Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HUMAN CANCER ANTIGEN OF TYROSINASE-RELATED PROTEIN 1 AND 2 AND GENES ENCODING SAME
Document Type and Number:
WIPO Patent Application WO/1997/029195
Kind Code:
A2
Abstract:
The present invention discloses that the normal melanogenic gene, gp75 gene, encodes a gene product, a 24 amino acid peptide of ORF3, which is processed to an antigenic cancer peptide recognized by T lymphocytes. The cancer peptide of the invention derived from ORF3 is recognized by cancer antigen specific T lymphocytes as a tumor rejection antigen. The present invention relates to the identification of a second tumor antigen recognized by an HLA-A31 restricted CTL clone derived from the TIL586 cell line. This antigen derived from the TRP-2 protein tumor antigen and peptides thereof are capable of sensitizing target cells for lysis by a CTL clone at 1 nM peptide concentration. Modified peptides were also recognized by the CTL clone. The products of these genes are promising candidates for immunotherapeutic strategies for the treatment and diagnosis of patients with cancer.

Inventors:
WANG RONG-FU
ROSENBERG STEVEN A
Application Number:
PCT/US1997/002186
Publication Date:
August 14, 1997
Filing Date:
February 06, 1997
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
US GOV HEALTH & HUMAN SERV (US)
International Classes:
C12N15/09; A01K67/027; A61K31/00; A61K35/14; A61K35/76; A61K39/00; A61K39/385; A61K45/08; A61K48/00; A61P35/00; C07H21/00; C07K7/06; C07K14/47; C07K14/705; C07K16/18; C07K16/30; C12N5/08; C12N5/10; C12N7/00; C12N9/00; C12N9/02; C12N9/90; C12N15/12; C12N15/52; C12N15/53; C12P21/02; C12Q1/02; C12Q1/68; G01N33/531; G01N33/554; G01N33/574; A61K38/00; C12R1/91; (IPC1-7): C12N15/53; C12N9/90; A61K38/17; G01N33/50; G01N33/60; C12N5/08; C07K16/40; C07K7/06; C12N15/61; C07K14/47; A61K38/44; G01N33/573; G01N33/68; C12N5/10; A61K48/00; C12N9/02; A61K38/08; A61K38/52; G01N33/574; C12Q1/68; C07K16/30; C12N15/86
Domestic Patent References:
WO1995023814A11995-09-08
WO1991014775A11991-10-03
Other References:
WANG ET AL.: "Identification of a gene encoding a melanoma tumor antigen recognized by HLA-A31-restricted tumor-infiltrating lymphocytes" THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 181, no. 2, 1 February 1995, pages 799-804, XP000674489 cited in the application
ORLOW ET AL.: "Changes in expression of putative antigens encoded by pigment genes in mouse melanomas at different stages of malignant progression" PROC. NATL. ACAD. SCI. USA, vol. 92, October 1995, pages 10152-10156, XP002040572
KAWAKAMI ET AL.: "Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes" THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 180, July 1994, pages 347-352, XP000565650 cited in the application
WANG ET AL.: "Utilization of an alternative open reading frame of a normal gene in generating a novel human cancer antigen" THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 183, no. 3, 1 March 1996, pages 1131-1140, XP000674490
WANG ET AL.: "Identification of TRP-2 as a human tumor antigen recognized by cytotoxic T lymphocytes" THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 184, no. 6, 1 December 1996, pages 2207-2216, XP000674491
BLOOM ET AL.: "Identification of Tyrosinase-related protein 2 as a tumor rejection antigen for the B16 melanoma" THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 185, no. 3, 3 February 1997, pages 453-459, XP002040588
COHEN ET AL.: "Nucleotide sequence of the cDNA encoding human tyrosinase-related protein" NUCLEIC ACIDS RESEARCH, vol. 18, no. 9, 1990, pages 2807-2808, XP002032549 cited in the application
YOKOYAMA ET AL.: "Molecular cloning and functional analysis of a cDNA coding for human DOPAchrome tautomerase / tyrosinase-related protein-2" BIOCHIMICA ET BIOPHYSICA ACTA. GENE STRUCTURE EXPRESSION, vol. 1217, no. 3, 1994, pages 317-321, XP002040983
Download PDF:
Claims:
WE CLAIM:
1. A cancer peptide, portion or derivative thereof encoded by an alternative open reading frame nucleic acid sequence from a gene other than an open reading frame sequence that encodes a normal protein or peptide from the same gene.
2. A cancer peptide, portion or derivative thereof according to claim 1 wherein the cancer peptide is immunologically recognized by MHC restricted T lymphocytes.
3. A cancer peptide, portion or derivative thereof according to claim 1 wherein the T lymphocytes are MHC class I restricted.
4. A cancer peptide, portion or derivative thereof according to claim 1 wherein the cancer peptide is derived from a cancer selected from the group consisting of: a nonHodgkins lymphoma, leukemia, Hodgkins lymphoma, lung cancer, liver cancer, metastases, melanoma, adenocarcinoma, thymoma, colon cancer, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer and sarcoma.
5. A cancer peptide, portion or derivative thereof according to claim 1 wherein the alternative open reading frame nucleic acid sequence is from a TRP1 gene.
6. A cancer peptide, portion or derivative thereof according to claim 1 wherein the cancer peptide is derived from a tumor associated antigen.
7. A cancer peptide, portion or derivative thereof according to claim 1 wherein the tumor associated antigen is selected from the group consisting of: oncofetal antigens, MART1, Mage1, Mage3, gplOO, tyrosinase, CEA, PSA, CA 171A, CA19A, CA125, erb2, P15 and /5galactosidase.
8. A cancer peptide, portion or derivative thereof according to claim 1 wherein the nucleic acid sequence comprises ORF3 depicted in Figure 2 as SEQ ID NO.: 5, portions or variants thereof.
9. A cancer peptide, portion or derivative thereof according to claim 1 wherein the alternative open reading frame nucleic acid sequence comprises: ATGTCACTGCAACGGCAATTTCTCAGG (SEQ ID NO.: 10) or portion or variant thereof.
10. A cancer peptide, portion or derivative thereof according to claim 1 wherein the cancer peptide comprises the amino acid sequence: MXaaLQRQFLRTQLWDVPSWLERSCL (SEQ ID NO. : 7), and fragments or derivatives thereof wherein Xaa = Ser or Ala.
11. A cancer peptide, portion or derivative thereof according to claim 1 wherein the cancer peptide comprises the amino acid sequence: MSLQRQFLR (SEQ ID NO.: 9), fragments or derivatives thereof.
12. A isolated tumor antigen having the amino acid sequence comprising SEQ ID NO:47 or a fragment or variant thereof.
13. An isolated tumor antigen encoded by a nucleic acid sequence comprising at least a fragment of SEQ ID NO:46 or a variant thereof.
14. A tumor antigen according to claim 12 wherein the tumor antigen is a cancer peptide, immunologically recognized by MHC restricted T lymphocytes.
15. A tumor antigen according to claim 14 wherein the T lymphocytes are HLAA31 restricted.
16. A tumor antigen according to claim 12 wherein the tumor antigen is derived from a cancer selected from the group consisting of: a non Hodgkins lymphoma, leukemia, Hodgkins lymphoma, lung cancer, liver cancer, metastases, melanoma, adenocarcinoma, thyoma, colon cancer, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer and sarcoma.
17. A tumor antigen according to claim 12 wherein the tumor antigen is a cancer peptide selected from the group consisting of SEQ ID NO:49 through SEQ ID NO:59.
18. A tumor antigen according to claim 13 further comprising the nucleic acid sequence: TTATTAGGACCAGGACGCCCCTACAGG or portion or variant thereof.
19. A tumor antigen according to claim 12 wherein the tumor antigen is a cancer peptide comprising the amino acid sequence: X'LLGPGRPYRX2, and variants thereof, wherein X1 and X2 are identical or different and each are one or more amino acids.
20. A tumor antigen according to claim 12 wherein the tumor antigen is a cancer peptide comprising the amino acid sequence: LLGPGRPYR or variants thereof.
21. A pharmaceutical composition comprising the cancer peptide, tumor antigen, portion or derivative thereof according to claims 119 or 20 and a pharmaceutically acceptable carrier.
22. A immunogen comprising the cancer peptide, tumor antigen, portion or derivative thereof according to claims 119 or 20 alone or in combination with at least one immunostimulatory molecule.
23. A immunogen according to claim 22 wherein the immunostimulatory molecule is an MHC molecule.
24. An isolated nucleic acid sequence comprising an alternative open reading frame nucleic acid sequence encoding a cancer peptide, portion or derivative thereof from a gene other than an open reading frame sequence that encodes a normal protein or peptide from the same gene.
25. An isolated nucleic acid sequence according to claim 24 wherein the cancer peptide is derived from a cancer selected from the group consisting of a nonHodgkins lymphoma, leukemia, Hodgkins lymphoma, lung cancer, liver cancer, metastases, melanoma, adenocarcinoma, thymoma, colon cancer, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer and sarcoma.
26. An isolated nucleic acid sequence according to claim 24 wherein the gene is a TRP1 gene.
27. An isolated nucleic acid sequence according to claim 24 wherein the nucleic acid sequence comprises ORF3 depicted in Figure 2 as SEQ ID NO.: 5, portions or variants thereof.
28. An isolated nucleic acid sequence according to claim 27 wherein the nucleic acid sequence comprises: ATGTCACTGCAACGGCAATTTCTCAGG (SEQ ID NO. : 10) or portion or variant thereof.
29. An isolated nucleic acid sequence according to claim 24 wherein the sequence encodes an amino acid sequence: MXaaLQRQFLRTQLWDVPSWLERSCL (SEQ ID NO.: 7), and fragments or derivatives thereof wherein Xaa = Ser or Ala.
30. An isolated nucleic acid sequence according to claim 29 wherein the sequence encodes an amino acid sequence: MSLQRQFLR (SEQ ID NO.: 9), fragments or derivatives thereof.
31. An isolated nucleic acid sequence comprising SEQ ID NO:46 or a fragment thereof encoding a tumor antigen.
32. An isolated nucleic acid sequence according to claim 31 wherein the nucleic acid sequence is derived from a cancer selected from the group consisting of a nonHodgkins lymphoma, leukemia, Hodgkins lymphoma, lung cancer, liver cancer, metastases, melanoma, adenocarcinoma, thymoma, colon cancer, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer and sarcoma.
33. An isolated nucleic acid sequence according to claim 31 wherein the nucleic acid sequence comprises TTATTAGGACCAGGACGCCCCTACAGG or variants thereof.
34. An isolated nucleic acid sequence according to claim 32 wherein the sequence encodes an amino acid sequence: LLGPGRPYR or variants thereof.
35. A recombinant expression vector comprising the nucleic acid sequence according to claims 2433 or 34.
36. A host organism transformed or transfected with a recombinant expression vector according to claim 35. 37.
37. An oligonucleotide comprising a nucleic acid sequence complementary to the nucleic acid sequence according to claims 2433 or 34.
38. A recombinant virus comprising a recombinant virus which has incorporated into a viral genome or portion thereof the nucleic acid sequence according to claims 2433 or 34. 39.
39. A recombinant virus according to claim 38 further comprising at least one gene encoding an immunostimulatory molecule.
40. The recombinant virus according to claim 38 wherein the virus is selected from the group consisting of retrovirus, baculovirus, Ankara virus, fowlpox, adenovirus, and vaccinia virus.
41. The recombinant virus according to claim 38 wherein the cancer peptide is derived from melanocytes.
42. A recombinant virus according to claim 39 wherein the immunostimulatory molecule is a MHC class I molecule.
43. A recombinant virus according to claim 38 wherein the nucleic acid sequence comprises: ATGTCACTGCAACGGCAATTTCTCAGG (SEQ ID NO.: 10) or variant thereof.
44. A recombinant virus according to claim 38 wherein the nucleic acid sequence encodes the cancer peptide comprising the amino acid sequence: MXaaLQRQFLRTQLWDVPSWLERSCL (SEQ ID NO. : 7), and fragments or derivatives thereof wherein Xaa = Ser or Ala.
45. A recombinant virus according to claim 38 wherein the nucleic acid sequence comprises: TTATTAGGACCAGGACGCCCCTACAGG .
46. A recombinant virus according to claim 38 wherein the nucleic acid sequence encodes the tumor antigen comprising the amino acid sequence: X'LLGPGRPYRX2, and variants thereof wherein X1 and X2 are identical or different and each are one or more amino acids.
47. An isolated antibody that binds the cancer peptide, or portion thereof encoded by an ORF3 of the TRP1 gene.
48. An isolated antibody that binds the cancer peptide, tumor antigen, or portion or variant thereof according to claims 119 or 20.
49. An isolated antibody that binds a tumor antigen encoded by SEQ ID NO:46 or a fragment thereof.
50. An isolated antibody that binds a tumor antigen comprising SEQ ID NO: 47 or a fragment thereof.
51. An isolated antibody that binds the tumor antigen or variant thereof according to claim 19.
52. A method of producing a recombinant cancer peptide or portion thereof comprising: a. inserting a nucleotide sequence of ORF3 depicted in Figure 2 as SEQ ID NO.: 5, or portion or variant thereof, into an expression vector; b. transferring the expression vector into a host cell; c. culturing the host cell under conditions appropriate for expression of the cancer peptide or portion thereof; and d. harvesting the recombinant cancer peptide, or portion thereof.
53. A method according to claim 52 further comprising in step (a) inserting a nucleotide sequence encoding an MHC class I molecule, or portion thereof into the expression vector.
54. A method of producing a recombinant TRP2 tumor antigen comprising: a. inserting a nucleotide sequence comprising at least a portion of SEQ ID NO:46, or variant thereof, into an expression vector; b. transferring the expression vector into a host cell; culturing the host cell under conditions appropriate for expression of the tumor antigen; and c. harvesting the recombinant tumor antigen.
55. A method according to claim 54 further comprising in step (a) inserting a nucleotide sequence encoding an MHC class I molecule, or portion thereof into the expression vector.
56. A method of detecting the presence of cancer or precancer in a mammal comprising: a. contacting a nucleic acid sequence of ORF3 depicted in Figure 2 as SEQ ID NO.: 5, or portion or variant thereof with a test biological sample of mRNA taken from the mammal under conditions allowing for a complex to form between the sequence and the mRNA; b. detecting the complex; c. comparing the amount of mRNA in the test sample with an amount of mRNA from a known normal biological sample, wherein an increased amount of mRNA from the test sample is indicative of cancer or precancer.
57. A method according to claim 56 wherein the cancer or precancer is melanoma.
58. A method of detecting the presence of cancer or precancer in a mammal comprising: contacting a nucleic acid sequence comprising at least a portion of SEQ ID NO:46 or a variant thereof with a test biological sample of mRNA taken from the mammal under conditions allowing for a complex to form between the sequence and the mRNA; and detecting the complex, presence of said complex being indicative of the presence of cancer or precancer.
59. A method according to claim 58 wherein the cancer or precancer is melanoma.
60. A method of detecting an ORF3 genomic nucleic acid sequence in a biological sample comprising: a. contacting the genomic nucleic acid sequence with ORF3 depicted in Figure 2 as SEQ ID NO.: 5, or portion or variant thereof under conditions to allow complexes to form between the genomic nucleic acid sequence and ORF 3; and b. detecting the complex.
61. A method of detecting the cancer peptide, tumor or antigen or portion or variant thereof according to claims 119 or 20 in a biological sample comprising: a. contacting the sample with antibodies specific for said cancer peptide under conditions to form an immune complex, and b. detecting the presence of the immune complex.
62. A method of preventing or inhibiting cancer in a mammal comprising: administering to the mammal an effective amount of the cancer peptide, tumor antigen or portion thereof according to claims 119 or 20, alone or in combination with an MHC molecular, said amount is effective in preventing or inhibiting the cancer in the mammal.
63. A method of inhibiting melanoma in a mammal comprising: a. exposing T lymphocytes in vitro to a cancer peptide, tumor antigen or portion thereof according to claims 1 19 or 20, alone or in combination with an MHC molecule for a time sufficient to elicite cancer peptide specific T lymphocytes; b. administering the cancer peptide specific T lymphocytes to the mammal in an amount sufficient to inhibit the melanoma.
64. A method of preventing or inhibiting cancer in a mammal comprising: administering to the mammal an effective amount of the cancer peptide or tumor antigen according to claims 119 or 20 alone, or in combination with an MHC molecule, said amount is effective in preventing or inhibiting cancer in a mammal.
65. A method of preventing or inhibiting cancer in a mammal comprising administering to the mammal an effective amount of a recombinant virus according to claims 3845 or 46 alone or in combination with an exogenous immunostimulatory molecule said amount is effective in preventing or inhibiting the cancer.
66. A pharmaceutical composition comprising the recombinant virus according to claims 3845 or 46 alone or in combination with an exogenous immunostimulatory molecule, chemotherapy drug, antibiotic, antifungal drug, antiviral drug or combination thereof and a pharmaceutically acceptable carrier.
67. A method of identifying an antigenic cancer epitope of a cancer antigen reactive with antigen specific T lymphocytes comprising: a. generation of nucleic acid deletion fragments from a gene encoding a cancer antigen; b. placing the deletion fragments in an expression vector; c. transfecting or transducing a host cell with the vector; d. determining a cancer antigen specific T lymphocyte response in the presence of the host expressing the deletion product.
68. A method according to claim 67 wherein the gene encoding the cancer antigen is TRP1 or TRP2.
69. A method of identifying an antigenic cancer epitope of a cancer antigen comprising: a. generating a multiplicity of synthetic peptides; b. pulsing cancer antigen presenting cells with the synthetic peptides; c. adding the peptidepulsed cells to cancer antigen specific T lymphocytes; and d. measuring a cancer antigen specific T lymphocyte response.
70. A method according to claim 69 wherein the cancer antigen is a melanoma antigen.
71. A transgenic animal carrying a gene encoding a tumor antigen comprising at least a fragment of SEQ ID NO:2 or SEQ ID NO:47.
72. A transgenic animal expressing a tumor antigen according to claim 71.
Description:
Human Cancer Antigen Of Tyrosinase-Related Protein 1 and 2 and Genes Encoding Same

Held of the Invention

The present invention relates to the area of cancer diagnostics and therapeutics including a cancer vaccine. More specifically, the invention relates to the isolation and purification of a novel cancer peptide and an alternative open reading frame DNA sequence encoding the cancer peptide. The invention further relates to the isolation and purification of a novel tumor antigen capable of acting as a tool in treating and preventing cancer and a DNA sequence encoding the cancer antige. The invention further relates novel cancer peptide encoded by an alternative open reading frame DNA sequence from within the tyrosinase-related protein 1 (TRP 1) gene. The invention also relates to novel cancer peptides from within the TRP2 gene. The invention further relates to methods of detecting and diagnosing and treating cancer and precancer in an individual.

Background of the Invention The adoptive transfer of tumor infiltrating lymphocytes (TIL) can mediate tumor regression in patients with metastatic melanoma, suggesting that tumor rejection antigens recognized by T cells exist on these tumor cells. The availability of such T cells has made it possible to clone and sequence the genes that encode human melanoma antigens. The antigens identified so far from human melanoma can be divided into two classes based on their expression pattern. The antigens of the first class are encoded by genes that are expressed only in tumor and testis, but not other normal human tissues. MAGE1 , MAGE3, GAGE and BAGE are examples of this class. The second class of antigens represents differentiation antigens encoded by genes that are expressed only in melanocytes, melanomas, and normal retina. MART- 1/Melan-A, gplOO and tyrosine are examples of this class. All these antigens are nonmutated self proteins. However, several mutated antigens were also identified to be recognized by T cells, including CDK 4, B-catenin and Mum-1. Identification of

the antigenic epitopes recognized by T cells derived from the corresponding gene products is important not only for understanding the mechanism of immune response to self antigens, but also for developing new, effective immunotherapeutic strategies with these antigens or synthetic peptides for the treatment of patients with cancer. Previous studies showed that the infusion of TIL586 plus IL-2 into the autologous patient with melanoma resulted in the objective regression of metastases. More recently, the gene, tyrosinase-related protein 1 (TRP-1 or gp75) was cloned which encodes the tumor antigen recognized by TIL586 in association with HLA-A31. Interestingly, the gene product, gp75, was originally identified as an antigen recognized by IgG antibodies in the serum from a patient with metastatic melanoma.

The gene was found to be expressed only in melanoma, normal melanocyte cell lines, and retina, but not in other normal tissues tested. Therefore, this gene is a member of the second class of antigens including MART-1/Melan-A, gplOO and tyrosinase.

In the art, it has been difficult to identify an epitope on a cancer cell which would be useful as an immunogen or vaccine to protect an individual from developing cancer. The present invention is the identification of a cancer peptide and the antigenic cancer epitope within the peptide encoded from an alternative open reading frame sequence within the TRP-1 gene which is specifically recognized by T cells. The present invention encompasses cancer peptides encoded by the TRP-2 gene. The cancer peptide of the invention is useful as an immunogen and vaccine to inhibit or prevent cancer in a mammal.

