NUCLEIC ACID AND CORRESPONDING PROTEIN ENTITLED 162P1E6 USEFUL IN TREATMENT AND DETECTION OF CANCER

Abstract
A novel gene (designated 125P5C8) and its encoded protein, and variants thereof, are described wherein 125P5C8 exhibits tissue specific expression in normal adult tissue, and is aberrantly expressed in the cancers listed in Table I. Consequently, 125P5C8 provides a diagnostic, prognostic, prophylactic and/or therapeutic target for cancer. The 125P5C8 gene or fragment thereof, or its encoded protein, or variants thereof, or a fragment thereof, can be used to elicit a humoral or cellular immune response; antibodies or T cells reactive with 125P5C8 can be used in active or passive immunization.
Description
STATEMENT OF RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH

Not applicable.


REFERENCE TO SEQUENCE LISTING SUBMITTED VIA EFS-WEB

The content of the following electronic submission of the sequence listing via the USPTO EFS-WEB server, as authorized and set forth in MPEP §1730 II.B.2(a)(C), is incorporated herein by reference in its entirety for all purposes. The sequence listing is identified on the electronically filed text file as follows:

















File Name
Date of Creation
Size (bytes)









511582003513Seqlist.txt
Jan. 25, 2010
123,621 bytes










FIELD OF THE INVENTION

The invention described herein relates to a gene and its encoded protein, termed 125P5C8, expressed in certain cancers, and to diagnostic and therapeutic methods and compositions useful in the management of cancers that express 125P5C8.


BACKGROUND OF THE INVENTION

Cancer is the second leading cause of human death next to coronary disease. Worldwide, millions of people die from cancer every year. In the United States alone, as reported by the American Cancer Society, cancer causes the death of well over a half-million people annually, with over 1.2 million new cases diagnosed per year. While deaths from heart disease have been declining significantly, those resulting from cancer generally are on the rise. In the early part of the next century, cancer is predicted to become the leading cause of death.


Worldwide, several cancers stand out as the leading killers. In particular, carcinomas of the lung, prostate, breast, colon, pancreas, and ovary represent the primary causes of cancer death. These and virtually all other carcinomas share a common lethal feature. With very few exceptions, metastatic disease from a carcinoma is fatal. Moreover, even for those cancer patients who initially survive their primary cancers, common experience has shown that their lives are dramatically altered. Many cancer patients experience strong anxieties driven by the awareness of the potential for recurrence or treatment failure. Many cancer patients experience physical debilitations following treatment. Furthermore, many cancer patients experience a recurrence.


Worldwide, prostate cancer is the fourth most prevalent cancer in men. In North America and Northern Europe, it is by far the most common cancer in males and is the second leading cause of cancer death in men. In the United States alone, well over 30,000 men die annually of this disease—second only to lung cancer. Despite the magnitude of these figures, there is still no effective treatment for metastatic prostate cancer. Surgical prostatectomy, radiation therapy, hormone ablation therapy, surgical castration and chemotherapy continue to be the main treatment modalities. Unfortunately, these treatments are ineffective for many and are often associated with undesirable consequences.


On the diagnostic front, the lack of a prostate tumor marker that can accurately detect early-stage, localized tumors remains a significant limitation in the diagnosis and management of this disease. Although the serum prostate specific antigen (PSA) assay has been a very useful tool, however its specificity and general utility is widely regarded as lacking in several important respects.


Progress in identifying additional specific markers for prostate cancer has been improved by the generation of prostate cancer xenografts that can recapitulate different stages of the disease in mice. The LAPC (Los Angeles Prostate Cancer) xenografts are prostate cancer xenografts that have survived passage in severe combined immune deficient (SCID) mice and have exhibited the capacity to mimic the transition from androgen dependence to androgen independence (Klein et al., 1997, Nat. Med. 3:402). More recently identified prostate cancer markers include PCTA-1 (Su et al., 1996, Proc. Natl. Acad. Sci. USA 93: 7252), prostate-specific membrane (PSM) antigen (Pinto et al., Clin Cancer Res 1996 September 2 (9): 1445-51), STEAP (Hubert, et al., Proc Natl Acad Sci USA. 1999 Dec. 7; 96(25): 14523-8) and prostate stem cell antigen (PSCA) (Reiter et al., 1998, Proc. Natl. Acad. Sci. USA 95: 1735).


While previously identified markers such as PSA, PSM, PCTA and PSCA have facilitated efforts to diagnose and treat prostate cancer, there is need for the identification of additional markers and therapeutic targets for prostate and related cancers in order to further improve diagnosis and therapy.


Renal cell carcinoma (RCC) accounts for approximately 3 percent of adult malignancies. Once adenomas reach a diameter of 2 to 3 cm, malignant potential exists. In the adult, the two principal malignant renal tumors are renal cell adenocarcinoma and transitional cell carcinoma of the renal pelvis or ureter. The incidence of renal cell adenocarcinoma is estimated at more than 29,000 cases in the United States, and more than 11,600 patients died of this disease in 1998. Transitional cell carcinoma is less frequent, with an incidence of approximately 500 cases per year in the United States.


Surgery has been the primary therapy for renal cell adenocarcinoma for many decades. Until recently, metastatic disease has been refractory to any systemic therapy. With recent developments in systemic therapies, particularly immunotherapies, metastatic renal cell carcinoma may be approached aggressively in appropriate patients with a possibility of durable responses. Nevertheless, there is a remaining need for effective therapies for these patients.


Of all new cases of cancer in the United States, bladder cancer represents approximately 5 percent in men (fifth most common neoplasm) and 3 percent in women (eighth most common neoplasm). The incidence is increasing slowly, concurrent with an increasing older population. In 1998, there was an estimated 54,500 cases, including 39,500 in men and 15,000 in women. The age-adjusted incidence in the United States is 32 per 100,000 for men and 8 per 100,000 in women. The historic male/female ratio of 3:1 may be decreasing related to smoking patterns in women. There were an estimated 11,000 deaths from bladder cancer in 1998 (7,800 in men and 3,900 in women). Bladder cancer incidence and mortality strongly increase with age and will be an increasing problem as the population becomes more elderly.


Most bladder cancers recur in the bladder. Bladder cancer is managed with a combination of transurethral resection of the bladder (TUR) and intravesical chemotherapy or immunotherapy. The multifocal and recurrent nature of bladder cancer points out the limitations of TUR. Most muscle-invasive cancers are not cured by TUR alone. Radical cystectomy and urinary diversion is the most effective means to eliminate the cancer but carry an undeniable impact on urinary and sexual function. There continues to be a significant need for treatment modalities that are beneficial for bladder cancer patients.


An estimated 130,200 cases of colorectal cancer occurred in 2000 in the United States, including 93,800 cases of colon cancer and 36,400 of rectal cancer. Colorectal cancers are the third most common cancers in men and women. Incidence rates declined significantly during 1992-1996 (−2.1% per year). Research suggests that these declines have been due to increased screening and polyp removal, preventing progression of polyps to invasive cancers. There were an estimated 56,300 deaths (47,700 from colon cancer, 8,600 from rectal cancer) in 2000, accounting for about 11% of all U.S. cancer deaths.


At present, surgery is the most common form of therapy for colorectal cancer, and for cancers that have not spread, it is frequently curative. Chemotherapy, or chemotherapy plus radiation, is given before or after surgery to most patients whose cancer has deeply perforated the bowel wall or has spread to the lymph nodes. A permanent colostomy (creation of an abdominal opening for elimination of body wastes) is occasionally needed for colon cancer and is infrequently required for rectal cancer. There continues to be a need for effective diagnostic and treatment modalities for colorectal cancer.


There were an estimated 164,100 new cases of lung and bronchial cancer in 2000, accounting for 14% of all U.S. cancer diagnoses. The incidence rate of lung and bronchial cancer is declining significantly in men, from a high of 86.5 per 100,000 in 1984 to 70.0 in 1996. In the 1990s, the rate of increase among women began to slow. In 1996, the incidence rate in women was 42.3 per 100,000.


Lung and bronchial cancer caused an estimated 156,900 deaths in 2000, accounting for 28% of all cancer deaths. During 1992-1996, mortality from lung cancer declined significantly among men (−1.7% per year) while rates for women were still significantly increasing (0.9% per year). Since 1987, more women have died each year of lung cancer than breast cancer, which, for over 40 years, was the major cause of cancer death in women. Decreasing lung cancer incidence and mortality rates most likely resulted from decreased smoking rates over the previous 30 years; however, decreasing smoking patterns among women lag behind those of men. Of concern, although the declines in adult tobacco use have slowed, tobacco use in youth is increasing again.


Treatment options for lung and bronchial cancer are determined by the type and stage of the cancer and include surgery, radiation therapy, and chemotherapy. For many localized cancers, surgery is usually the treatment of choice. Because the disease has usually spread by the time it is discovered, radiation therapy and chemotherapy are often needed in combination with surgery. Chemotherapy alone or combined with radiation is the treatment of choice for small cell lung cancer; on this regimen, a large percentage of patients experience remission, which in some cases is long lasting. There is however, an ongoing need for effective treatment and diagnostic approaches for lung and bronchial cancers.


An estimated 182,800 new invasive cases of breast cancer were expected to occur among women in the United States during 2000. Additionally, about 1,400 new cases of breast cancer were expected to be diagnosed in men in 2000. After increasing about 4% per year in the 1980s, breast cancer incidence rates in women have leveled off in the 1990s to about 110.6 cases per 100,000.


In the U.S. alone, there were an estimated 41,200 deaths (40,800 women, 400 men) in 2000 due to breast cancer. Breast cancer ranks second among cancer deaths in women. According to the most recent data, mortality rates declined significantly during 1992-1996 with the largest decreases in younger women, both white and black. These decreases were probably the result of earlier detection and improved treatment.


Taking into account the medical circumstances and the patient's preferences, treatment of breast cancer may involve lumpectomy (local removal of the tumor) and removal of the lymph nodes under the arm; mastectomy (surgical removal of the breast) and removal of the lymph nodes under the arm; radiation therapy; chemotherapy; or hormone therapy. Often, two or more methods are used in combination. Numerous studies have shown that, for early stage disease, long-term survival rates after lumpectomy plus radiotherapy are similar to survival rates after modified radical mastectomy. Significant advances in reconstruction techniques provide several options for breast reconstruction after mastectomy. Recently, such reconstruction has been done at the same time as the mastectomy.


Local excision of ductal carcinoma in situ (DCIS) with adequate amounts of surrounding normal breast tissue may prevent the local recurrence of the DCIS. Radiation to the breast and/or tamoxifen may reduce the chance of DCIS occurring in the remaining breast tissue. This is important because DCIS, if left untreated, may develop into invasive breast cancer. Nevertheless, there are serious side effects or sequalae to these treatments. There is, therefore, a need for efficacious breast cancer treatments.


There were an estimated 23,100 new cases of ovarian cancer in the United States in 2000. It accounts for 4% of all cancers among women and ranks second among gynecologic cancers. During 1992-1996, ovarian cancer incidence rates were significantly declining. Consequent to ovarian cancer, there were an estimated 14,000 deaths in 2000. Ovarian cancer causes more deaths than any other cancer of the female reproductive system.


Surgery, radiation therapy, and chemotherapy are treatment options for ovarian cancer. Surgery usually includes the removal of one or both ovaries, the fallopian tubes (salpingo-oophorectomy), and the uterus (hysterectomy). In some very early tumors, only the involved ovary will be removed, especially in young women who wish to have children. In advanced disease, an attempt is made to remove all intra-abdominal disease to enhance the effect of chemotherapy. There continues to be an important need for effective treatment options for ovarian cancer.


There were an estimated 28,300 new cases of pancreatic cancer in the United States in 2000. Over the past 20 years, rates of pancreatic cancer have declined in men. Rates among women have remained approximately constant but may be beginning to decline. Pancreatic cancer caused an estimated 28,200 deaths in 2000 in the United States. Over the past 20 years, there has been a slight but significant decrease in mortality rates among men (about −0.9% per year) while rates have increased slightly among women.


Surgery, radiation therapy, and chemotherapy are treatment options for pancreatic cancer. These treatment options can extend survival and/or relieve symptoms in many patients but are not likely to produce a cure for most. There is a significant need for additional therapeutic and diagnostic options for pancreatic cancer.


SUMMARY OF THE INVENTION

The present invention relates to a gene, designated 125P5C8, that has now been found to be over-expressed in the cancer(s) listed in Table I. Northern blot expression analysis of 125P5C8 gene expression in normal tissues shows a restricted expression pattern in adult tissues. The nucleotide (FIG. 2) and amino acid (FIG. 2, and FIG. 3) sequences of 125P5C8 are provided. The tissue-related profile of 125P5C8 in normal adult tissues, combined with the over-expression observed in the tissues listed in Table I, shows that 125P5C8 is aberrantly over-expressed in at least some cancers, and thus serves as a useful diagnostic, prophylactic, prognostic, and/or therapeutic target for cancers of the tissue(s) such as those listed in Table I.


The invention provides polynucleotides corresponding or complementary to all or part of the 125P5C8 genes, mRNAs, and/or coding sequences, preferably in isolated form, including polynucleotides encoding 125P5C8-related proteins and fragments of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 contiguous amino acids; at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100 or more than 100 contiguous amino acids of a 125P5C8-related protein, as well as the peptides/proteins themselves; DNA, RNA, DNA/RNA hybrids, and related molecules, polynucleotides or oligonucleotides complementary or having at least a 90% homology to the 125P5C8 genes or mRNA sequences or parts thereof, and polynucleotides or oligonucleotides that hybridize to the 125P5C8 genes, mRNAs, or to 125P5C8-encoding polynucleotides. Also provided are means for isolating cDNAs and the genes encoding 125P5C8. Recombinant DNA molecules containing 125P5C8 polynucleotides, cells transformed or transduced with such molecules, and host-vector systems for the expression of 125P5C8 gene products are also provided. The invention further provides antibodies that bind to 125P5C8 proteins and polypeptide fragments thereof, including polyclonal and monoclonal antibodies, murine and other mammalian antibodies, chimeric antibodies, humanized and fully human antibodies, and antibodies labeled with a detectable marker or therapeutic agent. In certain embodiments there is a proviso that the entire nucleic acid sequence of FIG. 2 is not encoded and/or the entire amino acid sequence of FIG. 2 is not prepared. In certain embodiments, the entire nucleic acid sequence of FIG. 2 is encoded and/or the entire amino acid sequence of FIG. 2 is prepared, either of which are in respective human unit dose forms.


The invention further provides methods for detecting the presence and status of 125P5C8 polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express 125P5C8. A typical embodiment of this invention provides methods for monitoring 125P5C8 gene products in a tissue or hematology sample having or suspected of having some form of growth dysregulation such as cancer.


The invention further provides various immunogenic or therapeutic compositions and strategies for treating cancers that express 125P5C8 such as cancers of tissues listed in Table I, including therapies aimed at inhibiting the transcription, translation, processing or function of 125P5C8 as well as cancer vaccines. In one aspect, the invention provides compositions, and methods comprising them, for treating a cancer that expresses 125P5C8 in a human subject wherein the composition comprises a carrier suitable for human use and a human unit dose of one or more than one agent that inhibits the production or function of 125P5C8. Preferably, the carrier is a uniquely human carrier. In another aspect of the invention, the agent is a moiety that is immunoreactive with 125P5C8 protein. Non-limiting examples of such moieties include, but are not limited to, antibodies (such as single chain, monoclonal, polyclonal, humanized, chimeric, or human antibodies), functional equivalents thereof (whether naturally occurring or synthetic), and combinations thereof. The antibodies can be conjugated to a diagnostic or therapeutic moiety. In another aspect, the agent is a small molecule as defined herein.


In another aspect, the agent comprises one or more than one peptide which comprises a cytotoxic T lymphocyte (CTL) epitope that binds an HLA class I molecule in a human to elicit a CTL response to 125P5C8 and/or one or more than one peptide which comprises a helper T lymphocyte (HTL) epitope which binds an HLA class II molecule in a human to elicit an HTL response. The peptides of the invention may be on the same or on one or more separate polypeptide molecules. In a further aspect of the invention, the agent comprises one or more than one nucleic acid molecule that expresses one or more than one of the CTL or HTL response stimulating peptides as described above. In yet another aspect of the invention, the one or more than one nucleic acid molecule may express a moiety that is immunologically reactive with 125P5C8 as described above. The one or more than one nucleic acid molecule may also be, or encodes, a molecule that inhibits production of 125P5C8. Non-limiting examples of such molecules include, but are not limited to, those complementary to a nucleotide sequence essential for production of 125P5C8 (e.g. antisense sequences or molecules that form a triple helix with a nucleotide double helix essential for 125P5C8 production) or a ribozyme effective to lyse 125P5C8 mRNA.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. The 125P5C8 SSH sequence (SEQ ID NO:1) of 287 nucleotides.



FIG. 2. The cDNA (SEQ ID NO:2) and amino acid sequence (SEQ ID. NO:3) of 125P5C8 variant 1 clone 125P5C8-BCP1 (also called “125P5C8 variant 1” or “125P5C8 var1” or “125P5C8 v.1”) is shown in FIG. 2A. The open reading frame extends from nucleic acid 183-2282 including the stop codon. The cDNA (SEQ ID. NO:4) and amino acid sequence (SEQ ID. NO:5) of 125P5C8 variant 2 (also called “125P5C8 var2” or “125P5C8 v.2”) is shown in FIG. 2B. The open reading frame extends from nucleic acid 183-2282 including the stop codon. The cDNA (SEQ ID. NO:6) and amino acid sequence (SEQ ID. NO:7) of 125P5C8 variant 3 (also called “125P5C8 var3” or “125P5C8 v.3”) is shown in FIG. 2C. The open reading frame extends from nucleic acid 183-2282 including the stop codon. The cDNA (SEQ ID. NO:8) and amino acid sequence (SEQ ID. NO:9) of 125P5C8 variant 4 (also called “125P5C8 var4” or “125P5C8 v.4”) is shown in FIG. 2D. The open reading frame extends from nucleic acid 183-2282 including the stop codon. The cDNA (SEQ ID. NO:10) and amino acid sequence (SEQ ID. NO:11) of 125P5C8 variant 5 (also called “125P5C8 var5” or “125P5C8 v.5”) is shown in FIG. 2E. The open reading frame extends from nucleic acid 183-2282 including the stop codon. The cDNA (SEQ ID. NO:12) and amino acid sequence (SEQ ID. NO:13) of 125P5C8 variant 6 (also called “125P5C8 var5a” or “125P5C8 v.6”) is shown in FIG. 2F. The open reading frame extends from nucleic acid 183-2282 including the stop codon. In FIGS. 2A to 2F, the start methionine is shown in bold text. Please note that a reference to 125P5C8 includes variants thereof.



FIG. 3. Amino acid sequence of 125P5C8 var1 (SEQ ID. NO:3) is shown in FIG. 3A; it has 699 amino acids. The amino acid sequence of 125P5C8 var2 (SEQ ID. NO:5) is shown in FIG. 3B; it has 699 amino acids. The amino acid sequence of 125P5C8 var3 (SEQ ID. NO:7) is shown in FIG. 3C; it has 699 amino acids. The amino acid sequence of 125P5C8 var4 (SEQ ID. NO:9) is shown in FIG. 3D; it has 699 amino acids. The amino acid sequence of 125P5C8 var5 (SEQ ID. NO:11) is shown in FIG. 3E; it has 699 amino acids. As used herein, a reference to 125P5C8 includes all variants thereof, including those shown in FIG. 11.



FIG. 4. Comparison of 125P5C8 (SEQ ID NO: 3) with known genes. The amino acid sequence alignment with human AK025164 protein (SEQ ID NO: 14) is shown in FIG. 4A. The amino acid sequence alignment with yeast YCR017L protein (SEQ ID NO: 15) is shown in FIG. 4B. The amino acid sequence alignment with human protein gi/3376644 (SEQ ID NO: 16) is shown in FIG. 4C. The amino acid sequence alignment with mouse homolog gi16741400 (SEQ ID NO:17) is shown in FIG. 4D. The comparison of consensus and 125P5C8 uridine kinase domain (SEQ ID NO:18) are shown in FIG. 4E.



FIG. 5. Hydrophilicity amino acid profile of 125P5C8 var1 determined by computer algorithm sequence analysis using the method of Hopp and Woods (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828) accessed on the Protscale website (www.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.



FIG. 6. Hydropathicity amino acid profile of 125P5C8 var1 determined by computer algorithm sequence analysis using the method of Kyte and Doolittle (Kyte J., Doolittle R. F., 1982. J. Mol. Biol. 157:105-132) accessed on the ProtScale website (www.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.



FIG. 7. Percent accessible residues amino acid profile of 125P5C8 var1 determined by computer algorithm sequence analysis using the method of Janin (Janin J., 1979 Nature 277:491-492) accessed on the ProtScale website (www.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.



FIG. 8. Average flexibility amino acid profile of 125P5C8 var1 determined by computer algorithm sequence analysis using the method of Bhaskaran and Ponnuswamy (Bhaskaran R., and Ponnuswamy P. K., 1988. Int. J. Pept. Protein Res. 32:242-255) accessed on the ProtScale website through the ExPasy molecular biology server.



FIG. 9. Beta-turn amino acid profile of 125P5C8 var1 determined by computer algorithm sequence analysis using the method of Deleage and Roux (Deleage, G., Roux B. 1987 Protein Engineering 1:289-294) accessed on the ProtScale website through the ExPasy molecular biology server. The numbers correspond to those of the first sequence (variant 1). The black boxes show similarity to 125P5C8 v.1. SNPs are indicated above the boxes.



FIG. 10. Nucleotide variants of 125P5C8. The black boxes show similarity to 125P5C8 v.1. Single amino acid differences are indicated above the box. Numbers correspond to those of the first variant.



FIG. 11. FIG. 11 shows the schematic alignment of protein variants, with variant nomenclature corresponding to nucleotide variants (see FIG. 10). Nucleotide variant 125P5C8 v.6 codes for the same protein as 125P5C8 v.1.



FIG. 12. 125P5C8 var1 expression by RT-PCR. First strand cDNA was prepared from vital pool 1 (liver, lung and kidney), vital pool 2 (pancreas, spleen and stomach), LAPC xenograft pool (LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI), prostate cancer pool, bladder cancer pool, kidney cancer pool, colon cancer pool, ovary cancer pool, breast cancer pool, and metastasis cancer pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi-quantitative PCR, using primers to 125P5C8, was performed at 26 and 30 cycles of amplification.



FIG. 13. Expression of 125P5C8 var1 in normal and cancer tissues. First strand cDNA was prepared from vital pool 1 (liver, lung and kidney), vital pool 2 (pancreas, spleen and stomach), LAPC xenograft pool (LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI), normal prostate pool, prostate cancer pool, bladder cancer pool, kidney cancer pool, colon cancer pool, and lung cancer pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi-quantitative PCR, using primers to 125P5C8, was performed at 26 and 30 cycles of amplification. Results show strong expression of 125P5C8 in the prostate cancer xenografts, normal prostate, and in prostate cancer. Expression of 125P5C8 is also detected in vital pool 1, bladder cancer pool, kidney cancer pool, and colon cancer pool.



FIG. 14. Expression of 125P5C8 var1 in normal tissues and in prostate cancer xenografts. Two multiple tissue northern blots (Clontech) and a LAPC xenograft blot both with 2 ug of mRNA/lane were probed with the 125P5C8 SSH fragment. Size standards in kilobases (kb) are indicated on the side. Results show strong expression of a 3 kb transcript in normal prostate with up-regulation in the LAPC-4AI and LAPC-9AI xenografts. Lower expression is detected in colon and kidney tissues.



FIG. 15. Expression of 125P5C8 in prostate cancer patient tissues. RNA was extracted from normal prostate (NP), prostate tumors (T) and their normal adjacent tissue (N) derived from prostate cancer patients, as well as from prostate xenograft tissues (LAPC-4AD, LAPC-4AI, LAPC-9AD, and LAPC-9AI) and from the prostate cancer cell lines PC3 and LNCaP. Northern blots with 10 ug of total RNA were probed with the 125P5C8 SSH fragment. Size standards in kilobases are on the side. Results show expression of 125P5C8 in the prostate cancer patient specimens.



FIG. 16. Expression of 125P5C8 var1 in kidney cancer patient tissues. RNA was extracted from normal kidney (NK), kidney cancer patient tumors (T) and their normal adjacent tissues (N). Northern blots with 10 ug of total RNA were probed with the 125P5C8 SSH fragment. Size standards in kilobases are on the side. Results show down-regulation of 125P5C8 in tumor tissues.



