drug may be administered in an amount of usually 0.0001 mg to 1000 mg per kg body weight, preferably 0.001 mg to 1000 mg per kg body weight, more preferably 0.1 mg to 10 mg per kg body weight.

When more than one active agent is incorporated in a single composition, they may be incorporated in an appropriately selected quantitative ratio depending on the disease to be treated, the condition of the subject, the route of administration, or other factors. For example, to treat a human subject by administering a composition comprising a peptide and/or compound described herein and an immunomodulator or other co-administered drug, the immunomodulator or co-administered drug may be used in an amount of 0.01 to 100 parts by weight relative to the peptide and/or compound.

The term "subject" as used herein includes both human and non-human mammals. Non-human mammals include, but are not limited to, non-human primates, sheep, dogs, cats, horses, and cattle. Human subjects, especially human subjects in need of an enhanced immune response are preferred.

The term "effective amount" as used herein refers to an amount of an active agent that completely or partially inhibits the progression of cancer or at least partially reduces one or more symptoms of cancer. The effective amount may be a therapeutically or prophylactically effective amount. The effective amount of an agent will be determined by the age or sex of the subject, the type or severity of the condition to be treated with the agent, the desired outcome of treatment with the agent, or other factors. One skilled in the art can determine an effective amount for a particular patient.

A compound or peptide of formula (1), or a pharmaceutically acceptable salt thereof, or a combination thereof, described herein, may be administered in combination with a non-drug therapy, or even more than one non-drug therapy, selected from, for example, surgery, radiation therapy, gene therapy, hyperthermia, cryotherapy, or laser cauterization therapy. For example, a compound or peptide of formula (1), or a pharmaceutically acceptable salt thereof, or a combination thereof described herein can be administered before or after a non-drug therapy, such as surgery, or before or after a combination of two or three non-drug therapies.

The compounds or peptides of formula (1), or pharmaceutically acceptable salts thereof, or combinations thereof described herein may be further used in combination with pharmaceutical agents to reduce undesirable side effects, if any, such as antiemetics, sleep-inducing agents, or anticonvulsants.

The disclosure also provides polynucleotides encoding the peptides described herein. The polynucleotide may be DNA or RNA. The polynucleotide has a nucleotide sequence corresponding to the amino acid sequence of the peptide encoded by the polynucleotide. The polynucleotide can be synthesized by a method for DNA or RNA synthesis or PCR.

The polynucleotides of the present disclosure include polynucleotides that are capable of hybridizing under stringent conditions to the complement of the polynucleotide encoding the peptide, and encode peptides having a similar ability to induce CTL or helper T cells as the peptide. Hybridization under stringent conditions can be a conventional hybridization as described in Sambrook J., Frisch E.F., Maniatis T., Molecular Cloning, 2 nd edition, Cold Spring Harbor Laboratory press. For example, "stringent conditions" can be generated in a solution of 6 XSSC (in this regard, 10 XSSC contains 1.5M NaCl and 0.15M trisodium citrate) with 50% formamide at 45 ℃ to form hybrids, and in a solution of 2 XSSC to wash hybrids (Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6).

The present invention also provides an expression vector comprising a polynucleotide of the present disclosure (hereinafter also referred to as WT1 expression vector). The expression vector may be of any suitable type and have any suitable sequence other than the polynucleotide sequences of the present disclosure, depending on the type of host to be transfected with the vector or any other particular factor. The vector may be a plasmid, a phage vector or a viral vector. For the transfection of E.coli, plasmid vectors such as pUC118, pUC119, pBR322 or pCR3, or phage vectors such as lambda ZAPII or lambda gt11 may be used. For transfection of yeast, pYES2 or pYEUra3 may be used. For transfection of insect cells, pAcSGHisNT-A may be used. For transfection of animal cells, plasmid vectors such as pKCR, pCDM8, pGL2, pcDNA3.1, pRc/RSV or pRc/CMV, or viral vectors such as retroviral vectors, adenoviral vectors or adeno-associated viral vectors can be used. The vector may further comprise a promoter for expression induction, a gene encoding a signal sequence, a marker gene for selection, or a terminator. The vector may also contain a sequence encoding a tag such as thioredoxin, His tag or GST (glutathione S-transferase) so that the protein is expressed together with the tag fused thereto, thereby facilitating the isolation or purification of the protein. Such vectors may comprise appropriate promoters (e.g., lac, tac, trc, trp, CMV, SV40 early promoters) depending on the host to be transfected with the vector, and may be GST-fused protein expression vectors (e.g., pGEX 4T), vectors comprising tag sequences such as Myc or His (e.g., pcdna3.1/Myc-His), or expression vectors for proteins fused with thioredoxin or His tags (e.g., pET32 a).

The expression vectors of the present disclosure express the peptides described herein in vitro or in vivo, and thus can be used to treat or prevent cancer.

The present disclosure further provides antibodies to the peptides described herein. The Antibodies may be polyclonal or monoclonal and may be prepared according to conventional methods for preparing polyclonal or monoclonal Antibodies (Current protocols in molecular biology, edited by Ausubel et al (1987) publish. John Wiley and sons. section 11.12-11.13, Antibodies; A Laboratory Manual, Lane, H, D. et al (edited), Cold Spring Harberorary Press, New York 1989). The antibody may be obtained as an antibody that recognizes the peptide of the present disclosure, or an antibody that recognizes and neutralizes the peptide of the present disclosure, from an animal immunized with the compound or peptide described herein by a conventional method. The antibodies may be used for affinity chromatography, or for immunodiagnosis based on e.g. immunoblotting, Radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA) or fluorescent or luminescent immunoassays, for cancer, especially cancer associated with WT1 gene expression or elevated levels of WT1 gene expression.

The compound of formula (1) of the present disclosure effectively induces CTLs, has good physicochemical stability, enables easy and simple controlled production, and can be widely used. In addition, the compounds of formula (1) of the present disclosure induce CTLs more effectively when combined with different cancer antigen peptides.

133页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:具有改进的效应子功能的人IgG Fc结构域变体

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!