Summary of the Invention

One object of the present invention is to provide a novel peptide and portions thereof recognized as a cancer antigen by T lymphocytes.

The cancer peptide of the present invention and the antigenic cancer epitope portion of the cancer peptide is encoded by an alternative open reading frame DNA sequence of a gene other than the open reading frame DNA sequence used to encode a normal protein or peptide from the same gene. A gene falling within this ambit is the TRP-1 gene.

Another aspect of the invention is a tumor antigen encoded by an alternative open reading frame DNA sequence of a gene other than the open reading frame DNA

sequence encoding a normal protein or peptide from the same gene. A gene falling within this ambit is the TRP-1 gene.

Another object of the present invention are peptide fragments of TRP-1 , TRP-2 and variants thereof, which function as cancer peptides. The tumor antigen of the present invention and the antigenic cancer peptides of the tumor antigen are encoded by all or a portion of the TRP-2 gene (SEQ ID NO:46). TRP-2 is a member of the tyrosinase related gene family. TRP-2 is presently identified as a new and potent tumor antigen capable of causing T cells to elicit an immune response. One aspect of the invention are cancer peptides encoded by the TRP-1 gene,

TRP-2 gene or variants thereof, which are useful as a cancer vaccine capable of protecting the recipient from development of cancer. The present invention also relates to a method of administering the cancer vaccine in an effective amount to prevent cancers. Another aspect of the present invention is a pharmaceutical composition comprising a cancer peptide or antigenic cancer epitope thereof alone or in combination with one or more immunostimulatory molecules. Another aspect of the present invention is a pharmaceutical composition comprising the TRP-1 cancer antigen, alone, TRP-2 tumor antigen alone or in combination with one or more co- immunostimulatory molecules. Another object of the present invention is a pharmaceutical composition comprising TRP-1 , TRP-2 peptides, or combinations thereof which stimulate T-cells to elicit an immunogenic response against tumors and cancers. The cancer peptide or antigenic cancer epitope thereof may be provided as an immunogen or as a vaccine for prevention or treatment of cancer. The pharmaceutical composition is useful in methods of treating or preventing cancer in a mammal. In the method of treatment, the pharmaceutical composition is administered to the mammal in an amount effective in preventing or inhibiting the cancer in the mammal.

Another object of the present invention is a method of generating cancer peptides and the antigenic cancer epitope within the peptide by translation of an alterative open reading frame DNA sequence from a gene other than the open reading frame DNA sequence encoding a normal protein from the same gene.

Yet another object of the invention is a method of detecting and identifying a cancer peptide gene product and portions thereof translated from an alternative open reading frame DNA sequence from a gene other than the gene product translated from the open reading frame DNA sequence encoding a normal protein or peptide. A further aspect of the invention is the alternative open reading frame DNA or RNA sequence that encodes a cancer peptide or portion thereof and the use of the DNA or RNA sequence in methods of producing the cancer peptide or portions thereof. The invention further provides oligonucleotides of the alternative open readings frame DNA or RNA sequence for use as probes or primers. Yet another object of the present invention are nucleic acid sequences encoding

TRP-2 and fragments thereof, which, when expressed in a cell produce tumor antigens.

The present invention further provides vectors comprising an alternative open reading frame DNA sequence encoding a cancer peptide or portions thereof alone or in combination with a second DNA sequence encoding at least one immunostimulatory molecule. The alternative open reading frame DNA sequence may be from the TRP-1 gene or variant thereof.

The invention also provides host cells transfected or transduced with a vector comprising an alternative open reading frame DNA sequence encoding a cancer peptide or portions thereof alone or in combination with a second DNA sequence encoding at least one immunostimulatory molecule.

The present invention further provides vectors comprising the TRP-2 gene or portions thereof encoding the tumor antigen alone or in combination with a second DNA sequence encoding at least one co-immunostimulatory molecule. The vectors and host cells may serve as vaccines in which expression of a tumor antigen or cancer peptides results in the stimulation of tumor antigen specific T lymphocytes in a mammal immunized with the vaccine.

The invention also provides host cells transfected or transduced with a vector comprising DNA encoding a TRP-2 tumor antigen or variant thereof alone or in combination with a second DNA sequence encoding at least one co-immunostimulatory molecule.

The vectors and host cells may serve as vaccines in which expression of a

TRP-2 tumor antigen results in the stimulation of antigen specific T lymphocytes in a mammal immunized with the vaccine.

The vectors and host cells may serve as vaccines in which expression of a cancer peptide or portion thereof results in the stimulation of cancer peptide specific T lymphocytes in a mammal immunized with the vaccine.

It is still another object of the invention to provide a method for diagnosing human pre-neoplastic and neoplastic cells and tissues.

The invention provides a method of diagnosis cancer or precancer in a mammal by detection of a cancer peptide or portions thereof encoded by an alternative reading frame nucleic acid sequence of a gene other than the open reading frame nucleic acid sequence used to encode a normal protein from the same gene wherein the cancer peptide or portion thereof is recognized by T lymphocytes. An alternative reading frame nucleic acid sequence may be from the TRP-1 gene or variant thereof.

It is still another object of the invention to provide a method for diagnosing human preneoplastic and neoplastic cells and tissues. In accordance with the invention, the method comprises isolating cells, tissues or extracts thereof from a human and detecting the alternative open reading frame DNA sequence, RNA sequence or portion thereof encoding a cancer peptide or detecting the cancer peptide or portions thereof expressed by the alternative open reading frame DNA sequence or RNA sequence, wherein detection of/or increase in the alternative open reading frame DNA sequence, RNA sequence or expression product is indicative of preneoplasia and neoplasia.

The invention provides a method of diagnosis cancer or precancer in a mammal by detection of the TRP-2 tumor antigen, wherein the tumor antigen, cancer peptide or variant thereof is recognized by T lymphocytes.

The invention provides a method of diagnosis cancer or precancer in a mammal by detection of the tumor antigen encoded by the TRP-2 gene or fragments thereof wherein the tumor antigen is recognized by T lymphocytes.

Still another object of the invention is to provide a transgenic animal which has incorporated into its genome one or more copies of the alternative open reading frame

DNA sequence encoding a cancer peptide or portion thereof. An alternative reading frame nucleic acid sequence may be from the TRP-1 gene or variant thereof. The

incorporation of the alternative open reading frame DNA sequence results in overexpression or expression of multiple forms or variants of the cancer peptide. Such transgenic animals are useful for screening of therapeutic agents useful in treating cancer. Still another object of the invention is to provide a transgenic animal which has incorporated into its genome one or more copies of the tumor antigen of the present invention thereof. The incorporation of the TRP-2 DNA sequence or fragment thereof results in overexpression or expression of the tumor antigen. Such transgenic animals are useful for screening of therapeutic agents useful in treating cancer. The invention also encompasses antisense oligonucleotides which specifically target and bind to the alternative open reading frame nucleic acid sequence or a TRP-2 tumor antigen nucleic acid sequence and inhibit the expression of the cancer peptide or tumor antigen without adversely affecting the expression of the normal protein from the same gene. Still another aspect of the invention are monoclonal and polyclonal antibodies reactive with the cancer peptide and antigenic cancer epitope thereof, including TRP-1 cancer peptide and TRP-2 tumor antigen for use in diagnostic and detection assays. The monoclonal and polyclonal antibodies may be provided in the form of a kit alone, or along with other reagents commonly used in diagnostic and detection assays. Brief Description of the Figures

These and other objects, features, and many of the attendant advantages of the invention will be better understood upon a reading of the following detailed description when considered in connection with the accompanying drawings wherein:

Fig. 1 A and IB show the location of the gp75 nucleotide sequence coding for the antigenic peptides recognized by TIL586.

Fig. IA shows the full length cDNA which comprises the 1584 bp open reading frame of gp75 is shown. Nucleotides are numbered from the start codon which translates into a protein consisting of a leader sequence and the mature gp75. pcDNA776 is a partial cDNA of gp75 which lacks the first 246 nucleotide coding region and was isolated from a cDNA library using an assay based on its ability to stimulate GM-CSF secretion by TIL586 when co-transfecting COS-7 along with the HLA-A31 gene. A series of deletion constructs and PCR DNA fragments were made.

pD776A is a derivative of pcDNA776 after digestion with the Apal restriction enzyme.

Fig. IB shows GM-CSF release by TIL586. GM-CSF secretion by TIL586 was measured after co-cultured with COS-7 co-transfected with the DNA fragments shown in Fig. IA and the HLA- A31 gene. Control stimulator cells include 586mel,

397mel /A31\ COS-7 alone, and COS-7 transfected either the HLA-A31 or the pcDNA776 cDNA.

Fig. 2 shows the nucleotide, amino acid sequence and open reading frames of the gp75 gene. The partial nucleotide and amino acid sequences of the first 157 amino acids was shown from the start codon for translation of ORF1 (gp75). The DNA fragment that conferred the ability to stimulate GM-CSF release from TIL586 is underlined. Two putative start codons, ATG (254-256) and ATG (294-296), are in bold and may result in the translation of ORF2 and ORF3, respectively. The peptide sequence recognized by TIL586 from ORF3 is in bold and underlined. Fig. 3A and 3B show an antigenic peptide and translation of an alternative open reading frame.

Fig. 3A shows the location and length of PCR fragments amplified by PCR. DNA fragments were obtained by PCR amplification and were then cloned into the pCR3 expression vector. Substitution of ATG at positions 294-296 with ATC was made as described in Material and Methods.

Fig. 3B shows the testing of DNA fragments and mutation constructs to stimulate cytokine release from TIL586. GM-CSF release assay was done as in Fig. 1.

Fig. 4A - 4C show characterization of the antigenic peptide recognized by TIL586. -

Fig. 4A shows GM-CSF release by the HLA-A31 restricted TIL586 when co- incubated with various stimulators. Transfection and cytokine assays were performed as Fig. 1 A and B. 586mel and 397mel were included as positive and negative controls for the reactivity of TIL586. The ORF3P peptide was incubated with 586EBV (A31 +) and T2 (non-A31) cells at a concentration of 1 μg/ml for 90 min. Stimulation of GM-CSF secretion by TIL586 significantly increased when co-incubated with autologous 586EBV and allogeneic 1510EBV (A31 +) cells pulsed with peptide

ORF3P, but not when co-incubated with either 586EBV alone or T2 (non-A31) cells loaded with the ORF3P peptide.

Fig. 4B shows cytotoxic lysis of the target cells by TIL586. 586mel (— ■— ) and 397mel (— D— ) were used as positive and negative controls, respectively. 586EBV B cells were incubated with ORF3P (pep) (—*—), with an irrelevant peptide (ipep) (— •— ) without peptide (~Δ~) and T2 cells pulsed with ORF3P (— O— ) as marked. After incubation, TIL586 was added and mixed with the target cells. Cytolytic activity of TIL586 was measured in a 4 h chromium release assay.

Fig. 4C shows titration of the peptide concentration to sensitize the target cells for lysis by TIL586. 586EBV cells were separately incubated with serial dilutions of

ORF3P (pep) ( — *— ) or irrelevant peptides (ipep) (— •— ) and T2 cells with the

ORF3P peptide (— O— ) for 90 min. The cytolytic activity of TIL586 was evaluated in a 4h 51 Cr release assay at an effector: target (E:T) ratio of 40: 1.

Figs. 5A and 5B show inhibition of lysis of 5, Cr-labeled target cells by non- labeled 586EBV cells loaded with the ORF3P peptide.

Fig. 5A shows 586mel cells were labeled with chromium for 90 min as a "hot" target. 586EBV cells pulsed with ORF3P (—*—), with irrelevant peptide (— Δ— ) and T2 cells loaded with ORF3P (— □— ) were used as "cold" target cells. After washing, "hot" and "cold" target cells were counted again and mixed at the "cold'Thot" ratio of 1 : 1 , 5: 1, 10: 1, and 20: 1. TIL586 was added at an effector: "hot" target (E:T) ratio of 20: 1. Chromium release was measured after 4 h incubation.

Fig. 5B shows lysis of 51 Cr-labeled 624mel ("hot" target) by TIL1200 which recognized gplOO was not inhibited by 586EBV cells pulsed with ORF3P (— * — ) compared to 586EBV cells pulsed with an irrelevant peptide (— Δ— ). "Cold" and "hot" target cells were mixed at the indicated ratios. TIL1200 was added at an effector: hot target (E:T) ratio of 30: 1. Cytolytic activity of TIL1200 was evaluated in a 4-h "Cr release assay.

Figures 6A-6D show recognition of the antigenic peptide T cell clones from the TIL586 cell line. T cell clones were generated from the TIL586 cell line. 586EBV B cells were pulsed with the ORF3P peptide or irrelevant peptide. T cell clone or TIL586 cells were added and coincubated. For 586 mel, 397 mel/A3T tumors and melanocyte NHEM680 cells, 1 X 10 5 cells per well were incubated with

1 x IO 5 cells to T cell clones, TIL586-C1 (Fig. 6A), TIL586-C4 (Fig. 6B) and TIL586-C6 (Fig. 6C) or TIL586 (Fig. 6D) for 18-24 h, respectively. GM-CSF assay was performed as described in Fig. IB.

Fig. 7. Recognition of various target cells and the antigenic peptide by CTL clones derived from TIL586. T cell clones were generated by limiting dilution (1 cell per well) from the TIL586 cell line and were further expanded in AIM-V medium containing 6000 IU /ml IL-2. GM-CSF secretion by CTL clone 586TIL-C1 and clone 4 was measured after coculturing with normal melanocyte cell line (HLA-A31 +), 586EBV B cells pulsed with the ORF3P peptide or irrelevant pepude, 397mel or 586mel cells.

Fig. 8. Identification of TRP-2 as a new tumor antigen recognized by CTL clone 4. GM-CSF release by CTL clone 4 was measured after co-culture with COS-7 cotransfected with the HLA-A31 cDNA along with genes encoding MART-1, gp75/TRP-l, gplOO, tyrosinase, pl5 and TRP-2. Control stimulator cells included 586mel, 397mel, COS-7 alone, and COS-7 transfected the HLA-A31 cDNA.

Fig. 9. Construction of deletions and subclones of the TRP-2 gene and T cell recognition. The full length cDNA of TRP-2 which comprises the 1557 bp open reading frame is shown. Nucleotides are numbered from the f— rst nucleotide from the 5' untranslated region of TRP-2 cDNA. A series of deletion constructs and subcloning of DNA fragments were made. T cell recognition of each construct was determined after co-culturing CTL clone 4 with COS-7 co-transfected with the DNA fragments shown above and the HLA- A31 gene.

Fig. 10. Antigenic peptide and partial coding sequence of TRP-2. The partial nucleotide and amino acid sequences of the TRP-2 gene are shown. The length and 3' terminus of the DNA fragments in pTD4, pTA, pTD3 and pTK are indicated by arrows and the restriction sites for Apa I, Pst I and Kpn I are marked. The antigenic peptide sequence recognized by CTL clone 4 is in bold and underlined.

Fig. 11A through 11C. Characterization of the Antigenic peptide recognized by CTL clone 4. Figure 11 (A). GM-CSF release by T cells at different peptide concentrations. 586EBV (A31 +) were pulsed with the TRP,,^ peptide ( *- ) and T2 (non- A31) cells were pulsed with the TRP 197 . M} ( -•- ) at various peptide concentrations for 90 min. ORF3P as a control peptide was pulsed onto 586EBV B

cells (~Δ~). GM-CSF release by CTL clone 4 was determined after co-incubate with 586EBV B cells pulsed with TRP W 205 and ORF3P, and T2 cells pulsed with TRP 197∞ . Figure 11 (B). Sensitization of the target cells for lysis by CTL clone 4 at different peptide concentrations. 586EBV B cells were incubated with TRP 197 . MJ ( -*- ), an irrelevant peptide ORF3P (-Δ~), and T2 cells pulsed with TRP,^ ( •* ) at various peptide concentrations. After peptide incubation, target cells were labelled for 30 min. Following washes, cytolytic activity of CTL clone 4 at an E: T ratio of 40: 1 was measured after a 4 h incubation of T cells with target cells. Figure 11 (C). Lysis of the target cells by CTL clone 4 at the different E: T ratios. Target 586EBV cells were separately incubated with TRP l97 .- o5 , ( * - ) or the irrelevant peptides ORF3P (~Δ~) and target T2 cells were incubated with the TRP I97a)J peptide ( •• ) for 90 min. 586mel ( π ) and 397mel ( » ) were used as positive and negative controls, respectively.

Detailed Description of the Invention

The present invention encompasses cancer peptides, tumor antigen and portion, derivatives or variants thereof which are immunologically recognized by T lymphocytes of the immune system. The present invention further encompasses the antigenic cancer epitope(s) which are contained in the cancer peptides or tumor antigen. The antigenic cancer epitope specifically causes a cellular mediated immune response by interaction with T cells of the immune system. This interaction between the antigenic cancer epitope and the T cells causes the T cells to respond against, and prevent, eliminate or reduce the cancer in a mammal, including humans.

In one embodiment, the cancer peptides and the antigenic cancer epitope contained within the cancer peptides of the present invention are distinguished from normal protein or peptides in that the cancer peptides are encoded by an alternative open reading frame of a gene other than the open reading frame that encodes the normal protein or peptide within the gene. The cancer peptide and portions thereof are characteristically absent from or present in very low levels from normal cells and are present in high levels from pre-cancer and cancer cells. Expression of the cancer peptide at high levels correlates with transformation of normal cells to a pre-cancer or cancer cell.

The cancer peptides of the present invention form part of, or are derived from, cancers including but not limited to primary or metastatic melanoma, thymoma, lymphoma, sarcoma, lung cancer, liver cancer, non-Hodgkin's lymphoma, Hodgkins lymphoma, leukemias, uterine cancer, cervical cancer, bladder cancer, kidney cancer and adenocarcinomas such as breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, and the like.

The term melanoma includes, but is not limited to, melanomas, metastatic melanomas, melanomas derived from either melanocytes or melanocyte related nevus cells, melanocarcinomas, melanoepitheliomas, melanosarcomas, melanoma in situ. superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral lentiginous melanoma, invasive melanoma or familial atypical mole and melanoma (FAM-M) syndrome.

Of particular interest are cancer peptides, fragments or derivatives thereof recognized by autologous CTL in patients with cancer, in particular melanoma. Of further interest are cancer peptides, fragments or derivatives thereof recognized by MHC restricted CTL, in particular MHC class I restricted CTLs.

The "tumor antigen" of the present invention encompasses the cancer or tumor protein and any portion or peptide of the cancer or tumor protein capable of eliciting an anti-tumor response in mammals. In one embodiment, the tumor antigen includes the full-length TRP-2 protein.

"Cancer peptides" as the term is used herein, encompasses any epitope or fragment of cancer or tumor protein, which acts as a tumor antigen.

"Fragment" as the term is used herein means any segment of a protein or gene, having at least 5 or 6 amino acids in the case of a protein fragment and at least 15-18 nucleotides in the case of a gene.

In one embodiment, the cancer peptides of the present invention arise from expression of an alternative open reading frame DNA sequence from a normal gene. Rather than the normal gene product being expressed, a cancer peptide is expressed which is capable of being immunologically recognized by T lymphocytes in an MHC restricted manner. The MHC restricted T lymphocytes are useful in identifying the alternative open reading frame gene product associated with cancer and pre-cancer.

Of particular interest are cancer peptides which are associated with TRP-1 (gp

75); the protein, pl9 ARF , which arises from an alternative reading frame of the mouse tumor suppressor INK4a gene (Quelle, D.E. et al £eJI Vol. 83, pp. 993-1000, 1995); the antigenic octapeptide SVVEFSSL, i.e. JAL8, an allogeneic peptide recognized by bml anti-B6 alloreactive bmlBZ19.4 T-cells (Malarkannan, S. et al J. Exp. Med. Vol. 182, pp. 1739-1750, 1995) and the like.

In one embodiment, a cancer peptide, fragment or derivative thereof of the present invention comprises antigenic cancer epitope immunologically recognized by tumor infiltrating lymphocytes (TIL) derived from a cancer tumor of a mammal. Of particular interest are antigenic cancer epitopes recognized by cancer antigen specific cytotoxic T cells (CD 8 + ).

In one embodiment of the present invention the cancer peptide comprises about 24 amino acids and is expressed by the alternative open reading frame 3 DNA sequence from the same gene that encodes tyrosinase-related protein 1 as depicted in Figure 2 or from homologs or variants thereof. In one embodiment, the cancer peptide of the present invention comprises the amino acid sequence:

MXaaLQRQFLRTQLWDVPSWLERSCL, (SEQ. ID NO: 7) and fragments, or derivatives thereof, wherein Xaa = Ser or Ala. Also encompassed in the ambit of the invention are cancer peptides or portions thereof that share partial sequence homology with SEQ. ID NO: 6. By partial amino acid sequence homology is meant a peptide having at least 85% sequence homology with SEQ. ID NO: 6, preferably at least 95% sequence homology or greater and has the biological function of stimulating cancer antigen specific T lymphocytes.