FIG. 17. Expression of 125P5C8 var1 in 293T cells following transfection. 293T cells were transfected with either 125P5C8.pcDNA3.1/mychis, or with pcDNA3.1/mychis vector control. Forty hours later, cell lysates (L) as well as culture supernatants (S) were collected. Samples were run on an SDS-PAGE acrylamide gel, blotted and stained with anti-his antibody. The blot was developed using the ECL chemiluminescence kit and visualized by autoradiography. Results show expression of 125P5C8 from the two clones of 125P5C8.pcDNA3.1/mychis but not from the vector control in the lysates of the transfected cells.



FIG. 18. 125P5C8 var1Expression in 293T Cells Following Transfection. 293T cells were transfected with either 125P5C8.pcDNA3.1/mychis or pcDNA3.1/mychis vector control. Forty hours later, cell lysates were collected. Samples were run on an SDS-PAGE acrylamide gel, blotted and stained with anti-his antibody. The blot was developed using the ECL chemiluminescence kit and visualized by autoradiography. Results show expression of 125P5C8 from the two clones of 125P5C8.pcDNA3.1/mychis in the lysates of 125P5C8.pcDNA3.1/mychis transfected cells.



FIG. 19. Fluorescence microscopy and flow cytometry showing cell surface expression of 125P5C8 var1 in 293 T-cells.



FIG. 20. Secondary structure predictions for 125P5C8 var1 (SEQ ID NO:3) are shown in FIGS. 20A-20C. Please note that, since variants 2-5 are SNPs, this data would be the same for all variants. The secondary structure of 125P5C8 shown in FIG. 20A was predicted using the HNN—Hierarchical Neural Network method (Guermeur, 1997) accessed from the ExPasy molecular biology server. This method predicts the presence and location of alpha helices, extended strands, and random coils from the primary protein sequence. The percent of the protein in a given secondary structure is also given.





A schematic representation of the probability of existence of transmembrane regions and orientation based on the TMpred algorithm which utilizes TMBASE is shown in FIG. 20B (K. Hofmann, W. Stoffel. TMBASE—A database of membrane spanning protein segments Biol. Chem. Hoppe-Seyler 374:166, 1993).


A schematic representation of the probability of the existence of transmembrane regions and the extracellular and intracellular orientation based on the TMHMM algorithm is shown in FIG. 20C (Erik L. L. Sonnhammer, Gunnar von Heijne, and Anders Krogh: A hidden Markov model for predicting transmembrane helices in protein sequences. In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p 175-182 Ed J. Glasgow, T. Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C. Sensen Menlo Park, Calif.: AAAI Press, 1998). The TMpred and TMHMM algorithms are accessed from the ExPasy molecular biology server. The results of the transmembrane prediction programs depict 125P5C8 var1 as containing 1 transmembrane domain.


DETAILED DESCRIPTION OF THE INVENTION

Outline of Sections


I.) Definitions


II.) 125P5C8 Polynucleotides

    • II.A.) Uses of 125P5C8 Polynucleotides
      • II.A.1.) Monitoring of Genetic Abnormalities
      • II.A.2.) Antisense Embodiments
      • II.A.3.) Primers and Primer Pairs
      • II.A.4.) Isolation of 125P5C8-Encoding Nucleic Acid Molecules
      • II.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems


III.) 125P5C8-related Proteins

    • III.A.) Motif-bearing Protein Embodiments
    • III.B.) Expression of 125P5C8-related Proteins
    • III.C.) Modifications of 125P5C8-related Proteins
    • III.D.) Uses of 125P5C8-related Proteins


IV.) 125P5C8 Antibodies


V.) 125P5C8 Cellular Immune Responses


VI.) 125P5C8 Transgenic Animals


VII.) Methods for the Detection of 125P5C8


VIII.) Methods for Monitoring the Status of 125P5C8-related Genes and Their Products


IX.) Identification of Molecules That Interact With 125P5C8


X.) Therapeutic Methods and Compositions

    • X.A.) Anti-Cancer Vaccines
    • X.B.) 125P5C8 as a Target for Antibody-Based Therapy
    • X.C.) 125P5C8 as a Target for Cellular Immune Responses
      • X.C.1. Minigene Vaccines
      • X.C.2. Combinations of CTL Peptides with Helper Peptides
      • X.C.3. Combinations of CTL Peptides with T Cell Priming Agents
      • X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides
    • X.D.) Adoptive Immunotherapy
    • X.E.) Administration of Vaccines for Therapeutic or Prophylactic Purposes


XI.) Diagnostic and Prognostic Embodiments of 125P5C8.


XII.) Inhibition of 125P5C8 Protein Function

    • XII.A.) Inhibition of 125P5C8 With Intracellular Antibodies
    • XII.B.) Inhibition of 125P5C8 with Recombinant Proteins
    • XII.C.) Inhibition of 125P5C8 Transcription or Translation
    • XII.D.) General Considerations for Therapeutic Strategies


XIII.) KITS


I.) Definitions:


Unless otherwise defined, all terms of art, notations and other scientific terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this invention pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. Many of the techniques and procedures described or referenced herein are well understood and commonly employed using conventional methodology by those skilled in the art, such as, for example, the widely utilized molecular cloning methodologies described in Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd. edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer defined protocols and/or parameters unless otherwise noted.


The terms “advanced prostate cancer”, “locally advanced prostate cancer”, “advanced disease” and “locally advanced disease” mean prostate cancers that have extended through the prostate capsule, and are meant to include stage C disease under the American Urological Association (AUA) system, stage C1-C2 disease under the Whitmore-Jewett system, and stage T3-T4 and N+disease under the TNM (tumor, node, metastasis) system. In general, surgery is not recommended for patients with locally advanced disease, and these patients have substantially less favorable outcomes compared to patients having clinically localized (organ-confined) prostate cancer. Locally advanced disease is clinically identified by palpable evidence of induration beyond the lateral border of the prostate, or asymmetry or induration above the prostate base. Locally advanced prostate cancer is presently diagnosed pathologically following radical prostatectomy if the tumor invades or penetrates the prostatic capsule, extends into the surgical margin, or invades the seminal vesicles.


“Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence 125P5C8 (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence 125P5C8. In addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.


The term “analog” refers to a molecule which is structurally similar or shares similar or corresponding attributes with another molecule (e.g. a 125P5C8-related protein). For example an analog of a 125P5C8 protein can be specifically bound by an antibody or T cell that specifically binds to 125P5C8.


The term “antibody” is used in the broadest sense. Therefore an “antibody” can be naturally occurring or man-made such as monoclonal antibodies produced by conventional hybridoma technology. Anti-125P5C8 antibodies comprise monoclonal and polyclonal antibodies as well as fragments containing the antigen-binding domain and/or one or more complementarity determining regions of these antibodies.


An “antibody fragment” is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, i.e., the antigen-binding region. In one embodiment it specifically covers single anti-125P5C8 antibodies and clones thereof (including agonist, antagonist and neutralizing antibodies) and anti-125P5C8 antibody compositions with polyepitopic specificity.


The term “codon optimized sequences” refers to nucleotide sequences that have been optimized for a particular host species by replacing any codons having a usage frequency of less than about 20%. Nucleotide sequences that have been optimized for expression in a given host species by elimination of spurious polyadenylation sequences, elimination of exon/intron splicing signals, elimination of transposon-like repeats and/or optimization of GC content in addition to codon optimization are referred to herein as an “expression enhanced sequences.”


The term “cytotoxic agent” refers to a substance that inhibits or prevents the expression activity of cells, function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof. Examples of cytotoxic agents include, but are not limited to maytansinoids, yttrium, bismuth, ricin, ricin A-chain, doxorubicin, daunorubicin, taxol, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, sapaonaria officinalis inhibitor, and glucocorticoid and other chemotherapeutic agents, as well as radioisotopes such as At211, I131, I125, Y90, Re186, Re188, Sm153, Bi212, P32 and radioactive isotopes of Lu. Antibodies may also be conjugated to an anti-cancer pro-drug activating enzyme capable of converting the pro-drug to its active form.


The term “homolog” refers to a molecule which exhibits homology to another molecule, by for example, having sequences of chemical residues that are the same or similar at corresponding positions.


“Human Leukocyte Antigen” or “HLA” is a human class I or class II Major Histocompatibility Complex (MHC) protein (see, e.g., Stites, et al., IMMUNOLOGY, 8TH ED., Lange Publishing, Los Altos, Calif. (1994).


The terms “hybridize”, “hybridizing”, “hybridizes” and the like, used in the context of polynucleotides, are meant to refer to conventional hybridization conditions, preferably such as hybridization in 50% formamide/6×SSC/0.1% SDS/100 □mg/ml ssDNA, in which temperatures for hybridization are above 37 degrees C. and temperatures for washing in 0.1×SSC/0.1% SDS are above 55 degrees C.


The phrases “isolated” or “biologically pure” refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state. Thus, isolated peptides in accordance with the invention preferably do not contain materials normally associated with the peptides in their in situ environment. For example, a polynucleotide is said to be “isolated” when it is substantially separated from contaminant polynucleotides that correspond or are complementary to genes other than the 125P5C8 genes or that encode polypeptides other than 125P5C8 gene product or fragments thereof. A skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated 125P5C8 polynucleotide. A protein is said to be “isolated,” for example, when physical, mechanical or chemical methods are employed to remove the 125P5C8 proteins from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated 125P5C8 protein. Alternatively, an isolated protein can be prepared by chemical means.


The term “mammal” refers to any organism classified as a mammal, including mice, rats, rabbits, dogs, cats, cows, horses and humans. In one embodiment of the invention, the mammal is a mouse. In another embodiment of the invention, the mammal is a human.


The terms “metastatic prostate cancer” and “metastatic disease” mean prostate cancers that have spread to regional lymph nodes or to distant sites, and are meant to include stage D disease under the AUA system and stage T×N×M+under the TNM system. As is the case with locally advanced prostate cancer, surgery is generally not indicated for patients with metastatic disease, and hormonal (androgen ablation) therapy is a preferred treatment modality. Patients with metastatic prostate cancer eventually develop an androgen-refractory state within 12 to 18 months of treatment initiation. Approximately half of these androgen-refractory patients die within 6 months after developing that status. The most common site for prostate cancer metastasis is bone. Prostate cancer bone metastases are often osteoblastic rather than osteolytic (i.e., resulting in net bone formation). Bone metastases are found most frequently in the spine, followed by the femur, pelvis, rib cage, skull and humorous. Other common sites for metastasis include lymph nodes, lung, liver and brain. Metastatic prostate cancer is typically diagnosed by open or laparoscopic pelvic lymphadenectomy, whole body radionuclide scans, skeletal radiography, and/or bone lesion biopsy.


The term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.


A “motif”, as in biological motif of a 125P5C8-related protein, refers to any pattern of amino acids forming part of the primary sequence of a protein, that is associated with a particular function (e.g. protein-protein interaction, protein-DNA interaction, etc) or modification (e.g. that is phosphorylated, glycosylated or amidated), or localization (e.g. secretory sequence, nuclear localization sequence, etc.) or a sequence that is correlated with being immunogenic, either humorally or cellularly. A motif can be either contiguous or capable of being aligned to certain positions that are generally correlated with a certain function or property. In the context of HLA motifs, “motif” refers to the pattern of residues in a peptide of defined length, usually a peptide of from about 8 to about 13 amino acids for a class I HLA motif and from about 6 to about 25 amino acids for a class II HLA motif, which is recognized by a particular HLA molecule. Peptide motifs for HLA binding are typically different for each protein encoded by each human HLA allele and differ in the pattern of the primary and secondary anchor residues.


A “pharmaceutical excipient” comprises a material such as an adjuvant, a carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservative, and the like.


“Pharmaceutically acceptable” refers to a non-toxic, inert, and/or composition that is physiologically compatible with humans or other mammals.


The term “polynucleotide” means a polymeric form of nucleotides of at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, and is meant to include single and double stranded forms of DNA and/or RNA. In the art, this term if often used interchangeably with “oligonucleotide”. A polynucleotide can comprise a nucleotide sequence disclosed herein wherein thymidine (T), as shown for example in FIG. 2, can also be uracil (U); this definition pertains to the differences between the chemical structures of DNA and RNA, in particular the observation that one of the four major bases in RNA is uracil (U) instead of thymidine (T).


The term “polypeptide” means a polymer of at least about 4, 5, 6, 7, or 8 amino acids. Throughout the specification, standard three letter or single letter designations for amino acids are used. In the art, this term is often used interchangeably with “peptide” or “protein”.


An HLA “primary anchor residue” is an amino acid at a specific position along a peptide sequence which is understood to provide a contact point between the immunogenic peptide and the HLA molecule. One to three, usually two, primary anchor residues within a peptide of defined length generally defines a “motif” for an immunogenic peptide. These residues are understood to fit in close contact with peptide binding groove of an HLA molecule, with their side chains buried in specific pockets of the binding groove. In one embodiment, for example, the primary anchor residues for an HLA class I molecule are located at position 2 (from the amino terminal position) and at the carboxyl terminal position of a 8, 9, 10, 11, or 12 residue peptide epitope in accordance with the invention. In another embodiment, for example, the primary anchor residues of a peptide that will bind an HLA class II molecule are spaced relative to each other, rather than to the termini of a peptide, where the peptide is generally of at least 9 amino acids in length. The primary anchor positions for each motif and supermotif are set forth in Table IV. For example, analog peptides can be created by altering the presence or absence of particular residues in the primary and/or secondary anchor positions shown in Table IV. Such analogs are used to modulate the binding affinity and/or population coverage of a peptide comprising a particular HLA motif or supermotif.


A “recombinant” DNA or RNA molecule is a DNA or RNA molecule that has been subjected to molecular manipulation in vitro.


Non-limiting examples of small molecules include compounds that bind or interact with 125P5C8, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit 125P5C8 protein function. Such non-limiting small molecules preferably have a molecular weight of less than about 10 kDa, more preferably below about 9, about 8, about 7, about 6, about 5 or about 4 kDa. In certain embodiments, small molecules physically associate with, or bind, 125P5C8 protein; are not found in naturally occurring metabolic pathways; and/or are more soluble in aqueous than non-aqueous solutions


“Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured nucleic acid sequences to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).


“Stringent conditions” or “high stringency conditions”, as defined herein, are identified by, but not limited to, those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 g/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC (sodium chloride/sodium. citrate) and 50% formamide at 55° C., followed by a high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C. “Moderately stringent conditions” are described by, but not limited to, those in Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and % SDS) less stringent than those described above. An example of moderately stringent conditions is overnight incubation at 37° C. in a solution comprising: 20% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 mg/mL denatured sheared salmon sperm DNA, followed by washing the filters in 1×SSC at about 37-50° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.


An HLA “supermotif” is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles.


As used herein “to treat” or “therapeutic” and grammatically related terms, refer to any improvement of any consequence of disease, such as prolonged survival, less morbidity, and/or a lessening of side effects which are the byproducts of an alternative therapeutic modality; full eradication of disease is not required.


A “transgenic animal” (e.g., a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, e.g., an embryonic stage. A “transgene” is a DNA that is integrated into the genome of a cell from which a transgenic animal develops.


As used herein, an HLA or cellular immune response “vaccine” is a composition that contains or encodes one or more peptides of the invention. There are numerous embodiments of such vaccines, such as a cocktail of one or more individual peptides; one or more peptides of the invention comprised by a polyepitopic peptide; or nucleic acids that encode such individual peptides or polypeptides, e.g., a minigene that encodes a polyepitopic peptide. The “one or more peptides” can include any whole unit integer from 1-699 or more, e.g., at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or 699 or more peptides of the invention. The peptides or polypeptides can optionally be modified, such as by lipidation, addition of targeting or other sequences. HLA class I peptides of the invention can be admixed with, or linked to, HLA class II peptides, to facilitate activation of both cytotoxic T lymphocytes and helper T lymphocytes. HLA vaccines can also comprise peptide-pulsed antigen presenting cells, e.g., dendritic cells.


The term “variant” refers to a molecule that exhibits a variation from a described type or norm, such as a protein that has one or more different amino acid residues in the corresponding position(s) of a specifically described protein (e.g. the 125P5C8 protein shown in FIG. 2 or FIG. 3. An analog is an example of a variant protein. Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.


The “125P5C8-related proteins” of the invention include those specifically identified herein, as well as allelic variants, conservative substitution variants, analogs and homologs that can be isolated/generated and characterized without undue experimentation following the methods outlined herein or readily available in the art. Fusion proteins that combine parts of different 125P5C8 proteins or fragments thereof, as well as fusion proteins of a 125P5C8 protein and a heterologous polypeptide are also included. Such 125P5C8 proteins are collectively referred to as the 125P5C8-related proteins, the proteins of the invention, or 125P5C8. The term “125P5C8-related protein” refers to a polypeptide fragment or a 125P5C8 protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or 699 or more amino acids.


II.) 125P5C8 Polynucleotides


One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a 125P5C8 gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a 125P5C8-related protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a 125P5C8 gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a 125P5C8 gene, mRNA, or to a 125P5C8 encoding polynucleotide (collectively, “125P5C8 polynucleotides”). In all instances when referred to in this section, T can also be U in FIG. 2.


Embodiments of a 125P5C8 polynucleotide include: a 125P5C8 polynucleotide having the sequence shown in FIG. 2, the nucleotide sequence of 125P5C8 as shown in FIG. 2 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2 where T is U. For example, embodiments of 125P5C8 nucleotides comprise, without limitation:


(I) a polynucleotide comprising, consisting essentially of, or consisting of a sequence as shown in FIG. 2, wherein T can also be U;


(II) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2, from nucleotide residue number 183 through nucleotide residue number 2282, including the stop codon, wherein T can also be U;


(III) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2B, from nucleotide residue number 183 through nucleotide residue number 2282, including the stop codon, wherein T can also be U;


(IV) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2C, from nucleotide residue number 183 through nucleotide residue number 2282, including the a stop codon, wherein T can also be U;


(V) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2D, from nucleotide residue number 183 through nucleotide residue number 2282, including the stop codon, wherein T can also be U;


(VI) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2E, from nucleotide residue number 183 through nucleotide residue number 2282, including the stop codon, wherein T can also be U;


(VII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2F, from nucleotide residue number 183 through nucleotide residue number 2282, including the stop codon, wherein T can also be U;


(VIII) a polynucleotide that encodes a 125P5C8-related protein that is at least 90% homologous to an entire amino acid sequence shown in FIG. 2A-F;


(IX) a polynucleotide that encodes a 125P5C8-related protein that is at least 90% identical to an entire amino acid sequence shown in FIG. 2A-F;


(X) a polynucleotide that encodes at least one peptide set forth in Tables V-XIX;


(XI) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of FIGS. 3A-3E in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5;


(XII) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of FIGS. 3A-3E in any whole number increment up to 699 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of FIG. 6;


(XIII) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of FIGS. 3A-3E in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7;


(XIV) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of FIGS. 3A-3E in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile on FIG. 8;


(XV) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of FIGS. 3A-3E in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of FIG. 9;


(XVI) a polynucleotide that encodes a 125P5C8-related protein whose sequence is encoded by the cDNAs contained in plasmid 125P5C8 Pro-pCR2.1, deposited with American Type Culture Collection (ATCC) as Accession No. PTA-3137, on Mar. 1, 2001.


(XVII) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XVI).


(XVIII) a peptide that is encoded by any of (I)-(XVII); and


(XIX) a polynucleotide of any of (I)-(XVII) or peptide of (XVIII) together with a pharmaceutical excipient and/or in a human unit dose form.


As used herein, a range is understood to specifically disclose all whole unit positions thereof.


Typical embodiments of the invention disclosed herein include 125P5C8 polynucleotides that encode specific portions of 125P5C8 mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example:


(a) 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or 699 or more contiguous amino acids of 125P5C8.


For example, representative embodiments of the invention disclosed herein include: polynucleotides and their encoded peptides themselves encoding about amino acid 1 to about amino acid 10 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 10 to about amino acid 20 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 20 to about amino acid 30 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 30 to about amino acid 40 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 40 to about amino acid 50 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 50 to about amino acid 60 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 60 to about amino acid 70 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 70 to about amino acid 80 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 80 to about amino acid 90 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 90 to about amino acid 100 of the 125P5C8 protein shown in FIG. 2 or FIG. 3, in increments of about 10 amino acids, ending at the carboxyl terminal amino acid set forth in FIG. 2 or FIG. 3. Accordingly polynucleotides encoding portions of the amino acid sequence (of about 10 amino acids), of amino acids 100 through the carboxyl terminal amino acid of the 125P5C8 protein are embodiments of the invention. Wherein it is understood that each particular amino acid position discloses that position plus or minus five amino acid residues.


Polynucleotides encoding relatively long portions of a 125P5C8 protein are also within the scope of the invention. For example, polynucleotides encoding from about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 30, or 40 or 50 etc.) of the 125P5C8 protein “or variant” shown in FIG. 2 or FIG. 3 can be generated by a variety of techniques well known in the art. These polynucleotide fragments can include any portion of the 125P5C8 sequence as shown in FIG. 2.


Additional illustrative embodiments of the invention disclosed herein include 125P5C8 polynucleotide fragments encoding one or more of the biological motifs contained within a 125P5C8 protein “or variant” sequence, including one or more of the motif-bearing subsequences of a 125P5C8 protein “or variant” set forth in Tables V-XIX. In another embodiment, typical polynucleotide fragments of the invention encode one or more of the regions of 125P5C8 protein or variant that exhibit homology to a known molecule. In another embodiment of the invention, typical polynucleotide fragments can encode one or more of the 125P5C8 protein or variant N-glycosylation sites, cAMP and cGMP-dependent protein kinase phosphorylation sites, casein kinase II phosphorylation sites or N-myristoylation site and amidation sites.


II.A.) Uses of 125P5C8 Polynucleotides


II.A.1.) Monitoring of Genetic Abnormalities


The polynucleotides of the preceding paragraphs have a number of different specific uses. The human 125P5C8 gene maps to the chromosomal location set forth in Example 3. For example, because the 125P5C8 gene maps to this chromosome, polynucleotides that encode different regions of the 125P5C8 proteins are used to characterize cytogenetic abnormalities of this chromosomal locale, such as abnormalities that are identified as being associated with various cancers. In certain genes, a variety of chromosomal abnormalities including rearrangements have been identified as frequent cytogenetic abnormalities in a number of different cancers (see e.g. Krajinovic et al., Mutat. Res. 382(3-4): 81-83 (1998); Johansson et al., Blood 86(10): 3905-3914 (1995) and Finger et al., P.N.A.S. 85(23): 9158-9162 (1988)). Thus, polynucleotides encoding specific regions of the 125P5C8 proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes 125P5C8 that may contribute to the malignant phenotype. In this context, these polynucleotides satisfy a need in the art for expanding the sensitivity of chromosomal screening in order to identify more subtle and less common chromosomal abnormalities (see e.g. Evans et al., Am. J. Obstet. Gynecol 171(4): 1055-1057 (1994)).


Furthermore, as 125P5C8 was shown to be highly expressed in bladder and other cancers, 125P5C8 polynucleotides are used in methods assessing the status of 125P5C8 gene products in normal versus cancerous tissues. Typically, polynucleotides that encode specific regions of the 125P5C8 proteins are used to assess the presence of perturbations (such as deletions, insertions, point mutations, or alterations resulting in a loss of an antigen etc.) in specific regions of the 125P5C8 gene, such as regions containing one or more motifs. Exemplary assays include both RT-PCR assays as well as single-strand conformation polymorphism (SSCP) analysis (see, e.g., Marrogi et al., J. Cutan. Pathol. 26(8): 369-378 (1999), both of which utilize polynucleotides encoding specific regions of a protein to examine these regions within the protein.


II.A.2.) Antisense Embodiments


Other specifically contemplated nucleic acid related embodiments of the invention disclosed herein are genomic DNA, cDNAs, ribozymes, and antisense molecules, as well as nucleic acid molecules based on an alternative backbone, or including alternative bases, whether derived from natural sources or synthesized, and include molecules capable of inhibiting the RNA or protein expression of 125P5C8. For example, antisense molecules can be RNAs or other molecules, including peptide nucleic acids (PNAs) or non-nucleic acid molecules such as phosphorothioate derivatives, that specifically bind DNA or RNA in a base pair-dependent manner. A skilled artisan can readily obtain these classes of nucleic acid molecules using the 125P5C8 polynucleotides and polynucleotide sequences disclosed herein.