In an embodiment of the present invention the cancer peptide may be represented by the formula:

Met Xaa Leu Gin Arg Gin Phe Leu Arg (SEQ. ID NO: 8) and fragments and derivatives thereof wherein Xaa = Ser or Ala.

In another embodiment the cancer peptide of the present invention comprises the amino acid sequence: MSLQRQFLR (SEQ. ID NO: 9) and fragments and derivatives thereof.

In another embodiment, the cancer peptides and the antigenic cancer epitope

contained within the tumor antigen of the present invention are derived from the TRP- 2 protein, which is expressed primarily in melanomas, normal melanocyte cell lines and retina. The tumor antigen of the present invention present in significantly lower levels in most normal cells than the elevated levels found in pre-cancer and cancer cells. Elevated expression of the tumor antigen correlates with transformation of normal cells to a pre-cancer or cancer cell. TRP-2 is located on the human chromosome 13 and has been shown to be a member of the tyrosinase-related gene family and shares a 40-45% amino acid sequence identity to tyrosinase and gp75 /TRP-1 (Yokoyama et al. (1994); Bouchard, et al. (1994)). TRP-2 encodes a protein with 519 amino acids and has been demonstrated to have DOPAchrome tautomerase activity involved in melanin synthesis (Bouchard et al. (1994)).

Of particular interest are cancer peptides of TRP-2, or variants thereof recognized by autologous CTL in patients with cancer, in particular melanoma. Of further interest are cancer peptides, fragments or derivatives thereof recognized by MHC restricted CTL, in particular MHC class I restricted CTLs. A preferred HLA subtype recognized by the cancer peptides are the HLA-A31 subtype. The present invention relates to the identification of TRP-2, a melanoma/melanocyte differentiation antigen of the tyrosinase protein family, as a potent tumor antigen recognized by HLA- A31 restricted T cells. The TRP-2 gene product is the second tumor antigen recognized by CTL clones isolated from TIL586.

In another embodiment of the present invention the tumor antigen is a cancer peptide comprising about 9 amino acids and is expressed from the gene that encodes tyrosinase-related protein-2 (SEQ ID NO:46) or from homologs or variants thereof depicted in Figure 10. In yet another embodiment, fragments of the TRP-2 protein or functionally equivalent variants thereof are used as cancer peptides. Preferably, the tumor antigen of the present invention comprises fragments of the TRP-2 protein containing at least a portion of amino acids 197-205. Most preferably, the cancer peptide of the present invention comprises the amino acid sequence: LLGPGRPYR and fragments, or derivatives thereof. Also encompassed in the ambit of the invention are cancer peptides or portions thereof that share partial sequence homology with the region of TRP-2 containing amino acids 197-205. By partial amino acid sequence homology is

meant a peptide having at least 85% sequence homology with LLGPGRPYR, preferably at least 95% sequence homology or greater and has the biological function of stimulating cancer antigen specific T lymphocytes.

Another embodiment of the present invention encompasses cancer peptides having sufficient homology to LLGPGRPYR to effectively act as cancer peptides. Such peptides may have conservative amino acid changes at one or more positions. By conservative amino acid changes is meant, an amino acid change at a particular position which is of the same type as originally present; i.e. a hydrophobic amino acid exchanged for a hydrophobic amino acid, a basic amino acid for a basic amino acid, etc. Such amino acid changes do not significantly alter the overall charge and configuration of the peptide and therefore such variants maintain the anti-cancer activity of a cancer peptide. Examples of such conservative changes are well-known to the skilled artisan and are within the scope of the present invention.

Yet another embodiment of the present invention relates to several cancer peptides which are derived from the LLGPGRPYR peptide, but contain non- conservative amino acid changes at one or more positions. Such peptides have been identified in the present invention and include, but art not limited to LSGPGRPYR, KLGPGRPYR, LLGPGFPYR and fragments and derivatives thereof.

The present invention relates to functionally equivalent variants of the TRP-1 or TRP-2 cancer peptides. "Functionally equivalent variants" includes peptides with partial sequence homology, peptides having one or more specific conservative and/or non-conservative amino acid changes, peptide conjugates, chimeric proteins, fusion proteins and peptide nucleic acids.

The cancer peptides, tumor antigen and their antigenic cancer epitopes may be purified and isolated from natural sources such as from primary clinical isolates, cell lines and the like. The cancer peptide and portions thereof are at least 90% pure, preferably at least 95% pure and as pure as 100%. The cancer peptides and their antigenic epitopes may also be obtained by chemical synthesis or by recombinant DNA techniques known in the arts. Techniques for chemical synthesis are described in J.M. Steward and J.D. Young, "Solid Phase Peptide Synthesis", W.H. Freeman & Co., San Francisco, 1969; M. Bodansky et al "Peptide Synthesis", John Wiley & Sons, Second Edition, 1976, and J. Meienhofer, "Hormonal Proteins and Peptides", Vol.

2, p. 46, Academic Press, New York, 1983 and E. Schroder and K. Kubke, "The Peptides", Vol. 1, Academic Press, New York, 1965.

The cancer peptides and their antigenic cancer epitopes may be formulated with pharmaceutically acceptable carriers into pharmaceutical compositions by methods known in the art. The composition is useful as a vaccine to prevent or treat cancer.

The composition may further comprise at least one immunostimulatory molecule.

Immunostimulatory molecules to be used in conjunction with the cancer peptide or portion thereof for stimulating antigen specific T cell responses include but are not limited to one or more major histocompatibility complex (MHC) molecules, such as class I and class II molecules, preferably a class I molecule. The composition may further comprise other stimulator molecules including B7.1 , B7.2, ICAM-1 , ICAM-2,

LFA-1, LFA-3, CD72 and the like, and cytokines which include but are not limited to IL-1 through IL-15, TNFα, IFNγ, RANTES, G-CSF, M-CSF, IFNα, CTAP III,

ENA-78, GRO, 1-309, PF-4, IP-10, LD-78, MGSA, MlP-l , MIP-1/3, or combination thereof, and the like for immunopotentiation.

The stimulatory molecule may be provided as a physically separate entity or it may be provided in the membrane of an antigen presenting cell such as B-cell, macrophage or dendritic cell, in the membrane of a liposome, or expressed on the surface of a transduced or transfected cell. DNA sequences of MHC immunostimulatory molecules are available from GenBank and the like.

The cancer peptides, tumor antigen and their antigenic cancer epitopes are useful in methods of preventing or treating cancer and useful in diagnostic assay for detecting cancer or precancer in a mammal, including humans. The cancer peptides or portions thereof may be in the form of a derivative in which other constituents are attached thereto such as radiolabels, biotin, fluorescein. A targeting agent may also be attached to the tumor antigen, cancer peptides or portions thereof that allow for specific targeting to a specific organ, tumor or cell types. Such targeting agents may be hormones, cytokines, cellular receptors and the like. The cancer peptide, tumor antigen and portions thereof may be prepared in the form of a kit, alone or in combination with other reagents.

Another aspect of the invention is a vaccine useful in inducing tumor-specific cell-mediated immunity against cancer.

Approaches to cancer immunotherapy can be divided into active or passive categories. Active immunotherapy involves the direct immunization of cancer patients with cancer antigens in an attempt to boost immune responses against the tumor. Passive immunotherapy refers to the administration of immune reagents, such as immune cells or antibodies with antitumor reactivity with the goal of directly mediating antitumor responses.

Most prior attempts at active immunotherapy utilized either intact cancer cells or cancer cell extracts with the expectation that these materials contained tumor antigens in an amount and form capable of stimulating immune responses. The molecular identification of cancer antigens however, has open new possibilities for developing immunotherapies for the treatment of human cancer. A summary of some of these approaches is presented in Table 1.

- 17 -

Table 1 Cancer Therapies Based on the Molecular Identification of Cancer Antigens

1. Active immunotherapy with: a. Immunodominant peptides 1) alone

2) combined with adjuvants

3) linked to helper peptides, lipids or liposomes

4) pulsed onto antigen presenting cells b. Immunodominant peptides with amino acids substitutions to increase binding to MHC molecules c. Proteins alone or combined with adjuvants d. "Naked" DNA encoding cancer antigens

1) "gene gun" for intradermal injection

2) intramuscular injection 3) linked to lipids e. Recombinant viruses such as vaccinia, fowlpox or adenovirus encoding

1) cancer antigens alone

2) cancer antigens plus genes encoding cytokines, costimulatory molecules, or other genes to enhance the immune response f. Recombinant bacteria such as 6CG, Salmonella or Listeria encoding cancer antigens alone or in combination with immunostimulatory molecules

2. Active immunotherapy (above) followed by the administration of immunostimulatory cytokines.

1. IL-2

2. IL-6 3. IL-10

4. IL-12

5. IL-15, and the like.

3. Passive immunotherapy with anti-tumor lymphocytes raised by in vitro sensitization of TIL or PBL to 1. immunodominant peptides pulsed onto antigen presenting cells (raise CD8 cells)

2. antigenic proteins coincubated with antigen presenting cells (exogenous antigen presenting pathway to raise CD4 cells).

The insertion of the gene encoding cancer antigens into high efficiency expression systems such as E. coli. yeast or baculovirus and the like provides the opportunity to obtain large amounts of purified tumor antigen for use in immunization. Alternatively, the immunodominant peptides from these tumor antigens could readily be synthesized in vitro and purified in large amounts for immunization alone or in a form intended to improve their immunogenicity such as in combination with adjuvant, linkage to lipid s/liposomes or helper peptides, or pulsed onto antigen presenting cells. Modification of individual amino acids of the immunodominant peptides to improve binding efficiency to MHC antigens can potentially increase immunogenicity compared to the native peptide.

Recent techniques utilizing "naked" DNA injected directly into muscle or into the skin have been shown to raise both cellular and humoral immune reactions to encoded antigens (Cooney, E.L., A.C. Collier, P.D. Greenberg, R.W. Coombs, J. Zarling, D.E. Arditti, M.C. Hoffman, S.L. Hu and L. Correy, 1991, Lancet 337:567; Wolff, J.A., R.W. Malone, P. Williams, W. Chong, G. Acsadi, A. Jani, and P.L. Feigner, 1990, Science 247: 1465; Davis, H.L., R.G. Whalen, and B.A. Demeniex, 1993, Hum. Gene Ther. 4: 151; Yang, N.S., J. Burkholder, B. Roberts, B. Martinelli, and D. McCabe, 1990, Proc. Natl. Acad. Sci. USA 87:9568; Williams, R.S., S.A. Johnston, M. Riedy, M.J. DeVit, S.G. McElligott, and J.C. Sanford, 1991 , Proc. Natl. Acad. Sci. USA 88:2726; Fynan, E.R. , Webster, D.H. Fuller, J.R. Haynes, J.C. Santoro, and H.L. Robinson, 1995, Proc. Natl. Acad. Sci. USA 90:11478; Eisenbraum, M.D., D.H. Fuller, and J.R. Haynes, 1993, DNA and Cell Bio. 12:791; Fuller, D.H. and J.R. Haynes, 1994, AIDS Res. Hum. Retrovir. 10(11): 1433; Acsadi, G., G. Dickson, D.R. Love, A. Jani, F.S. Walsh, A. Gurusinghe, J.A. Wolff, and K.E. Davies, 1991 , Nature 352:815). Techniques using nonviable DNA vectors have the advantage of ease of preparation and safety of administration. The alternative nucleic acid sequence of the present invention is useful as an immunogen and as a DNA vaccine against cancer. The alternative open reading frame nucleic acid sequence of the present invention of TRP-1 or the DNA sequence as encoding TRP-2 protein or peptides of the present invention may be administered using a gene gun in amounts to elicite a cellular response against a cancer cell. Nonogram quantities are useful for such purposes.

An effective form of immunization involves the incorporation of genes encoding immunogenic molecules into recombinant bacteria such as BCG, Salmonella or Listeria or into recombinant viruses such as vaccinea, fowlpox or adenovirus and the like. The genes encoding cancer antigens can be expressed either alone or in combination with genes encoding immunostimulatory molecules or other genes which can enhance the immune response following infection. Studies with model tumor antigens in murine models have shown that incorporation of the gene for interleukin-2 (IL-2) or B7.1 can increase the immunogenicity of model tumor antigens and even mediate the regression of established lung metastases bearing these antigens and even mediate the regression of established lung metastases bearing these antigens. Active immunotherapy followed by the exogenous administration of immunostimulatory cytokines such as IL-2, IL-6, IL-10, IL-12, or IL-15 may also be used to improve immune responses.

Passive immunotherapy with genetically modified immune cells (commonly referred to as adoptive immunotherapy) capable of recognizing human tumor antigens is effective in mediating the regression of cancer in selected patients with metastatic melanoma. In vitro techniques have been developed in which human lymphocytes are sensitized in vitro to tumor antigen immunodominant peptides presented on antigen presenting cells. By repetitive in vitro stimulation cells can be derived with a far greater capacity to recognize human tumor antigens than the TIL that were used to clone the genes encoding these antigens. Thus by repeated in vitro sensitization with the cancer peptides, lymphocytes could be derived with 50 to 100 times more potency of TIL. The adoptive transfer of these cells may be more effective in mediating tumor regression in vivo than are conventionally grown TIL. In the methods of preventing or inhibiting cancer, the cancer peptides or portions thereof may be administered via one of several routes including but not limited to intravenous, intramuscular, subcutaneous, intradermal, intraperitoneal, intrathecal, intrapleural, intrauterine, rectal, vaginal, topical, intratumor and the like.

Administration may be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic

acid derivatives. In addition, detergents may be used to facilitate permeation. Transmucosal administration may be by nasal sprays, for example, or suppositories. For oral administration, the cancer peptide, tumor antigen, portion or variant thereof is formulated into conventional oral administration form such as capsules, tablets and toxics.

In general, it is desirable to provide the recipient with a dosage of cancer peptide or portion thereof of at least about lpg per Kg bodyweight, preferably at least about Ing per Kg bodyweight, more preferably at least about lμg or greater per Kg bodyweight of the recipient. A range of from about Ing per Kg bodyweight to about lOOmg per Kg bodyweight is preferred although a lower or higher dose may be administered. The dose is effective to prime, stimulate and/or cause the clonal expansion of cancer antigen specific T lymphocytes, preferably cytotoxic T lymphocytes, which in turn are capable of preventing or inhibiting cancer in the recipient. The dose is administered at least once and may be provided as a bolus or a continuous administration. Multiple administrations of the dose over a period of several weeks to months may be preferable. Subsequent doses may be administered as indicated.

In a method of treatment, a vaccine comprising the cancer peptide or portion thereof is administered to a mammal in an amount effective to prevent cancer in the mammals. Of particular interest is a vaccine comprising the cancer peptide or portion thereof encoded by ORF3 of the TRP-1 gene for prevention of melanoma. Also of particular interest is a vaccine comprising one or more peptides encoded by fragments of the TRP-2 gene for prevention of melanoma. In a method of reducing tumor burden in animals having tumors the method comprises administration of an effective amount of a antigenic cancer epitope at a site of tumor burden, said amount is effective to reduce the size of the tumor at the site.

In another method of treatment, autologous cytotoxic lymphocytes or tumor infiltrating lymphocytes may be obtained from a patient with cancer. The lymphocytes are grown in culture and cancer antigen specific lymphocytes expanded by culturing in the presence of specific cancer peptides or antigenic cancer epitopes alone or in combination with at least one immunostimulatory molecule with cytokines. The

antigen specific lymphocytes are then infused back into the patient in an amount effective to reduce or eliminate the tumors in the patient.

After immunization the efficacy of the vaccine can be assessed by production of immune cells that recognize the cancer antigen, as assessed by specific lytic activity, specific cytokine production, tumor regression or combination of these. If the mammal to be immunized is already afflicted with cancer or metastasis cancer the vaccine can be administered in conjunction with other therapeutic treatments such as immunomodulators, for example, IL-2, IL-6, IL-10, IL-12, IL-15, interferon, tumor necrosis factor and the like, chemotherapeutic drugs such as cisplatinum, antiviral such as gancyclovir, amphotericin B, antibiotics and the like.

Another aspect of the invention is an alternative open reading frame DNA sequence of a gene other than the open reading frame DNA sequence encoding a normal protein or peptide wherein the alternative open reading frame DNA sequence encodes cancer peptides and portions thereof which are immunologically recognized by T cells of the immune system.

Alternative open reading frame DNA sequence include but are not limited to DNA sequences from the TRP-1 gene, the TRP-2 gene, the INK4a gene and the like.

Of interest are alternative open reading frame DNA sequence from a melanogenic gene. Melanogenic genes include but are not limited to genes encoding MART-1/Melan A, tyrosinase, gp 100, gp 75 (TRP-1), TRP-2 (Halahan, R. et al 1993 J. Invest. Dermatol. 100 (Suppl.): 176S-185S), and the like.

One embodiment of the invention is an alternative open reading frame DNA sequence or portion thereof encoding a cancer peptide from within the gene sequence that encodes tyrosinase-related protein. The gene sequence for TRP1 has been disclosed through the EMBL data bank under accession number X51455 as described by Vijayasaradhi, S. et al (1990, J. Exp. Med. 171 : 1375-80) and EMBL accession number X51420 as described by Cohen, T. et al 1990 Nucleic Acids Research. Vol. 18:2807.

In one embodiment, the alternative open reading frame DNA sequence comprises ORF3 depicted in Figure 2 having SEQ. ID NO.: 5, portions thereof and functionally equivalent sequence variant thereof that encode a cancer peptide or portions thereof recognized by cancer antigen specific T lymphocytes including tumor

infiltrating lymphocytes. Also encompassed by the present invention are nucleic acid sequences complementary, as well as anticomplementary to ORF3 depicted in Fig. 2. In another embodiment, the alternative open reading frame DNA sequence comprises: ATGTCACTGCAACGGCAATTTCTCAGG (SEQ. ID NO: 10).

One embodiment of the invention are portions of the TRP-2 encoding one or more cancer peptides. The gene sequence for TRP-2 has been disclosed through the Genbank under accession number D 17547 as described by Yokoyama et al. (1994) and Genbank accession number S69231 as described by Bouchard et al. (1994). In one embodiment, TRP-2 gene fragments encoding LLGPGRPYR and functionally equivalent sequence variants thereof for a cancer peptide recognized by cancer antigen specific T lymphocytes including tumor infiltrating lymphocytes. Also encompassed by the present invention are nucleic acid sequences complementary, as well as anticomplementary to a sequence encoding LLGPGRPYR and equivalent sequence variants thereof.

In another embodiment, the DNA sequence encoding TRP-2 protein expresses all or more portions thereof. A preferred fragment of the TRP-2 gene comprises a region between a PstI site at nucleotide position 947 and a Kpnl site at nucleotide position 1080. Another preferred fragment of TRP-2 gene comprises:

TTATTAGGACCAGGACGCCCCTACAGG.

Due to degeneracy in the generic code, variations in the DNA sequence will result in translation of an equivalent cancer peptide. As a result, substitutions are included in the ambit of the invention as long as the substitution results in expression of a cancer peptide that is recognized by cancer antigen MHC-restricted T cells. One substitution encompassed in the present invention is the substitution of TCA encoding Ser for GCT, GCC, GCA or GCG encoding Ala. Homologs from other mammalian species is included within the ambit of the invention.

All or part of the alternative open reading frame DNA sequence such as from the TRP-1 gene and the like may be used as probes to identify and isolate the homologs of the cancer peptide in other mammalian species. Similarly, all or part of the TRP-2 gene may be used as probes to identify and isolate the homologs of the

cancer peptides in other mammalian species. In one embodiment, a murine cDNA sequence is used to screen a mammalian cDNA library for a human homolog nucleic acid sequence. Positive clones are selected and sequenced. Examples of tissue sources from which the cDNA library can be synthesized include but are not limited to dermis, epidermis, solid tumors, melanomas, melanocytes, and the like. One skilled in the art will understand the appropriate hybridization conditions to be used to detect the homologs. Conventional methods for nucleic acid hybridization construction of libraries and cloning techniques are described in Sambrook et al, (eds) (1989) in "Molecular Cloning. A Laboratory Manual" Cold Spring Harbor Press, Plainview, New York and Ausubel et al (eds) in "Current Protocols in Molecular Biology" (1987), John Wiley and Sons, New York, New York.

Another aspect of the invention are nucleic acid probes for the detection and quantification of RNA that transcribes the cancer peptides such as TRP-1, TRP-2 cancer peptides and the like in biologic samples isolated from a mammal with cancer. Alterations in the level of RNA relative to a control RNA sample is useful in diagnosis and prognosis of the disease in the mammal.

In one embodiment, mRNA is derived from tissue of a patient suspected of having cancer or precancer and compared with mRNA derived from a healthy control subject. A quantitative and/or qualitative increase of the alternative open reading frame mRNA encoding a cancer peptide in the patient, as compared to the control, is indicative of cancer or precancer in the patient. The mRNA may be detected using oligonucleotide probes hybridizable with the mRNA. In one embodiment the probe is hybridizable with the transcription product of ORF3 of TRP-1.