Antisense technology entails the administration of exogenous oligonucleotides that bind to a target polynucleotide located within the cells. The term “antisense” refers to the fact that such oligonucleotides are complementary to their intracellular targets, e.g., 125P5C8. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988). The 125P5C8 antisense oligonucleotides of the present invention include derivatives such as S-oligonucleotides (phosphorothioate derivatives or S-oligos, see, Jack Cohen, supra), which exhibit enhanced cancer cell growth inhibitory action. S-oligos (nucleoside phosphorothioates) are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom. The S-oligos of the present invention can be prepared by treatment of the corresponding O-oligos with 3H-1,2-benzodithiol-3-one-1,1-dioxide, which is a sulfur transfer reagent. See, e.g., Iyer, R. P. et al., J. Org. Chem. 55:4693-4698 (1990); and Iyer, R. P. et al., J. Am. Chem. Soc. 112:1253-1254 (1990). Additional 125P5C8 antisense oligonucleotides of the present invention include morpholino antisense oligonucleotides known in the art (see, e.g., Partridge et al., 1996, Antisense & Nucleic Acid Drug Development 6: 169-175).


The 125P5C8 antisense oligonucleotides of the present invention typically can be RNA or DNA that is complementary to and stably hybridizes with the first 100 5′ codons or last 100 3′ codons of a 125P5C8 genomic sequence or the corresponding mRNA. Absolute complementarity is not required, although high degrees of complementarity are preferred. Use of an oligonucleotide complementary to this region allows for the selective hybridization to 125P5C8 mRNA and not to mRNA specifying other regulatory subunits of protein kinase. In one embodiment, 125P5C8 antisense oligonucleotides of the present invention are 15 to 30-mer fragments of the antisense DNA molecule that have a sequence that hybridizes to 125P5C8 mRNA. Optionally, 125P5C8 antisense oligonucleotide is a 30-mer oligonucleotide that is complementary to a region in the first 10 5′ codons or last 10 3′ codons of 125P5C8. Alternatively, the antisense molecules are modified to employ ribozymes in the inhibition of 125P5C8 expression, see, e.g., L. A. Couture & D. T. Stinchcomb; Trends Genet. 12: 510-515 (1996).


II.A.3.) Primers and Primer Pairs


Further specific embodiments of this nucleotides of the invention include primers and primer pairs, which allow the specific amplification of polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof. Probes can be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme. Such probes and primers are used to detect the presence of a 125P5C8 polynucleotide in a sample and as a means for detecting a cell expressing a 125P5C8 protein.


Examples of such probes include polypeptides comprising all or part of the human 125P5C8 cDNA sequence shown in FIG. 2. Examples of primer pairs capable of specifically amplifying 125P5C8 mRNAs are also described in the Examples. As will be understood by the skilled artisan, a great many different primers and probes can be prepared based on the sequences provided herein and used effectively to amplify and/or detect a 125P5C8 mRNA.


The 125P5C8 polynucleotides of the invention are useful for a variety of purposes, including but not limited to their use as probes and primers for the amplification and/or detection of the 125P5C8 gene(s), mRNA(s), or fragments thereof; as reagents for the diagnosis and/or prognosis of prostate cancer and other cancers; as coding sequences capable of directing the expression of 125P5C8 polypeptides; as tools for modulating or inhibiting the expression of the 125P5C8 gene(s) and/or translation of the 125P5C8 transcript(s); and as therapeutic agents.


The present invention includes the use of any probe as described herein to identify and isolate a 125P5C8 or 125P5C8 related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence per se, which would comprise all or most of the sequences found in the probe used.


II.A.4.) Isolation of 125P5C8-Encoding Nucleic Acid Molecules


The 125P5C8 cDNA sequences described herein enable the isolation of other polynucleotides encoding 125P5C8 gene product(s), as well as the isolation of polynucleotides encoding 125P5C8 gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a 125P5C8 gene product as well as polynucleotides that encode analogs of 125P5C8-related proteins. Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a 125P5C8 gene are well known (see, for example, Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press, New York, 1989; Current Protocols in Molecular Biology. Ausubel et al., Eds., Wiley and Sons, 1995). For example, lambda phage cloning methodologies can be conveniently employed, using commercially available cloning systems (e.g., Lambda ZAP Express, Stratagene). Phage clones containing 125P5C8 gene cDNAs can be identified by probing with a labeled 125P5C8 cDNA or a fragment thereof. For example, in one embodiment, a 125P5C8 cDNA (e.g., FIG. 2) or a portion thereof can be synthesized and used as a probe to retrieve overlapping and full-length cDNAs corresponding to a 125P5C8 gene. A 125P5C8 gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with 125P5C8 DNA probes or primers.


II.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems


The invention also provides recombinant DNA or RNA molecules containing a 125P5C8 polynucleotide, a fragment, analog or homologue thereof, including but not limited to phages, plasmids, phagemids, cosmids, YACs, BACs, as well as various viral and non-viral vectors well known in the art, and cells transformed or transfected with such recombinant DNA or RNA molecules. Methods for generating such molecules are well known (see, for example, Sambrook et al., 1989, supra).


The invention further provides a host-vector system comprising a recombinant DNA molecule containing a 125P5C8 polynucleotide, fragment, analog or homologue thereof within a suitable prokaryotic or eukaryotic host cell. Examples of suitable eukaryotic host cells include a yeast cell, a plant cell, or an animal cell, such as a mammalian cell or an insect cell (e.g., a baculovirus-infectible cell such as an Sf9 or HighFive cell). Examples of suitable mammalian cells include various prostate cancer cell lines such as DU145 and TsuPr1, other transfectable or transducible prostate cancer cell lines, primary cells (PrEC), as well as a number of mammalian cells routinely used for the expression of recombinant proteins (e.g., COS, CHO, 293, 293T cells). More particularly, a polynucleotide comprising the coding sequence of 125P5C8 or a fragment, analog or homolog thereof can be used to generate 125P5C8 proteins or fragments thereof using any number of host-vector systems routinely used and widely known in the art.


A wide range of host-vector systems suitable for the expression of 125P5C8 proteins or fragments thereof are available, see for example, Sambrook et al., 1989, supra; Current Protocols in Molecular Biology, 1995, supra). Preferred vectors for mammalian expression include but are not limited to pcDNA 3.1 myc-His-tag (Invitrogen) and the retroviral vector pSRαtkneo (Muller et al., 1991, MCB 11:1785). Using these expression vectors, 125P5C8 can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPr1. The host-vector systems of the invention are useful for the production of a 125P5C8 protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of 125P5C8 and 125P5C8 mutations or analogs.


Recombinant human 125P5C8 protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a 125P5C8-related nucleotide. For example, 293T cells can be transfected with an expression plasmid encoding 125P5C8 or fragment, analog or homolog thereof, a 125P5C8-related protein is expressed in the 293T cells, and the recombinant 125P5C8 protein is isolated using standard purification methods (e.g., affinity purification using anti-125P5C8 antibodies). In another embodiment, a 125P5C8 coding sequence is subcloned into the retroviral vector pSRαMSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPr1, 293 and rat-1 in order to establish 125P5C8 expressing cell lines. Various other expression systems well known in the art can also be employed. Expression constructs encoding a leader peptide joined in frame to a 125P5C8 coding sequence can be used for the generation of a secreted form of recombinant 125P5C8 protein.


As discussed herein, redundancy in the genetic code permits variation in 125P5C8 gene sequences. In particular, it is known in the art that specific host species often have specific codon preferences, and thus one can adapt the disclosed sequence as preferred for a desired host. For example, preferred analog codon sequences typically have rare codons (i.e., codons having a usage frequency of less than about 20% in known sequences of the desired host) replaced with higher frequency codons. Codon preferences for a specific species are calculated, for example, by utilizing codon usage tables available on the INTERNET such as at URL www.dna.affrc.go.jp/˜nakamura/codon.html.


Additional sequence modifications are known to enhance protein expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon/intron splice site signals, transposon-like repeats, and/or other such well-characterized sequences that are deleterious to gene expression. The GC content of the sequence is adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. Where possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures. Other useful modifications include the addition of a translational initiation consensus sequence at the start of the open reading frame, as described in Kozak, Mol. Cell. Biol., 9:5073-5080 (1989). Skilled artisans understand that the general rule that eukaryotic ribosomes initiate translation exclusively at the 5′ proximal AUG codon is abrogated only under rare conditions (see, e.g., Kozak PNAS 92(7): 2662-2666, (1995) and Kozak NAR 15(20): 8125-8148 (1987)).


III.) 125P5C8-Related Proteins


Another aspect of the present invention provides 125P5C8-related proteins. Specific embodiments of 125P5C8 proteins comprise a polypeptide having all or part of the amino acid sequence of human 125P5C8 as shown in FIG. 2 or FIG. 3. Alternatively, embodiments of 125P5C8 proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of 125P5C8 shown in FIG. 2 or FIG. 3.


In general, naturally occurring allelic variants of human 125P5C8 share a high degree of structural identity and homology (e.g., 90% or more homology). Typically, allelic variants of a 125P5C8 protein contain conservative amino acid substitutions within the 125P5C8 sequences described herein or contain a substitution of an amino acid from a corresponding position in a homologue of 125P5C8. One class of 125P5C8 allelic variants are proteins that share a high degree of homology with at least a small region of a particular 125P5C8 amino acid sequence, but further contain a radical departure from the sequence, such as a non-conservative substitution, truncation, insertion or frame shift. In comparisons of protein sequences, the terms, similarity, identity, and homology each have a distinct meaning as appreciated in the field of genetics. Moreover, orthology and paralogy can be important concepts describing the relationship of members of a given protein family in one organism to the members of the same family in other organisms.


Amino acid abbreviations are provided in Table II. Conservative amino acid substitutions can frequently be made in a protein without altering either the conformation or the function of the protein. Proteins of the invention can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 conservative substitutions. Such changes include substituting any of isoleucine (I), valine (V), and leucine (L) for any other of these hydrophobic amino acids; aspartic acid (D) for glutamic acid (E) and vice versa; glutamine (Q) for asparagine (N) and vice versa; and serine (S) for threonine (T) and vice versa. Other substitutions can also be considered conservative, depending on the environment of the particular amino acid and its role in the three-dimensional structure of the protein. For example, glycine (G) and alanine (A) can frequently be interchangeable, as can alanine (A) and valine (V). Methionine (M), which is relatively hydrophobic, can frequently be interchanged with leucine and isoleucine, and sometimes with valine. Lysine (K) and arginine (R) are frequently interchangeable in locations in which the significant feature of the amino acid residue is its charge and the differing pK's of these two amino acid residues are not significant. Still other changes can be considered “conservative” in particular environments (see, e.g. Table III herein; pages 13-15 “Biochemistry” 2nd ED. Lubert Stryer ed (Stanford University); Henikoff et al., PNAS 1992 Vol 89 10915-10919; Lei et al., J Biol Chem 1995 May 19; 270(20):11882-6).


Embodiments of the invention disclosed herein include a wide variety of art-accepted variants or analogs of 125P5C8 proteins such as polypeptides having amino acid insertions, deletions and substitutions. 125P5C8 variants can be made using methods known in the art such as site-directed mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. Acids Res., 10:6487 (1987)), cassette mutagenesis (Wells et al., Gene, 34:315 (1985)), restriction selection mutagenesis (Wells et al., Philos. Trans. R. Soc. London SerA, 317:415 (1986)) or other known techniques can be performed on the cloned DNA to produce the 125P5C8 variant DNA.


Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence that is involved in a specific biological activity such as a protein-protein interaction. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions (Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976)). If alanine substitution does not yield adequate amounts of variant, an isosteric amino acid can be used.


As defined herein, 125P5C8 variants, analogs or homologs, have the distinguishing attribute of having at least one epitope that is “cross reactive” with a 125P5C8 protein having an amino acid sequence of FIG. 3. As used in this sentence, “cross reactive” means that an antibody or T cell that specifically binds to a 125P5C8 variant also specifically binds to a 125P5C8 protein having an amino acid sequence set forth in FIG. 3. A polypeptide ceases to be a variant of a protein shown in FIG. 3, when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting 125P5C8 protein. Those skilled in the art understand that antibodies that recognize proteins bind to epitopes of varying size, and a grouping of the order of about four or five amino acids, contiguous or not, is regarded as a typical number of amino acids in a minimal epitope. See, e.g., Nair et al., J. Immunol. 2000 165(12): 6949-6955; Hebbes et al., Mol Immunol (1989) 26(9):865-73; Schwartz et al., J Immunol (1985) 135(4):2598-608.


Other classes of 125P5C8-related protein variants share 70%, 75%, 80%, 85% or 90% or more similarity with an amino acid sequence of FIG. 3, or a fragment thereof. Another specific class of 125P5C8 protein variants or analogs comprise one or more of the 125P5C8 biological motifs described herein or presently known in the art. Thus, encompassed by the present invention are analogs of 125P5C8 fragments (nucleic or amino acid) that have altered functional (e.g. immunogenic) properties relative to the starting fragment. It is to be appreciated that motifs now or which become part of the art are to be applied to the nucleic or amino acid sequences of FIG. 2 or FIG. 3.


As discussed herein, embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a 125P5C8 protein shown in FIG. 2 or FIG. 3. For example, representative embodiments of the invention comprise peptides/proteins having any 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids of a 125P5C8 protein shown in FIG. 2 or FIG. 3.


Moreover, representative embodiments of the invention disclosed herein include polypeptides consisting of about amino acid 1 to about amino acid 10 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 10 to about amino acid 20 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 20 to about amino acid 30 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 30 to about amino acid 40 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 40 to about amino acid 50 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 50 to about amino acid 60 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 60 to about amino acid 70 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 70 to about amino acid 80 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 80 to about amino acid 90 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 90 to about amino acid 100 of a 125P5C8 protein shown in FIG. 2 or FIG. 3, etc. throughout the entirety of a 125P5C8 amino acid sequence. Moreover, polypeptides consisting of about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 130, or 140 or 150 etc.) of a 125P5C8 protein shown in FIG. 2 or FIG. 3 are embodiments of the invention. It is to be appreciated that the starting and stopping positions in this paragraph refer to the specified position as well as that position plus or minus 5 residues.


125P5C8-related proteins are generated using standard peptide synthesis technology or using chemical cleavage methods well known in the art. Alternatively, recombinant methods can be used to generate nucleic acid molecules that encode a 125P5C8-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a 125P5C8 protein (or variants, homologs or analogs thereof).


III.A.) Motif-Bearing Protein Embodiments


Additional illustrative embodiments of the invention disclosed herein include 125P5C8 polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a 125P5C8 polypeptide sequence set forth in FIG. 2 or FIG. 3. Various motifs are known in the art, and a protein can be evaluated for the presence of such motifs by a number of publicly available Internet sites (see, e.g., Epimatrix™ and Epimer™, Brown University, and BIMAS).


Motif bearing subsequences of all 125P5C8 variant proteins are set forth and identified in Tables V-XIX.


Table XX sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu/). The columns of Table XX list (1) motif name abbreviation, (2) percent identity found amongst the different member of the motif family, (3) motif name or description and (4) most common function; location information is included if the motif is relevant for location.


Polypeptides comprising one or more of the 125P5C8 motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the 125P5C8 motifs discussed above are associated with growth dysregulation and because 125P5C8 is overexpressed in certain cancers (See, e.g., Table I). Casein kinase II, cAMP and camp-dependent protein kinase, and Protein Kinase C, for example, are enzymes known to be associated with the development of the malignant phenotype (see e.g. Chen et al., Lab Invest., 78(2): 165-174 (1998); Gaiddon et al., Endocrinology 136(10): 4331-4338 (1995); Hall et al., Nucleic Acids Research 24(6): 1119-1126 (1996); Peterziel et al., Oncogene 18(46): 6322-6329 (1999) and O'Brian, Oncol. Rep. 5(2): 305-309 (1998)). Moreover, both glycosylation and myristoylation are protein modifications also associated with cancer and cancer progression (see e.g. Dennis et al., Biochem. Biophys. Acta 1473(1):21-34 (1999); Raju et al., Exp. Cell Res. 235(1): 145-154 (1997)). Amidation is another protein modification also associated with cancer and cancer progression (see e.g. Treston et al., J. Natl. Cancer Inst. Monogr. (13): 169-175 (1992)).


In another embodiment, proteins of the invention comprise one or more of the immunoreactive epitopes identified in accordance with art-accepted methods, such as the peptides set forth in Tables V-XVIII. CTL epitopes can be determined using specific algorithms to identify peptides within a 125P5C8 protein that are capable of optimally binding to specified HLA alleles (e.g., Table IV; Epimatrix™ and Epimer™, Brown University, and BIMAS) Moreover, processes for identifying peptides that have sufficient binding affinity for HLA molecules and which are correlated with being immunogenic epitopes, are well known in the art, and are carried out without undue experimentation. In addition, processes for identifying peptides that are immunogenic epitopes, are well known in the art, and are carried out without undue experimentation either in vitro or in vivo.


Also known in the art are principles for creating analogs of such epitopes in order to modulate immunogenicity. For example, one begins with an epitope that bears a CTL or HTL motif (see, e.g., the HLA Class I and HLA Class II motifs/supermotifs of Table IV). The epitope is analoged by substituting out an amino acid at one of the specified positions, and replacing it with another amino acid specified for that position. For example, one can substitute out a deleterious residue in favor of any other residue, such as a preferred residue as defined in Table IV; substitute a less-preferred residue with a preferred residue as defined in Table IV; or substitute an originally-occurring preferred residue with another preferred residue as defined in Table IV. Substitutions can occur at primary anchor positions or at other positions in a peptide; see, e.g., Table IV.


A variety of references reflect the art regarding the identification and generation of epitopes in a protein of interest as well as analogs thereof. See, for example, WO 9733602 to Chesnut et al.; Sette, Immunogenetics 1999 50(3-4): 201-212; Sette et al., J. Immunol. 2001 166(2): 1389-1397; Sidney et al., Hum. Immunol. 1997 58(1): 12-20; Kondo et al., Immunogenetics 1997 45(4): 249-258; Sidney et al., J. Immunol. 1996 157(8): 3480-90; and Falk et al., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et al., J. Immunol. 152:163-75 (1994)); Kast et al., 1994 152(8): 3904-12; Borras-Cuesta et al., Hum. Immunol. 2000 61(3): 266-278; Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., PMID: 7895164, UI: 95202582; O'Sullivan et al., J. Immunol. 1991 147(8): 2663-2669; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res. 1998 18(2): 79-92.


Related embodiments of the invention include polypeptides comprising combinations of the different motifs set forth in Table XXI, and/or, one or more of the predicted CTL epitopes of Tables V-XIX, and/or, one or more of the T cell binding motifs known in the art. Preferred embodiments contain no insertions, deletions or substitutions either within the motifs or the intervening sequences of the polypeptides. In addition, embodiments which include a number of either N-terminal and/or C-terminal amino acid residues on either side of these motifs may be desirable (to, for example, include a greater portion of the polypeptide architecture in which the motif is located). Typically the number of N-terminal and/or C-terminal amino acid residues on either side of a motif is between about 1 to about 100 amino acid residues, preferably 5 to about 50 amino acid residues.


125P5C8-related proteins are embodied in many forms, preferably in isolated form. A purified 125P5C8 protein molecule will be substantially free of other proteins or molecules that impair the binding of 125P5C8 to antibody, T cell or other ligand. The nature and degree of isolation and purification will depend on the intended use. Embodiments of a 125P5C8-related proteins include purified 125P5C8-related proteins and functional, soluble 125P5C8-related proteins. In one embodiment, a functional, soluble 125P5C8 protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.


The invention also provides 125P5C8 proteins comprising biologically active fragments of a 125P5C8 amino acid sequence shown in FIG. 2 or FIG. 3. Such proteins exhibit properties of the starting 125P5C8 protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting 125P5C8 protein; to be bound by such antibodies; to elicit the activation of HTL or CTL; and/or, to be recognized by HTL or CTL that also specifically bind to the starting protein.


125P5C8-related polypeptides that contain particularly interesting structures can be predicted and/or identified using various analytical techniques well known in the art, including, for example, the methods of Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis, or on the basis of immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-125P5C8 antibodies, or T cells or in identifying cellular factors that bind to 125P5C8. For example, hydrophilicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Hopp, T. P. and Woods, K. R., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Kyte, J. and Doolittle, R. F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated, and immunogenic peptide fragments identified, using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated, and immunogenic peptide fragments identified, using the method of Bhaskaran R., Ponnuswamy P. K., 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated, and immunogenic peptide fragments identified, using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294.


CTL epitopes can be determined using specific algorithms to identify peptides within a 125P5C8 protein that are capable of optimally binding to specified HLA alleles (e.g., by using the SYFPEITHI site at World Wide Web, the listings in Table IV(A)-(E); Epimatrix™ and Epimer™, Brown University, and BIMAS). Illustrating this, peptide epitopes from 125P5C8 that are presented in the context of human MHC Class I molecules, e.g., HLA-A1, A2, A3, A11, A24, B7 and B35 were predicted (Tables V-XIX). Specifically, the complete amino acid sequence of the 125P5C8 protein and relevant portions of other variants, i.e., for HLA Class I predictions 9 flanking residues on either side of a point mutation, and for HLA Class II predictions 14 flanking residues on either side of a point mutation, were entered into the HLA Peptide Motif Search algorithm found in the Bioinformatics and Molecular Analysis Section (BIMAS) web site listed above; for HLA Class II the site SYFPEITHI was used.


The HLA peptide motif search algorithm was developed by Dr. Ken Parker based on binding of specific peptide sequences in the groove of HLA Class I molecules, in particular HLA-A2 (see, e.g., Falk et al., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et al., J. Immunol. 152:163-75 (1994)). This algorithm allows location and ranking of 8-mer, 9-mer, and 10-mer peptides from a complete protein sequence for predicted binding to HLA-A2 as well as numerous other HLA Class I molecules. Many HLA class I binding peptides are 8-, 9-, 10 or 11-mers. For example, for Class I HLA-A2, the epitopes preferably contain a leucine (L) or methionine (M) at position 2 and a valine (V) or leucine (L) at the C-terminus (see, e.g., Parker et al., J. Immunol. 149:3580-7 (1992)). Selected results of 125P5C8 predicted binding peptides are shown in Tables V-XVIII herein. In Tables V-XVIII, the top 50 ranking candidates, 9-mers and 10-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score. The binding score corresponds to the estimated half time of dissociation of complexes containing the peptide at 37° C. at pH 6.5. Peptides with the highest binding score are predicted to be the most tightly bound to HLA Class I on the cell surface for the greatest period of time and thus represent the best immunogenic targets for T-cell recognition.


Actual binding of peptides to an HLA allele can be evaluated by stabilization of HLA expression on the antigen-processing defective cell line T2 (see, e.g., Xue et al., Prostate 30:73-8 (1997) and Peshwa et al., Prostate 36:129-38 (1998)). Immunogenicity of specific peptides can be evaluated in vitro by stimulation of CD8+ cytotoxic T lymphocytes (CTL) in the presence of antigen presenting cells such as dendritic cells.


It is to be appreciated that every epitope predicted by the BIMAS site, Epimer™ and Epimatrix™ sites, or specified by the HLA class I or class II motifs available in the art or which become part of the art such as set forth in Table IV (or determined using World Wide Web site URL syfpeithi.bmi-heidelberg.com/, or BIMAS, bimas.dcrt.nih.gov/) are to be “applied” to a 125P5C8 protein in accordance with the invention. As used in this context “applied” means that a 125P5C8 protein is evaluated, e.g., visually or by computer-based patterns finding methods, as appreciated by those of skill in the relevant art. Every subsequence of a 125P5C8 protein of 8, 9, 10, or 11 amino acid residues that bears an HLA Class I motif, or a subsequence of 9 or more amino acid residues that bear an HLA Class II motif are within the scope of the invention.


III.B.) Expression of 125P5C8-Related Proteins


In an embodiment described in the examples that follow, 125P5C8 can be conveniently expressed in cells (such as 293T cells) transfected with a commercially available expression vector such as a CMV-driven expression vector encoding 125P5C8 with a C-terminal 6×His and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville Tenn.). The Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted 125P5C8 protein in transfected cells. The secreted HIS-tagged 125P5C8 in the culture media can be purified, e.g., using a nickel column using standard techniques.


III.C.) Modifications of 125P5C8-Related Proteins


Modifications of 125P5C8-related proteins such as covalent modifications are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of a 125P5C8 polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues of a 125P5C8 protein. Another type of covalent modification of a 125P5C8 polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of a protein of the invention. Another type of covalent modification of 125P5C8 comprises linking a 125P5C8 polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. No. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.


The 125P5C8-related proteins of the present invention can also be modified to form a chimeric molecule comprising 125P5C8 fused to another, heterologous polypeptide or amino acid sequence. Such a chimeric molecule can be synthesized chemically or recombinantly. A chimeric molecule can have a protein of the invention fused to another tumor-associated antigen or fragment thereof. Alternatively, a protein in accordance with the invention can comprise a fusion of fragments of a 125P5C8 sequence (amino or nucleic acid) such that a molecule is created that is not, through its length, directly homologous to the amino or nucleic acid sequences shown in FIG. 2 or FIG. 3. Such a chimeric molecule can comprise multiples of the same subsequence of 125P5C8. A chimeric molecule can comprise a fusion of a 125P5C8-related protein with a polyhistidine epitope tag, which provides an epitope to which immobilized nickel can selectively bind, with cytokines or with growth factors. The epitope tag is generally placed at the amino- or carboxyl-terminus of a 125P5C8 protein. In an alternative embodiment, the chimeric molecule can comprise a fusion of a 125P5C8-related protein with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also referred to as an “immunoadhesin”), such a fusion could be to the Fc region of an IgG molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a 125P5C8 polypeptide in place of at least one variable region within an Ig molecule. In a preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgGI molecule. For the production of immunoglobulin fusions see, e.g., U.S. Pat. No. 5,428,130 issued Jun. 27, 1995.


III.D.) Uses of 125P5C8-Related Proteins


The proteins of the invention have a number of different specific uses. As 125P5C8 is highly expressed in prostate and other cancers, 125P5C8-related proteins are used in methods that assess the status of 125P5C8 gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a 125P5C8 protein are used to assess the presence of perturbations (such as deletions, insertions, point mutations etc.) in those regions (such as regions containing one or more motifs). Exemplary assays utilize antibodies or T cells targeting 125P5C8-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a 125P5C8 polypeptide sequence in order to evaluate the characteristics of this region in normal versus cancerous tissues or to elicit an immune response to the epitope. Alternatively, 125P5C8-related proteins that contain the amino acid residues of one or more of the biological motifs in a 125P5C8 protein are used to screen for factors that interact with that region of 125P5C8.


125P5C8 protein fragments/subsequences are particularly useful in generating and characterizing domain-specific antibodies (e.g., antibodies recognizing an extracellular or intracellular epitope of a 125P5C8 protein), for identifying agents or cellular factors that bind to 125P5C8 or a particular structural domain thereof, and in various therapeutic and diagnostic contexts, including but not limited to diagnostic assays, cancer vaccines and methods of preparing such vaccines.


Proteins encoded by the 125P5C8 genes, or by analogs, homologs or fragments thereof, have a variety of uses, including but not limited to generating antibodies and in methods for identifying ligands and other agents and cellular constituents that bind to a 125P5C8 gene product. Antibodies raised against a 125P5C8 protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of 125P5C8 protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers. 125P5C8-related nucleic acids or proteins are also used in generating HTL or CTL responses.


Various immunological assays useful for the detection of 125P5C8 proteins are used, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), immunocytochemical methods, and the like. Antibodies can be labeled and used as immunological imaging reagents capable of detecting 125P5C8-expressing cells (e.g., in radioscintigraphic imaging methods). 125P5C8 proteins are also particularly useful in generating cancer vaccines, as further described herein.


IV.) 125P5C8 Antibodies


Another aspect of the invention provides antibodies that bind to 125P5C8-related proteins. Preferred antibodies specifically bind to a 125P5C8-related protein and do not bind (or bind weakly) to peptides or proteins that are not 125P5C8-related proteins. For example, antibodies that bind 125P5C8 can bind 125P5C8-related proteins such as the homologs or analogs thereof.


125P5C8 antibodies of the invention are particularly useful in cancer (see, e.g., Table I) diagnostic and prognostic assays, and imaging methodologies. Similarly, such antibodies are useful in the treatment, diagnosis, and/or prognosis of other cancers, to the extent 125P5C8 is also expressed or overexpressed in these other cancers. Moreover, intracellularly expressed antibodies (e.g., single chain antibodies) are therapeutically useful in treating cancers in which the expression of 125P5C8 is involved, such as advanced or metastatic prostate cancers.


The invention also provides various immunological assays useful for the detection and quantification of 125P5C8 and mutant 125P5C8-related proteins. Such assays can comprise one or more 125P5C8 antibodies capable of recognizing and binding a 125P5C8-related protein, as appropriate. These assays are performed within various immunological assay formats well known in the art, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), and the like.


Immunological non-antibody assays of the invention also comprise T cell immunogenicity assays (inhibitory or stimulatory) as well as major histocompatibility complex (MHC) binding assays.


In addition, immunological imaging methods capable of detecting prostate cancer and other cancers expressing 125P5C8 are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled 125P5C8 antibodies. Such assays are clinically useful in the detection, monitoring, and prognosis of 125P5C8 expressing cancers such as prostate cancer.


125P5C8 antibodies are also used in methods for purifying a 125P5C8-related protein and for isolating 125P5C8 homologues and related molecules. For example, a method of purifying a 125P5C8-related protein comprises incubating a 125P5C8 antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a 125P5C8-related protein under conditions that permit the 125P5C8 antibody to bind to the 125P5C8-related protein; washing the solid matrix to eliminate impurities; and eluting the 125P5C8-related protein from the coupled antibody. Other uses of 125P5C8 antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a 125P5C8 protein.


Various methods for the preparation of antibodies are well known in the art. For example, antibodies can be prepared by immunizing a suitable mammalian host using a 125P5C8-related protein, peptide, or fragment, in isolated or immunoconjugated form (Antibodies: A Laboratory Manual, CSH Press, Eds., Harlow, and Lane (1988); Harlow, Antibodies, Cold Spring Harbor Press, NY (1989)). In addition, fusion proteins of 125P5C8 can also be used, such as a 125P5C8 GST-fusion protein. In a particular embodiment, a GST fusion protein comprising all or most of the amino acid sequence of FIG. 2 or FIG. 3 is produced, then used as an immunogen to generate appropriate antibodies. In another embodiment, a 125P5C8-related protein is synthesized and used as an immunogen.


In addition, naked DNA immunization techniques known in the art are used (with or without purified 125P5C8-related protein or 125P5C8 expressing cells) to generate an immune response to the encoded immunogen (for review, see Donnelly et al., 1997, Ann. Rev. Immunol. 15: 617-648).


The amino acid sequence of a 125P5C8 protein as shown in FIG. 2 or FIG. 3 can be analyzed to select specific regions of the 125P5C8 protein for generating antibodies. For example, hydrophobicity and hydrophilicity analyses of a 125P5C8 amino acid sequence are used to identify hydrophilic regions in the 125P5C8 structure. Regions of a 125P5C8 protein that show immunogenic structure, as well as other regions and domains, can readily be identified using various other methods known in the art, such as Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis. Hydrophilicity profiles can be generated using the method of Hopp, T. P. and Woods, K. R., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity profiles can be generated using the method of Kyte, J. and Doolittle, R. F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated using the method of Bhaskaran R., Ponnuswamy P. K., 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294. Thus, each region identified by any of these programs or methods is within the scope of the present invention. Methods for the generation of 125P5C8 antibodies are further illustrated by way of the examples provided herein. Methods for preparing a protein or polypeptide for use as an immunogen are well known in the art. Also well known in the art are methods for preparing immunogenic conjugates of a protein with a carrier, such as BSA, KLH or other carrier protein. In some circumstances, direct conjugation using, for example, carbodiimide reagents are used; in other instances linking reagents such as those supplied by Pierce Chemical Co., Rockford, Ill., are effective. Administration of a 125P5C8 immunogen is often conducted by injection over a suitable time period and with use of a suitable adjuvant, as is understood in the art. During the immunization schedule, titers of antibodies can be taken to determine adequacy of antibody formation.


125P5C8 monoclonal antibodies can be produced by various means well known in the art. For example, immortalized cell lines that secrete a desired monoclonal antibody are prepared using the standard hybridoma technology of Kohler and Milstein or modifications that immortalize antibody-producing B cells, as is generally known. Immortalized cell lines that secrete the desired antibodies are screened by immunoassay in which the antigen is a 125P5C8-related protein. When the appropriate immortalized cell culture is identified, the cells can be expanded and antibodies produced either from in vitro cultures or from ascites fluid.


The antibodies or fragments of the invention can also be produced, by recombinant means. Regions that bind specifically to the desired regions of a 125P5C8 protein can also be produced in the context of chimeric or complementarity determining region (CDR) grafted antibodies of multiple species origin. Humanized or human 125P5C8 antibodies can also be produced, and are preferred for use in therapeutic contexts. Methods for humanizing murine and other non-human antibodies, by substituting one or more of the non-human antibody CDRs for corresponding human antibody sequences, are well known (see for example, Jones et al., 1986, Nature 321: 522-525; Riechmann et al., 1988, Nature 332: 323-327; Verhoeyen et al., 1988, Science 239: 1534-1536). See also, Carter et al., 1993, Proc. Natl. Acad. Sci. USA 89: 4285 and Sims et al., 1993, J. Immunol. 151: 2296.


Methods for producing fully human monoclonal antibodies include phage display and transgenic methods (for review, see Vaughan et al., 1998, Nature Biotechnology 16: 535-539). Fully human 125P5C8 monoclonal antibodies can be generated using cloning technologies employing large human Ig gene combinatorial libraries (i.e., phage display) (Griffiths and Hoogenboom, Building an in vitro immune system: human antibodies from phage display libraries. In: Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man, Clark, M. (Ed.), Nottingham Academic, pp 45-64 (1993); Burton and Barbas, Human Antibodies from combinatorial libraries. Id., pp 65-82). Fully human 125P5C8 monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application WO98/24893, Kucherlapati and Jakobovits et al., published Dec. 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 7(4): 607-614; U.S. Pat. Nos. 6,162,963 issued 19 Dec. 2000; 6,150,584 issued 12 Nov. 2000; and, 6,114,598 issued 5 Sep. 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.


Reactivity of 125P5C8 antibodies with a 125P5C8-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 125P5C8-related proteins, 125P5C8-expressing cells or extracts thereof. A 125P5C8 antibody or fragment thereof can be labeled with a detectable marker or conjugated to a second molecule. Suitable detectable markers include, but are not limited to, a radioisotope, a fluorescent compound, a bioluminescent compound, chemiluminescent compound, a metal chelator or an enzyme. Further, bi-specific antibodies specific for two or more 125P5C8 epitopes are generated using methods generally known in the art. Homodimeric antibodies can also be generated by cross-linking techniques known in the art (e.g., Wolff et al., Cancer Res. 53: 2560-2565).


V.) 125P5C8 Cellular Immune Responses


The mechanism by which T cells recognize antigens has been delineated. Efficacious peptide epitope vaccine compositions of the invention induce a therapeutic or prophylactic immune responses in very broad segments of the world-wide population. For an understanding of the value and efficacy of compositions of the invention that induce cellular immune responses, a brief review of immunology-related technology is provided.


A complex of an HLA molecule and a peptidic antigen acts as the ligand recognized by HLA-restricted T cells (Buus, S. et al., Cell 47:1071, 1986; Babbitt, B. P. et al., Nature 317:359, 1985; Townsend, A. and Bodmer, H., Annu. Rev. Immunol. 7:601, 1989; Germain, R. N., Annu. Rev. Immunol. 11:403, 1993). Through the study of single amino acid substituted antigen analogs and the sequencing of endogenously bound, naturally processed peptides, critical residues that correspond to motifs required for specific binding to HLA antigen molecules have been identified and are set forth in Table IV (see also, e.g., Southwood, et al., J. Immunol. 160:3363, 1998; Rammensee, et al., Immunogenetics 41:178, 1995; Rammensee et al., SYFPEITHI, access via World Wide Web at URL syfpeithi.bmi-heidelberg.com/; Sette, A. and Sidney, J. Curr. Opin. Immunol. 10:478, 1998; Engelhard, V. H., Curr. Opin. Immunol. 6:13, 1994; Sette, A. and Grey, H. M., Curr. Opin. Immunol. 4:79, 1992; Sinigaglia, F. and Hammer, J. Curr. Biol. 6:52, 1994; Ruppert et al., Cell 74:929-937, 1993; Kondo et al., J. Immunol. 155:4307-4312, 1995; Sidney et al., J. Immunol. 157:3480-3490, 1996; Sidney et al., Human Immunol. 45:79-93, 1996; Sette, A. and Sidney, J. Immunogenetics 1999 November; 50(3-4):201-12, Review).


Furthermore, x-ray crystallographic analyses of HLA-peptide complexes have revealed pockets within the peptide binding cleft/groove of HLA molecules which accommodate, in an allele-specific mode, residues borne by peptide ligands; these residues in turn determine the HLA binding capacity of the peptides in which they are present. (See, e.g., Madden, D. R. Annu. Rev. Immunol. 13:587, 1995; Smith, et al., Immunity 4:203, 1996; Fremont et al., Immunity 8:305, 1998; Stern et al., Structure 2:245, 1994; Jones, E. Y. Curr. Opin. Immunol. 9:75, 1997; Brown, J. H. et al., Nature 364:33, 1993; Guo, H. C. et al., Proc. Natl. Acad. Sci. USA 90:8053, 1993; Guo, H. C. et al., Nature 360:364, 1992; Silver, M. L. et al., Nature 360:367, 1992; Matsumura, M. et al., Science 257:927, 1992; Madden et al., Cell 70:1035, 1992; Fremont, D. H. et al., Science 257:919, 1992; Saper, M. A., Bjorkman, P. J. and Wiley, D.C., J. Mol. Biol. 219:277, 1991.)


Accordingly, the definition of class I and class II allele-specific HLA binding motifs, or class I or class II supermotifs allows identification of regions within a protein that are correlated with binding to particular HLA antigen(s).


Thus, by a process of HLA motif identification, candidates for epitope-based vaccines have been identified; such candidates can be further evaluated by HLA-peptide binding assays to determine binding affinity and/or the time period of association of the epitope and its corresponding HLA molecule. Additional confirmatory work can be performed to select, amongst these vaccine candidates, epitopes with preferred characteristics in terms of population coverage, and/or immunogenicity.


Various strategies can be utilized to evaluate cellular immunogenicity, including:


1) Evaluation of primary T cell cultures from normal individuals (see, e.g., Wentworth, P. A. et al., Mol. Immunol. 32:603, 1995; Celis, E. et al., Proc. Natl. Acad. Sci. USA 91:2105, 1994; Tsai, V. et al., J. Immunol. 158:1796, 1997; Kawashima, I. et al., Human Immunol. 59:1, 1998). This procedure involves the stimulation of peripheral blood lymphocytes (PBL) from normal subjects with a test peptide in the presence of antigen presenting cells in vitro over a period of several weeks. T cells specific for the peptide become activated during this time and are detected using, e.g., a lymphokine- or 51Cr-release assay involving peptide sensitized target cells.


2) Immunization of HLA transgenic mice (see, e.g., Wentworth, P. A. et al., J. Immunol. 26:97, 1996; Wentworth, P. A. et al., Int. Immunol. 8:651, 1996; Alexander, J. et al., J. Immunol. 159:4753, 1997). For example, in such methods peptides in incomplete Freund's adjuvant are administered subcutaneously to HLA transgenic mice. Several weeks following immunization, splenocytes are removed and cultured in vitro in the presence of test peptide for approximately one week. Peptide-specific T cells are detected using, e.g., a 51Cr-release assay involving peptide sensitized target cells and target cells expressing endogenously generated antigen.


3) Demonstration of recall T cell responses from immune individuals who have been either effectively vaccinated and/or from chronically ill patients (see, e.g., Rehermann, B. et al., J. Exp. Med. 181:1047, 1995; Doolan, D. L. et al., Immunity 7:97, 1997; Bertoni, R. et al., J. Clin. Invest. 100:503, 1997; Threlkeld, S. C. et al., J. Immunol. 159:1648, 1997; Diepolder, H. M. et al., J. Virol. 71:6011, 1997). Accordingly, recall responses are detected by culturing PBL from subjects that have been exposed to the antigen due to disease and thus have generated an immune response “naturally”, or from patients who were vaccinated against the antigen. PBL from subjects are cultured in vitro for 1-2 weeks in the presence of test peptide plus antigen presenting cells (APC) to allow activation of “memory” T cells, as compared to “naive” T cells. At the end of the culture period, T cell activity is detected using assays including 51Cr release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.


VI.) 125P5C8 Transgenic Animals


Nucleic acids that encode a 125P5C8-related protein can also be used to generate either transgenic animals or “knock out” animals that, in turn, are useful in the development and screening of therapeutically useful reagents. In accordance with established techniques, cDNA encoding 125P5C8 can be used to clone genomic DNA that encodes 125P5C8. The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode 125P5C8. Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 issued 12 Apr. 1988, and 4,870,009 issued 26 Sep. 1989. Typically, particular cells would be targeted for 125P5C8 transgene incorporation with tissue-specific enhancers.


Transgenic animals that include a copy of a transgene encoding 125P5C8 can be used to examine the effect of increased expression of DNA that encodes 125P5C8. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression. In accordance with this aspect of the invention, an animal is treated with a reagent and a reduced incidence of a pathological condition, compared to untreated animals that bear the transgene, would indicate a potential therapeutic intervention for the pathological condition.


Alternatively, non-human homologues of 125P5C8 can be used to construct a 125P5C8 “knock out” animal that has a defective or altered gene encoding 125P5C8 as a result of homologous recombination between the endogenous gene encoding 125P5C8 and altered genomic DNA encoding 125P5C8 introduced into an embryonic cell of the animal. For example, cDNA that encodes 125P5C8 can be used to clone genomic DNA encoding 125P5C8 in accordance with established techniques. A portion of the genomic DNA encoding 125P5C8 can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5′ and 3′ ends) are included in the vector (see, e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors). The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected (see, e.g., Li et al., Cell, 69:915 (1992)). The selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras (see, e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal, and the embryo brought to term to create a “knock out” animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knock out animals can be characterized, for example, for their ability to defend against certain pathological conditions or for their development of pathological conditions due to absence of a 125P5C8 polypeptide.


VII.) Methods for the Detection of 125P5C8


Another aspect of the present invention relates to methods for detecting 125P5C8 polynucleotides and 125P5C8-related proteins, as well as methods for identifying a cell that expresses 125P5C8. The expression profile of 125P5C8 makes it a diagnostic marker for metastasized disease. Accordingly, the status of 125P5C8 gene products provides information useful for predicting a variety of factors including susceptibility to advanced stage disease, rate of progression, and/or tumor aggressiveness. As discussed in detail herein, the status of 125P5C8 gene products in patient samples can be analyzed by a variety protocols that are well known in the art including immunohistochemical analysis, the variety of Northern blotting techniques including in situ hybridization, RT-PCR analysis (for example on laser capture micro-dissected samples), Western blot analysis and tissue array analysis.


More particularly, the invention provides assays for the detection of 125P5C8 polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like. Detectable 125P5C8 polynucleotides include, for example, a 125P5C8 gene or fragment thereof, 125P5C8 mRNA, alternative splice variant 125P5C8 mRNAs, and recombinant DNA or RNA molecules that contain a 125P5C8 polynucleotide. A number of methods for amplifying and/or detecting the presence of 125P5C8 polynucleotides are well known in the art and can be employed in the practice of this aspect of the invention.


In one embodiment, a method for detecting a 125P5C8 mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a 125P5C8 polynucleotides as sense and antisense primers to amplify 125P5C8 cDNAs therein; and detecting the presence of the amplified 125P5C8 cDNA. Optionally, the sequence of the amplified 125P5C8 cDNA can be determined.


In another embodiment, a method of detecting a 125P5C8 gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using 125P5C8 polynucleotides as sense and antisense primers; and detecting the presence of the amplified 125P5C8 gene. Any number of appropriate sense and antisense probe combinations can be designed from a 125P5C8 nucleotide sequence (see, e.g., FIG. 2) and used for this purpose.


The invention also provides assays for detecting the presence of a 125P5C8 protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like. Methods for detecting a 125P5C8-related protein are also well known and include, for example, immunoprecipitation, immunohistochemical analysis, Western blot analysis, molecular binding assays, ELISA, ELIFA and the like. For example, a method of detecting the presence of a 125P5C8-related protein in a biological sample comprises first contacting the sample with a 125P5C8 antibody, a 125P5C8-reactive fragment thereof, or a recombinant protein containing an antigen binding region of a 125P5C8 antibody; and then detecting the binding of 125P5C8-related protein in the sample.


Methods for identifying a cell that expresses 125P5C8 are also within the scope of the invention. In one embodiment, an assay for identifying a cell that expresses a 125P5C8 gene comprises detecting the presence of 125P5C8 mRNA in the cell. Methods for the detection of particular mRNAs in cells are well known and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled 125P5C8 riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for 125P5C8, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like). Alternatively, an assay for identifying a cell that expresses a 125P5C8 gene comprises detecting the presence of 125P5C8-related protein in the cell or secreted by the cell. Various methods for the detection of proteins are well known in the art and are employed for the detection of 125P5C8-related proteins and cells that express 125P5C8-related proteins.


125P5C8 expression analysis is also useful as a tool for identifying and evaluating agents that modulate 125P5C8 gene expression. For example, 125P5C8 expression is significantly upregulated in prostate cancer, and is expressed in cancers of the tissues listed in Table I. Identification of a molecule or biological agent that inhibits 125P5C8 expression or over-expression in cancer cells is of therapeutic value. For example, such an agent can be identified by using a screen that quantifies 125P5C8 expression by RT-PCR, nucleic acid hybridization or antibody binding.


VIII.) Methods for Monitoring the Status of 125P5C8-Related Genes and Their Products


Oncogenesis is known to be a multistep process where cellular growth becomes progressively dysregulated and cells progress from a normal physiological state to precancerous and then cancerous states (see, e.g., Alers et al., Lab Invest. 77(5): 437-438 (1997) and Isaacs et al., Cancer Surv. 23:19-32 (1995)). In this context, examining a biological sample for evidence of dysregulated cell growth (such as aberrant 125P5C8 expression in cancers) allows for early detection of such aberrant physiology, before a pathologic state such as cancer has progressed to a stage that therapeutic options are more limited and or the prognosis is worse. In such examinations, the status of 125P5C8 in a biological sample of interest can be compared, for example, to the status of 125P5C8 in a corresponding normal sample (e.g. a sample from that individual or alternatively another individual that is not affected by a pathology). An alteration in the status of 125P5C8 in the biological sample (as compared to the normal sample) provides evidence of dysregulated cellular growth. In addition to using a biological sample that is not affected by a pathology as a normal sample, one can also use a predetermined normative value such as a predetermined normal level of mRNA expression (see, e.g., Grever et al., J. Comp. Neurol. 1996 Dec. 9; 376(2): 306-14 and U.S. Pat. No. 5,837,501) to compare 125P5C8 status in a sample.


The term “status” in this context is used according to its art accepted meaning and refers to the condition or state of a gene and its products. Typically, skilled artisans use a number of parameters to evaluate the condition or state of a gene and its products. These include, but are not limited to the location of expressed gene products (including the location of 125P5C8 expressing cells) as well as the level, and biological activity of expressed gene products (such as 125P5C8 mRNA, polynucleotides and polypeptides). Typically, an alteration in the status of 125P5C8 comprises a change in the location of 125P5C8 and/or 125P5C8 expressing cells and/or an increase in 125P5C8 mRNA and/or protein expression.


125P5C8 status in a sample can be analyzed by a number of means well known in the art, including without limitation, immunohistochemical analysis, in situ hybridization, RT-PCR analysis on laser capture micro-dissected samples, Western blot analysis, and tissue array analysis. Typical protocols for evaluating the status of a 125P5C8 gene and gene products are found, for example in Ausubel et al. eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis). Thus, the status of 125P5C8 in a biological sample is evaluated by various methods utilized by skilled artisans including, but not limited to genomic Southern analysis (to examine, for example perturbations in a 125P5C8 gene), Northern analysis and/or PCR analysis of 125P5C8 mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of 125P5C8 mRNAs), and, Western and/or immunohistochemical analysis (to examine, for example alterations in polypeptide sequences, alterations in polypeptide localization within a sample, alterations in expression levels of 125P5C8 proteins and/or associations of 125P5C8 proteins with polypeptide binding partners). Detectable 125P5C8 polynucleotides include, for example, a 125P5C8 gene or fragment thereof, 125P5C8 mRNA, alternative splice variants, 125P5C8 mRNAs, and recombinant DNA or RNA molecules containing a 125P5C8 polynucleotide.