Another aspect of the invention are nucleic acid probes for the detection and quantification of RNA that transcribes the TRP-2 tumor antigen in biologic samples isolated from a mammal with cancer. Alterations in the level of RNA relative to a control RNA sample is useful in diagnosis and prognosis of the disease in the mammal.

In one embodiment, TRP-2 mRNA is derived from tissue of a patient suspected of having cancer or precancer and compared with TRP-2 mRNA derived from a healthy control subject. A quantitative and/or qualitative increase of the TRP-2 mRNA in the patient, as compared to the control, is indicative of cancer or precancer

in the patient. The mRNA may be detected using oligonucleotide probes.

Combinations of oligonucleotides pairs based on the sequence encoding the cancer peptide or portions thereof may be used as PCR primers to detect mRNA in biological samples using the reverse transcriptase polymerase chain reaction (RT-PCR) process for amplifying selected RNA sequences. The present invention also encompasses in situ PCR and in situ RT-PCR for detection of DNA and RNA encoding the cancer peptides or portions thereof. The technique is preferred when the copy number of a target nucleic acid is very low, or when different forms of nucleic acids must be distinguished. The method is especially useful in detecting and differentiating precancer and cancer cells from normal cells.

The present invention includes a method of identifying an antigenic cancer epitope reactive with antigen specific T cells comprising the generation of nucleic acid deletion fragments from a gene. The deletion fragments are placed in an appropriate vector which in turn are transfected or transduced into a host cell for the expression of the nucleic acid product. Optionally, the host cell may also express an immunostimulatory molecule. Cancer antigen specific T-cell responses are determined in the presence of the host cell expressing the deletion product.

In the case where the host cell expresses only the deletion product, a immunostimulatory molecule may be provided by an antigen presenting cell such as a B cell, macrophage, dendritic cell and the like or by a cell transfected with a stimulatory molecule. In one embodiment, the immunostimulatory molecule is a MHC class I molecule.

By mapping using this approach, the alternative open reading frame DNA sequence encoding the cancer peptide or the antigenic cancer epitope is determined. An alternative method of identifying the cancer antigen and the antigenic cancer epitope is by generating synthetic peptides, pulsing antigen presenting cells with the synthetic peptides and adding the peptide pulsed antigen presenting cells with antigen specific T cells and measuring the antigen specific response of T cells in the presence of the peptide pulsed antigen presenting cells. The synthetic peptides that result in antigen specific T cell responses contains the antigenic cancer epitope of the present invention.

The present invention also encompassed vector comprising the alternative open

reading frame DNA sequence encoding cancer peptides or the antigenic cancer epitope. Optionally the vector may also comprise a DNA sequence encoding at least one immunostimulatory molecule. In one embodiment the vector comprises ORF-3 of the TRP- 1 gene. The present invention also encompasses a vector comprising the TRP-2 gene and fragments thereof encoding the tumor antigen of the present invention. Optionally the vector may also comprise a DNA sequence encoding at least one co- immunostimulatory molecule.

Eukaryotic expression vectors include but are not limited to retroviral vectors, vaccinia virus vectors, adenovirus vectors, herpes virus vectors, fowlpox virus vectors, baculovirus vectors, human papillomavirus vectors, equine encephalitis vectors, influenza virus vectors and the like.

The present invention encompasses novel recombinant virus expressing a cancer peptide or portion thereof encoded by an alternative open reading frame nucleic acid sequence of a gene other than the open reading frame nucleic acid sequence used to encode a normal protein or peptide from the same gene. In another embodiment, the present invention encompasses novel recombinant virus expressing the TRP-2 tumor antigen encoded by nucleic acid sequence of the TRP-2 gene or fragments or variant thereof. The recombinant virus may also express at least one immunostimulatory molecule. The recombinant virus is capable of eliciting or upregulating a cell-mediate immune response in a mammal for the purpose of preventing or treating cancer in the mammal, particularly humans.

The recombinant virus has incorporated into its genome or portion thereof a nucleic acid sequence encoding a cancer peptide, portion thereof, or antigenic cancer epitope, alone, or in combination with one or more genes encoding an immunostimulatory molecule. In one embodiemnt, the recombinant virus has incorporated into its genome a nucleic acid sequence encoding a TRP- 1 cancer peptide or portion thereof. In another embodiment, the recombinant virus has incorporated into its genome or portion thereof a nucleic acid sequence encoding a TRP-2 tumor antigen or variant thereof, alone, or in combination with one or more genes encoding an co-immunostimulatory molecule. A host cell infected with the recombinant virus expresses the cancer peptide, portion thereof, or antigenic cancer epitope, alone or in

combination with at least one immunostimulatory molecule.

Methods for constructing and expressing exogenous gene products from recombinant vaccinia virus vectors are disclosed by Perkus et al Science 229:981-984, 1985, Kaufman et al Int. J. Cancer 48:900-907. 1991, Moss Science 252: 1662. 1991 , Smith and Moss BioTechniques Nov/Dec, p. 306-312, 1984, and U.S. Patent No. 4,738,846. Suiter and Moss (Proc. Nat'l Acad. Sci. U.S.A. 89: 10847-10851 , 1992) and Sutter et al (Virology 1994) disclose the construction and use as a vector, the non- replicating recombinant Ankara virus (MVA, modified vaccinia Ankara) which may be used as a viral vector in the present invention. Baxby and Paoletti (Vaccine 10:8- 9, 1992) disclose the construction and use as a vector, a non-replicating proxvirus, including canarypox virus, fowlpox virus and other avian species for use as a viral vector in the present invention.

The vectors of the present invention may be placed in an appropriate host cell for the expression of the cancer peptide or antigenic cancer epitope. Eukaryotic host cell lines include, but are not limited to COS cells, CHO cells, Hela cells, NIH/3T3 cells, insect cells, antigen presenting cells such as dendritic cells and the like. Optionally the host cell may also express a stimulatory molecule. In the case where the host cells express both the cancer peptide or antigenic cancer epitope in combination with at least one MHC molecule, it is preferable that a eukaryotic expression system be used to allow for proper glycosylation. The expression of both the cancer antigen and the immunostimulatory molecule by the host cell provides the necessary MHC restricted peptide to specific T cells and the appropriate signal to the T cell to aid in antigen recognition and proliferation or clonal expansion of antigen specific T cells. The overall result is an upregulation of the immune system. The upregulation of the immune response is manifest by an increase in cancer antigen specific cytotoxic lymphocytes which are able to kill or inhibit the growth of cancer or precancer cells.

The DNA may be inserted into the host cell by transfection, transduction, liposomes and the like by methods known in the art. (Sambrook et al, 1989, in: "Molecular Cloning A Laboratory Manual", Cold Spring Harbor press, Plainview, New York). For liposomes, cationic lipids are preferred, for example, polycationic lipid, dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium (DMRIE) complexed

with the neutral phospholipid dioleoyl phosphatidyl-ethanolamine (DOPE) as disclosed by Nabel, E.G. et al, 1992, Hum. Gene. Ther. 3:367-275; Nabel, G.J. et al, 1992, Hum. Gene Ther. 3:649-656; Stewart, M.J. et al 1992 Hum. Gene Ther. 3:399-410; Nabel, G.J. et al 1993 Proc. Natl. Acad. Sci. USA 90: 11307-11311 ; and Harrison, G.S. et al 1995 Bio Techniques 19:816-823.

The recombinant cancer protein, tumor antigen or antigenic cancer epitope expressed by the host cells may be purified from cell lysates or cell supernatants by standard protein purification procedures known in the art. These include but are not limited to molecular sieve chromatography, ion-exchange chromatography, isoelectric focusing, gel electrophoresis, affinity chromatography, HPLC, reverse phase HPLC and the like. (Ausubel et al, 1987, in Current Protocols in Molecular Biology. John Wiley and Sons, New York, NY). Immunoaffinity chromatography may also be used for purification using anti-cancer protein antibodies or antigen binding fragments thereof as described herein, as the immunoaffinity agent. The recombinant virus may also be used as a therapeutic or vaccine. In such uses it is desirable to provide the recipient with a dosage of recombinant virus in the range of from about IO 5 to about 10'° plaque forming units/mg mammal, although a lower or higher dose may be administered.

The recombinant viral vector may be introduced into a mammal either prior to any evidence of cancer such as melanoma or to mediate regression of the disease in a mammal afflicted with a cancer such as melanoma. Examples of methods for administering the viral vector into mammals include, but are not limited to, exposure of cells to the recombinant virus ex vivo, or injection of the recombinant virus into the affected tissue or intravenous, subcutaneous, intradermal, intramuscular and the like administration of the virus. Alternatively, the recombinant viral vector or combination of recombinant viral vectors may be administered locally by direct injection into the cancerous lesion or topical application in a suitable pharmaceutically acceptable carrier. The quantity of recombinant viral vector, carrying the nucleic acid sequence of interest is based on the titer of virus particles. A preferred range for immunization is about IO 5 to 10 10 virus particles per mammal, preferably a human. Cancer antigen epitope of the present invention which is involved in tumor rejection is not limited to the ones specifically disclosed herein. Using the methods

disclosed in the present invention other cancer antigen epitopes contained in alternative open reading frame products may be identified from other tumor associated antigens (Van der Bruggen, P. et al 1991 Science 254: 1643-47; Gaugler, B.et.al. 1994 J. Exp. Med. 179:921-30; Boel, P. et al 1995 Immunity. 2: 167-75; Brichard, V. et al 1993, J Exp. Med. 178:489-95; Robbins, P. F. et al. 1994, Cancer Research 54:3124-26; Kawakami, Y. et al 1994, Proc. Natl. Acad. Sci. U. S. A. 91 :3515-19; Coulie, P. G. et al 1994 J. Exp. Med. 180:35-42; Bakker, A. et al 1994, J. Exp. Med. 179: 1005-09) such as from MART-1/Melan A (Kawakami etal J. Exp. Med. 180:347- 352, 1994), MAGE-3 (Gaugler et al J. Exp. Med. 179:921-930, 1994), gp 100 (Kawakami et al Proc. Nat'l Acad. Sci. U.S.A. 91:6458-6462, 1994), tyrosinase (Brichard et al J. Exp. Med. 178:489, 1993), TRP-2, CEA, CA-19-A, CA-125, PSA, erb-2 (Boon et al Ann. Rev. Immunol. 12:337, 1994).

Tumor infiltrating lymphocytes (TILs) derived from tumor-bearing patients recognize tumor associated antigens presented by major histocompatibility complex (MHC) class I molecules. The infusion of TIL586 along with interleukin-2 (IL-2) into the autologous patient with metastatic melanoma resulted in the objective regression of tumor. A gene encoding a tumor antigen recognized by TIL586 was recently isolated and shown to encode gp75. The present invention is the identification and isolation of an antigenic peptide, MSLQRQFLR (SEQ. ID NO. : 9), recognized by TIL586, which is not derived from the normal gp75 protein. Instead this nonamer peptide resulted from translation of an alternative open reading frame of the same gene. Thus, the gp75 gene encodes two completely different polypeptides, gp75 as an antigen recognized by IgG antibodies in sera from a patient with cancer, and a 24 amino acid product as a tumor rejection antigen recognized by T cells. This represents the first demonstration that a human tumor rejection antigen can be generated from a normal cellular gene using an open reading frame other than that used to encode the normal protein. These finding revealed a novel mechanism for generating tumor antigens, which may be useful as vaccines to induce tumor-specific cell-mediated immunity against cancer. The method of ExoIII/S 1 deletion analysis localized the cancer epitope in a small DNA fragment of the gp 75 gene. The cancer epitope was absent from the normal gp75 protein. The cancer peptide of the present invention recognized by

TIL586 was derived from the gene product translated from an alternative open reading frame of the same gene encoding the normal gp 75 protein. Substitution of ATG with ATC at nucleotides 294-296 resulted in a complete loss of the ability to stimulate cytokine release from TIL586. Cold target inhibition experiments indicated that the identified cancer epitope was capable of competing for T cell recognition with a naturally processed peptide present on the tumor cells . Six T cell clones generated from the TIL586 cell line were capable of recognizing 586mel tumor cells, 586EBV B cells pulsed with this peptide and normal melanocytes in a HLA- A31 restricted fashion, also suggesting that the gene product encoded by the alternative open reading frame might be present in the tumor cells as well as the normal melanocytes.

The present invention also relates to a gene encoding a tumor antigen recognized by TIL586 and shown to encode TRP-2. The present invention also relates to the identification and isolation of an antigenic peptide, LLGPGRPYR, derived from TRP-2 and active as a cancer vaccine. The invention provides a transgenic animal which has incorporated into its genome one or more copies of the alternative open reading frame DNA sequence encoding a cancer peptide or portion thereof. The invention also provides a transgenic animal which has incorporated into its genome one or more copies of a DNA sequence encoding TRP-2 tumor antigens or portion thereof. The general method of producing transgenic animals is described in Krimpenfort et al U.S. Patent No. 5, 175,384, Leder et al U.S. Patent No. 5,175,383, Wagner et al U.S. Patent No. 5,175,385, Evans et al U.S. Patent No. 4,870,009 and Berns U.S. Patent No. 5, 174,986. The incorporation of the gene results in overexpression, altered expression or expression of multiple forms or variants of the cancer peptides. The resulting transgenic animal are useful in studies of the development of cancer or tumor antigen of the present invention. The animal model is useful in screening vaccines and chemotherapeutic drugs for cancer treatment. The transgenic animal is also useful in studies of the development of cancer.

This invention further comprises an antibody or antigen binding portion thereof elicited by immunization of the cancer peptide or antigenic cancer epitope of the present invention. In the case where the cancer peptide or antigenic cancer epitope is comprised of only a few amino acids, the cancer peptide or antigenic cancer epitope

ay be conjugated to a carrier protein in order to elicite an antibody response. Carrier proteins such as KLH, tetanus toxoid and the like and methods of conjugation are known in the art. The antibody has specificity for and reacts or binds with the cancer peptide and the antigenic cancer epitope of the present invention. Exemplary antibody molecules are intact immunoglobulin molecules, substantially intact immunoglobulin molecules or these portions of an immunoglobulin molecule that contain the antigen binding site, including those portions of immunoglobulin molecules known in the art as F (ab), F (ab'), F (ab' , humanized chimeric antibody, and F (v). Polyclonal or monoclonal antibodies may be produced by methods known in the art. (Kohler and Milstein (1975) Nature 256, 495-497; Campbell "Monoclonal Antibody Technology, the Production and Characterization of Rodent and Human Hybridomas" in Burdon et al (eds.) (1985) "Laboratory Techniques in Biochemistry and Molecular Biology", Vol. 13, Elsevier Science Publishers, Amsterdam). The antibodies or antigen binding fragments may also be produced by genetic engineering. The technology for expression of both heavy and light chain genes is the subject of the PCT patent applications: publication number WO 901443, WO 9014424, Huse et al (1989) Science 246: 1275-1281 , and U.S. Patent No. 4,946,778.

In one embodiment, the antibodies of the invention are used in immunoassays to detect cancer peptides including TRP-1 peptides and TRP-2 tumor antigen or portions thereof in biological samples. The antibodies or antigen binding fragments thereof may be used to detect cancer peptides in tissue biopsy samples from a mammal afflicted with cancer. Assessment of the cancer antigen in a diseased tissue can be used to prognose the progression of the disease in a mammal or may diagnose the efficacy of a treatment. The immunoassay may be a radioimmunoassay, Western blot assay, immunofluorescent assay, enzyme immunoassay, chemiluminescent assay, immunohistochemical assay and the like and may be performed in vitro, in vivo or in situ. Standard techniques known in the art for ELISA are described in "Methods in Immunodiagnosis", 2nd Edition, Rose and Bigazzi, eds. John Wiley & Sons, 1980; Campbell et al "Methods and Immunology", W.A. Benjamin, Inc., 1964; and Oellerich, M. 1984, J. Clin. Chem. Clin. Biochem. 22:895-904. Conventional methods for immunohistochemistry are described in Harlow and Lane (eds) (1988) In

"Antibodies A Laboratory Manual", Cold Spring Harbor Press, Cold Spring Harbor, New York; Ausbel et al (eds) (1987) In Current Protocols In Molecular Biology, John Wiley and Sons (New York, NY). Biological samples appropriate for such detection assays include but are not limited to cells, tissue biopsy, whole blood, plasma, serum, sputum, cerebrospinal fluid, pleural fluid, urine and the like.

The antibodies or antigen binding fragments of the present invention may also be used in immunotherapy. The antibodies or antigen binding fragment thereof is provided to a mammal in an amount sufficient to prevent, lessen or attenuate the severity, extent or duration of the cancer. All articles and patents referred to are incorporated herein by reference.

While the invention is described above in relation to certain specific embodiments, it will be understood that many variations are possible, and that alternative materials and reagents can be used without departing from the invention. In some cases such variations and substitutions may require some experimentation, but will only involve routine testing.

The foregoing description of the specific embodiments will so fully reveal the general nature of the invention and others can, by applying current knowledge, readily modify and/or adopt for various applications such specific embodiments without departing from the generic concept, and therefore such adaptations and modifications are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments.

Example 1 Materials and Methods Chemicals and Reagents

The following chemicals and reagents were purchased from the sources indicated: RPMI 1640, AIM-V media, Lipofectamine, G418 (GIBCO BRL, Gaithersberg, MD); the eukaryotic expression vector pCR3 (Invitrogen, San Diego, CA); anti-HLA-A31 monoclonal antibody (One lambda, Canoga Park, CA); anti- immunoglobulin M antibody conjugated with fluorescein isothiocyanate (Vector Laboratories, Inc., Burlingame, CA).

Cytotoxic T lymphocytes (CTLs) and cell Unes

TIL586 were isolated from the tumor specimen of a patient with metastatic melanoma and grown in medium containing IL-2 (6000 IU/ml) (Chiron) for 32-60 days as previously described (Topalian, S., D. Solomon, F. P. Avis, A. E. Chang, D. L. Freeksen, W. M. Linehan, M. T. Lotze, C. N. Robertson, C. A. Seipp, P. Simon, C. G. Simpson, and S. A. Rosenberg, 1988, J. Clin. Oncol. 6:839-53). TIL586 were predominantly CD8 + T cells. TIL1200 were grown under the same conditions as described for TIL586. The T cell clones were generated by the limiting dilution method from the TIL586 cell line, and then expanded in AIM-V medium containing 6000 IU/ml IL-2.

Melanoma cell lines 397mel, 397mel/A31, 586mel, 624mel, and EBV transformed B- cell lines 586EBV and 1510EBV were established in this laboratory and cultured in RPMI 1640 medium containing 10% fetal calf serum (FCS). Normal cultured melanocytes derived from infant foreskin (NHEM680 purchased from Clonetics, CA) were cultured in melanocyte growth medium (MGM; Clonetics, CA). The COS-7 cell line was provided by Dr. W. Leonard (NIH). GM-CSF Secretion Assay

DNA transfection and GM-CSF assay were done as previously described (Wang, R. F., P. F. Robbins, Y. Kawakami, X. Q. Kang, and S. A. Rosenberg, 1995, J. Exp. Med. 181:799-804). Briefly, 200 μg of DNA carrying a different fragment and 50 ng of the HLA- A31 DNA were mixed with 2 μl of lipofectamine in 100 μl of DMEM for 15-45 min. The DNA/lipofectamine mixture was then added to the COS-7 (5 X IO 4 ) cells and incubated overnight. The following day, cells were washed twice with DMEM medium. TIL586 was added at a concentration of 1 X 10 5 cells/ well in AIM-V medium containing 120 IU/ml of IL-2. After 18-24 h incubation, 100 μl of supernatant was collected and GM-CSF was measured in a standard ELISA assay (R + D Systems, Minneapolis, MN). For peptides, 586EBV, 1510EBV and T2 cells were incubated with peptides at 37°C for 90 min, and then washed three times with AIM-V medium containing 120 IU/ml of IL-2. TIL586 was added and incubated for additional 18-24 h, 100 μl of supernatant was collected for GM-CSF assay.

Exo 111/ SI deletion constructions and PCR fragments

To make a series of deletions, the pcDNA776 plasmid DNA was digested with Xba I and filled in with alpha-phosphorothioate deoxyribonucleotide triphosphates to block Exo III nuclease digestion. The pcDNA776 plasmid is a derivative of the pcDNA3 vector containing a 2.4 kb DNA fragment and a CMV promoter for directing transcription. The linearized DNA was subjected to the second restriction enzyme Xho I digestion to generate one end sensitive to Exo III. Exo III nuclease / Mung bean nuclease deletion was performed according to the manufacture's instructions (Stratagene, CA). The detailed protocol is as follows: 1. lOμg DNA was digested with Xbal and filled in with alpha- phosphorothioate deoxyribonucleotide .

2. The DNA was digested with the second enzyme Xhol followed by phenol extraction and ethanol precipitation.

3. DNA pellet was dried and suspended in 125ul 2X Exo Buffer, 25 ul lOOmM ϊ-mercaptoethanol, lOOul water.

4. When all aliquots had been removed and placed on dry ice, the tubes were heated at 68°C for 15 minutes, and then place on ice.