The expression profile of 125P5C8 makes it a diagnostic marker for local and/or metastasized disease, and provides information on the growth or oncogenic potential of a biological sample. In particular, the status of 125P5C8 provides information useful for predicting susceptibility to particular disease stages, progression, and/or tumor aggressiveness. The invention provides methods and assays for determining 125P5C8 status and diagnosing cancers that express 125P5C8, such as cancers of the tissues listed in Table I. For example, because 125P5C8 mRNA is so highly expressed in prostate and other cancers relative to normal prostate tissue, assays that evaluate the levels of 125P5C8 mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with 125P5C8 dysregulation, and can provide prognostic information useful in defining appropriate therapeutic options.


The expression status of 125P5C8 provides information including the presence, stage and location of dysplastic, precancerous and cancerous cells, predicting susceptibility to various stages of disease, and/or for gauging tumor aggressiveness. Moreover, the expression profile makes it useful as an imaging reagent for metastasized disease. Consequently, an aspect of the invention is directed to the various molecular prognostic and diagnostic methods for examining the status of 125P5C8 in biological samples such as those from individuals suffering from, or suspected of suffering from a pathology characterized by dysregulated cellular growth, such as cancer.


As described above, the status of 125P5C8 in a biological sample can be examined by a number of well-known procedures in the art. For example, the status of 125P5C8 in a biological sample taken from a specific location in the body can be examined by evaluating the sample for the presence or absence of 125P5C8 expressing cells (e.g. those that express 125P5C8 mRNAs or proteins). This examination can provide evidence of dysregulated cellular growth, for example, when 125P5C8-expressing cells are found in a biological sample that does not normally contain such cells (such as a lymph node), because such alterations in the status of 125P5C8 in a biological sample are often associated with dysregulated cellular growth. Specifically, one indicator of dysregulated cellular growth is the metastases of cancer cells from an organ of origin (such as the prostate) to a different area of the body (such as a lymph node). In this context, evidence of dysregulated cellular growth is important for example because occult lymph node metastases can be detected in a substantial proportion of patients with prostate cancer, and such metastases are associated with known predictors of disease progression (see, e.g., Murphy et al., Prostate 42(4): 315-317 (2000);Su et al., Semin. Surg. Oncol. 18(1): 17-28 (2000) and Freeman et al., J Urol 1995 August 154(2 Pt 1):474-8).


In one aspect, the invention provides methods for monitoring 125P5C8 gene products by determining the status of 125P5C8 gene products expressed by cells from an individual suspected of having a disease associated with dysregulated cell growth (such as hyperplasia or cancer) and then comparing the status so determined to the status of 125P5C8 gene products in a corresponding normal sample. The presence of aberrant 125P5C8 gene products in the test sample relative to the normal sample provides an indication of the presence of dysregulated cell growth within the cells of the individual.


In another aspect, the invention provides assays useful in determining the presence of cancer in an individual, comprising detecting a significant increase in 125P5C8 mRNA or protein expression in a test cell or tissue sample relative to expression levels in the corresponding normal cell or tissue. The presence of 125P5C8 mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I. The presence of significant 125P5C8 expression in any of these tissues is useful to indicate the emergence, presence and/or severity of a cancer, since the corresponding normal tissues do not express 125P5C8 mRNA or express it at lower levels.


In a related embodiment, 125P5C8 status is determined at the protein level rather than at the nucleic acid level. For example, such a method comprises determining the level of 125P5C8 protein expressed by cells in a test tissue sample and comparing the level so determined to the level of 125P5C8 expressed in a corresponding normal sample. In one embodiment, the presence of 125P5C8 protein is evaluated, for example, using immunohistochemical methods. 125P5C8 antibodies or binding partners capable of detecting 125P5C8 protein expression are used in a variety of assay formats well known in the art for this purpose.


In a further embodiment, one can evaluate the status of 125P5C8 nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules. These perturbations can include insertions, deletions, substitutions and the like. Such evaluations are useful because perturbations in the nucleotide and amino acid sequences are observed in a large number of proteins associated with a growth dysregulated phenotype (see, e.g., Marrogi et al., 1999, J. Cutan. Pathol. 26(8):369-378). For example, a mutation in the sequence of 125P5C8 may be indicative of the presence or promotion of a tumor. Such assays therefore have diagnostic and predictive value where a mutation in 125P5C8 indicates a potential loss of function or increase in tumor growth.


A wide variety of assays for observing perturbations in nucleotide and amino acid sequences are well known in the art. For example, the size and structure of nucleic acid or amino acid sequences of 125P5C8 gene products are observed by the Northern, Southern, Western, PCR and DNA sequencing protocols discussed herein. In addition, other methods for observing perturbations in nucleotide and amino acid sequences such as single strand conformation polymorphism analysis are well known in the art (see, e.g., U.S. Pat. Nos. 5,382,510 issued 7 Sep. 1999, and 5,952,170 issued 17 Jan. 1995).


Additionally, one can examine the methylation status of a 125P5C8 gene in a biological sample. Aberrant demethylation and/or hypermethylation of CpG islands in gene 5′ regulatory regions frequently occurs in immortalized and transformed cells, and can result in altered expression of various genes. For example, promoter hypermethylation of the pi-class glutathione S-transferase (a protein expressed in normal prostate but not expressed in >90% of prostate carcinomas) appears to permanently silence transcription of this gene and is the most frequently detected genomic alteration in prostate carcinomas (De Marzo et al., Am. J. Pathol. 155(6): 1985-1992 (1999)). In addition, this alteration is present in at least 70% of cases of high-grade prostatic intraepithelial neoplasia (PIN) (Brooks et al., Cancer Epidemiol. Biomarkers Prev., 1998, 7:531-536). In another example, expression of the LAGE-I tumor specific gene (which is not expressed in normal prostate but is expressed in 25-50% of prostate cancers) is induced by deoxy-azacytidine in lymphoblastoid cells, suggesting that tumoral expression is due to demethylation (Lethe et al., Int. J. Cancer 76(6): 903-908 (1998)). A variety of assays for examining methylation status of a gene are well known in the art. For example, one can utilize, in Southern hybridization approaches, methylation-sensitive restriction enzymes that cannot cleave sequences that contain methylated CpG sites to assess the methylation status of CpG islands. In addition, MSP (methylation specific PCR) can rapidly profile the methylation status of all the CpG sites present in a CpG island of a given gene. This procedure involves initial modification of DNA by sodium bisulfite (which will convert all unmethylated cytosines to uracil) followed by amplification using primers specific for methylated versus unmethylated DNA. Protocols involving methylation interference can also be found for example in Current Protocols In Molecular Biology, Unit 12, Frederick M. Ausubel et al. eds., 1995.


Gene amplification is an additional method for assessing the status of 125P5C8. Gene amplification is measured in a sample directly, for example, by conventional Southern blotting or Northern blotting to quantitate the transcription of mRNA (Thomas, 1980, Proc. Natl. Acad. Sci. USA, 77:5201-5205), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies are employed that recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn are labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.


Biopsied tissue or peripheral blood can be conveniently assayed for the presence of cancer cells using for example, Northern, dot blot or RT-PCR analysis to detect 125P5C8 expression. The presence of RT-PCR amplifiable 125P5C8 mRNA provides an indication of the presence of cancer. RT-PCR assays are well known in the art. RT-PCR detection assays for tumor cells in peripheral blood are currently being evaluated for use in the diagnosis and management of a number of human solid tumors. In the prostate cancer field, these include RT-PCR assays for the detection of cells expressing PSA and PSM (Verkaik et al., 1997, Urol. Res. 25:373-384; Ghossein et al., 1995, J. Clin. Oncol. 13:1195-2000; Heston et al., 1995, Clin. Chem. 41:1687-1688).


A further aspect of the invention is an assessment of the susceptibility that an individual has for developing cancer. In one embodiment, a method for predicting susceptibility to cancer comprises detecting 125P5C8 mRNA or 125P5C8 protein in a tissue sample, its presence indicating susceptibility to cancer, wherein the degree of 125P5C8 mRNA expression correlates to the degree of susceptibility. In a specific embodiment, the presence of 125P5C8 in prostate or other tissue is examined, with the presence of 125P5C8 in the sample providing an indication of prostate cancer susceptibility (or the emergence or existence of a prostate tumor). Similarly, one can evaluate the integrity 125P5C8 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations in 125P5C8 gene products in the sample is an indication of cancer susceptibility (or the emergence or existence of a tumor).


The invention also comprises methods for gauging tumor aggressiveness. In one embodiment, a method for gauging aggressiveness of a tumor comprises determining the level of 125P5C8 mRNA or 125P5C8 protein expressed by tumor cells, comparing the level so determined to the level of 125P5C8 mRNA or 125P5C8 protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of 125P5C8 mRNA or 125P5C8 protein expression in the tumor sample relative to the normal sample indicates the degree of aggressiveness. In a specific embodiment, aggressiveness of a tumor is evaluated by determining the extent to which 125P5C8 is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors. Another embodiment is the evaluation of the integrity of 125P5C8 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations indicates more aggressive tumors.


Another embodiment of the invention is directed to methods for observing the progression of a malignancy in an individual over time. In one embodiment, methods for observing the progression of a malignancy in an individual over time comprise determining the level of 125P5C8 mRNA or 125P5C8 protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of 125P5C8 mRNA or 125P5C8 protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of 125P5C8 mRNA or 125P5C8 protein expression in the tumor sample over time provides information on the progression of the cancer. In a specific embodiment, the progression of a cancer is evaluated by determining 125P5C8 expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity 125P5C8 nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like, where the presence of one or more perturbations indicates a progression of the cancer.


The above diagnostic approaches can be combined with any one of a wide variety of prognostic and diagnostic protocols known in the art. For example, another embodiment of the invention is directed to methods for observing a coincidence between the expression of 125P5C8 gene and 125P5C8 gene products (or perturbations in 125P5C8 gene and 125P5C8 gene products) and a factor that is associated with malignancy, as a means for diagnosing and prognosticating the status of a tissue sample. A wide variety of factors associated with malignancy can be utilized, such as the expression of genes associated with malignancy (e.g. PSA, PSCA and PSM expression for prostate cancer etc.) as well as gross cytological observations (see, e.g., Bocking et al., 1984, Anal. Quant. Cytol. 6(2):74-88; Epstein, 1995, Hum. Pathol. 26(2):223-9; Thorson et al., 1998, Mod. Pathol. 11(6):543-51; Baisden et al., 1999, Am. J. Surg. Pathol. 23(8):918-24). Methods for observing a coincidence between the expression of 125P5C8 gene and 125P5C8 gene products (or perturbations in 125P5C8 gene and 125P5C8 gene products) and another factor that is associated with malignancy are useful, for example, because the presence of a set of specific factors that coincide with disease provides information crucial for diagnosing and prognosticating the status of a tissue sample.


In one embodiment, methods for observing a coincidence between the expression of 125P5C8 gene and 125P5C8 gene products (or perturbations in 125P5C8 gene and 125P5C8 gene products) and another factor associated with malignancy entails detecting the overexpression of 125P5C8 mRNA or protein in a tissue sample, detecting the overexpression of PSA mRNA or protein in a tissue sample (or PSCA or PSM expression), and observing a coincidence of 125P5C8 mRNA or protein and PSA mRNA or protein overexpression (or PSCA or PSM expression). In a specific embodiment, the expression of 125P5C8 and PSA mRNA in prostate tissue is examined, where the coincidence of 125P5C8 and PSA mRNA overexpression in the sample indicates the existence of prostate cancer, prostate cancer susceptibility or the emergence or status of a prostate tumor.


Methods for detecting and quantifying the expression of 125P5C8 mRNA or protein are described herein, and standard nucleic acid and protein detection and quantification technologies are well known in the art. Standard methods for the detection and quantification of 125P5C8 mRNA include in situ hybridization using labeled 125P5C8 riboprobes, Northern blot and related techniques using 125P5C8 polynucleotide probes, RT-PCR analysis using primers specific for 125P5C8, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like. In a specific embodiment, semi-quantitative RT-PCR is used to detect and quantify 125P5C8 mRNA expression. Any number of primers capable of amplifying 125P5C8 can be used for this purpose, including but not limited to the various primer sets specifically described herein. In a specific embodiment, polyclonal or monoclonal antibodies specifically reactive with the wild-type 125P5C8 protein can be used in an immunohistochemical assay of biopsied tissue.


IX.) Identification of Molecules that Interact with 125P5C8


The 125P5C8 protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with 125P5C8, as well as pathways activated by 125P5C8 via any one of a variety of art accepted protocols. For example, one can utilize one of the so-called interaction trap systems (also referred to as the “two-hybrid assay”). In such systems, molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed. Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. Nos. 5,955,280 issued 21 Sep. 1999, 5,925,523 issued 20 Jul. 1999, 5,846,722 issued 8 Dec. 1998 and 6,004,746 issued 21 Dec. 1999. Algorithms are also available in the art for genome-based predictions of protein function (see, e.g., Marcotte, et al., Nature 402: 4 Nov. 1999, 83-86).


Alternatively one can screen peptide libraries to identify molecules that interact with 125P5C8 protein sequences. In such methods, peptides that bind to 125P5C8 are identified by screening libraries that encode a random or controlled collection of amino acids. Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the 125P5C8 protein(s).


Accordingly, peptides having a wide variety of uses, such as therapeutic, prognostic or diagnostic reagents, are thus identified without any prior information on the structure of the expected ligand or receptor molecule. Typical peptide libraries and screening methods that can be used to identify molecules that interact with 125P5C8 protein sequences are disclosed for example in U.S. Pat. Nos. 5,723,286 issued 3 Mar. 1998 and 5,733,731 issued 31 Mar. 1998.


Alternatively, cell lines that express 125P5C8 are used to identify protein-protein interactions mediated by 125P5C8. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun. 1999, 261:646-51). 125P5C8 protein can be immunoprecipitated from 125P5C8-expressing cell lines using anti-125P5C8 antibodies. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of 125P5C8 and a His-tag (vectors mentioned above). The immunoprecipitated complex can be examined for protein association by procedures such as Western blotting, 35S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.


Small molecules and ligands that interact with 125P5C8 can be identified through related embodiments of such screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with 125P5C8's ability to mediate phosphorylation and de-phosphorylation, interaction with DNA or RNA molecules as an indication of regulation of cell cycles, second messenger signaling or tumorigenesis. Similarly, small molecules that modulate 125P5C8-related ion channel, protein pump, or cell communication functions are identified and used to treat patients that have a cancer that expresses 125P5C8 (see, e.g., Hille, B., Ionic Channels of Excitable Membranes 2nd Ed., Sinauer Assoc., Sunderland, Mass., 1992). Moreover, ligands that regulate 125P5C8 function can be identified based on their ability to bind 125P5C8 and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued 27 Jul. 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule. In an illustrative embodiment, cells engineered to express a fusion protein of 125P5C8 and a DNA-binding protein are used to co-express a fusion protein of a hybrid ligand/small molecule and a cDNA library transcriptional activator protein. The cells further contain a reporter gene, the expression of which is conditioned on the proximity of the first and second fusion proteins to each other, an event that occurs only if the hybrid ligand binds to target sites on both hybrid proteins. Those cells that express the reporter gene are selected and the unknown small molecule or the unknown ligand is identified. This method provides a means of identifying modulators which activate or inhibit 125P5C8.


An embodiment of this invention comprises a method of screening for a molecule that interacts with a 125P5C8 amino acid sequence shown in FIG. 2 or FIG. 3, comprising the steps of contacting a population of molecules with a 125P5C8 amino acid sequence, allowing the population of molecules and the 125P5C8 amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the 125P5C8 amino acid sequence, and then separating molecules that do not interact with the 125P5C8 amino acid sequence from molecules that do. In a specific embodiment, the method further comprises purifying, characterizing and identifying a molecule that interacts with the 125P5C8 amino acid sequence. The identified molecule can be used to modulate a function performed by 125P5C8. In a preferred embodiment, the 125P5C8 amino acid sequence is contacted with a library of peptides.


X.) Therapeutic Methods and Compositions


The identification of 125P5C8 as a protein that is normally expressed in a restricted set of tissues, but which is also expressed in prostate and other cancers, opens a number of therapeutic approaches to the treatment of such cancers. As contemplated herein, 125P5C8 functions as a transcription factor involved in activating tumor-promoting genes or repressing genes that block tumorigenesis.


Accordingly, therapeutic approaches that inhibit the activity of a 125P5C8 protein are useful for patients suffering from a cancer that expresses 125P5C8. These therapeutic approaches generally fall into two classes. One class comprises various methods for inhibiting the binding or association of a 125P5C8 protein with its binding partner or with other proteins. Another class comprises a variety of methods for inhibiting the transcription of a 125P5C8 gene or translation of 125P5C8 mRNA.


X.A.) Anti-Cancer Vaccines


The invention provides cancer vaccines comprising a 125P5C8-related protein or 125P5C8-related nucleic acid. In view of the expression of 125P5C8, cancer vaccines prevent and/or treat 125P5C8-expressing cancers with minimal or no effects on non-target tissues. The use of a tumor antigen in a vaccine that generates humoral and/or cell-mediated immune responses as anti-cancer therapy is well known in the art and has been employed in prostate cancer using human PSMA and rodent PAP immunogens (Hodge et al., 1995, Int. J. Cancer 63:231-237; Fong et al., 1997, J. Immunol. 159:3113-3117).


Such methods can be readily practiced by employing a 125P5C8-related protein, or a 125P5C8-encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the 125P5C8 immunogen (which typically comprises a number of antibody or T cell epitopes). Skilled artisans understand that a wide variety of vaccine systems for delivery of immunoreactive epitopes are known in the art (see, e.g., Heryln et al., Ann Med 1999 February 31(1):66-78; Maruyama et al., Cancer Immunol Immunother 2000 June 49(3): 123-32) Briefly, such methods of generating an immune response (e.g. humoral and/or cell-mediated) in a mammal, comprise the steps of: exposing the mammal's immune system to an immunoreactive epitope (e.g. an epitope present in a 125P5C8 protein shown in FIG. 3 or analog or homolog thereof) so that the mammal generates an immune response that is specific for that epitope (e.g. generates antibodies that specifically recognize that epitope). In a preferred method, a 125P5C8 immunogen contains a biological motif, see e.g., Tables V-XVIII, or a peptide of a size range from 125P5C8 indicated in FIG. 5, FIG. 6, FIG. 7, FIG. 8, and FIG. 9.


The entire 125P5C8 protein, immunogenic regions or epitopes thereof can be combined and delivered by various means. Such vaccine compositions can include, for example, lipopeptides (e.g., Vitiello, A. et al., J. Clin. Invest. 95:341, 1995), peptide compositions encapsulated in poly(DL-lactide-co-glycolide) (“PLG”) microspheres (see, e.g., Eldridge, et al., Molec. Immunol. 28:287-294, 1991: Alonso et al., Vaccine 12:299-306, 1994; Jones et al., Vaccine 13:675-681, 1995), peptide compositions contained in immune stimulating complexes (ISCOMS) (see, e.g., Takahashi et al., Nature 344:873-875, 1990; Hu et al., Clin Exp Immunol. 113:235-243, 1998), multiple antigen peptide systems (MAPs) (see e.g., Tam, J. P., Proc. Natl. Acad. Sci. U.S.A. 85:5409-5413, 1988; Tam, J. P., J. Immunol. Methods 196:17-32, 1996), peptides formulated as multivalent peptides; peptides for use in ballistic delivery systems, typically crystallized peptides, viral delivery vectors (Perkus, M. E. et al., In: Concepts in vaccine development, Kaufmann, S. H. E., ed., p. 379, 1996; Chakrabarti, S. et al., Nature 320:535, 1986; Hu, S. L. et al., Nature 320:537, 1986; Kieny, M.-P. et al., AIDS Bio/Technology 4:790, 1986; Top, F. H. et al., J. Infect. Dis. 124:148, 1971; Chanda, P. K. et al., Virology 175:535, 1990), particles of viral or synthetic origin (e.g., Kofler, N. et al., J. Immunol. Methods. 192:25, 1996; Eldridge, J. H. et al., Sem. Hematol. 30:16, 1993; Falo, L. D., Jr. et al., Nature Med. 7:649, 1995), adjuvants (Warren, H. S., Vogel, F. R., and Chedid, L. A. Annu. Rev. Immunol. 4:369, 1986; Gupta, R. K. et al., Vaccine 11:293, 1993), liposomes (Reddy, R. et al., J. Immunol. 148:1585, 1992; Rock, K. L., Immunol. Today 17:131, 1996), or, naked or particle absorbed cDNA (Ulmer, J. B. et al., Science 259:1745, 1993; Robinson, H. L., Hunt, L. A., and Webster, R. G., Vaccine 11:957, 1993; Shiver, J. W. et al., In: Concepts in vaccine development, Kaufmann, S. H. E., ed., p. 423, 1996; Cease, K. B., and Berzofsky, J. A., Annu. Rev. Immunol. 12:923, 1994 and Eldridge, J. H. et al., Sem. Hematol. 30:16, 1993). Toxin-targeted delivery technologies, also known as receptor mediated targeting, such as those of Avant Immunotherapeutics, Inc. (Needham, Mass.) may also be used.


In patients with 125P5C8-associated cancer, the vaccine compositions of the invention can also be used in conjunction with other treatments used for cancer, e.g., surgery, chemotherapy, drug therapies, radiation therapies, etc. including use in combination with immune adjuvants such as IL-2, IL-12, GM-CSF, and the like.


Cellular Vaccines:


CTL epitopes can be determined using specific algorithms to identify peptides within 125P5C8 protein that bind corresponding HLA alleles (see e.g., Table IV; Epimer™ and Epimatrix™, Brown University and, BIMAS, and SYFPEITHI). In a preferred embodiment, a 125P5C8 immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables V-XIX or a peptide of 8, 9, 10 or 11 amino acids specified by an HLA Class I motif/supermotif (e.g., Table IV (A), Table IV (D), or Table IV (E)) and/or a peptide of at least 9 amino acids that comprises an HLA Class II motif/supermotif (e.g., Table IV (B) or Table IV (C)). As is appreciated in the art, the HLA Class I binding groove is essentially closed ended so that peptides of only a particular size range can fit into the groove and be bound, generally HLA Class I epitopes are 8, 9, 10, or 11 amino acids long. In contrast, the HLA Class II binding groove is essentially open ended; therefore a peptide of about 9 or more amino acids can be bound by an HLA Class II molecule. Due to the binding groove differences between HLA Class I and II, HLA Class I motifs are length specific, i.e., position two of a Class I motif is the second amino acid in an amino to carboxyl direction of the peptide. The amino acid positions in a Class II motif are relative only to each other, not the overall peptide, i.e., additional amino acids can be attached to the amino and/or carboxyl termini of a motif-bearing sequence. HLA Class II epitopes are often 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids long, or longer than 25 amino acids.


Antibody-Based Vaccines


A wide variety of methods for generating an immune response in a mammal are known in the art (for example as the first step in the generation of hybridomas). Methods of generating an immune response in a mammal comprise exposing the mammal's immune system to an immunogenic epitope on a protein (e.g. a 125P5C8 protein) so that an immune response is generated. A typical embodiment consists of a method for generating an immune response to 125P5C8 in a host, by contacting the host with a sufficient amount of at least one 125P5C8 B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the 125P5C8 B cell or cytotoxic T-cell epitope or analog thereof. A specific embodiment consists of a method of generating an immune response against a 125P5C8-related protein or a man-made multiepitopic peptide comprising: administering 125P5C8 immunogen (e.g. a 125P5C8 protein or a peptide fragment thereof, a 125P5C8 fusion protein or analog etc.) in a vaccine preparation to a human or another mammal. Typically, such vaccine preparations further contain a suitable adjuvant (see, e.g., U.S. Pat. No. 6,146,635) or a universal helper epitope such as a PADRE™ peptide (Epimmune Inc., San Diego, Calif.; see, e.g., Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res. 1998 18(2): 79-92). An alternative method comprises generating an immune response in an individual against a 125P5C8 immunogen by: administering in vivo to muscle or skin of the individual's body a DNA molecule that comprises a DNA sequence that encodes a 125P5C8 immunogen, the DNA sequence operatively linked to regulatory sequences which control the expression of the DNA sequence; wherein the DNA molecule is taken up by cells, the DNA sequence is expressed in the cells and an immune response is generated against the immunogen (see, e.g., U.S. Pat. No. 5,962,428). Optionally a genetic vaccine facilitator such as anionic lipids; saponins; lectins; estrogenic compounds; hydroxylated lower alkyls; dimethyl sulfoxide; and urea is also administered. In addition, an antiidiotypic antibody can be administered that mimics 125P5C8, in order to generate a response to the target antigen.