5. 15 U of Mung Bean Nuclease was added (previously diluted with IX Mung Bean Dilution Buffer) to each time point tube and incubated for 30 minutes at 30°C.

6. The following were added:

4μl of 20% SDS 20μl 8M LiCl 250μl buffer-equilibrated phenol: chloroform

7. Samples were vortexed, spun 1 minute in microfuge, upper aqueous layer was removed and extracted with chloroform to extract Mung Bean protein away from DNA.

8. 25μl 3M NaOAc pH 7.0 was added to the aqueous phase. tRNA to a final concentration of lOng/μl was used as a carrier for the precipitation.

9. 650μl of cold ethanol was added. Samples were chilled on dry ice 10

minutes and spun in a microfuge for 20 minutes.

10. The supernatant was drained off and the pellets washed with 80% ethanol.

11. The pellet was dried. 12. The DNA pellet was redissolved in 15μl of lOmM Tris-Cl pH 7.5,

O. lmM EDTA. B. Ligation

13. DNA deletions were ligated using the following conditions: 1.Oμl Exo/Mung treated DNA 2.0μl 10X Ligation Buffer

500 mM Tris-HCl, pH 7.5 70 mM MgCl 2 10 mM DTT 2.0μl 5 mM ATP, pH 7.0-7.5 2.0μl T4 DNA Ligase (Cat # 600011 ; provided in kit)

13.0μl H 2 O

20.0μl Total Reaction Volume Incubated at room temperature

Stratagene offers a DNA Ligation Kit (Cat # 203003) for ligating inserts to vectors.

14. 7μl of the remaining 14μl of Exo/Mung treated DNA was used for gel electrophoresis analysis.

15. lμl of the ligation reaction was used to transform lOOμl of Epicurian coli RecA-JM109 or XLl-Blue cells and plate on LB/AMP plates. C. Low Melting Agarose Technique

To minimize screening of deletions, a portion of the deletion was run in low melting point agarose, the band of interest excised and ligation continued. The agarose level was kept below 0.5% in the ligation reaction.

PCR amplification was performed at 94 °C for 2 min followed by 25 cycles of 94°C for 1 min, 55°C for 45 sec and 72°C for 1 min. Primers gpN

(5'AGAATGAGTGCTCCTAAACTCCTCTCTCTGGG) (SEQ. ID NO: 42) and gpl lB (5'CATGTGAGA AAAGCTGGTCCCTCA) (SEQ. ID NO: 43) were used to

generate the DNA fragment (1-667) and then cloned into the pCR3 expression vector to produce pPCRHO. Plasmids ρPCR210 and pPCR220 were pCR3 vectors containing DNA insertion fragments amplified by using primers gp-1 (5' TGGATATGGCAAAGCGC ACAACTC) (SEQ. ID NO: 44) and gpl lB, gp-1 and gp22 (5' TAAATGGAA ATGTTCTCAAATTGT GGCGTG) (SEQ. ID NO: 45), respectively. Cytotoxic lysis assays

Cytolytic assay was done as previously described (Kawakami, Y et al., 1994, Proc. Natl. Acad. Sci. USA 91 :6458-62). Briefly, the target cells were labeled with chromium for 90 min. After washing three times, the cells were incubated with peptides at a concentration of 1 μg/ml for 90 min. The cells were washed again, counted, and then mixed with TIL586 at the indicated ratio of effector : targets (E : T). Chromium release was measured after 4 h incubation. The peptides were synthesized by a solid-phase method using a peptide synthesizer (Model AMS 422, Gilson Co., Inc., Worthington, OH). Some peptides were purified by HPLC and had greater than 98% in purity. For titration of the ORF3P peptide recognized by TIL586, 586EBV B cells were incubated with various concentrations of the purified ORF3P peptide. Percentage of specific lysis was determined from the equation (A- B)/(C-B) X 100 where A is lysis of 586EBV B cells by TIL586 in the presence of a peptide, B is spontaneous release from 586EBV B cells in presence of the same peptide but in the absence of effector cells, and C is the maximum chromium release. Cold target inhibition of cytolysis was performed using "Cr-labeled 586mel or 624mel cells as "hot" targets and 586EBV B and T2 cells pulsed with peptides as "cold" targets. Site-directed mutagenesis

For construction of site-directed mutagenesis, mutated primers GPMUT1 were used (5'GCCATGGGCAGAGATGATCGGGAGGTCTGGCCCTTGCGCTTCTTC AATAGGACATCTCACTGCAAC) (SEQ. ID NO: 11) and GPA1 to generate a PCR fragment containing a mutation (G to C) at nucleotide 296. The wild-type DNA fragments were ampl i fied by the use of pri mers GPF 1 (5'GAAGATCTGCCATGGGCAGAGATGATCGGGAGG TCTG) (SEQ. ID NO: 12), GPE1 (5' GAATTCGTTG TGT CCTGAGAAATTGCCGTTG) (SEQ. ID NO: 13),

GPE2 (5'GA ATTCGACTATGAGAACCCTCTGGTCACAGGC) (SEQ. ID NO: 14) and GPA1 (5'AAGATCTGGGCC CGGACAAAGTGGTTCTTTTC) (SEQ. ID NO: 15) as indicated by arrowheads in Fig. 3A. The purified PCR products were then cloned into the pCR3 expression vector. All plasmids containing PCR fragments were sequenced to confirm the orientation and nucleotide sequence.

Example 2 Localization of the antigenic peptide (s) recognized by TIL586 In order to identify the antigenic epitope from gp75, we generated a series of nested deletions of gp75 gene were generated from the 3' end using Exo III/S1 nuclease as well as additional DNA fragments from gp75 by PCR amplification (Fig. IA) . The reason pcDNA776 was chosen as a starting material for deletion studies is that this clone was initially identified by a library screening and conferred the ability to stimulate cytokine release from TIL586. Plasmid pcDNA776 was depositied with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, MD under the terms of the Budapest Treaty on January 19, 1996 and was assigned the accession number, ATCC 97423. Since the goal of this study was to identify the epitope recognized by TIL586, fragments as short as possible were used (the truncated form of gp75, instead of full length cDNA) such that the epitope in a relative small DNA fragment could quickly be located. These deletions constructs were then transfected into COS-7 cells together with the pBK-CMV plasmid containing the HLA- A31 gene. (Wang, R.F. et al., 1995, J. Exp. Med. 181 :799-804). After 24 hours, the transfected COS-7 cells were tested to determine which construct could stimulate cytokine release by TIL586. A small truncated DNA fragment ranging from nucleotide 247 to 771 , which lacked the normal gp75 initiation codon, retained the ability tostimulate GM-CSF release by TIL586, suggesting that the epitope recognized by TIL586 was located in the DNA fragment containing nucleotides from 247 to 771. Since there is an ATG start codon in a relative good context of Kozak sequence (GATATGG) (SEQ. ID NO: 16) located at nucleotides 445-447 and is in the same frame as gp75 open reading frame, it was reasoned that the epitope recognized by TIL586 might be located in the region from nucleotide 445-771. Therefore, pPCR210 and pPCR220 were constructed, which were derivatives of the pCR3 expression vector and contained an internal ATG codon in frame with gp75 (GATATGG) (SEQ. ID NO:

16) located at 445 bp as a start codon for translation of the truncated normal gp75 protein. However, neither pPCR210 nor pPCR220 conferred the ability to stimulate cytokine secretion from TIL586 after co-transfection of COS-7 with the HLA- A31 gene (Fig. IB), suggesting that the epitope was located upstream of these fragments. Therefore, an additional plasmid pD776A was constructed, which contained the nucleotide sequence from 247-442 and did not have any ATG codon in the same frame as gp75, but did contain two ATG codons in different open reading frames relative to gp75. This plasmid strongly stimulated cytokine release from TIL586 when co- transfected with A31 cDNA into COS-7 cells. The plasmid pPCRHO containing the authentic start codon of gp75 stimulated several fold lower cytokine release than did pDel 5 or pD776A when co-transfected with the HLA-A31 gene (Fig. IB). These results suggested that the epitope(s) recognized by TIL586 were located in the region from nucleotides 247 to 442.

Although this region (nucleotides 247-442) did not have any ATG start codon in the normal gp75 open reading frame, initiation of translation from the non- ATG codons such as ACG, CTG and GTG had been reported in some cases (Hann, S.R. 1994, Biochimie 76:880-86; Muralidhar, S. et al J Virol. 1994, 68: 170-76). To identify the epitope in this region, synthetic peptides were made based upon the peptide binding motif of HLA-A31 (hydrophobic residue at position 2 and positively charged residue at the C-terminus) (Fig. 2) (Falk, K. et al lmmunogenetics 1994, 40:238-41). The majority of the peptides selected for this study were nonamers, although some were lOmers and l lmers. These peptides were pulsed onto 586EBV B cells and the ability of these cells to stimulate cytokine release by TIL586 (Table 1). One peptide, AACDQRVLIVRR (SEQ. ID NO: 25), very weakly induced GM- CSF release from TIL586. However, this peptide failed to sensitize peptide-loaded 586EBV B for lysis by TIL586 (Table 1).

Tab|e I Table 1. Screening of synthetic peptides with reactivity to TIL586

Target cells pulsed with peptide TIL586

% Specific

GM-CSF release lvsis (E:T 20: n

Peotides from QRFl (»D751

586EBV + peptide DDREVWPLR (Seq. ID No: 17) <50 < 10 586EBV + peptide VWPLRFFNR (Seq.ID No: 18) <50 < 10

586EBV + peptide SGHNCOTCR (Seq.ID No: 19) <50 < 10

586EBV + peptide CGTCRPGWR (Seq.ID No: 20) <50 < 10

586EBV + peptide ACDQRVLIVR (Seq.ID No: 21) <50 < 10

586EBV + peptide ACDQRVLTVRR (Seq.ID No. 22) <50 < I0 586EBV + peptide LWDVPSWLER (Seq.ID No. 23) <50 < 10

586EBV + peptide AISQDTTVGR (Seq.ID No. 24) <50 < ιo

586EBV + peptide AACDQRVLIVR (Seq.ID No. 25) 250 < 10

586EBV + peptide DQRVLIVRR (Seq.ID No. 26) <50 < 10

S86EBV + peptide IVRRNI DLSK (Seq.ID No. 27) <50 < 10 586EBV + pepude LSKEEKNHFVR (Seq.ID No. 28) <50 < 10

586EBV + none <50 < 10

586mel -I- none >5000 45

586EBV cells were incubated with individual peptide at a concentration of 1 Mg/ml for 90 min. GM-CSF release was measured after co-incubation of peptide-loaded 586EBV cells with TIL586. GM-CSF secretion by TIL586 alone without stimulators was subtracted. S86EBV was a EBV transformed B cell line expressing HLA-A-31 . Cytotoxic lysis of peptide-pulsed 586EBV by T1L586 was done in a 4-h chromium release assay.

Because this peptide weakly stimulated cytokine release from TIL586 only when incubated 586EBV B cells at high concentrations (> lμg/ml) and did not sensitize the target cells for lysis by TIL586 even at 10 μg/ml of peptide concentration (data not shown), it may not represent the predominant T cell epitope recognized by TIL586.

To further define the region containing the predominant T cell epitope, two additional plasmids were constructed containing PCR fragments amplified by primers GPF1, GPE1 and GPE2, respectively (Fig. 3 A). As shown in Fig. 3B, both plasmids containing an ATG start codon at the beginning of the smaller PCR fragment conferred the ability to stimulate cytokine release by TIL586 in association with HLA- A31, suggesting that the epitope recognized by T cells was encoded within an 82

nucleotide sequence between bases 247 and 329 of the gp75 cDNA. Twenty eight overlapping peptides (9 mers or 10 mers) in ORFl were synthesized based upon the amino acid sequence of gp75 in this region, but none were found to stimulate GM- CSF release or sensitize 586EBV B cells for lysis by TIL586 (data not shown). Example 3

Antigenic peptides resulted from translation of an alternative open reading frame of the gp75 gene

The failure to identify an epitope recognized by TIL586 in this small region suggested that alternative open reading frames might be translated. Two ATG codons in the relatively good context were present in this region (nucleotides 247-442) in different open reading frames relative to the gp75 open reading frame (ORFl) (Fig. 2). Translation from the first ATG (251 GAGATGA257) (SEQ. ID NO: 29) resulted in a open reading frame encoding 45 amino acids (ORF2) while translation starting from the ATG located between nucleotides 294-296 (GACATGT) (SEQ. ID NO: 30) generated a 24 amino acid gene product (ORF3) (Fig. 2). Three peptides derived from ORF2 and two peptides from ORF3 were selected and synthesized on the basis of the HLA-A31 binding motif (Table 2 and Fig. 2). Suφrisingly, one peptide MSLQRQFLR (SEQ. ID NO: 9) (designated as ORF3P) which derived from ORF3 was strongly recognized by TIL586 when pulsed onto 586EBV B cells (Table 2). The recognition of the ORF3P peptide (MSLQRQFLR) (SEQ. ID NO: 9) by TIL586 was observed only when the peptide was pulsed onto autologous 586EBV B cells and 1510EBV B (A31 +) cells, but not when peptide was loaded onto T2 (non-HLA-A31) cells (Fig. 4A), suggesting that recognition of this peptide by TIL586 was HLA- A31 restricted. The peptide mass of ORF3P was confirmed by mass spectrometry analysis. As shown in Fig. 4B, TIL586 lysed 586EBV B cells pulsed with the ORF3P peptide, but failed to lyse 586EBV B cells pulsed with irrelevant peptide which met the criterion of the peptide binding motif of HLA-A31 , but was not recognized by TIL586 or T2 cells pulsed with the ORF3P peptide. Sensitization for lysis by the peptide showed maximal effect at 100 nM, though lytic activity was detected even at 1 nM of peptide concentration (Fig. 4C). TIL586 did not recognize either peptides MSLQRQFLRT (SEQ. ID NO: 33) or SLQRQFLRT (SEQ. ID NO: 34), or modified peptides (substitution of anchor residues at positions 2, 6 and 9) MLLQRQFLR (SEQ.

ID NO: 36), MRLQRQFLR (SEQ. ID NO: 37), MSLQRLFLR (SEQ. ID NO: 38), MSLQRQFLE (SEQ. ID NO: 39), MSLQRQFLR (SEQ. ID NO: 40) derived from MSLQRQFLR (SEQ. ID NO: 9) (Table 2). TIL586 only recognized the peptide MALQRQFLR (SEQ. ID NO: 35) with a substitution Ser with Ala at position 2 compared to the peptide MSLQRQFLR (SEQ. ID NO: 9) (Table 2).

Table 2. Identification of antigenic peptides with reactivity to TIL586

Target cells pulsed with peptide TIL586

GM-CSF release % Specific lysis (E:T 20: n

Experiment A

Peptides derived fro QRF2

586EBV + peptide ISQDTTVGR (Seq.ID No: 31) <50 < 10

586EBV + peptide AISQDTTVGR (Seq.ID No. 24) <50 < 10 586EBV + peptide AGEELPVTR (Seq.ID No. 32) <50 < 10

Peptides derived from QRF3

586EBV + peptide LWDVPSWLER (Seq.ID No. 23) <50 < 10

586EBV + peptide MSLQRQFLR (Seq.ID No. 9) >8000 60

586EBV + None <50 < 10 586 mel + None >5000 47

Experiment B

Modified peptides of MSLQRQFLR

586EBV + peptide MSLQRQFLR (Seq.ID No. 9) >8000 63

586EBV + peptide MSLQRQFLRT (Seq.ID No. 33) <50 < 10

586EBV + peptide SLQRQFLRT (Seq.ID No. 34) <50 < 10

586EBV + peptide MALQRQFLR (Seq.ID No. 35) >5000 54

586EBV + peptide MLLQRQFLR (Seq.ID No. 36) <50 < 10

586EBV + peptide MRLQRQFLR (Seq.ID No. 37) <50 < 10

586EBV + peptide MSLQRLFLR (Seq.ID. No. 38) <50 < 10

586EBV + peptide MSLQRQFLE (Seq.ID No. 39) <50 < 10

586EBV + peptide MSLQRQFLR (Seq.ID No. 40) <50 < 10

586EBV + None <50 < 10

586mel + None >5000 48

Conditions for peptide incubation with 586EBV B cells and GM-CSF release assay were the same as described in Table 1. GM- CSF secretion by TIL586 alone without stimulators was subtracted. Modified peptides were made by substitution of amino acid at the positions 2, 6 and 9 relative lo MSLQRQFLR (Seq. ID No. 9). Cytotoxic lysis of peptide-pulsed 586EBV by TIL586 was done in a 4-h chromium release assay.

Example 3 Translation is necessary for generating the naturally processed antigenic peptide

Since there was a stop codon TAG (288-290) located in the six nucleotides upstream of the ATG start codon of ORF3 (294-296) (Fig. 2), it was unlikely that the ORF3P peptide resulted from a frameshift. The DNA sequence analysis also

confirmed that there was no deletion or insertion in the upstream region. To investigate if the ATG located at nucleotides 294-296 played an important role in translating the 24 amino acid product, ATGT (294-297) to ATC.T were mutated (294- 297) to eliminate the translation of ORF3, which would result in a change of Cys (UGU) to Ser (UCU) in ORFl (gp75) (Fig. 3A). A plasmid containing the mutated gene (pGFMUTl) was tested for its ability to confer recognition by TIL586 when co- transfected into COS-7 along with the HLA- A31 cDNA. Fig. 3B showed that the mutated gene completely lost the ability to stimulate GM-CSF release by TIL586 compared to the construct containing the wild type gene. This observation indicated that ATG in ORF3 in the nucleotide positions 294-296 was required for translation of the 24 amino acid product, and therefore was essential for generating the T cell epitope recognized by TIL586.

Since the Met (ATG) is in position 1 of the peptide epitope and the mutation of ATG to ATC at nucleotides 294-296 resulted in a change of Met to He in position 1 of the peptide, the possibility that the loss of recognition of the mutated gene by TIL586 could be due to the loss of the ability of the mutated peptide to bind to MHC class I molecules was investigated. A synthetic peptide (ISLQRQFLR) (SEQ. ID NO: 41) with the same amino acid sequence as that encoded by the mutated gene was made and tested for recognition by TIL586. It was found that the synthetic mutated peptide was still recognized by TIL586 at comparable concentrations to that of the wild-type peptide. Furthermore, when the same mutation was introduced into the full length cDNA, no reactivity to TIL586 was observed whereas the wild-type cDNA was capable of stimulating cytokine release from TIL586 at a level similar to pPCRHO. This is in agreement with the deletion data, indicating that TIL586 did not recognized peptide(s) in other regions of the gene. These results suggested that the loss of recognition of the mutated gene (ATG to ATC at nucleotides 294-296) by T cells was due to inactivation of translation initiation of ORF3.

Example 4 Recognition of the antigenic peptide on tumor cells as well as melanocytes To address the question of whether TIL586 recognized a naturally processed peptide which is similar or identical to the ORF3P peptide on the tumor cells, the ability of the ORF3P peptide pulsed 586EBV B cells to inhibit lysis of 51 Cr-labeled

586mel in a cold-target inhibition assay was examined. Significant inhibition of lysis of 5, Cr-labeled 586mel was observed by 586EBV B cells pulsed with the ORF3P peptide but not with 586EBV B cells pulsed with irrelevant peptide or T2 cells pulsed with the ORF3P peptide (Fig. 5A), indicating that this peptide epitope was capable of competing with a naturally processed peptide on tumor cells for T cell recognition. As predicted, the ORF3P loaded 586EBV B cells did not inhibit lysis of target 624mel by TIL1200, which recognizes the gplOO antigen in the HLA-A2 context, compared to 586EBV B alone (Fig. 5B). To further test if T cell clones can recognize both the ORF3P peptide pulsed 586EBV B cells and tumor cell lines, T cell clones were generated from the TIL586 cell line by limiting dilution (1 cell /well in 96-well round bottom microplate) and further expanded in culture. T cell clones were generated by limiting dilution (1 cell per well) from the TIL586 cell line. T cell clones were further expanded in AIM-V medium containing 6000 IU/ml IL-2. 586EBV B cells were pulsed with the ORF3P peptide or irrelevant peptide for 90 min at 31°C. After washing three times, T cell clone or TIL586 cells were added and coincubated for additional 18-24 h. For 586 mel, 397 mel/A31 + tumors and melanocyte NHEM680 cells, 1 X 10 s cells per well were incubated with 1 x 10 s cells to T cell clones, TIL586-C1 (Fig. 6A), TIL586-C4 (Fig. 6B) and TIL586-C6 (Fig. 6C) or TIL586 (Fig. 6D) for 18-24 h, respectively. GM-CSF assay was performed as described in Fig. IB Six T cell clones were capable of recognizing 586mel tumor cells, 586EBV B cells pulsed with the ORF3P peptide, and HLA-A31 positive melanocytes, but not 397mel/A31 or 586EBV B cells alone. Representative data is shown in Figures 6A- 6D. These results suggested that T cell clones probably recognized a naturally processed peptide either similar or identical to the ORF3P peptide on tumor cells and normal melanocytes.

Since there is a 40-45% amino acid sequence identity of gp75 to tyrosinase, gplOO and TRP-2, the possibility that the peptide recognized by the T cell clones was not derived from gp75, but from one of these other proteins was tested. COS-7 cells were transfected with HLA-A31 plus tyrosinase, gplOO or TRP-2 cDNAs, respectively, and found that none could be recognized by the six T clones while the COS-7 transfected HLA-A31 and gp75 cDNA stimulated GM-CSF release from these clones (data not shown). A computer database search also indicated that no known

proteins including tyrosinase, gplOO and TRP-2 in the database contained amino acid sequences with the peptide binding motif of HLA- A31 and significant similarity to the pepude epitope recognized by TIL586.

Example 5 In vivo Protection Assay

For in vivo protection studies, MHL-A31 + transgenic mice are immunized with 0, lpg, Ing, lμg, lmg or lOOmg of cancer peptide (SEQ. ID NO: 9), intravenously at day zero and day 14 before a subcutaneous challenge with IO 4 Trp-1 + B16 mouse melanoma cells or intravenous challenge with 5 x 10 s Trp-2 + B16 mouse melanoma cells. Mice receiving tumor cells subcutaneously are observed twice a week for tumor development and the size determined. Mice receiving tumor cells intravenously are euthanized on day 12 and the number of lung metastases determined as described by Houghton, A.N. 1994 J. Exp. Med. 180: 1-40.

Example 6 In vivo Treatment Assay

For in vivo treatment, MHL-A31 + transgenic mice are challenged with either 1 x 10 5 or 5 x 10 s Trp-1 + B16 mouse melanoma cells intravenously in order to establish pulmonary metastases. Mice are subsequently vaccinated with a recombinant virus expressing cancer peptide (SEQ. ID NO: 9) at 10 5 PFU/mg body weight. Mice are euthanized on day 12 and the number of pulmonary metastases in vaccinated mice vs. non-vaccinated mice determined.

Example 7

Cancer Antigen Specific T Lymphocytes

Immunotherapy T-lymphocytes presensitized to a melanoma antigen may be effective in therapeutically treating mammals afflicted with a melanoma. T-lymphocytes are isolated from peripheral blood or melanoma tumor suspensions and cultured in vitro

(Kawakami, Y. et al, 1988, J. Exp. Med. 168:2183-2191).

The T lymphocytes are exposed to the cancer peptide (SEQ. ID NO: 6) at a concentration of 1 μg/ml alone or in the presence of IL-2, resensitized and expanded in culture. T-lymphocytes exposed to the cancer peptide are administered to a mammal at about IO 9 to 10 12 lymphocytes per mammal. The lymphocytes are

administered either intravenously, intraperitoneally or intralesionally. The treatment may be administered concurrently with other therapeutic treatments such as cytokines, surgical excision of melanoma lesions and chemotherapeutic drugs.

Example 8 Treatment of Patients with Metastatic Melanoma

In this protocol, patients with advanced melanoma are immunized with an antigenic cancer epitope.

Patients eligible for the trial must have evidence of measurable or evaluable metastatic melanoma that has failed standard effective therapy. Patients must have tumors that express the TPI-1 antigen as evidenced by PCR or Northern Blot analysis of tumor cell RNA.

Patients receive either Ing, lμg, lmg or 500mg/kg body weight of a cancer peptide (SEQ. ID NO: 6) via intravenously at day zero, day 7 and day 14 alone or in combination with IL2 and/or an immunostimulatory molecule. Patients are evaluated for toxicity, immunologic effects and therapeutic efficacy.

Lymphocytes taken from the treated patients are tested for specific response to the cancer antigen comprising the amino acid sequence MSLQRQFLR (SEQ. ID NO: 6).

A complete response is defined as the disappearance of all clinical evidence of disease that lasts at least four weeks. A partial response is a 50% or greater decrease in the sum of the products of the perpendicular diameter of all measurable lesions for at least four weeks with no appearance of new lesions or increase in any lesions. Minor responses are defined as 25-49% decrease in the sum of the products of the perpendicular diameters of all measurable lesions with no appearance of new lesions and no increase in any lesions. Any patient with less than a partial response is considered a non-responder. The appearance of new lesions or greater than 25 % increase in the product of perpendicular diameters of prior lesions following a partial or complete response is considered as a relapse.

EXAMPLE 9 Materials and Methods For TRP-2

Chemicals and Reagents

The following chemicals and reagents were purchased from the sources

indicated: RPMI1640, AIM-V media, Lipofectamine, G418 (GIBCO BRL, Gaithersberg, MD); the eukaryotic expression vector pCR3 (Invitrogen, San Diego, CA); anti-HLA-A31 monoclonal antibody (One lambda, Canoga Park, CA); anti-immunoglobulin M antibody conjugated with fluorescein isothiocyanate (Vector Laboratories, Inc., Burlingame, CA).

T cell clones and lines

The T cell clones were generated by limiting dilution methods (at 1 cells/well) from the TIL586 cell line, and used allogenic PBL (IX IO 3 cells/well) as feeder cells in RPMI1640 containing 10% human AB sera and 500 IU IL-2. After 12 days, T cell clones were then expanded in AIM-V medium containing 6000 IU/ml IL-2. To obtain an optimal expansion, we used the OKT3 expansion method described by S. Riddell (Walter et al. 1995 N. Engl. J. Med. 333: 1038-1044). Briefly, on day 0, 5x10' 5x10* T cells were cocultured with HLA-A31 PBL (500: 1, PBL: T cell ratio) and 586EBV B cells (100: 1 , EBV: T cell ratio) in 25 ml RPMI 1640 containing 11 % human sera, 30 ng/ml OKT3 antibody and antibiotics. On day 1 , IL-2 was added at final concentration of 180 IU/ml. The media were changed with fresh media containing 11 % human sera, IL-2 180 IU/ml on day 5. The media were then changed every three days. On day 12-14, T cells were harvested, counted and cryopreserved. TIL586 were isolated from the tumor specimen of a patient with metastatic melanoma and grown in medium containing IL-2 (6000 IU/ml) (Chiron) for 32-60 days as previously described (Topalian et al. 1988, J. Clin. Oncol. 6:839-853). TIL586 were predominantly CD8 + T cells.

Melanoma cell lines 397mel, 397mel/A31, 586mel, 624mel, 624mel/A31 and EBV transformed B-cell lines 586EBV and 1510EBV were established in our laboratory and cultured in RPMI 1640 medium containing 10% fetal calf serum (FCS). Normal cultured melanocytes derived from infant foreskin (NHEM680 purchased from Clonetics, CA) were cultured in melanocyte growth medium (MOM; Clonetics, CA). The COS-7 cell line was provided by Dr. W. Leonard (NIH).

GM-CSF Secretion Assay

DNA transfection and GM-CSF assays were performed as previously described (Wang et al. 1995, J. Exp. Med. 181:799-804). Briefly, 200 ng of DNA encoding antigens and 50 ng of the HLA- A31 DNA were mixed with 2μl of lipofectamine in 100 ml of DMEM for 15-45 min. The DNA/lipofectamine mixture was then added to the COS-7 (5 X 104) cells and incubated ovemight. The following day, cells were washed twice with DMEM medium. TIL586 was added at a concentration of 1 X 105 cells/well in AIM-V medium containing 120 IU/ml of IL-2. For T cell clones, only 1-2 X 10" cells/well were added. After 18-24 h incubation, 100 μl of supernatant was collected and GM-CSF was measured in a standard ELISA assay (R + D Systems, Minneapolis, MN). For testing peptides recognition, 586EBV or T2 cells were incubated with peptides at 37° C for 90 min, and then washed three times with AIM-V medium containing 120 IU/ml of IL-2. T cells were added and incubated for an additional 18-24 h, 100 μl of supernatant was collected for GM-CSF assay.

Exo III /SI deletion constructions and subcloning

TRP-2 cDNA was a gift of Dr. Shibahara (Yokoyama et al. 1994, Biochim. Biophvs. Acta 1217:317-321) and subcloned into the pCR3 vector with a CMV promoter for expression. To make a series of deletions, the plasmid pCR3 containing TRP-2 cDNA was digested with Xba I and filled in with alpha-phosphorothioate deoxyribonucleotide triphosphates to block Exo III nuclease digestion. The linearized DNA was subjected to the second restriction enzyme digestion to generate one end sensitive to Exo III Exo III nuclease/Mung bean nuclease deletion was performed according to the manufacture's instructions (Stratagene, CA). All deletion constructs were sequenced to determine the location of DNA sequence being removed. pTA plasmid was a derivative of pCR3-TRP2, in which an Apa I DNA fragment was deleted from the 3' end of TRP-2 gene. pTK was created after removal of a Kpnl DNA fragment from the 3' end of the TRP-2 gene. pTP was generated by deleting an internal Pst I fragment and religation.

Northern Blot Analysis

Total RNA was isolated by the guanidine isothiocyanate/cesium chloride centrifugation method. Total RNA from human normal tissue was purchased from Clontech, CA. Twenty μg of total RNA was subjected to electrophoresis in a 1.2% agarose formaldehyde gel and transferred to a nylon membrane. A 2.0 kb DNA fragment of the TRP-2 gene was labeled with 32 P-α-CTP by the random priming method. Prehybridization and hybridization were performed according to the QuickHyb protocol (Stratagene). Membranes were washed twice with 2 X SSC/0.1 % SDS at room temperature for 15 min and twice with 0.1 X SSC/0.1 % SDS at 60 °C for 30 min. The autoradiography was performed at -70 "C.

Cytotoxicity assays

Cytolysis was determined by use of Calcein AM (Molecular Probes, Eugene, OR). Briefly, T2 or 586EBV B cells were pulsed with peptides in RPMI 1640/5 %FCS for 90 min. Tumor cells and the peptide pulsed EBV B cells were labelled with Calcein AM (15 μl of 1 mg/ml Calcein AM for every lxlO 6 cells) for 30 min at 37 * C. Following incubation, cells were washed three times with AIM V/120 IU IL-2. lxlO 3 target cells were mixed with T cells at various E:T ratio. After 4 h incubation at 37° C, 5 μl of bovine hemoglobin quench solution containing ethidium bromide was added. The plate was read by Lambda scan. The percentage of lysis was calculated from the following equation: [1-(A-B)/(C-B)] X 100 where A is the reading of non-lysed cells in the presence of T cells, B is background signal value and C is the maximum signal value from target cells.

The peptides were synthesized by a solid-phase method using a peptide synthesizer (Model AMS 422, Gilson Co. , Inc., Worthington, OH). Some peptides were purified by HPLC and had greater than 98% in purity. The peptide mass of some peptides was confirmed by mass spectrometry analysis.

EXAMPLE 10 Recognition of new antigens on tumor cells by CTL clones

In previous studies, we have isolated a number of T cell clones from the TIL586 cell line by the limiting dilution method (Wang et al. 1996b, J. Exp. Med.

183: 1131-1140). Among them, six clones recognized 586EBV B cells pulsed with the ORF3P peptide derived from a gene product translated from an alternative open reading of TRP-l/gp75 gene, and the autologous 586mel tumor cells, but did not recognize 586EBV B cells pulsed with an irrelevant peptide. TIL586-C1 was chosen as one representative for these T cell clones as shown in Fig. 7. However, several T cell clones isolated from the same TIL586 cell line recognized neither 586EBV B cells pulsed with the TRP-1 peptide ORF3P nor COS cells transfected with TRP-1 and HLA- A31 cDNAs, but were capable of recognizing 586mel as well as HLA-A31 + melanocytes (Fig. 7). These results suggested that these T cell clones recognized additional tumor antigens on the 586mel tumor cells. These T cell clones were then expanded to obtain enough cells for screening cDNA libraries or testing other cDNAs for recognition by methods described in the Materials and Methods section (Example 9). One of clones, CTL clone 4, was successfully expanded and used for further studies as described below.

EXAMPLE 11

Identification of a cDNA encoding a tumor antigen recognized by T cell clones To determine the HLA molecule responsible for presenting antigen to CTL clone 4, we transfected HLA-A31 cDNA into A31 -negative tumor lines such as 397mel and 624mel and tested for recognition by the CTL clone. Transfectants of 397mel and 624mel expressing HLA-A31 were significantly recognized by CTL clone 4 (Table 3). Furthermore, these T cells were also capable of recognizing the HLA- A31 positive allogeneic tumor line 1353mel, indicating that recognition of the tumor antigen by CTL clone 4 was HLA- A31 restricted.

Table 3: Specific secretion of GM-CSF by CTL clone 4 is HLA-A31 -restricted Stimulators Cell lines Transfected gene HLA-A31 GM-CSF expression secretion

(pg/ml)

None none < 10

none 23

397mel

(TRP1-/TRP2+)

397mel HLA-A31 2840

(TRP1-/TRP2+)

624mel none 39

624mel HLA-A31 + 670

(TRP1 +/TRP2+)

1353mel none 879

(TRP1 +/TRP2+)

586mel none > 4000

586EBVB none + 29

COS-7 none 35

COS-7 HLA- A31 30

GM-CSF in the supernatant was measured after 24 h incubation of 2 x 10 4 CTL clone 4 cells with either melanoma cell lines or COS-7 transfected with the HLA- A31 cDNA.

Since only a limited number of T cells were available, we first tested whether or not these T cells recognized previously identified tumor antigens or melanocyte-lineage differentiation proteins. Recognition of COS-7 cells transfected with HLA- A31 cDNA and genes encoding the known tumor antigens or putative antigens including MART-1 (Kawakami et al. 1994a, Proc. Nat'l Acad. Sci USA 91:3515-3519), gp75 (Wang et al. 1995, J. Exp. Med. 181:799-804), gplOO (Kawakami et al. 1994b, Proc. Nat'l Acad. Sci. USA 91:6458-6462), tyrosinase (Brichard et al. 1993, J. Exp. Med. 178:489-495), plS (Robbins et al. 1995, Immunol. 154:5944-5950) and TRP-2 (Yokoyama et al. 1994, Biochi . Biophys. Acta 1217:317-321; Bouchard et al. 1994, Eur. J. Biochem. 219: 127-134) by CTL clone 4 was tested. COS cells transfected with HLA-A31 alone or TRP-2 alone did not confer recognition by the T cell clones. However, COS cells transfected with HLA-A31 and TRP-2 cDNA stimulated GM-CSF release from T cells, whereas COS cells transfected with HLA- A31 and other genes did not, indicating that the T cell

clone 4 recognized TRP-2 as a tumor antigen in an HLA- A31 restricted manner.

Analysis of the structural similarities in HLA- A3, Al l, A31, A33 and A68 and their peptide binding motif has suggested the existence of the A3-like supermotif (Sidney et al. 1996, Human Immunol. 45:79-93). A single epitope peptide could cross-react with HLA- A3, Al l , A31 , A33 and A68 molecules which are cumulatively expressed in about 40-50% of the general population. It has been reported that the same peptide epitope derived from Hepatitis B virus nucleocapsid protein could be presented by HLA- A31 and -A68 molecules and recognized by the corresponding HLA- A31 or -A68 restricted CTL (Missale et al. 1993, J. Exp. Med. 177:751-762). We tested whether HLA-A31 restricted T cells recognized TRP-1 and TRP-2 epitopes when pulsed onto HLA-A3 positive EBV B cells. Interestingly, weak recognition was detected based on GM-CSF release from T cells. However, no recognition of HLA-A3 positive tumor cells was detected.

EXAMPLE 12

Expression of the TRP-2 gene

Northern blot analyses were performed using TRP-2 cDNA as a probe to evaluate the expression pattern of TRP-2 in different tissues. Normal retinal tissue was shown to be the only positive in the expression of TRP-2 among the normal human tissues tested. The expression pattern of TRP-2 in melanoma cell lines and other cell lines is listed in Table 4 below. Twenty five of thirty melanoma cell lines were found to express TRP-2. The Burkitt's B cell line Daudi and the breast cancer cell line MDA23 1 were negative, in agreement with previous results (Bouchard et al. 1994, Eur. J. Biochem. 219: 127-134). Thus, like tyrosinase, TRP-1, gplOO and MART-1, the expression pattern of this gene appeared to be restricted to melanomas, normal melanocyte cell lines and retina.

Table 4: Expression of TRP-2 in different cell lines and human tissues tested

Melanoma cell lines Expression of Cell lines/tissues E E:xpression of TRP-2

TRP-2

397mel + A375 - 526mel + 586EBVB -

397mel + Melanocytes 50 lmel + FM906 + 537mel - F 680 + 553Bmel + 586mel + Other tumor lines

624mel + Daudi - 677mel + MDA231 - 679mel 697mel + Normal tissues 729mel - Retina +

894mel + Testis - 836mel - Brain - 888mel + Spleen - 928mel + Liver - 1290mel + Fetal liver -

1300mel + Thymus - 952mel + Lung - HT144mel + 101 lmel + 1088mel +

SK23 + SK28 + Maisel + groves + WN266 +

Expression of TRP-2 was tested by Northern blot analysis with 10-20 ~g of total RNA and probed with the TRP-2 cDNA fragment. Daudi is a Burkitt's B cell line and MDA231 is a breast cancer cell line.

EX AMPLE 13 The peptide epitopes recognized by T cells

To determine the antigenic epitopes from TRP-2, a series of nested deletions of the TRP-2 gene from the 3' end using Exo III/Sl nuclease as well as DNA fragments encoding the truncated form of TRP-2 were generated. These deletions and subcloned constructs were transfected into COS-7 cells along with the pBK-CMV plasmid containing the HLA-A31 cDNA. Recognition of the transfected COS cells were tested with CTL clone by measuring GM-CSF cytokine release from the CTL clone. Fig. 9 indicated that pTDl, 2 and 3 constructs retained the ability to stimulate cytokine release from the CTL clone 4, but pTD4 and pTD5 lost the stimulating activity to the CTL clone 4, indicating that the epitope(s) recognized by the CTL clone 4 was located in the region of nucleotides 836-1045. This was consistent with results obtained by the subcloning experiments. Although pTA and pTP lost the ability to stimulate cytokine release from CTL clone 4, pTK still remain positive in the cytokine release assay. Therefore, the epitopes resided in a DNA fragment flanked by the first PstI and Kpnl sites as shown in Fig. 10.

To identify the epitopes from the coding region of this small DNA fragment, five synthetic peptides were based on the synthesized peptide binding motif for HLA- A31 (hydrophobic residues at position 2 and positively charged residues at position 9) (Rammensee et al. 1995, Immunogenetics 41: 178-228). These peptides were pulsed onto 586EBV B cells and tested for their ability to stimulate cytokine release by CTL clone 4. As shown in Table 5, peptide TRP,,^ was strongly recognized by CTL clone 4 when pulsed on 586EBV B cells. The recognition of this peptide by CTL clone 4 was observed only when the peptide was pulsed onto HLA-A31 + EBV B cells such as 586EBV and 1510EBV, but not onto HLA-A31 negative T2 cells. CTL clone 4 did not the ORF3P peptide derived from the alternative open reading frame of the TRP-1 gene. These results demonstrated that TIL586-C1 specifically recognized the ORF3P peptide derived from TRP-1 and CTL clone 4 specifically recognized the peptide derived from TRP2. No cross reactivity was observed while both ORF3P and TRP2-pl97 were presented to T cells by HLA- A31 molecules.

Table 5. Identification of synthetic peptides with reactivity to T cell clones

Target cells pulsed GM-CSF release with peptide CTL clone 1 (TRP-1) CTL clone 4 (TRP-2)

586EBV + TRP 186 . 1W VWLHYYSVR(TRP-2) <50 <50

586EBV + FVWLHYYSVR(TRP-2) <50 <50

586EBV + TRP. M . J K RDTLLGPGR(TRP-2) <50 <50

586EBV + TRP., 7 . 205 LLGPGRPYR(TRP-2) <50 >4000

586EBV + TRP 2U . 2 -, GPAFVTWHR(TRP-2) <50 <50

586EBV + ORF3P MSLQRQFLR(TRP-1) > 8000 <50

1510EBV + LLGPGRPYR(TRP-2) <50 > 4000

1510EBV + ORF3P MSLQRQFLR(TRP-l) >6000 <50

T2 + TRPi . 3 (, j LLGPGRPYR(TRP-2) <50 <50

T2 + ORF3P MSLQRQFLR(TRP-l) <50 <50

586EBV + None <50 <50

586EBV + None >5000 > 3000

586EBV cells were incubated with individual peptides at a concentration of l ~g/ml for 90 min. GM-CSF release was measured after co-incubation of peptide-loaded 586EBV cells with T cell clones recognizing either TRP- 1 or TRP-2. GM-CSF secretion by T cells alone without stimulators was subtracted. 586EBV and 1510EBV were EBV transformed B cell lines expressing HLA-A3 1.