Nucleic Acid Vaccines:


Vaccine compositions of the invention include nucleic acid-mediated modalities. DNA or RNA that encode protein(s) of the invention can be administered to a patient. Genetic immunization methods can be employed to generate prophylactic or therapeutic humoral and cellular immune responses directed against cancer cells expressing 125P5C8. Constructs comprising DNA encoding a 125P5C8-related protein/immunogen and appropriate regulatory sequences can be injected directly into muscle or skin of an individual, such that the cells of the muscle or skin take-up the construct and express the encoded 125P5C8 protein/immunogen. Alternatively, a vaccine comprises a 125P5C8-related protein. Expression of the 125P5C8-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a 125P5C8 protein. Various prophylactic and therapeutic genetic immunization techniques known in the art can be used. Nucleic acid-based delivery is described, for instance, in Wolff et. al., Science 247:1465 (1990) as well as U.S. Pat. Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; WO 98/04720. Examples of DNA-based delivery technologies include “naked DNA”, facilitated (bupivacaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated (“gene gun”) or pressure-mediated delivery (see, e.g., U.S. Pat. No. 5,922,687).


For therapeutic or prophylactic immunization purposes, proteins of the invention can be expressed via viral or bacterial vectors. Various viral gene delivery systems that can be used in the practice of the invention include, but are not limited to, vaccinia, fowlpox, canarypox, adenovirus, influenza, poliovirus, adeno-associated virus, lentivirus, and sindbis virus (see, e.g., Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Inst. 87:982-990 (1995)). Non-viral delivery systems can also be employed by introducing naked DNA encoding a 125P5C8-related protein into the patient (e.g., intramuscularly or intradermally) to induce an anti-tumor response.


Vaccinia virus is used, for example, as a vector to express nucleotide sequences that encode the peptides of the invention. Upon introduction into a host, the recombinant vaccinia virus expresses the protein immunogenic peptide, and thereby elicits a host immune response. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al., Nature 351:456-460 (1991). A wide variety of other vectors useful for therapeutic administration or immunization of the peptides of the invention, e.g. adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, and the like, will be apparent to those skilled in the art from the description herein.


Thus, gene delivery systems are used to deliver a 125P5C8-related nucleic acid molecule. In one embodiment, the full-length human 125P5C8 cDNA is employed. In another embodiment, 125P5C8 nucleic acid molecules encoding specific cytotoxic T lymphocyte (CTL) and/or antibody epitopes are employed.


Ex Vivo Vaccines


Various ex vivo strategies can also be employed to generate an immune response. One approach involves the use of antigen presenting cells (APCs) such as dendritic cells (DC) to present 125P5C8 antigen to a patient's immune system. Dendritic cells express MHC class I and II molecules, B7 co-stimulator, and IL-12, and are thus highly specialized antigen presenting cells. In prostate cancer, autologous dendritic cells pulsed with peptides of the prostate-specific membrane antigen (PSMA) are being used in a Phase I clinical trial to stimulate prostate cancer patients' immune systems (Tjoa et al., 1996, Prostate 28:65-69; Murphy et al., 1996, Prostate 29:371-380). Thus, dendritic cells can be used to present 125P5C8 peptides to T cells in the context of MHC class I or II molecules. In one embodiment, autologous dendritic cells are pulsed with 125P5C8 peptides capable of binding to MHC class I and/or class II molecules. In another embodiment, dendritic cells are pulsed with the complete 125P5C8 protein. Yet another embodiment involves engineering the overexpression of a 125P5C8 gene in dendritic cells using various implementing vectors known in the art, such as adenovirus (Arthur et al., 1997, Cancer Gene Ther. 4:17-25), retrovirus (Henderson et al., 1996, Cancer Res. 56:3763-3770), lentivirus, adeno-associated virus, DNA transfection (Ribas et al., 1997, Cancer Res. 57:2865-2869), or tumor-derived RNA transfection (Ashley et al., 1997, J. Exp. Med. 186:1177-1182). Cells that express 125P5C8 can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.


X.B.) 125P5C8 as a Target for Antibody-based Therapy


125P5C8 is an attractive target for antibody-based therapeutic strategies. A number of antibody strategies are known in the art for targeting both extracellular and intracellular molecules (see, e.g., complement and ADCC mediated killing as well as the use of intrabodies). Because 125P5C8 is expressed by cancer cells of various lineages relative to corresponding normal cells, systemic administration of 125P5C8-immunoreactive compositions are prepared that exhibit excellent sensitivity without toxic, non-specific and/or non-target effects caused by binding of the immunoreactive composition to non-target organs and tissues. Antibodies specifically reactive with domains of 125P5C8 are useful to treat 125P5C8-expressing cancers systemically, either as conjugates with a toxin or therapeutic agent, or as naked antibodies capable of inhibiting cell proliferation or function.


125P5C8 antibodies can be introduced into a patient such that the antibody binds to 125P5C8 and modulates a function, such as an interaction with a binding partner, and consequently mediates destruction of the tumor cells and/or inhibits the growth of the tumor cells. Mechanisms by which such antibodies exert a therapeutic effect can include complement-mediated cytolysis, antibody-dependent cellular cytotoxicity, modulation of the physiological function of 125P5C8, inhibition of ligand binding or signal transduction pathways, modulation of tumor cell differentiation, alteration of tumor angiogenesis factor profiles, and/or apoptosis.


Those skilled in the art understand that antibodies can be used to specifically target and bind immunogenic molecules such as an immunogenic region of a 125P5C8 sequence shown in FIG. 2 or FIG. 3. In addition, skilled artisans understand that it is routine to conjugate antibodies to cytotoxic agents (see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)). When cytotoxic and/or therapeutic agents are delivered directly to cells, such as by conjugating them to antibodies specific for a molecule expressed by that cell (e.g. 125P5C8), the cytotoxic agent will exert its known biological effect (i.e. cytotoxicity) on those cells.


A wide variety of compositions and methods for using antibody-cytotoxic agent conjugates to kill cells are known in the art. In the context of cancers, typical methods entail administering to an animal having a tumor a biologically effective amount of a conjugate comprising a selected cytotoxic and/or therapeutic agent linked to a targeting agent (e.g. an anti-125P5C8 antibody) that binds to a marker (e.g. 125P5C8) expressed, accessible to binding or localized on the cell surfaces. A typical embodiment is a method of delivering a cytotoxic and/or therapeutic agent to a cell expressing 125P5C8, comprising conjugating the cytotoxic agent to an antibody that immunospecifically binds to a 125P5C8 epitope, and, exposing the cell to the antibody-agent conjugate. Another illustrative embodiment is a method of treating an individual suspected of suffering from metastasized cancer, comprising a step of administering parenterally to said individual a pharmaceutical composition comprising a therapeutically effective amount of an antibody conjugated to a cytotoxic and/or therapeutic agent.


Cancer immunotherapy using anti-125P5C8 antibodies can be done in accordance with various approaches that have been successfully employed in the treatment of other types of cancer, including but not limited to colon cancer (Arlen et al., 1998, Crit. Rev. Immunol. 18:133-138), multiple myeloma (Ozaki et al., 1997, Blood 90:3179-3186, Tsunenari et al., 1997, Blood 90:2437-2444), gastric cancer (Kasprzyk et al., 1992, Cancer Res. 52:2771-2776), B-cell lymphoma (Funakoshi et al., 1996, J. Immunother. Emphasis Tumor Immunol. 19:93-101), leukemia (Zhong et al., 1996, Leuk. Res. 20:581-589), colorectal cancer (Moun et al., 1994, Cancer Res. 54:6160-6166; Velders et al., 1995, Cancer Res. 55:4398-4403), and breast cancer (Shepard et al., 1991, J. Clin. Immunol. 11:117-127). Some therapeutic approaches involve conjugation of naked antibody to a toxin or radioisotope, such as the conjugation of Y91 or I131 to anti-CD20 antibodies (e.g., Zevalin™, IDEC Pharmaceuticals Corp. or Bexxar™, Coulter Pharmaceuticals), while others involve co-administration of antibodies and other therapeutic agents, such as Herceptin™ (trastuzumab) with paclitaxel (Genentech, Inc.). The antibodies can be conjugated to a therapeutic agent. To treat prostate cancer, for example, 125P5C8 antibodies can be administered in conjunction with radiation, chemotherapy or hormone ablation. Also, antibodies can be conjugated to a toxin such as calicheamicin (e.g., Mylotarg™, Wyeth-Ayerst, Madison, N.J., a recombinant humanized IgG4 kappa antibody conjugated to antitumor antibiotic calicheamicin) or a maytansinoid (e.g., taxane-based Tumor-Activated Prodrug, TAP, platform, ImmunoGen, Cambridge, Mass., also see e.g., U.S. Pat. No. 5,416,064).


Although 125P5C8 antibody therapy is useful for all stages of cancer, antibody therapy can be particularly appropriate in advanced or metastatic cancers. Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy. Alternatively, antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment. Additionally, antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well. Fan et al. (Cancer Res. 53:4637-4642, 1993), Prewett et al. (International J. of Onco. 9:217-224, 1996), and Hancock et al. (Cancer Res. 51:4575-4580, 1991) describe the use of various antibodies together with chemotherapeutic agents.


Although 125P5C8 antibody therapy is useful for all stages of cancer, antibody therapy can be particularly appropriate in advanced or metastatic cancers. Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy. Alternatively, antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment. Additionally, antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well.


Cancer patients can be evaluated for the presence and level of 125P5C8 expression, preferably using immunohistochemical assessments of tumor tissue, quantitative 125P5C8 imaging, or other techniques that reliably indicate the presence and degree of 125P5C8 expression. Immunohistochemical analysis of tumor biopsies or surgical specimens is preferred for this purpose. Methods for immunohistochemical analysis of tumor tissues are well known in the art.


Anti-125P5C8 monoclonal antibodies that treat prostate and other cancers include those that initiate a potent immune response against the tumor or those that are directly cytotoxic. In this regard, anti-125P5C8 monoclonal antibodies (mAbs) can elicit tumor cell lysis by either complement-mediated or antibody-dependent cell cytotoxicity (ADCC) mechanisms, both of which require an intact Fc portion of the immunoglobulin molecule for interaction with effector cell Fc receptor sites on complement proteins. In addition, anti-125P5C8 mAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express 125P5C8. Mechanisms by which directly cytotoxic mAbs act include: inhibition of cell growth, modulation of cellular differentiation, modulation of tumor angiogenesis factor profiles, and the induction of apoptosis. The mechanism(s) by which a particular anti-125P5C8 mAb exerts an anti-tumor effect is evaluated using any number of in vitro assays that evaluate cell death such as ADCC, ADMMC, complement-mediated cell lysis, and so forth, as is generally known in the art.


In some patients, the use of murine or other non-human monoclonal antibodies, or human/mouse chimeric mAbs can induce moderate to strong immune responses against the non-human antibody. This can result in clearance of the antibody from circulation and reduced efficacy. In the most severe cases, such an immune response can lead to the extensive formation of immune complexes which, potentially, can cause renal failure. Accordingly, preferred monoclonal antibodies used in the therapeutic methods of the invention are those that are either fully human or humanized and that bind specifically to the target 125P5C8 antigen with high affinity but exhibit low or no antigenicity in the patient.


Therapeutic methods of the invention contemplate the administration of single anti-125P5C8 mAbs as well as combinations, or cocktails, of different mAbs. Such mAb cocktails can have certain advantages inasmuch as they contain mAbs that target different epitopes, exploit different effector mechanisms or combine directly cytotoxic mAbs with mAbs that rely on immune effector functionality. Such mAbs in combination can exhibit synergistic therapeutic effects. In addition, anti-125P5C8 mAbs can be administered concomitantly with other therapeutic modalities, including but not limited to various chemotherapeutic agents, androgen-blockers, immune modulators (e.g., IL-2, GM-CSF), surgery or radiation. The anti-125P5C8 mAbs are administered in their “naked” or unconjugated form, or can have a therapeutic agent(s) conjugated to them.


Anti-125P5C8 antibody formulations are administered via any route capable of delivering the antibodies to a tumor cell. Routes of administration include, but are not limited to, intravenous, intraperitoneal, intramuscular, intratumor, intradermal, and the like. Treatment generally involves repeated administration of the anti-125P5C8 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight. In general, doses in the range of 10-1000 mg mAb per week are effective and well tolerated.


Based on clinical experience with the Herceptin™ mAb in the treatment of metastatic breast cancer, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-125P5C8 mAb preparation represents an acceptable dosing regimen. Preferably, the initial loading dose is administered as a 90 minute or longer infusion. The periodic maintenance dose is administered as a 30 minute or longer infusion, provided the initial dose was well tolerated. As appreciated by those of skill in the art, various factors can influence the ideal dose regimen in a particular case. Such factors include, for example, the binding affinity and half life of the Ab or mAbs used, the degree of 125P5C8 expression in the patient, the extent of circulating shed 125P5C8 antigen, the desired steady-state antibody concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.


Optionally, patients should be evaluated for the levels of 125P5C8 in a given sample (e.g. the levels of circulating 125P5C8 antigen and/or 125P5C8 expressing cells) in order to assist in the determination of the most effective dosing regimen, etc. Such evaluations are also used for monitoring purposes throughout therapy, and are useful to gauge therapeutic success in combination with the evaluation of other parameters (for example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).


Anti-idiotypic anti-125P5C8 antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a 125P5C8-related protein. In particular, the generation of anti-idiotypic antibodies is well known in the art; this methodology can readily be adapted to generate anti-idiotypic anti-125P5C8 antibodies that mimic an epitope on a 125P5C8-related protein (see, for example, Wagner et al., 1997, Hybridoma 16: 33-40; Foon et al., 1995, J. Clin. Invest. 96:334-342; Herlyn et al., 1996, Cancer Immunol. Immunother. 43:65-76). Such an anti-idiotypic antibody can be used in cancer vaccine strategies.


X.C.) 125P5C8 as a Target for Cellular Immune Responses


Vaccines and methods of preparing vaccines that contain an immunogenically effective amount of one or more HLA-binding peptides as described herein are further embodiments of the invention. Furthermore, vaccines in accordance with the invention encompass compositions of one or more of the claimed peptides. A peptide can be present in a vaccine individually. Alternatively, the peptide can exist as a homopolymer comprising multiple copies of the same peptide, or as a heteropolymer of various peptides. Polymers have the advantage of increased immunological reaction and, where different peptide epitopes are used to make up the polymer, the additional ability to induce antibodies and/or CTLs that react with different antigenic determinants of the pathogenic organism or tumor-related peptide targeted for an immune response. The composition can be a naturally occurring region of an antigen or can be prepared, e.g., recombinantly or by chemical synthesis.


Carriers that can be used with vaccines of the invention are well known in the art, and include, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acids such as poly L-lysine, poly L-glutamic acid, influenza, hepatitis B virus core protein, and the like. The vaccines can contain a physiologically tolerable (i.e., acceptable) diluent such as water, or saline, preferably phosphate buffered saline. The vaccines also typically include an adjuvant. Adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide, or alum are examples of materials well known in the art. Additionally, as disclosed herein, CTL responses can be primed by conjugating peptides of the invention to lipids, such as tripalmitoyl-S-glycerylcysteinlyseryl-serine (P3CSS). Moreover, an adjuvant such as a synthetic cytosine-phosphorothiolated-guanine-containing (CpG) oligonucleotides has been found to increase CTL responses 10- to 100-fold. (see, e.g. Davila and Celis J. Immunol. 165:539-547 (2000))


Upon immunization with a peptide composition in accordance with the invention, via injection, aerosol, oral, transdermal, transmucosal, intrapleural, intrathecal, or other suitable routes, the immune system of the host responds to the vaccine by producing large amounts of CTLs and/or HTLs specific for the desired antigen. Consequently, the host becomes at least partially immune to later development of cells that express or overexpress 125P5C8 antigen, or derives at least some therapeutic benefit when the antigen was tumor-associated.


In some embodiments, it may be desirable to combine the class I peptide components with components that induce or facilitate neutralizing antibody and or helper T cell responses directed to the target antigen. A preferred embodiment of such a composition comprises class I and class II epitopes in accordance with the invention. An alternative embodiment of such a composition comprises a class I and/or class II epitope in accordance with the invention, along with a cross reactive HTL epitope such as PADRE™ (Epimmune, San Diego, Calif.) molecule (described e.g., in U.S. Pat. No. 5,736,142).


A vaccine of the invention can also include antigen-presenting cells (APC), such as dendritic cells (DC), as a vehicle to present peptides of the invention. Vaccine compositions can be created in vitro, following dendritic cell mobilization and harvesting, whereby loading of dendritic cells occurs in vitro. For example, dendritic cells are transfected, e.g., with a minigene in accordance with the invention, or are pulsed with peptides. The dendritic cell can then be administered to a patient to elicit immune responses in vivo. Vaccine compositions, either DNA- or peptide-based, can also be administered in vivo in combination with dendritic cell mobilization whereby loading of dendritic cells occurs in vivo.


Preferably, the following principles are utilized when selecting an array of epitopes for inclusion in a polyepitopic composition for use in a vaccine, or for selecting discrete epitopes to be included in a vaccine and/or to be encoded by nucleic acids such as a minigene. It is preferred that each of the following principles be balanced in order to make the selection. The multiple epitopes to be incorporated in a given vaccine composition may be, but need not be, contiguous in sequence in the native antigen from which the epitopes are derived.


1.) Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance. For HLA Class I this includes 3-4 epitopes that come from at least one tumor associated antigen (TAA). For HLA Class II a similar rationale is employed; again 3-4 epitopes are selected from at least one TAA (see, e.g., Rosenberg et al., Science 278:1447-1450). Epitopes from one TAA may be used in combination with epitopes from one or more additional TAAs to produce a vaccine that targets tumors with varying expression patterns of frequently-expressed TAAs.


2.) Epitopes are selected that have the requisite binding affinity established to be correlated with immunogenicity: for HLA Class I an IC50 of 500 nM or less, often 200 nM or less; and for Class II an IC50 of 1000 nM or less.


3.) Sufficient supermotif bearing-peptides, or a sufficient array of allele-specific motif-bearing peptides, are selected to give broad population coverage. For example, it is preferable to have at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess the breadth, or redundancy of, population coverage.


4.) When selecting epitopes from cancer-related antigens it is often useful to select analogs because the patient may have developed tolerance to the native epitope.


5.) Of particular relevance are epitopes referred to as “nested epitopes.” Nested epitopes occur where at least two epitopes overlap in a given peptide sequence. A nested peptide sequence can comprise B cell, HLA class I and/or HLA class II epitopes. When providing nested epitopes, a general objective is to provide the greatest number of epitopes per sequence. Thus, an aspect is to avoid providing a peptide that is any longer than the amino terminus of the amino terminal epitope and the carboxyl terminus of the carboxyl terminal epitope in the peptide. When providing a multi-epitopic sequence, such as a sequence comprising nested epitopes, it is generally important to screen the sequence in order to insure that it does not have pathological or other deleterious biological properties.


6.) If a polyepitopic protein is created, or when creating a minigene, an objective is to generate the smallest peptide that encompasses the epitopes of interest. This principle is similar, if not the same as that employed when selecting a peptide comprising nested epitopes. However, with an artificial polyepitopic peptide, the size minimization objective is balanced against the need to integrate any spacer sequences between epitopes in the polyepitopic protein. Spacer amino acid residues can, for example, be introduced to avoid junctional epitopes (an epitope recognized by the immune system, not present in the target antigen, and only created by the man-made juxtaposition of epitopes), or to facilitate cleavage between epitopes and thereby enhance epitope presentation. Junctional epitopes are generally to be avoided because the recipient may generate an immune response to that non-native epitope. Of particular concern is a junctional epitope that is a “dominant epitope.” A dominant epitope may lead to such a zealous response that immune responses to other epitopes are diminished or suppressed.


7.) Where the sequences of multiple variants of the same target protein are present, potential peptide epitopes can also be selected on the basis of their conservancy. For example, a criterion for conservancy may define that the entire sequence of an HLA class I binding peptide or the entire 9-mer core of a class II binding peptide be conserved in a designated percentage of the sequences evaluated for a specific protein antigen.


X.C.1. Minigene Vaccines


A number of different approaches are available which allow simultaneous delivery of multiple epitopes. Nucleic acids encoding the peptides of the invention are a particularly useful embodiment of the invention. Epitopes for inclusion in a minigene are preferably selected according to the guidelines set forth in the previous section. A preferred means of administering nucleic acids encoding the peptides of the invention uses minigene constructs encoding a peptide comprising one or multiple epitopes of the invention.


The use of multi-epitope minigenes is described below and in, Ishioka et al., J. Immunol. 162:3915-3925, 1999; An, L. and Whitton, J. L., J. Virol. 71:2292, 1997; Thomson, S. A. et al., J. Immunol. 157:822, 1996; Whitton, J. L. et al., J. Virol. 67:348, 1993; Hanke, R. et al., Vaccine 16:426, 1998. For example, a multi-epitope DNA plasmid encoding supermotif- and/or motif-bearing epitopes derived 125P5C8, the PADRE® universal helper T cell epitope (or multiple HTL epitopes from 125P5C8), and an endoplasmic reticulum-translocating signal sequence can be engineered. A vaccine may also comprise epitopes that are derived from other TAAs.


The immunogenicity of a multi-epitopic minigene can be confirmed in transgenic mice to evaluate the magnitude of CTL induction responses against the epitopes tested. Further, the immunogenicity of DNA-encoded epitopes in vivo can be correlated with the in vitro responses of specific CTL lines against target cells transfected with the DNA plasmid. Thus, these experiments can show that the minigene serves to both: 1.) generate a CTL response and 2.) that the induced CTLs recognized cells expressing the encoded epitopes.


For example, to create a DNA sequence encoding the selected epitopes (minigene) for expression in human cells, the amino acid sequences of the epitopes may be reverse translated. A human codon usage table can be used to guide the codon choice for each amino acid. These epitope-encoding DNA sequences may be directly adjoined, so that when translated, a continuous polypeptide sequence is created. To optimize expression and/or immunogenicity, additional elements can be incorporated into the minigene design. Examples of amino acid sequences that can be reverse translated and included in the minigene sequence include: HLA class I epitopes, HLA class II epitopes, antibody epitopes, a ubiquitination signal sequence, and/or an endoplasmic reticulum targeting signal. In addition, HLA presentation of CTL and HTL epitopes may be improved by including synthetic (e.g. poly-alanine) or naturally-occurring flanking sequences adjacent to the CTL or HTL epitopes; these larger peptides comprising the epitope(s) are within the scope of the invention.


The minigene sequence may be converted to DNA by assembling oligonucleotides that encode the plus and minus strands of the minigene. Overlapping oligonucleotides (30-100 bases long) may be synthesized, phosphorylated, purified and annealed under appropriate conditions using well known techniques. The ends of the oligonucleotides can be joined, for example, using T4 DNA ligase. This synthetic minigene, encoding the epitope polypeptide, can then be cloned into a desired expression vector.


Standard regulatory sequences well known to those of skill in the art are preferably included in the vector to ensure expression in the target cells. Several vector elements are desirable: a promoter with a down-stream cloning site for minigene insertion; a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g. ampicillin or kanamycin resistance). Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.


Additional vector modifications may be desired to optimize minigene expression and immunogenicity. In some cases, introns are required for efficient gene expression, and one or more synthetic or naturally-occurring introns could be incorporated into the transcribed region of the minigene. The inclusion of mRNA stabilization sequences and sequences for replication in mammalian cells may also be considered for increasing minigene expression.


Once an expression vector is selected, the minigene is cloned into the polylinker region downstream of the promoter. This plasmid is transformed into an appropriate E. coli strain, and DNA is prepared using standard techniques. The orientation and DNA sequence of the minigene, as well as all other elements included in the vector, are confirmed using restriction mapping and DNA sequence analysis. Bacterial cells harboring the correct plasmid can be stored as a master cell bank and a working cell bank.


In addition, immunostimulatory sequences (ISSs or CpGs) appear to play a role in the immunogenicity of DNA vaccines. These sequences may be included in the vector, outside the minigene coding sequence, if desired to enhance immunogenicity.


In some embodiments, a bi-cistronic expression vector which allows production of both the minigene-encoded epitopes and a second protein (included to enhance or decrease immunogenicity) can be used. Examples of proteins or polypeptides that could beneficially enhance the immune response if co-expressed include cytokines (e.g., IL-2, IL-12, GM-CSF), cytokine-inducing molecules (e.g., LeIF), costimulatory molecules, or for HTL responses, pan-DR binding proteins (PADRE™, Epimmune, San Diego, Calif.). Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction. In contrast to HTL or CTL induction, specifically decreasing the immune response by co-expression of immunosuppressive molecules (e.g. TGF-β) may be beneficial in certain diseases.


Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli, followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, Calif.). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.


Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as “naked DNA,” is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be desirable. A variety of methods have been described, and new techniques may become available. Cationic lipids, glycolipids, and fusogenic liposomes can also be used in the formulation (see, e.g., as described by WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Felgner, et al., Proc. Nat'l Acad. Sci. USA 84:7413 (1987). In addition, peptides and compounds referred to collectively as protective, interactive, non-condensing compounds (PINC) could also be complexed to purified plasmid DNA to influence variables such as stability, intramuscular dispersion, or trafficking to specific organs or cell types.


Target cell sensitization can be used as a functional assay for expression and HLA class I presentation of minigene-encoded CTL epitopes. For example, the plasmid DNA is introduced into a mammalian cell line that is suitable as a target for standard CTL chromium release assays. The transfection method used will be dependent on the final formulation. Electroporation can be used for “naked” DNA, whereas cationic lipids allow direct in vitro transfection. A plasmid expressing green fluorescent protein (GFP) can be co-transfected to allow enrichment of transfected cells using fluorescence activated cell sorting (FACS). These cells are then chromium-51 (51Cr) labeled and used as target cells for epitope-specific CTL lines; cytolysis, detected by 51Cr release, indicates both production of, and HLA presentation of, minigene-encoded CTL epitopes. Expression of HTL epitopes may be evaluated in an analogous manner using assays to assess HTL activity.


In vivo immunogenicity is a second approach for functional testing of minigene DNA formulations. Transgenic mice expressing appropriate human HLA proteins are immunized with the DNA product. The dose and route of administration are formulation dependent (e.g., IM for DNA in PBS, intraperitoneal (i.p.) for lipid-complexed DNA). Twenty-one days after immunization, splenocytes are harvested and restimulated for one week in the presence of peptides encoding each epitope being tested. Thereafter, for CTL effector cells, assays are conducted for cytolysis of peptide-loaded. 51Cr-labeled target cells using standard techniques. Lysis of target cells that were sensitized by HLA loaded with peptide epitopes, corresponding to minigene-encoded epitopes, demonstrates DNA vaccine function for in vivo induction of CTLs. Immunogenicity of HTL epitopes is confirmed in transgenic mice in an analogous manner.


Alternatively, the nucleic acids can be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253. Using this technique, particles comprised solely of DNA are administered. In a further alternative embodiment, DNA can be adhered to particles, such as gold particles.


Minigenes can also be delivered using other bacterial or viral delivery systems well known in the art, e.g., an expression construct encoding epitopes of the invention can be incorporated into a viral vector such as vaccinia.


X.C.2. Combinations of CTL Peptides with Helper Peptides


Vaccine compositions comprising CTL peptides of the invention can be modified, e.g., analoged, to provide desired attributes, such as improved serum half life, broadened population coverage or enhanced immunogenicity.


For instance, the ability of a peptide to induce CTL activity can be enhanced by linking the peptide to a sequence which contains at least one epitope that is capable of inducing a T helper cell response. Although a CTL peptide can be directly linked to a T helper peptide, often CTL epitope/HTL epitope conjugates are linked by a spacer molecule. The spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present spacer need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues and sometimes 10 or more residues. The CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide. The amino terminus of either the immunogenic peptide or the T helper peptide may be acylated.


In certain embodiments, the T helper peptide is one that is recognized by T helper cells present in a majority of a genetically diverse population. This can be accomplished by selecting peptides that bind to many, most, or all of the HLA class II molecules. Examples of such amino acid bind many HLA Class II molecules include sequences from antigens such as tetanus toxoid at positions 830-843 (QYIKANSKFIGITE; SEQ ID NO:19), Plasmodium falciparum circumsporozoite (CS) protein at positions 378-398 (DIEKKIAKMEKASSVFNVVNS; SEQ ID NO: 20), and Streptococcus 18 kD protein at positions 116-131 (GAVDSILGGVATYGAA; SEQ ID NO: 21). Other examples include peptides bearing a DR 1-4-7 supermotif, or either of the DR3 motifs.


Alternatively, it is possible to prepare synthetic peptides capable of stimulating T helper lymphocytes, in a loosely HLA-restricted fashion, using amino acid sequences not found in nature (see, e.g., PCT publication WO 95/07707). These synthetic compounds called Pan-DR-binding epitopes (e.g., PADRE™, Epimmune, Inc., San Diego, Calif.) are designed to most preferably bind most HLA-DR (human HLA class II) molecules. For instance, a pan-DR-binding epitope peptide having the formula: aKXVAAWTLKAAa (SEQ ID NO: 22), where “X” is either cyclohexylalanine, phenylalanine, or tyrosine, and a is either D-alanine or L-alanine, has been found to bind to most HLA-DR alleles, and to stimulate the response of T helper lymphocytes from most individuals, regardless of their HLA type. An alternative of a pan-DR binding epitope comprises all “L” natural amino acids and can be provided in the form of nucleic acids that encode the epitope.


HTL peptide epitopes can also be modified to alter their biological properties. For example, they can be modified to include D-amino acids to increase their resistance to proteases and thus extend their serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase their biological activity. For example, a T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.


X.C.3. Combinations of CTL Peptides with T Cell Priming Agents


In some embodiments it may be desirable to include in the pharmaceutical compositions of the invention at least one component which primes B lymphocytes or T lymphocytes. Lipids have been identified as agents capable of priming CTL in vivo. For example, palmitic acid residues can be attached to the ε- and α-amino groups of a lysine residue and then linked, e.g., via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic peptide. The lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant, e.g., incomplete Freund's adjuvant. In a preferred embodiment, a particularly effective immunogenic composition comprises palmitic acid attached to ε- and α-amino groups of Lys, which is attached via linkage, e.g., Ser-Ser, to the amino terminus of the immunogenic peptide.


As another example of lipid priming of CTL responses, E. coli lipoproteins, such as tripalmitoyl-S-glycerylcysteinlyseryl-serine (P3CSS) can be used to prime virus specific CTL when covalently attached to an appropriate peptide (see, e.g., Deres, et al., Nature 342:561, 1989). Peptides of the invention can be coupled to P3CSS, for example, and the lipopeptide administered to an individual to specifically prime an immune response to the target antigen. Moreover, because the induction of neutralizing antibodies can also be primed with P3CSS-conjugated epitopes, two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses.


X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides


An embodiment of a vaccine composition in accordance with the invention comprises ex vivo administration of a cocktail of epitope-bearing peptides to PBMC, or isolated DC therefrom, from the patient's blood. A pharmaceutical to facilitate harvesting of DC can be used, such as Progenipoietin™ (Pharmacia-Monsanto, St. Louis, Mo.) or GM-CSF/IL-4. After pulsing the DC with peptides and prior to reinfusion into patients, the DC are washed to remove unbound peptides. In this embodiment, a vaccine comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces.


The DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to 125P5C8. Optionally, a helper T cell (HTL) peptide, such as a natural or artificial loosely restricted HLA Class II peptide, can be included to facilitate the CTL response. Thus, a vaccine in accordance with the invention is used to treat a cancer which expresses or overexpresses 125P5C8.


X.D. Adoptive Immunotherapy


Antigenic 125P5C8-related peptides are used to elicit a CTL and/or HTL response ex vivo, as well. The resulting CTL or HTL cells, can be used to treat tumors in patients that do not respond to other conventional forms of therapy, or will not respond to a therapeutic vaccine peptide or nucleic acid in accordance with the invention. Ex vivo CTL or HTL responses to a particular antigen are induced by incubating in tissue culture the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of antigen-presenting cells (APC), such as dendritic cells, and the appropriate immunogenic peptide. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cell (e.g., a tumor cell). Transfected dendritic cells may also be used as antigen presenting cells.


X.E. Administration of Vaccines for Therapeutic or Prophylactic Purposes


Pharmaceutical and vaccine compositions of the invention are typically used to treat and/or prevent a cancer that expresses or overexpresses 125P5C8. In therapeutic applications, peptide and/or nucleic acid compositions are administered to a patient in an amount sufficient to elicit an effective B cell, CTL and/or HTL response to the antigen and to cure or at least partially arrest or slow symptoms and/or complications. An amount adequate to accomplish this is defined as “therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the particular composition administered, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician.


For pharmaceutical compositions, the immunogenic peptides of the invention, or DNA encoding them, are generally administered to an individual already bearing a tumor that expresses 125P5C8. The peptides or DNA encoding them can be administered individually or as fusions of one or more peptide sequences. Patients can be treated with the immunogenic peptides separately or in conjunction with other treatments, such as surgery, as appropriate.


For therapeutic use, administration should generally begin at the first diagnosis of 125P5C8-associated cancer. This is followed by boosting doses until at least symptoms are substantially abated and for a period thereafter. The embodiment of the vaccine composition (i.e., including, but not limited to embodiments such as peptide cocktails, polyepitopic polypeptides, minigenes, or TAA-specific CTLs or pulsed dendritic cells) delivered to the patient may vary according to the stage of the disease or the patient's health status. For example, in a patient with a tumor that expresses 125P5C8, a vaccine comprising 125P5C8-specific CTL may be more efficacious in killing tumor cells in patient with advanced disease than alternative embodiments.


It is generally important to provide an amount of the peptide epitope delivered by a mode of administration sufficient to effectively stimulate a cytotoxic T cell response; compositions which stimulate helper T cell responses can also be given in accordance with this embodiment of the invention.


The dosage for an initial therapeutic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1,000 μg and the higher value is about 10,000; 20,000; 30,000; or 50,000 μg. Dosage values for a human typically range from about 500 μg to about 50,000 μg per 70 kilogram patient. Boosting dosages of between about 1.0 μg to about 50,000 μg of peptide pursuant to a boosting regimen over weeks to months may be administered depending upon the patient's response and condition as determined by measuring the specific activity of CTL and HTL obtained from the patient's blood. Administration should continue until at least clinical symptoms or laboratory tests indicate that the neoplasia, has been eliminated or reduced and for a period thereafter. The dosages, routes of administration, and dose schedules are adjusted in accordance with methodologies known in the art.


In certain embodiments, the peptides and compositions of the present invention are employed in serious disease states, that is, life-threatening or potentially life threatening situations. In such cases, as a result of the minimal amounts of extraneous substances and the relative nontoxic nature of the peptides in preferred compositions of the invention, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these peptide compositions relative to these stated dosage amounts.


The vaccine compositions of the invention can also be used purely as prophylactic agents. Generally the dosage for an initial prophylactic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1000 μg and the higher value is about 10,000; 20,000; 30,000; or 50,000 μg. Dosage values for a human typically range from about 500 μg to about 50,000 μg per 70 kilogram patient. This is followed by boosting dosages of between about 1.0 μg to about 50,000 μg of peptide administered at defined intervals from about four weeks to six months after the initial administration of vaccine. The immunogenicity of the vaccine can be assessed by measuring the specific activity of CTL and HTL obtained from a sample of the patient's blood.


The pharmaceutical compositions for therapeutic treatment are intended for parenteral, topical, oral, nasal, intrathecal, or local (e.g. as a cream or topical ointment) administration. Preferably, the pharmaceutical compositions are administered parentally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly. Thus, the invention provides compositions for parenteral administration which comprise a solution of the immunogenic peptides dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.


A variety of aqueous carriers may be used, e.g., water, buffered water, 0.8% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well-known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.


The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.


The concentration of peptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.


A human unit dose form of a composition is typically included in a pharmaceutical composition that comprises a human unit dose of an acceptable carrier, in one embodiment an aqueous carrier, and is administered in a volume/quantity that is known by those of skill in the art to be used for administration of such compositions to humans (see, e.g., Remington's Pharmaceutical Sciences, 17th Edition, A. Gennaro, Editor, Mack Publishing Co., Easton, Pa., 1985). For example a peptide dose for initial immunization can be from about 1 to about 50,000 μg, generally 100-5,000 μg, for a 70 kg patient. For example, for nucleic acids an initial immunization may be performed using an expression vector in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 μg) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5×109 pfu.


For antibodies, a treatment generally involves repeated administration of the anti-125P5C8 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1 to about 10 mg/kg body weight. In general, doses in the range of 10-500 mg mAb per week are effective and well tolerated. Moreover, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-125P5C8 mAb preparation represents an acceptable dosing regimen. As appreciated by those of skill in the art, various factors can influence the ideal dose in a particular case. Such factors include, for example, half life of a composition, the binding affinity of an Ab, the immunogenicity of a substance, the degree of 125P5C8 expression in the patient, the extent of circulating shed 125P5C8 antigen, the desired steady-state concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient. Non-limiting preferred human unit doses are, for example, 500 μg-1 mg, 1 mg-50 mg, 50 mg-100 mg, 100 mg-200 mg, 200 mg-300 mg, 400 mg-500 mg, 500 mg-600 mg, 600 mg-700 mg, 700 mg-800 mg, 800 mg-900 mg, 900 mg-1 g, or 1 mg-700 mg. In certain embodiments, the dose is in a range of 2-5 mg/kg body weight, e.g., with follow on weekly doses of 1-3 mg/kg; 0.5 mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg/kg body weight followed, e.g., in two, three or four weeks by weekly doses; 0.5-10 mg/kg body weight, e.g., followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400 mg m2 of body area weekly; 1-600 mg m2 of body area weekly; 225-400 mg m2 of body area weekly; these does can be followed by weekly doses for 2, 3, 4, 5, 6, 7, 8, 9, 19, 11, 12 or more weeks.


In one embodiment, human unit dose forms of polynucleotides comprise a suitable dosage range or effective amount that provides any therapeutic effect. As appreciated by one of ordinary skill in the art a therapeutic effect depends on a number of factors, including the sequence of the polynucleotide, molecular weight of the polynucleotide and route of administration. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. Generally, for a polynucleotide of about 20 bases, a dosage range may be selected from, for example, an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg. For example, a dose may be about any of the following: 0.1 to 100 mg/kg, 0.1 to 50 mg/kg, 0.1 to 25 mg/kg, 0.1 to 10 mg/kg, 1 to 500 mg/kg, 100 to 400 mg/kg, 200 to 300 mg/kg, 1 to 100 mg/kg, 100 to 200 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 1000 mg/kg, 500 to 5000 mg/kg, or 500 to 10,000 mg/kg. Generally, parenteral routes of administration may require higher doses of polynucleotide compared to more direct application to the nucleotide to diseased tissue, as do polynucleotides of increasing length.


In one embodiment, human unit dose forms of T-cells comprise a suitable dosage range or effective amount that provides any therapeutic effect. As appreciated by one of ordinary skill in the art, a therapeutic effect depends on a number of factors. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. A dose may be about 104 cells to about 106 cells, about 106 cells to about 108 cells, about 108 to about 1011 cells, or about 108 to about 5×1010 cells. A dose may also about 106 cells/m2 to about 1010 cells/m2, or about 106 cells/m2 to about 108 cells/m2.


Proteins(s) of the invention, and/or nucleic acids encoding the protein(s), can also be administered via liposomes, which may also serve to: 1) target the proteins(s) to a particular tissue, such as lymphoid tissue; 2) to target selectively to diseases cells; or, 3) to increase the half-life of the peptide composition. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations, the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes either filled or decorated with a desired peptide of the invention can be directed to the site of lymphoid cells, where the liposomes then deliver the peptide compositions. Liposomes for use in accordance with the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid liability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka, et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), and U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.


For targeting cells of the immune system, a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells. A liposome suspension containing a peptide may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.


For solid compositions, conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For oral administration, a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, and more preferably at a concentration of 25%-75%.


For aerosol administration, immunogenic peptides are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of peptides are about 0.01%-20% by weight, preferably about 1%-10%. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such agents are the esters or partial esters of fatty acids containing from about 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. The surfactant may constitute about 0.1%-20% by weight of the composition, preferably about 0.25-5%. The balance of the composition is ordinarily propellant. A carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery.


XI.) Diagnostic and Prognostic Embodiments of 125P5C8.


As disclosed herein, 125P5C8 polynucleotides, polypeptides, reactive cytotoxic T cells (CTL), reactive helper T cells (HTL) and anti-polypeptide antibodies are used in well known diagnostic, prognostic and therapeutic assays that examine conditions associated with dysregulated cell growth such as cancer, in particular the cancers listed in Table I (see, e.g., both its specific pattern of tissue expression as well as its overexpression in certain cancers as described for example in Example 4).


125P5C8 can be analogized to a prostate associated antigen PSA, the archetypal marker that has been used by medical practitioners for years to identify and monitor the presence of prostate cancer (see, e.g., Merrill et al., J. Urol. 163(2): 503-5120 (2000); Polascik et al., J. Urol. August; 162(2):293-306 (1999) and Fortier et al., J. Nat. Cancer Inst. 91(19): 1635-1640 (1999)). A variety of other diagnostic markers are also used in similar contexts including p53 and K-ras (see, e.g., Tulchinsky et al., Int J Mol Med 1999 July 4(1):99-102 and Minimoto et al., Cancer Detect Prev 2000; 24(1):1-12). Therefore, this disclosure of 125P5C8 polynucleotides and polypeptides (as well as 125P5C8 polynucleotide probes and anti-125P5C8 antibodies used to identify the presence of these molecules) and their properties allows skilled artisans to utilize these molecules in methods that are analogous to those used, for example, in a variety of diagnostic assays directed to examining conditions associated with cancer.


Typical embodiments of diagnostic methods which utilize the 125P5C8 polynucleotides, polypeptides, reactive T cells and antibodies are analogous to those methods from well-established diagnostic assays which employ, e.g., PSA polynucleotides, polypeptides, reactive T cells and antibodies. For example, just as PSA polynucleotides are used as probes (for example in Northern analysis, see, e.g., Sharief et al., Biochem. Mol. Biol. Int. 33(3):567-74 (1994)) and primers (for example in PCR analysis, see, e.g., Okegawa et al., J. Urol. 163(4): 1189-1190 (2000)) to observe the presence and/or the level of PSA mRNAs in methods of monitoring PSA overexpression or the metastasis of prostate cancers, the 125P5C8 polynucleotides described herein can be utilized in the same way to detect 125P5C8 overexpression or the metastasis of prostate and other cancers expressing this gene. Alternatively, just as PSA polypeptides are used to generate antibodies specific for PSA which can then be used to observe the presence and/or the level of PSA proteins in methods to monitor PSA protein overexpression (see, e.g., Stephan et al., Urology 55(4):560-3 (2000)) or the metastasis of prostate cells (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3):233-7 (1996)), the 125P5C8 polypeptides described herein can be utilized to generate antibodies for use in detecting 125P5C8 overexpression or the metastasis of prostate cells and cells of other cancers expressing this gene.


Specifically, because metastases involves the movement of cancer cells from an organ of origin (such as the lung or prostate gland etc.) to a different area of the body (such as a lymph node), assays which examine a biological sample for the presence of cells expressing 125P5C8 polynucleotides and/or polypeptides can be used to provide evidence of metastasis. For example, when a biological sample from tissue that does not normally contain 125P5C8-expressing cells (lymph node) is found to contain 125P5C8-expressing cells such as the 125P5C8 expression seen in LAPC4 and LAPC9, xenografts isolated from lymph node and bone metastasis, respectively, this finding is indicative of metastasis.


Alternatively 125P5C8 polynucleotides and/or polypeptides can be used to provide evidence of cancer, for example, when cells in a biological sample that do not normally express 125P5C8 or express 125P5C8 at a different level are found to express 125P5C8 or have an increased expression of 125P5C8 (see, e.g., the 125P5C8 expression in the cancers listed in Table I and in patient samples etc. shown in the accompanying Figures). In such assays, artisans may further wish to generate supplementary evidence of metastasis by testing the biological sample for the presence of a second tissue restricted marker (in addition to 125P5C8) such as PSA, PSCA etc. (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)).


Just as PSA polynucleotide fragments and polynucleotide variants are employed by skilled artisans for use in methods of monitoring PSA, 125P5C8 polynucleotide fragments and polynucleotide variants are used in an analogous manner. In particular, typical PSA polynucleotides used in methods of monitoring PSA are probes or primers which consist of fragments of the PSA cDNA sequence. Illustrating this, primers used to PCR amplify a PSA polynucleotide must include less than the whole PSA sequence to function in the polymerase chain reaction. In the context of such PCR reactions, skilled artisans generally create a variety of different polynucleotide fragments that can be used as primers in order to amplify different portions of a polynucleotide of interest or to optimize amplification reactions (see, e.g., Caetano-Anolles, G. Biotechniques 25(3): 472-476, 478-480 (1998); Robertson et al., Methods Mol. Biol. 98:121-154 (1998)). An additional illustration of the use of such fragments is provided in Example 4, where a 125P5C8 polynucleotide fragment is used as a probe to show the expression of 125P5C8 RNAs in cancer cells. In addition, variant polynucleotide sequences are typically used as primers and probes for the corresponding mRNAs in PCR and Northern analyses (see, e.g., Sawai et al., Fetal Diagn. Ther. 1996 November-December 11(6):407-13 and Current Protocols In Molecular Biology, Volume 2, Unit 2, Frederick M. Ausubel et al. eds., 1995)). Polynucleotide fragments and variants are useful in this context where they are capable of binding to a target polynucleotide sequence (e.g., a 125P5C8 polynucleotide shown in FIG. 2 or variant thereof) under conditions of high stringency.


Furthermore, PSA polypeptides which contain an epitope that can be recognized by an antibody or T cell that specifically binds to that epitope are used in methods of monitoring PSA. 125P5C8 polypeptide fragments and polypeptide analogs or variants can also be used in an analogous manner. This practice of using polypeptide fragments or polypeptide variants to generate antibodies (such as anti-PSA antibodies or T cells) is typical in the art with a wide variety of systems such as fusion proteins being used by practitioners (see, e.g., Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubel et al. eds., 1995). In this context, each epitope(s) functions to provide the architecture with which an antibody or T cell is reactive. Typically, skilled artisans create a variety of different polypeptide fragments that can be used in order to generate immune responses specific for different portions of a polypeptide of interest (see, e.g., U.S. Pat. No. 5,840,501 and U.S. Pat. No. 5,939,533). For example it may be preferable to utilize a polypeptide comprising one of the 125P5C8 biological motifs discussed herein or a motif-bearing subsequence which is readily identified by one of skill in the art based on motifs available in the art. Polypeptide fragments, variants or analogs are typically useful in this context as long as they comprise an epitope capable of generating an antibody or T cell specific for a target polypeptide sequence (e.g. a 125P5C8 polypeptide shown in FIG. 3).


As shown herein, the 125P5C8 polynucleotides and polypeptides (as well as the 125P5C8 polynucleotide probes and anti-125P5C8 antibodies or T cells used to identify the presence of these molecules) exhibit specific properties that make them useful in diagnosing cancers such as those listed in Table I. Diagnostic assays that measure the presence of 125P5C8 gene products, in order to evaluate the presence or onset of a disease condition described herein, such as prostate cancer, are used to identify patients for preventive measures or further monitoring, as has been done so successfully with PSA. Moreover, these materials satisfy a need in the art for molecules having similar or complementary characteristics to PSA in situations where, for example, a definite diagnosis of metastasis of prostatic origin cannot be made on the basis of a test for PSA alone (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)), and consequently, materials such as 125P5C8 polynucleotides and polypeptides (as well as the 125P5C8 polynucleotide probes and anti-125P5C8 antibodies used to identify the presence of these molecules) need to be employed to confirm a metastases of prostatic origin.


Finally, in addition to their use in diagnostic assays, the 125P5C8 polynucleotides disclosed herein have a number of other utilities such as their use in the identification of ontogenetic associated chromosomal abnormalities in the chromosomal region to which the 125P5C8 gene maps (see Example 3 below). Moreover, in addition to their use in diagnostic assays, the 125P5C8-related proteins and polynucleotides disclosed herein have other utilities such as their use in the forensic analysis of tissues of unknown origin (see, e.g., Takahama K Forensic Sci Int 1996 Jun. 28; 80(1-2): 63-9).


Additionally, 125P5C8-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of 125P5C8. For example, the amino acid or nucleic acid sequence of FIG. 2 or FIG. 3, or fragments of either, can be used to generate an immune response to a 125P5C8 antigen. Antibodies or other molecules that react with 125P5C8 can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.


XII.) Inhibition of 125P5C8 Protein Function


The invention includes various methods and compositions for inhibiting the binding of 125P5C8 to its binding partner or its association with other protein(s) as well as methods for inhibiting 125P5C8 function.