EXAMPLE 14 Characterization of TRP ιnLχβ peptide

Titration experiments demonstrated that 1 nM of peptide was sufficient to stimulate GM-CSF release from the T cell clone 4 and the stimulation reached a plateau at 500 nM (Fig. 10A). Lysis of 586EBV B cells pulsed with TRP 197 aoJ by CTL clone 4 was also determined at various peptide concentrations (Fig. 10B), and similar to cytokine release assays, lysis of target cells by the CTL clone 4 was detected at 1 nM peptide concentration. Maximum lysis was seen at 100 nM of peptide concentration. CTL clone 4 was capable of lysing 586EBV pulsed with TRP2-pl97 and 586mel tumor cells even at low E:T ratio, but failed to lyse 586EBV B cells alone or pulsed with the control peptide ORF3P nor the HLA-A31 negative 397mel line (Fig. 10).

The majority of human melanoma antigens identified to date are

non-mutated self-antigens and the T cell recognition and binding affinity of these self-peptides to the corresponding MHC molecules has in some instances been improved by substitution of amino acids at anchor residues. A number of synthetic peptides including 8mer or lOmer and modified peptides as indicated in Table 6 were made and tested for recognition by CTL clone 4 when pulsed onto 586EBV B cells. The lOmer TLLGPGRPYR, in which one amino acid was extended at the N-terminus of TRP^. aa , was still recognized by CTL clone 4 when pulsed on 586EBV B cells, but the reactivity was about 60% of the overlapping 9mer LLGPGRPYR. Based on the binding motif of HLA-A31 , a few modified peptides were generated with substitution of amino acids at anchor residues positions 2 and 9 as well as at other positions. The Arg residue at position 9 in the C-terminus could be substituted with the Lys residue and the modified peptide retained at least 60% of the activity of the parental peptide. Substitution of a Leu residue at position 2 with either Ser, He or Val residues retained the same activity or reduced the activity to 60% of the parental peptide while substitution with Ala or Phe at this position reduced the ability to stimulate cytokine release from T cells (Table 6). Other modifications provided variable cancer peptide activity in T cell recognition.

Table 6. Comparison of T cell reactivity of modified peptides

Target cells pulsed with peptides CTL clone 4

GM-CSF release

586EBV + LLGPGRPYR 3450 586EBV + TLLGPGRPYR 2100

586EBV + LLGPGRPYRA 545

586EBV + LGPGRPYR <50

586EBV + LIGPGRPYR 2545

586EBV + LVGPGRPYR 2100 586EBV + LSGPGRPYR 3300

586EBV + LΔGPGRPYR 550

586EBV + LFGPGRPYR <50

586EBV + LLGPGRPYR 2000

586EBV + LLGPGRPYH <50 586EBV + ALGPGRPYR < 50

586EBV + gLGPGRPYR <50

586EBV + KLGPGRPYR 420

586EBV + LLLPGRPYR < 50

586EBV + LLFPGRPYR < 50 586EBV + LLAPGRPYR <50

586EBV + LLGPGFPYR 738

586EBV + LLGPGAPYR <50

586EBV + LLGPGIPYR < 50

586EBV + LLGPGVPYR <50 586EBV + LLGPGKPYR 321

586EBV < 50

586mel > 3000 586EBV cells were incubated with individual peptides at a concentration of 0.5 —g/ml for 90 min. GM-CSF release was measured after co-incubation of peptide-loaded 586EBV cells with the CTL clone 4 cells. GM-CSF secretion by T cells alone without stimulators was subtracted. 586EBV was a EBV transformed B cell line expressing HLA- A31.

The TRP-2 protein contains two putative copper-binding sites, cysteine-rich regions and a transmembrane domain. Human TRP-2 has been mapped to chromosome 13 while the mouse counterpart has been mapped to chromosome 14 in the region of the coat color mutation, slaty. There is about a 40% amino acid sequence identity between TRP-2 and tyrosinase or TRP- l/gp75, but no CTL line or clone was found thus far that recognizes a common peptide epitope among the tyrosinase protein family.

The 9 mer TRP I972M peptide recognized by CTL clone 4 is located at one of the copper binding sites in the coding region of TRP-2. This peptide most efficiently stimulated cytokine release from T cells compared with other peptides including motified peptides tested in this study. This was in agreement with the predicted HLA- A31 binding motif, which indicates that Leu at position 2 and Arg at position 9 are the favorable residues. Although Leu at position 2 and Arg at position

9 could be replaced with He and Ser at position 2 and Lys at position 9, respectively, with little or minor loss of reactivity to T cell recognition, substitutions of amino acids at positions 1 , 3 or 6 led to some loss of reactivity.

EXAMPLE 15

In vivo Treatment Assay

For in vivo treatment, MHL-A31 4 transgenic mice are challenged with either 1 x IO 5 or 5 x 10 5 Trp-1 + B16 mouse melanoma cells intravenously in order to establish pulmonary metastases. Mice are subsequently vaccinated with a recombinant virus expressing cancer peptide, LLGPGRPYR at IO 5 PFU/mg body weight. Mice are euthanized on day 12 and the number of pulmonary metastases in vaccinated mice vs. non-vaccinated mice determined. EXAMPLE 16

Cancer Antigen Specific T Lymphocytes Immunotherapy

T-lymphocytes presensitized to a melanoma antigen may be effective in therapeutically treating mammals afflicted with a melanoma. T-lymphocytes are isolated from peripheral blood or melanoma tumor suspensions and cultured in vitro (Kawakami et al 1988, J. Exp. Med. 168:2183-2191).

The T lymphocytes are exposed to the cancer peptide, LLGPGRPYR

at a concentration of 1 μg/ml alone or in the presence of IL-2, resensitized and expanded in culture. T-lymphocytes exposed to the cancer peptide are administered to a mammal at about IO 9 to IO' 2 lymphocytes per mammal. The lymphocytes are administered either intravenously, intraperitoneally or intralesionally. The treatment may be administered concurrently with other therapeutic treatments such as cytokines, surgical excision of melanoma lesions and chemotherapeutic drugs.

EXAMPLE 17 Treatment of Patients with Metastatic Melanoma In this protocol, patients with advanced melanoma are immunized with an antigenic cancer epitope.

Patients eligible for the trial must have evidence of measurable or evaluable metastatic melanoma that has failed standard effective therapy. Patients must have tumors that express the TPI-2 antigen as evidenced by PCR or Northern Blot analysis of tumor cell RNA. Patients receive either 1 ng, 1 μg, 1 mg or 500 mg/kg body weight of a cancer peptide LLGPGRPYR via intravenously at day zero, day 7 and day 14 alone or in combination with IL-2 and/or a co-immunostimulatory molecule. Patients are evaluated for toxicity, immunologic effects and therapeutic efficacy.

Lymphocytes taken from the treated patients are tested for specific response to the cancer antigen comprising the amino acid sequence LLGPGRPYR.

A complete response is defined as the disappearance of all clinical evidence of disease that lasts at least four weeks. A partial response is a 50% or greater decrease in the sum of the products of the perpendicular diameter of all measurable lesions for at least four weeks with no appearance of new lesions or increase in any lesions. Minor responses are defined as 25-49% decrease in the sum of the products of the perpendicular diameters of all measurable lesions with no appearance of new lesions and no increase in any lesions. Any patient with less than a partial response is considered a non-responder. The appearance of new lesions or greater than 25% increase in the product of perpendicular diameters of prior lesions following a partial or complete response is considered as a relapse.

Discussion

Several antigenic T-cell epitopes derived from the normal open reading frame of the corresponding non-mutated shared melanoma antigens such as tyrosinase, MART-1/Melan-A and gplOO have been identified. In this study, it was demonstrated that the antigenic peptide recognized by TIL586 was derived from a second gene product of the gp75 gene. To our knowledge, this is the first example that T cells recognize an antigenic peptide resulting from the translation of an overlapping open reading frame of the same gene and the only example in eukaryotic cells that two completely different proteins and/or peptides can be translated from overlapping open reading frames of a single cellular gene. The ORF3 of the gp75 gene encodes a short protein of 24 amino acid. The antigenic peptide recognized by TIL586 is encoded by the sequence located immediately behind the ATG (294-296) start codon of the alternative open reading frame.

Although gp75 shares a 40-45% sequence homology to tyrosinase, gplOO and TRP-2, co-transfection of HLA-A31 and tyrosinase, gplOO or TRP-2 cDNAs, respectively, into COS-7 cells failed to stimulate GM-CSF release from T cell clones derived from TIL586. A database search did not reveal any proteins that had the HLA-A31 peptide binding motif and significant sequence homology to the peptide epitope recognized by TIL586 and its derived T cell clones. In addition, previous studies showed that melanoma transfectants (gp757A31 + ) conferred the ability to stimulate GM-CSF release from TIL586, but gp75 melanoma transfectants (gp75 /A31 + ) did not (Wang, R.F. et al 1995 J. Exp. Med. 181:799-804). Similar results were obtained with additional melanoma cell lines (gp757A31 + ). Since the ORF3P peptide was only epitope identified from the gp75 gene and was recognized by six T cell clones derived from TIL586, this peptide may be identical or similar to the naturally processed peptide on tumor cells and melanocytes. This was further supported by cold-target inhibition experiments since this peptide was capable of competing for T cell recognition with a natural peptide on tumor cells.

It was reported that T-cell epitope peptides derived from the frameshift of the mutated adenomatosis polyposis coli (APC) gene in colon cancer were recognized by CTLs generated from vaccinated BALB/c mice (Townsend, A. et al 1994 Nature 371:662). The results in Fig. 3 indicated that the ATG at nucleotides

294-296 was required for translation of the 24 amino acid product which, in turn, was processed to the antigenic peptide recognized by TIL586. TIL586 still recognized the mutated synthetic peptide pulsed on 586EBV B cells, but not the mutated gene when co-transfected into COS-7 cells with HLA- A31 cDNA, indicating that the loss of recognition of the mutated (ATG to ATC) gene by TIL586 resulted from elimination of translation of the ORF3 product. These results plus DNA sequence analysis ruled out the possibility that the antigenic peptide recognized by TIL586 was derived from the frameshift product of gp75. It is possible, therefore, that multiple peptides or proteins are often translated from overlapping open reading frames of a single eukaryotic gene, but that means to detect these alternate products have not been available. The exquisite sensitivity of T cells to detect naturally processed peptides may reveal many other examples of this phenomenon.

The mechanism by which the overlapping open reading frame 3 (ORF3) is translated in vivo is currently unclear. Although examples have been reported of cellular mRNAs that initiate at more than one AUG codon, and that, in some rare cases, initiate at both AUG and non-AUG codons such as CUG to generate N- terminally extended identical sequences, the use of overlapping open reading frames (i.e. translating two completely dissimilar peptides) from a single eukaryotic cellular mRNA has never been described to our knowledge. Several examples of translation of overlapping reading frames from a single mRNA have been described, but exclusively limited to viral genes. The detection of the products of overlapping reading frames in viral genes have been possible because of the existence of reactive antibodies in the sera of virally infected hosts. In the present invention a T cell assay was used to identify the epitope peptide recognized by T cells. This approach is very different from and more sensitive than conventional Western blots or immunoprecipitation analyses. Although there are five ATG codons between the authentic start codon and the start codon of ORF3, the construct pPCRHO covering the N-terminal part of ORFl (gp75), the entire ORF2 and ORF3 (nucleotides 1-667) still retained the ability to stimulate cytokine release from TIL586. The level of stimulation, however, was several fold lower than that stimulated by the 5' truncated (lacking the first 246 nucleotides) form of gp75 (Fig. 1 A and IB), suggesting that the upstream ATG codons may have partially inhibited the expression of ORF3. Several

factors have made it possible to detect the expression of the ORF3 product in this system. First, the upstream ATG codons proceeding the ATG start codon of ORF3 did not appear to be in the optimal context, which may allow to use the downstream ATG as start codons by the leaky scanning model. Second, the relative high expression of transfected genes in COS-7 cells and the availability of the T cell assay as an extremely sensitive means may allow detection of very low levels of the translated products.

Interestingly, the ORF3 product was detected by T cells in the tumor cells as well as normal melanocytes (Figures 6A-6D), strongly suggesting that the ORF3 protein was not a gene product resulted from genetic alterations in tumor cells. In the previous studies, it was shown that TIL586 recognized multiple tumor cell lines (gp75 + /A3T) tested, suggesting that TIL586 recognizes a non-mutated, shared tumor antigen (Wang R.F. et al, ibid). Since the gp75 gene is highly expressed in melanomas based on Northern blot and PCR analyses (Wang R.F. et al, ibid) and its gene product gp75 protein is the most abundant intracellular glycoprotein expressed in melanoma cells and melanocytes (Tai, T. , Eisinger, M., Ogata, S. and Lloyd, K. O, 1983, Cancer Res. 43: 2773-2779; Thomson, T. N., Mattes, J. M., Roux, L., Old, L. J. and Lloyd, K. O. 1985, J. Invest. Dermat. 85, 169-174; Thomson, T. N. , real, F. X., Mutakami, S. , Cordon-cardo, C, Old, L. J. and Houghton, A. N. 1988, J. Invest. Dermat. 90, 459-466), it is not surprising that the T cell clones recognized the ORF3P peptide when pulsed onto 586EBV B cells (A31 + ), melanoma (gp75 + /A31 + ) as well as A31 + melanocytes, but not gp75V A31 + melanoma cells or ORF3P pulsed on non-A31 T2 cells. Another possibility to explain the peptide expression in tumor cells and melanocytes is that the ORF3 may be translated from a separate mRNA transcript(s) generated by an alternative splicing of gp75 mRNA or a different promoter. To our knowledge, mRNA transcripts generated by alternative splicing are translated into isoform proteins by use of the same open reading frames. In our case, however, if a separate transcript was generated and used as a template for translation, but a completely different open reading frame relative to gp75 was used to translate ORF3 product. Further experiments are needed to clarify the mechanisms for the translation of the ORF3 protein in vivo. Nevertheless, these possibilities mentioned above are not mutually exclusive.

SEQUENCE LISTING

(1) GENERAL INFORMATION:

(i) APPLICANTS: THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY OF HEALTH AND HUMAN SERVICES

(ii) TITLE OF INVENTION: NOVEL HUMAN CANCER

ANTIGEN OF TYROSINASE-RELATED PROTEIN 1 AND 2 AND GENES ENCODING SAME

(iii) NUMBER OF SEQUENCES: 60

(iv) CORRESPONDENCE ADDRESS:

(A) ADDRESSEE: MORGAN & FINNEGAN, L.L.P.

(B) STREET: 345 PARK AVENUE

(C) CITY: NEW YORK

(D) STATE: NEW YORK

(E) COUNTRY: USA

(F) ZIP: 10154

(V) COMPUTER READABLE FORM:

(A) MEDIUM TYPE: 3.5-INCH, 1.44MB STORAGE

(B) COMPUTER: IBM PC COMPATIBLE

(C) OPERATING SYSTEM: PC-DOS/MS-DOS

(D) SOFTWARE: WORDPERFECT 5.1

(vi) CURRENT APPLICATION DATA:

(A) FILING DATE: 06-FEB-1997

(vii) PRIOR APPLICATION DATA:

(A) APPLICATION NUMBER: 08/599,602 (A) FILING DATE: 09-FEB-1996

(vii) PRIOR APPLICATION DATA:

(A) APPLICATION NUMBER: 08/725,736 (A) FILING DATE: 04-OCT-1996

(Viii) ATTORNEY/AGENT INFORMATION:

(A) NAME: KATHRYN M. BROWN

(B) REGISTRATION NUMBER: 34,556

(C) REFERENCE/DOCKET NUMBER: 2026-4209PC

(ix) TELECOMMUNICATION INFORMATION:

(A) TELEPHONE: (212) 758-4800

(B) TELEFAX: (212) 751-6849

(C) TELEX: 421792

(2) INFORMATION FOR SEQ ID NO:l:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 471 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(X) PUBLICATION INFORMATION:

(A) AUTHORS: COHEN, T. ; MULLE , R.M. ; TOMITA, Y.; SHIBAHARA, S.

(B) TITLE: NUCLEOTIDE SEQUENCE OF THE cDNA ENCODING HUMAN TYROSINASE-RELATED PROTEIN

(C) JOURNAL: NUCLEIC ACIDS RESEARCH

(D) VOLUME: 18

(E) ISSUE: 9

(F) PAGES: 2807-2808

(G) DATE: 11 MAY 1990

(H) RELEVANT RESIDUES IN SEQ ID NO:l: FROM 1 TO 471

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:

ATG AGT GCT CCT AAA CTC CTC TCT CTG GGC TGT ATC 36

Met Ser Ala Pro Lys Leu Leu Ser Leu Gly Cys lie 1 5 10

TTC TTC CCC TTG CTA CTT TTT CAG CAG GCC CGG GCT 72

Phe Phe Pro Leu Leu Leu Phe Gin Gin Ala Arg Ala 15 20

CAA TTC CCA AGA CAG TGT GCC ACT GTT GAG GCT TTG 108 Gin Phe Pro Arg Gin Cys Ala Thr Val Glu Ala Leu 25 30 35

AGA AGT GGT ATG TGT TGC CCA GAC CTG TCC CCT GTG 144 Arg Ser Gly Met Cys Cys Pro Asp Leu Ser Pro Val 40 45

TCT GGG CCT GGG ACA GAC CGC TGT GGC TCA TCA TCA 180 Ser Gly Pro Gly Thr Asp Arg Cys Gly Ser Ser Ser 50 55 60

GGG AGG GGC AGA TGT GAG GCA GTG ACT GCA GAC TCC 216 Gly Arg Gly Arg Cys Glu Ala Val Thr Ala Asp Ser

65 70

CGG CCC CAC AGC CCT CAG TAT CCC CAT GAT GGC AGA 252 Arg Pro His Ser Pro Gin Tyr Pro His Asp Gly Arg 75 80

GAT GAT CGG GAG GTC TGG CCC TTG CGC TTC TTC AAT 288 Asp Asp Arg Glu Val Trp Pro Leu Arg Phe Phe Asn 85 90 95

AGG ACA TGT CAC TGC AAC GGC AAT TTC TCA GGA CAC 324

Arg Thr Cys His Cys Asn Gly Asn Phe Ser Gly His

100 105

AAC TGT GGG ACG TGC CGT CCT GGC TGG AGA GGA GCT 360

Asn Cys Gly Thr Cys Arg Pro Gly Trp Arg Gly Ala

110 115 120

GCC TGT GAC CAG AGG GTT CTC ATA GTC AGG AGA AAT 396 Ala Cys Asp Gin Arg Val Leu lie Val Arg Arg Asn

125 130

CTT CTG GAC TTA AGT AAA GAA GAA AAG AAC CAC TTT 432 Leu Leu Asp Leu Ser Lys Glu Glu Lys Asn His Phe 135 140

GTC CGG GCC CTG GAT ATG GCA AAG CGC ACA ACT CAC 468 Val Arg Ala Leu Asp Met Ala Lys Arg Thr Thr His 145 150 155

CCT 471 Pro

(2) INFORMATION FOR SEQ ID NO:2:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 157 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PROTEIN

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:

Met Ser Ala Pro Lys Leu Leu Ser Leu Gly Cys lie 1 5 10

Phe Phe Pro Leu Leu Leu Phe Gin Gin Ala Arg Ala 15 20

Gin Phe Pro Arg Gin Cys Ala Thr Val Glu Ala Leu 25 30 35

Arg Ser Gly Met Cys Cys Pro Asp Leu Ser Pro Val 40 45

Ser Gly Pro Gly Thr Asp Arg Cys Gly Ser Ser Ser 50 55 60

Gly Arg Gly Arg Cys Glu Ala Val Thr Ala Asp Ser

65 70

Arg Pro His Ser Pro Gin Tyr Pro His Asp Gly Arg 75 80

Asp Asp Arg Glu Val Trp Pro Leu Arg Phe Phe Asn 85 90 95

Arg Thr Cys His Cys Asn Gly Asn Phe Ser Gly His 100 105

Asn Cys Gly Thr Cys Arg Pro Gly Trp Arg Gly Ala 110 115 120

Ala Cys Asp Gin Arg Val Leu lie Val Arg Arg Asn

125 130

Leu Leu Asp Leu Ser Lys Glu Glu Lys Asn His Phe 135 140

Val Arg Ala Leu Asp Met Ala Lys Arg Thr Thr His 145 150 155

Pro

(2) INFORMATION FOR SEQ ID NO:3:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 129 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: YES

(iv) ANTI-SENSE: NO

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:

ATG ATC GGG AGG TCT GGC CCT TGC GCT TCT TCA ATA 36

Met lie Gly Arg Ser Gly Pro Cys Ala Ser Ser lie 1 5 10

GGA CAT GTC ACT GCA ACG GCA ATT TCT CAG GAC ACA 72 Gly His Val Thr Ala Thr Ala lie Ser Gin Asp Thr 15 20

ACT GTG GGA CGT GCC GTC CTG GCT GGA GAG GAG CTG 108 Thr Val Gly Arg Ala Val Leu Ala Gly Glu Glu Leu 25 30 35

CCT GTG ACC AGA GGG TTC TCA 129

Pro Val Thr Arg Gly Phe Ser 40

(2) INFORMATION FOR SEQ ID NO:4:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 43 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PROTEIN

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:

Met lie Gly Arg Ser Gly Pro Cys Ala Ser Ser lie 1 5 10

Gly His Val Thr Ala Thr Ala lie Ser Gin Asp Thr 15 20

Thr Val Gly Arg Ala Val Leu Ala Gly Glu Glu Leu 25 30 35

Pro Val Thr Arg Gly Phe Ser 40

(2) INFORMATION FOR SEQ ID NO:5:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 72 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: YES

(iv) ANTI-SENSE: NO

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:

ATG TCA CTG CAA CGG CAA TTT CTC AGG ACA CAA CTG 36 Met Ser Leu Gin Arg Gin Phe Leu Arg Thr Gin Leu 1 5 10

TGG GAC GTG CCG TCC TGG CTG GAG AGG AGC TGC CTG 72 Trp Asp Val Pro Ser Trp Leu Glu Arg Ser Cys Leu 15 20

(2) INFORMATION FOR SEQ ID NO:6:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:

Met Ser Leu Gin Arg Gin Phe Leu Arg Thr Gin Leu 1 5 10

Trp Asp Val Pro Ser Trp Leu Glu Arg Ser Cys Leu 15 20

(2) INFORMATION FOR SEQ ID NO:7:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(ix) FEATURE:

(A) NAME/KEY: XAA

(B) LOCATION: 1 TO 2

(C) IDENTIFICATION METHOD: BY EXPERIMENT

(D) OTHER INFORMATION: XAA = SER OR ALA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:

Met Xaa Leu Gin Arg Gin Phe Leu Arg Thr Gin Leu 1 5 10

Trp Asp Val Pro Ser Trp Leu Glu Arg Ser Cys Leu 15 20

(2) INFORMATION FOR SEQ ID NO:8:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(ix) FEATURE:

(A) NAME/KEY: XAA

(B) LOCATION: 1 TO 2

(C) IDENTIFICATION METHOD: BY EXPERIMENT

(D) OTHER INFORMATION: XAA = SER OR ALA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:

Met Xaa Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:9:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:

Met Ser Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:10:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 27 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10: ATGTCACTGC AACGGCAATT TCTCAGG 27

(2) INFORMATION FOR SEQ ID NO:11:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 66 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: CDNA

(iii) HYPOTHETICAL: YES

(iv) ANTI-SENSE: NO

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: GCCATGGGCA GAGATGATCG GGAGGTCTGG CCCTTGCGCT 40

TCTTCAATAG GACATCTCAC TGCAAC 66

(2) INFORMATION FOR SEQ ID NO:12:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 37 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: CDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: GAAGATCTGC CATGGGCAGA GATGATCGGG AGGTCTG 37

(2) INFORMATION FOR SEQ ID NO:13:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 31 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: CDNA

(iii) HYPOTHETICAL: YES

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:

GAATTCGTTG TGTCCTGAGA AATTGCCGTT G 31

(2) INFORMATION FOR SEQ ID NO:14:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 33 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: GAATTCGACT ATGAGAACCC TCTGGTCACA GGC 33

(2) INFORMATION FOR SEQ ID NO:15:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 32 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:

AAGATCTGGG CCCGGACAAA GTGGTTCTTT TC 32

(2) INFORMATION FOR SEQ ID NO:16:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 7 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16: GATATGG

(2) INFORMATION FOR SEQ ID NO:17:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:

Asp Asp Arg Glu Val Trp Pro Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:18:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:

Val Trp Pro Leu Arg Phe Phe Asn Arg

1 5

(2) INFORMATION FOR SEQ ID NO:19:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:

Ser Gly His Asn Cys Gly Thr Cys Arg 1 5

(2) INFORMATION FOR SEQ ID NO:20:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20.

Cys Gly Thr Cys Arg Pro Gly Trp Arg 1 5

(2) INFORMATION FOR SEQ ID NO:21:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 10 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:21:

Ala Cys Asp Gin Arg Val Leu lie Val Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:22:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 11 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:

Ala Cys Asp Gin Arg Val Leu lie Val Arg Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:23:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 10 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23:

Leu Trp Asp Val Pro Ser Trp Leu Glu Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:24:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 10 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:

Ala lie Ser Gin Asp Thr Thr Val Gly Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:25:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 11 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:

Ala Ala Cys Asp Gin Arg Val Leu lie Val Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:26:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:26:

Asp Gin Arg Val Leu lie Val Arg Arg 1 5

(2) INFORMATION FOR SEQ ID NO:27:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 11 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:27: lie Val Arg Arg Asn Leu Leu Asp Leu Ser Lys 1 5 10

(2) INFORMATION FOR SEQ ID NO:28:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 11 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:28:

Leu Ser Lys Glu Glu Lys Asn His Phe Val Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:29:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 7 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: YES

(iv) ANTI-SENSE: NO

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:29: GAGATGA

(2) INFORMATION FOR SEQ ID NO:30:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 7 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: CDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30: GACATGT

(2) INFORMATION FOR SEQ ID NO:31:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31: lie Ser Gin Asp Thr Thr Val Gly Arg 1 5

(2) INFORMATION FOR SEQ ID NO:32:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:32:

Ala Gly Glu Glu Leu Pro Val Thr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:33:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 10 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:33:

Met Ser Leu Gin Arg Gin Phe Leu Arg Thr 1 5 10

(2) INFORMATION FOR SEQ ID NO:34:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:34:

Ser Leu Gin Arg Gin Phe Leu Arg Thr 1 5

(2) INFORMATION FOR SEQ ID NO:35:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:35:

Met Ala Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:36:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:36:

Met Leu Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:37:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:37:

Met Arg Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:38:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:38

Met Ser Leu Gin Arg Leu Phe Leu Arg

1 5

(2) INFORMATION FOR SEQ ID NO:39:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 39:

Met Ser Leu Gin Arg Gin Phe Leu Glu 1 5

(2) INFORMATION FOR SEQ ID NO:40:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:40:

Met Ser Leu Gin Arg Gin Phe Leu Lys 1 5

(2) INFORMATION FOR SEQ ID NO:41:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9 AMINO ACIDS

(B) TYPE: AMINO ACID (D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: PEPTIDE

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:41: lie Ser Leu Gin Arg Gin Phe Leu Arg 1 5

(2) INFORMATION FOR SEQ ID NO:42:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 32 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42: AGAATGAGTG CTCCTAAACT CCTCTCTCTG GG 32

(2) INFORMATION FOR SEQ ID NO:43:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:43: CATGTGAGAA AAGCTGGTCC CTCA 24

(2) INFORMATION FOR SEQ ID NO:44:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:44: TGGATATGGC AAAGCGCACA ACTC 24

(2) INFORMATION FOR SEQ ID NO:45:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 30 BASE PAIRS

(B) TYPE: NUCLEIC ACID

(C) STRANDNESS: SINGLE

(D) TOPOLOGY: LINEAR

(ii) MOLECULE TYPE: CDNA (iii) HYPOTHETICAL: YES (iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:45: TAAATGGAAA TGTTCTCAAA TTGTGGCGTG 30

(2) INFORMATION FOR SEQ ID NO:46:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 2291

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: DOUBLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: cDNA

(X) PUBLICATION INFORMATION:

(A) AUTHORS: YOKAYAMA, ET AL.

(B) TITLE: MOLECULAR CLONING AND FUNCTIONAL ANALYSIS OF A cDNA CODING FOR HUMAN DOPACHROME TAUTOMERASE/TYROSINASE- RELATED PROTEIN-2.

(C) JOURNAL: BIOCHIM. BIOPHSY. ACTA.

(D) VOLUME: 1217

(E) ISSUE:

(F) PAGES: 317-321

(G) DATE: 1994

(xi) SEQUENCE DESCRIPTION:SEQ ID NO:46: GCAATTAAAG TCAAGAGCTA AGGAGGGAGG GAGAGGGTTT 40

AGAAATACCA GCATAATAAG TAGTATGACT GGGTGCTCTG 80 TAAATTAACT CAATTAGACA AAGCCTGACT TAACGGGGGA 120 AGATGGTGAG AAGCGCTACC CTCATTAAAT TTGGTTGTTA 160 GAGGCGCTTC TAAGGAAATT AAGTCTGTTA GTTGTTTGAA 200 TCACATAAAA TTGTGTGTGC ACGTTCATGT ACACATGTGC 240 ACACATGTAA CCTCTGTGAT TCTTGTGGGT ATTTTTTTAA 280 GAAGAAAGGA ATAGAAAGCA AAGAAAAATA AAAAATACTG 320 AAAAGAAAAG ACTGAAAGAG TAGAAGATAA GGAGAAAAGT 360 ACGACAGAGA CAAGGAAAGT AAGAGAGAGA GAGAGCTCTC 400 CCAATTATAA AGCCATGAGC CCCCTTTGGT GGGGGTTTCT 440 GCTCAGTTGC TTGGGCTGCA AAATCCTGCC AGGAGCCCAG 480 GGTCAGTTCC CCCGAGTCTG CATGACGGTG GACAGCCTAG 520 TGAACAAGGA GTGCTGCCCA CGCCTGGGTG CAGAGTCGGC 560 CAATGTCTGT GGCTCTCAGC AAGGCCGGGG GCAGTGCACA 600 GAGGTGCGAG CCGACACAAG GCCCTGGAGT GGTCCCTACA 640 TCCTACGAAA CCAGGATGAC CGTGAGCTGT GGCCAAGAAA 680 ATTCTTCCAC CGGACCTGCA AGTGCACAGG AAACTTTGCC 720 GGCTATAATT GTGGAGACTG CAAGTTTGGC TGGACCGGTC 760 CCAACTGCGA GCGGAAGAAA CCACCAGTGA TTCGGCAGAA 800 CATCCATTCC TTGAGTCCTC AGGAAAGAGA GCAGTTCTTG 840 GGCGCCTTAG ATCTCGCGAA GAAGAGAGTA CACCCCGACT 880 ACGTGATCAC CACACAACAC TGGCTGGGCC TGCTTGGGCC 920 CAATGGAACC CAGCCGCAGT TTGCCAACTG CAGTGTTTAT 960 GATTTTTTTG TGTGGCTCCA TTATTATTCT GTTAGAGATA 1000 CATTATTAGG ACCAGGACGC CCCTACAGGG CCATAGATTT 1040 CTCACATCAA GGACCTGCAT TTGTTACCTG GCACCGGTAC 1080 CATTTGTTGT GTCTGGAAAG AGATCTCCAG CGACTCATTG 1120 GCAATGAGTC TTTTGCTTTG CCCTACTGGA ACTTTGCCAC 1160 TGGGAGGAAC GAGTGTGATG TGTGTACAGA CCAGCTGTTT 1200 GGGGCAGCGA GACCAGACGA TCCGACTCTG ATTAGTCGGA 1240 ACTCAAGATT CTCCAGCTGG GAAACTGTCT GTGATAGCTT 1280 GGATGACTAC AACCACCTGG TCACCTTGTG CAATGGAACC 1320 TATGAAGGTT TGCTGAGAAG AAATCAAATG GGAAGAAACA 1360 GCATGAAATT GCCAACCTTA AAAGACATAC GAGATTGCCT 1400 GTCTCTCCAG AAGTTTGACA ATCCTCCCTT CTTCCAGAAC 1440 TCTACCTTCA GTTTCAGGAA TGCTTTGGAA GGGTTTGATA 1480 AAGCAGATGG GACTCTGGAT TCTCAAGTGA TGAGCCTTCA 1520 TAATTTGGTT CATTCCTTCC TGAACGGGAC AAACGCTTTG 1560 CCACATTCAG CCGCCAATGA TCCCATTTTT GTGGTTCTTC 1600 ATTCCTTTAC TGATGCCATC TTTGATGAGT GGATGAAAAG 1640 ATTTAATCCT CCTGCAGATG CCTGGCCTCA GGAGCTGGCC 1680 CCTATTGGTC ACAATCGGAT GTACAACATG GTTCCTTTCT 1720 TCCCTCCAGT GACTAATGAA GAACTCTTTT TAACCTCAGA 1760 CCAACTTGGC TACAGCTATG CCATCGATCT GCCAGTTTCA 1800 GTTGAAGAAA CTCCAGGTTG GCCCACAACT CTCTTAGTAG 1840 TCATGGGAAC ACTGGTGGCT TTGGTTGGTC TTTTTGTGCT 1880 GTTGGCTTTT CTTCAATATA GAAGACTTCG AAAAGGATAT 1920 ACACCCCTAA TGGAGACACA TTTAAGCAGC AAGAGATACA 1960 CAGAAGAAGC CTAGGGTGCT CATGCCTTAC CTAAGAGAAG 2000 AGGCTGGCCA AGCCACAGTT CTGACGCTGA CAATAAAGGA 2040 ACTAATCCTC ACTGTTCCTT CTTGAGTTGA AGATCTTTGA 2080 CATAGGTTCT TCTATAGTGA TGATGATCTC ATTCAGAAGA 2120 TGCTTAGCTG TAGTTTCCGC TTTGCTTGCT TGTTTAACAA 2160 ACCCAACTAA AGTGCTTGAG GCTACCTCTA CCTTCAAATA 2200

AAGATAGACC TGACAATTTG TGATATCTAA TAATAACCCC 2240

CCCCCCAATA TTGATTAAGC CTCCTCCTTT TCTGAAAGCA 2280

TTTAAAAAAA A 2291

(2) INFORMATION FOR SEQ ID NO:47:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 519

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PROTEIN

(X) PUBLICATION INFORMATION:

(A) AUTHORS: YOKAYAMA, ET AL.

(B) TITLE: MOLECULAR CLONING AND FUNCTIONAL ANALYSIS OF A cDNA CODING FOR HUMAN DOPACHROME TAUTOMERASE/TYROSINASE- RELATED PROTEIN-2.

(C) JOURNAL: BIOCHIM. BIOPHSY. ACTA.

(D) VOLUME: 1217

(E) ISSUE:

(F) PAGES: 317-321

(G) DATE: 1994

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:47:

Met Ser Pro Leu Trp Trp Gly Phe Leu Leu Ser Cys

1 5 10

Leu Gly Cys Lys lie Leu Pro Gly Ala Gin Gly Gin

15 20

Phe Pro Arg Val Cys Met Thr Val Asp Ser Leu Val

25 30 35

Asn Lys Glu Cys Cys Pro Arg Leu Gly Ala Glu Ser

40 45

Ala Asn Val Cys Gly Ser Gin Gin Gly Arg Gly Gin 50 55 60

Cys Thr Glu Val Arg Ala Asp Thr Arg Pro Trp Ser

65 70

Gly Pro Tyr lie Leu Arg Asn Gin Asp Asp Arg Glu

75 80

Leu Trp Pro Arg Lys Phe Phe His Arg Thr Cys Lys

85 90 95

Cys Thr Gly Asn Phe Ala Gly Tyr Asn Cys Gly Asp

100 105

Cys Lys Phe Gly Trp Thr Gly Pro Asn Cys Glu Arg 110 115 120

Lys Lys Pro Pro Val lie Arg Gin Asn lie His Ser

125 130

Leu Ser Pro Gin Glu Arg Glu Gin Phe Leu Gly Ala 135 140

Leu Asp Leu Ala Lys Lys Arg Val His Pro Asp Tyr

145 150 155

Val He Thr Thr Gin His Trp Leu Gly Leu Leu Gly 160 165

Pro Asn Gly Thr Gin Pro Gin Phe Ala Asn Cys Ser 170 175 180

Val Tyr Asp Phe Phe Val Trp Leu His Tyr Tyr Ser 185 190

Val Arg Asp Thr Leu Leu Gly Pro Gly Arg Pro Tyr

195 200

Arg Ala He Asp Phe Ser His Gin Gly Pro Ala Phe

205 210 215

Val Thr Trp His Arg Tyr His Leu Leu Cys Leu Glu 220 225

Arg Asp Leu Gin Arg Leu He Gly Asn Glu Ser Phe 230 235 240

Ala Leu Pro Tyr Trp Asn Phe Ala Thr Gly Arg Asn 245 250

Gl Cys Asp Val Cys Thr Asp Gin Leu Phe Gly Ala 255 260

Ala Arg Pro Asp Asp Pro Thr Leu He Ser Arg Asn

265 270 275

Ser Arg Phe Ser Ser Trp Glu Thr Val Cys Asp Ser 280 285

Leu Asp Asp Tyr Asn His Leu Val Thr Leu Cys Asn 290 295 300

Gly Thr Tyr Glu Gly Leu Leu Arg Arg Asn Gin Met 305 310

Gly Arg Asn Ser Met Lys Leu Pro Thr Leu Lys Asp

315 320 lie Arg Asp Cys Leu Ser Leu Gin Lys Phe Asp Asn

325 330 335

Pro Pro Phe Phe Gin Asn Ser Thr Phe Ser Phe Arg 340 345

Asn Ala Leu Glu Gly Phe Asp Lys Ala Asp Gly Thr 350 355 360

Leu Asp Ser Gin Val Met Ser Leu His Asn Leu Val 365 370

His Ser Phe Leu Asn Gly Thr Asn Ala Leu Pro His 375 380

Ser Ala Ala Asn Asp Pro He Phe Val Val Leu His

385 390 395

Ser Phe Thr Asp Ala He Phe Asp Glu Trp Met Lys 400 405

Arg Phe Asn Pro Pro Ala Asp Ala Trp Pro Gin Glu 410 415 420

Leu Ala Pro He Gly His Asn Arg Met Tyr Asn Met 425 430

Val Pro Phe Phe Pro Pro Val Thr Asn Glu Glu Leu

435 440

Phe Leu Thr Ser Asp Gin Leu Gly Tyr Ser Tyr Ala

445 450 455

He Asp Leu Pro Val Ser Val Glu Glu Thr Pro Gly 460 465

Trp Pro Thr Thr Leu Leu Val Val Met Gly Thr Leu

470 475 480

Val Ala Leu Val Gly Leu Phe Val Leu Leu Ala Phe

485 490

Leu Gin Tyr Arg Arg Leu Arg Lys Gly Tyr Thr Pro

495 500

Leu Met Glu Thr His Leu Ser Ser Lys Arg Tyr Thr 505 510 515

Glu Glu Ala

(2) INFORMATION FOR SEQ ID NO:48:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 11

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(ix) FEATURE:

(A) NAME/KEY: TRP-2 PEPTIDE

(B) LOCATION: 1 to 11

(C) IDENTIFICATION METHOD: By Experiment

(D) OTHER INFORMATION: WHEREIN Xaa CAN BE THE SAME OR DIFFERENT AND MAY BE ONE OR MORE AMINO ACIDS.

(xi) SEQUENCE DESCRIPTION:SEQ ID NO:48:

Xaa Leu Leu Gly Pro Gly Arg Pro Tyr Arg Xaa 1 5 10

(2) INFORMATION FOR SEQ ID NO:49:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:49:

Leu Leu Gly Pro Gly Arg Pro Tyr Arg 1 5

( 2 ) INFORMATION FOR SEQ ID NO : 50 :

( i) SEQUENCE CHARACTERISTICS :

(A) LENGTH: 10

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(xi) SEQUENCE DESCRIPTION:SEQ ID NO:50:

Thr Leu Leu Gly Pro Gly Arg Pro Tyr Arg 1 5 10

(2) INFORMATION FOR SEQ ID NO:51:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 10

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:51:

Leu Leu Gly Pro Gly Arg Pro Tyr Arg Ala 1 5 10

(2) INFORMATION FOR SEQ ID NO:52:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:52:

Leu He Gly Pro Gly Arg Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:53:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(xi) SEQUENCE DESCRIPTION:SEQ ID NO:53:

Leu Val Gly Pro Gly Arg Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:54:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:54:

Leu Ser Gly Pro Gly Arg Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:55:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:55:

Leu Ala Gly Pro Gly Arg Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:56:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:56:

Leu Leu Gly Pro Gly Arg Pro Tyr Lys 1 5

(2) INFORMATION FOR SEQ ID NO:57:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:57:

Lys Leu Gly Pro Gly Arg Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:58:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(xi) SEQUENCE DESCRIPTION:SEQ ID NO:58:

Leu Leu Gly Pro Gly Phe Pro Tyr Arg 1 5

(2) INFORMATION FOR SEQ ID NO:59:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 9

(B) TYPE: AMINO ACID

(C) STRANDEDNESS: SINGLE

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: PEPTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:59:

Leu Leu Gly Pro Gly Lys Pro Tyr Arg l 5

(2) INFORMATION FOR SEQ ID NO:60:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 27

(B) TYPE: NUCLEIC ACID

(C) STRANDEDNESS: UNKNOWN

(D) TOPOLOGY: UNKNOWN

(ii) MOLECULE TYPE:

(A) DESCRIPTION: OLIGONUCLEOTIDE

(Xi) SEQUENCE DESCRIPTION:SEQ ID NO:60:

TTATTAGGAC CAGGACGCCC CTACAGG 27