XII.A.) Inhibition of 125P5C8 with Intracellular Antibodies


In one approach, a recombinant vector that encodes single chain antibodies that specifically bind to 125P5C8 are introduced into 125P5C8 expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti-125P5C8 antibody is expressed intracellularly, binds to 125P5C8 protein, and thereby inhibits its function. Methods for engineering such intracellular single chain antibodies are well known. Such intracellular antibodies, also known as “intrabodies”, are specifically targeted to a particular compartment within the cell, providing control over where the inhibitory activity of the treatment is focused. This technology has been successfully applied in the art (for review, see Richardson and Marasco, 1995, TIBTECH vol. 13). Intrabodies have been shown to virtually eliminate the expression of otherwise abundant cell surface receptors (see, e.g., Richardson et al., 1995, Proc. Natl. Acad. Sci. USA 92: 3137-3141; Beerli et al., 1994, J. Biol. Chem. 289: 23931-23936; Deshane et al., 1994, Gene Ther. 1: 332-337).


Single chain antibodies comprise the variable domains of the heavy and light chain joined by a flexible linker polypeptide, and are expressed as a single polypeptide. Optionally, single chain antibodies are expressed as a single chain variable region fragment joined to the light chain constant region. Well-known intracellular trafficking signals are engineered into recombinant polynucleotide vectors encoding such single chain antibodies in order to precisely target the intrabody to the desired intracellular compartment. For example, intrabodies targeted to the endoplasmic reticulum (ER) are engineered to incorporate a leader peptide and, optionally, a C-terminal ER retention signal, such as the KDEL amino acid motif. Intrabodies intended to exert activity in the nucleus are engineered to include a nuclear localization signal. Lipid moieties are joined to intrabodies in order to tether the intrabody to the cytosolic side of the plasma membrane. Intrabodies can also be targeted to exert function in the cytosol. For example, cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.


In one embodiment, intrabodies are used to capture 125P5C8 in the nucleus, thereby preventing its activity within the nucleus. Nuclear targeting signals are engineered into such 125P5C8 intrabodies in order to achieve the desired targeting. Such 125P5C8 intrabodies are designed to bind specifically to a particular 125P5C8 domain. In another embodiment, cytosolic intrabodies that specifically bind to a 125P5C8 protein are used to prevent 125P5C8 from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing 125P5C8 from forming transcription complexes with other factors).


In order to specifically direct the expression of such intrabodies to particular cells, the transcription of the intrabody is placed under the regulatory control of an appropriate tumor-specific promoter and/or enhancer. In order to target intrabody expression specifically to prostate, for example, the PSA promoter and/or promoter/enhancer can be utilized (See, for example, U.S. Pat. No. 5,919,652 issued 6 Jul. 1999).


XII.B.) Inhibition of 125P5C8 with Recombinant Proteins


In another approach, recombinant molecules bind to 125P5C8 and thereby inhibit 125P5C8 function. For example, these recombinant molecules prevent or inhibit 125P5C8 from accessing/binding to its binding partner(s) or associating with other protein(s). Such recombinant molecules can, for example, contain the reactive part(s) of a 125P5C8 specific antibody molecule. In a particular embodiment, the 125P5C8 binding domain of a 125P5C8 binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two 125P5C8 ligand binding domains linked to the Fc portion of a human IgG, such as human IgG1. Such IgG portion can contain, for example, the CH2 and CH3 domains and the hinge region, but not the CH1 domain. Such dimeric fusion proteins are administered in soluble form to patients suffering from a cancer associated with the expression of 125P5C8, whereby the dimeric fusion protein specifically binds to 125P5C8 and blocks 125P5C8 interaction with a binding partner. Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.


XII.C.) Inhibition of 125P5C8 Transcription or Translation


The present invention also comprises various methods and compositions for inhibiting the transcription of the 125P5C8 gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of 125P5C8 mRNA into protein.


In one approach, a method of inhibiting the transcription of the 125P5C8 gene comprises contacting the 125P5C8 gene with a 125P5C8 antisense polynucleotide. In another approach, a method of inhibiting 125P5C8 mRNA translation comprises contacting a 125P5C8 mRNA with an antisense polynucleotide. In another approach, a 125P5C8 specific ribozyme is used to cleave a 125P5C8 message, thereby inhibiting translation. Such antisense and ribozyme based methods can also be directed to the regulatory regions of the 125P5C8 gene, such as 125P5C8 promoter and/or enhancer elements. Similarly, proteins capable of inhibiting a 125P5C8 gene transcription factor are used to inhibit 125P5C8 mRNA transcription. The various polynucleotides and compositions useful in the aforementioned methods have been described above. The use of antisense and ribozyme molecules to inhibit transcription and translation is well known in the art.


Other factors that inhibit the transcription of 125P5C8 by interfering with 125P5C8 transcriptional activation are also useful to treat cancers expressing 125P5C8. Similarly, factors that interfere with 125P5C8 processing are useful to treat cancers that express 125P5C8. Cancer treatment methods utilizing such factors are also within the scope of the invention.


XII.D.) General Considerations for Therapeutic Strategies


Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing 125P5C8 (i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 125P5C8 inhibitory molecules). A number of gene therapy approaches are known in the art. Recombinant vectors encoding 125P5C8 antisense polynucleotides, ribozymes, factors capable of interfering with 125P5C8 transcription, and so forth, can be delivered to target tumor cells using such gene therapy approaches.


The above therapeutic approaches can be combined with any one of a wide variety of surgical, chemotherapy or radiation therapy regimens. The therapeutic approaches of the invention can enable the use of reduced dosages of chemotherapy (or other therapies) and/or less frequent administration, an advantage for all patients and particularly for those that do not tolerate the toxicity of the chemotherapeutic agent well.


The anti-tumor activity of a particular composition (e.g., antisense, ribozyme, intrabody), or a combination of such compositions, can be evaluated using various in vitro and in vivo assay systems. In vitro assays that evaluate therapeutic activity include cell growth assays, soft agar assays and other assays indicative of tumor promoting activity, binding assays capable of determining the extent to which a therapeutic composition will inhibit the binding of 125P5C8 to a binding partner, etc.


In vivo, the effect of a 125P5C8 therapeutic composition can be evaluated in a suitable animal model. For example, xenogenic prostate cancer models can be used, wherein human prostate cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al., 1997, Nature Medicine 3: 402-408). For example, PCT Patent Application WO98/16628 and U.S. Pat. No. 6,107,540 describe various xenograft models of human prostate cancer capable of recapitulating the development of primary tumors, micrometastasis, and the formation of osteoblastic metastases characteristic of late stage disease. Efficacy can be predicted using assays that measure inhibition of tumor formation, tumor regression or metastasis, and the like.


In vivo assays that evaluate the promotion of apoptosis are useful in evaluating therapeutic compositions. In one embodiment, xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition.


The therapeutic compositions used in the practice of the foregoing methods can be formulated into pharmaceutical compositions comprising a carrier suitable for the desired delivery method. Suitable carriers include any material that when combined with the therapeutic composition retains the anti-tumor function of the therapeutic composition and is generally non-reactive with the patient's immune system. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like (see, generally, Remington's Pharmaceutical Sciences 16th Edition, A. Osal., Ed., 1980).


Therapeutic formulations can be solubilized and administered via any route capable of delivering the therapeutic composition to the tumor site. Potentially effective routes of administration include, but are not limited to, intravenous, parenteral, intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like. A preferred formulation for intravenous injection comprises the therapeutic composition in a solution of preserved bacteriostatic water, sterile unpreserved water, and/or diluted in polyvinylchloride or polyethylene bags containing 0.9% sterile Sodium Chloride for Injection, USP. Therapeutic protein preparations can be lyophilized and stored as sterile powders, preferably under vacuum, and then reconstituted in bacteriostatic water (containing for example, benzyl alcohol preservative) or in sterile water prior to injection.


Dosages and administration protocols for the treatment of cancers using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.


XIII.) Kits


For use in the diagnostic and therapeutic applications described herein, kits are also within the scope of the invention. Such kits can comprise a carrier, package or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in the method. For example, the container(s) can comprise a probe that is or can be detectably labeled. Such probe can be an antibody or polynucleotide specific for a 125P5C8-related protein or a 125P5C8 gene or message, respectively. Where the method utilizes nucleic acid hybridization to detect the target nucleic acid, the kit can also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence and/or a container comprising a reporter-means, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, florescent, or radioisotope label. The kit can include all or part of the amino acid sequence of FIG. 2 or FIG. 3 or analogs thereof, or a nucleic acid molecules that encodes such amino acid sequences.


The kit of the invention will typically comprise the container described above and one or more other containers comprising materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.


A label can be present on the container to indicate that the composition is used for a specific therapy or non-therapeutic application, and can also indicate directions for either in vivo or in vitro use, such as those described above. Directions and or other information can also be included on an insert which is included with the kit.


EXAMPLES

Various aspects of the invention are further described and illustrated by way of the several examples that follow, none of which are intended to limit the scope of the invention.


Example 1
SSH-Generated Isolation of a cDNA Fragment of the 125P5C8 Gene

Suppression Subtractive Hybridization (SSH) was used to identify cDNAs corresponding to genes that may be differentially expressed in prostate cancer. The SSH reaction utilized cDNA from two LAPC-9 AD xenografts. Specifically, to isolate genes that are involved in the progression of androgen dependent (AD) prostate cancer to androgen independent (AI) cancer, we conducted an experiment with the LAPC-9 AD xenograft in male SCID mice. Mice that harbored LAPC-9 AD xenografts were castrated when tumors reached a size of 1 cm in diameter. The tumors regressed in size and temporarily stopped producing the androgen dependent protein PSA. Seven to fourteen days post-castration, PSA levels were detectable again in the blood of the mice. Eventually the tumors develop an AI phenotype and start growing again in the castrated males. Tumors were harvested at different time points after castration to identify genes that are turned on or off during the transition to androgen independence.


The gene 125P5C8 was derived from an LAPC-9 AD (14 days post-castration) minus LAPC-9 AD subtraction. The SSH DNA sequence of 287 bp (FIG. 1) was designated 125P5C8.


The 125P5C8 SSH cDNA of 287 bp is listed in FIG. 1. The full length 125P5C8 cDNAs and ORFs are described in FIG. 2 with the protein sequences listed in FIGS. 2 and 3.


Materials and Methods


LAPC Xenografts and Human Tissues:


LAPC xenografts were obtained from Dr. Charles Sawyers (UCLA) and generated as described (Klein et al, 1997, Nature Med. 3: 402-408; Craft et al., 1999, Cancer Res. 59: 5030-5036). Androgen dependent and independent LAPC-4 xenografts LAPC-4 AD and AI, respectively) and LAPC-9 AD and AI xenografts were grown in male SCID mice and were passaged as small tissue chunks in recipient males. LAPC-4AI and LAPC-9AI xenografts were derived from LAPC-4D or LAPC-9AD tumors, respectively. To generate the AI xenografts, male mice bearing AD tumors were castrated and maintained for 2-3 months. After the tumors re-grew, the tumors were harvested and passaged in castrated males or in female SCID mice.


RNA Isolation:


Tumor tissues were homogenized in Trizol reagent (Life Technologies, Gibco BRL) using 10 ml/g tissue or 10 ml/108 cells to isolate total RNA. Poly A RNA was purified from total RNA using Qiagen's Oligotex mRNA Mini and Midi kits. Total and mRNA were quantified by spectrophotometric analysis (O.D. 260/280 nm) and analyzed by gel electrophoresis.


Oligonucleotides:


The following HPLC purified oligonucleotides were used.











DPNCDN (cDNA synthesis primer):







(SEQ ID NO: 23)









5′TTTTGATCAAGCTT303′







Adaptor 1:







(SEQ ID NO: 24)









5′CTAATACGACTCACTATAGGGCTCGAGCGGCCGCCCGGGCAG3′











(SEQ ID NO: 25)









3′GGCCCGTCCTAG5′







Adaptor 2:







(SEQ ID NO: 26)









5′GTAATACGACTCACTATAGGGCAGCGTGGTCGCGGCCGAG3′











(SEQ ID NO: 27)









3′CGGCTCCTAG5′







PCR primer 1:







(SEQ ID NO: 28)









5′CTAATACGACTCACTATAGGGC3′







Nested primer (NP)1:







(SEQ ID NO: 29)









5′TCGAGCGGCCGCCCGGGCAGGA3′







Nested primer (NP)2:







(SEQ ID NO: 30)









5′AGCGTGGTCGCGGCCGAGGA3′






Suppression Subtractive Hybridization:


Suppression Subtractive Hybridization (SSH) was used to identify cDNAs corresponding to genes that may be differentially expressed in prostate cancer. The SSH reaction utilized cDNA from prostate cancer xenograft LAPC-9AD. The gene 125P5C8 was derived from an LAPC-9 AD minus LAPC-9 AD (28 days post-castration) subtraction. The SSH DNA sequence (FIG. 1) was identified.


The cDNA derived from prostate cancer xenograft LAPC-9AD tissue was used as the source of the “driver” cDNA, while the cDNA from prostate cancer xenograft LAPC-9AD (28 days post-castration) was used as the source of the “tester” cDNA. Double stranded cDNAs corresponding to tester and driver cDNAs were synthesized from 2 μg of poly(A)+ RNA isolated from the relevant tissue, as described above, using CLONTECH's PCR-Select cDNA Subtraction Kit and 1 ng of oligonucleotide DPNCDN as primer. First- and second-strand synthesis were carried out as described in the Kit's user manual protocol (CLONTECH Protocol No. PT1117-1, Catalog No. K1804-1). The resulting cDNA was digested with Dpn II for 3 hrs at 37° C. Digested cDNA was extracted with phenol/chloroform (1:1) and ethanol precipitated.


Tester cDNA was generated by diluting 11 of Dpn II digested cDNA from the relevant tissue source (see above) (400 ng) in 5 μl of water. The diluted cDNA (2 μl, 160 ng) was then ligated to 2 μl of Adaptor 1 and Adaptor 2 (10 μM), in separate ligation reactions, in a total volume of 10 μl at 16° C. overnight, using 400 U of T4 DNA ligase (CLONTECH). Ligation was terminated with 1 μl of 0.2 M EDTA and heating at 72° C. for 5 min.


The first hybridization was performed by adding 1.5 μl (600 ng) of driver cDNA to each of two tubes containing 1.5 μl (20 ng) Adaptor 1- and Adaptor 2-ligated tester cDNA. In a final volume of 4 μl, the samples were overlaid with mineral oil, denatured in an MJ Research thermal cycler at 98° C. for 1.5 minutes, and then were allowed to hybridize for 8 hrs at 68° C. The two hybridizations were then mixed together with an additional 1 μl of fresh denatured driver cDNA and were allowed to hybridize overnight at 68° C. The second hybridization was then diluted in 2001 of 20 mM Hepes, pH 8.3, 50 mM NaCl, 0.2 mM EDTA, heated at 70° C. for 7 min. and stored at −20° C.


PCR Amplification, Cloning and Sequencing of Gene Fragments Generated from SSH:


To amplify gene fragments resulting from SSH reactions, two PCR amplifications were performed. In the primary PCR reaction 1 μl of the diluted final hybridization mix was added to 1 μl of PCR primer 1 (10 μM), 0.5 μl dNTP mix (10 μM), 2.5 μl 10× reaction buffer (CLONTECH) and 0.5 μl 50×Advantage cDNA polymerase Mix (CLONTECH) in a final volume of 25 μl. PCR 1 was conducted using the following conditions: 75° C. for 5 min., 94° C. for 25 sec., then 27 cycles of 94° C. for 10 sec, 66° C. for 30 sec, 72° C. for 1.5 min. Five separate primary PCR reactions were performed for each experiment. The products were pooled and diluted 1:10 with water. For the secondary PCR reaction, 1 μl from the pooled and diluted primary PCR reaction was added to the same reaction mix as used for PCR 1, except that primers NP1 and NP2 (10 μM) were used instead of PCR primer 1. PCR 2 was performed using 10-12 cycles of 94° C. for 10 sec, 68° C. for 30 sec, and 72° C. for 1.5 minutes. The PCR products were analyzed using 2% agarose gel electrophoresis.


The PCR products were inserted into pCR2.1 using the T/A vector cloning kit (Invitrogen). Transformed E. coli were subjected to blue/white and ampicillin selection. White colonies were picked and arrayed into 96 well plates and were grown in liquid culture overnight. To identify inserts, PCR amplification was performed on 1 ml of bacterial culture using the conditions of PCR1 and NP1 and NP2 as primers. PCR products were analyzed using 2% agarose gel electrophoresis.


Bacterial clones were stored in 20% glycerol in a 96 well format. Plasmid DNA was prepared, sequenced, and subjected to nucleic acid homology searches of the GenBank, dBest, and NCI-CGAP databases.


RT-PCR Expression Analysis:


First strand cDNAs can be generated from 1 μg of mRNA with oligo (dT)12-18 priming using the Gibco-BRL Superscript Preamplification system. The manufacturer's protocol was used which included an incubation for 50 min at 42° C. with reverse transcriptase followed by RNAse H treatment at 37° C. for 20 min. After completing the reaction, the volume can be increased to 200 μl with water prior to normalization.


Normalization of the first strand cDNAs from multiple tissues was performed by using the primers 5′atatcgccgcgctcgtcgtcgacaa 3′ (SEQ ID NO: 31) and 5′agccacacgcagctcattgtagaagg 3′ (SEQ ID NO: 32) to amplify β-actin. First strand cDNA (5 μl) were amplified in a total volume of 50 μl containing 0.4 μM primers, 0.2 μM each dNTPs, 1×PCR buffer (Clontech, 10 mM Tris-HCL, 1.5 mM MgCl2, 50 mM KCl, pH8.3) and 1× Klentaq DNA polymerase (Clontech). Five μl of the PCR reaction can be removed at 18, 20, and 22 cycles and used for agarose gel electrophoresis. PCR was performed using an MJ Research thermal cycler under the following conditions: Initial denaturation can be at 94° C. for 15 sec, followed by a 18, 20, and 22 cycles of 94° C. for 15, 65° C. for 2 min, 72° C. for 5 sec. A final extension at 72° C. was carried out for 2 min. After agarose gel electrophoresis, the band intensities of the 283 bp β-actin bands from multiple tissues were compared by visual inspection. Dilution factors for the first strand cDNAs were calculated to result in equal β-actin band intensities in all tissues after 22 cycles of PCR. Three rounds of normalization can be required to achieve equal band intensities in all tissues after 22 cycles of PCR.


To determine expression levels of the 125P5C8 gene, 5 μl of normalized first strand cDNA were analyzed by PCR using 26, and 30 cycles of amplification. Semi-quantitative expression analysis can be achieved by comparing the PCR products at cycle numbers that give light band intensities.


A typical RT-PCR expression analysis is shown in FIG. 12. First strand cDNA was prepared from vital pool 1 (liver, lung and kidney), vital pool 2 (pancreas, colon and stomach), LAPC xenograft pool (LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI), prostate cancer pool, bladder cancer pool, kidney cancer pool, colon cancer pool, ovary cancer pool, breast cancer pool, and metastasis cancer pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi-quantitative PCR, using primers to 125P5C8, was performed at 26 and 30 cycles of amplification. 125P5C8 Expression was observed in normal prostate, prostate cancer xenografts, prostate cancer pool, bladder cancer pool, kidney cancer pool, colon cancer pool, and cancer metastasis pool. Expression was also detected in vital pool 1 and vital pool 2, albeit to lower levels than xenograft cancer pool and prostate cancer pool.


Example 2
Full Length Cloning of 125P5C8 and Homology Comparison to Known Sequences

To isolate genes that are involved in the progression of androgen dependent (AD) prostate cancer to androgen independent (AI) cancer, we conducted an experiment with the LAPC-9AD xenograft in male SCID mice. Mice that harbored LAPC-9AD xenografts were castrated when the tumors reached a size of 1 cm in diameter. The tumors regressed in size and temporarily stopped producing the androgen dependent protein PSA. Seven to fourteen days post-castration, PSA levels were detectable again in the blood of the mice. Eventually the tumors develop an AI phenotype and start growing again in the castrated males. Tumors were harvested at different time points after castration to identify genes that are turned on or off during the transition to androgen independence.


The gene 125P5C8 was derived from an LAPC-9AD (14 days post-castration) minus LAPC-9AD subtraction. The SSH DNA sequence of 287 bp (FIG. 1) was designated 125P5C8.


The full-length 125P5C8 cDNA clone 125P5C8-Pro-pCR2.1 (FIG. 2) was identified by assembling EST fragments homologous to 125P5C8 into a large contiguous sequence with an ORF and amplifying the ORF by PCR using normal prostate first strand cDNA. The 125P5C8 v.1 cDNA clone 125P5C8-Pro-pCR2.1 encodes a 699 amino acid ORF with 10 transmembrane domains predicted at the cell surface based on PSORT analysis (http://psort.nibb.acjp:8800/form.html). Further variants of 125P5C8 were identified to have single nucleotide polymorphisms and are shown in FIGS. 2-3, and FIG. 10-11.


The 125P5C8 v.1 clone 125P5C8-Pro-pCR2.1 protein is the same as the GenBank protein AK025164 with one amino acid difference (FIG. 4). This amino acid difference at amino acid position 689 may be significant since it is located in the long extracellular C-terminal region that may be involved in ligand binding, may affect the stability of the protein, or may be involved in the binding of the 125P5C8 protein to itself or other proteins. In addition, 125P5C8 is homologous to several yeast proteins, one of which is predicted to be localized to the cell surface and involved in drug sensitivity (FIG. 4).


As provided above, the 125P5C8 v.1 cDNA was deposited with the ATCC as plasmid 125P5C8-Pro-pCR2.1, and has been assigned Accession No. PTA-3137.


Example 3
Chromosomal Localization of 125P5C8

Chromosomal localization can implicate genes in disease pathogenesis. Several chromosome mapping approaches are available, including fluorescent in situ hybridization (FISH), human/hamster radiation hybrid (RH) panels (Walter et al., 1994; Nature Genetics 7:22; Research Genetics, Huntsville Ala.), human-rodent somatic cell hybrid panels such as is available from the Coriell Institute (Camden, N.J.), and genomic viewers utilizing BLAST homologies to sequenced and mapped genomic clones (NCBI, Bethesda, Md.).


Using 125P5C8 sequence and the NCBI BLAST tool, 125P5C8 maps to chromosome 6q23, between D6S1040 and D6S457, a genomic region found to be rearranged in certain cancers.


Example 4
Expression Analysis of 125P5C8 in Normal Tissues and Patient Specimens

Expression analysis by RT-PCR demonstrated that 125P5C8 is strongly expressed in prostate cancer xenografts, normal prostate and in prostate cancer patient specimens (FIGS. 12 and 13). Expression is also detected in patient kidney cancer pool, bladder cancer pool, colon cancer pool, ovary cancer pool, breast cancer pool, cancer metastasis pool, as well as vital pool 1 and vital pool 2, albeit to lower levels.


Extensive Northern blot analysis of 125P5C8 in 16 human normal tissues is shown in FIG. 14. An approximately 3 kb transcript is detected in prostate and to lower levels in colon and kidney. 125P5C8 expression was also shown in prostate cancer xenografts.


Northern blot analysis on patient tumor specimens shows expression of 125P5C8 in all prostate tumor tissues tested (FIG. 15). However, expression of 125P5C8 seems to be lower in kidney cancers compared to normal kidney (FIG. 16).


The restricted expression of 125P5C8 in normal tissues and the expression detected in prostate cancer, bladder cancer, colon cancer, kidney cancer, ovarian cancer, and breast cancer suggest that 125P5C8 is a potential therapeutic target and a diagnostic marker for human cancers.


Example 5
Splice Variants of 125P5C8

Splice variants are alternatively spliced transcripts. When a gene is transcribed from genomic DNA, the initial RNA is generally spliced to produce functional mRNA, which has only exons and is used for translation into an amino acid sequence. Accordingly, a given gene can have zero to many alternatively spliced mRNA products. Alternative transcripts each have a unique exon makeup, and can have different coding and/or non-coding (5′ or 3′ end) portions, from the original transcript. Alternative transcripts can code for similar proteins with the same or a similar function or may encode proteins with different functions, and may be expressed in the same tissue at the same time, or at different tissue at different times, proteins encoded by alternative transcripts can have similar or different cellular or extracellular localizations, e.g., be secreted.


Splice variants are identified by a variety of art-accepted methods. For example, splice variants are identified by use of EST data. First, all human ESTs were grouped into clusters which show direct or indirect identity with each other. Second, ESTs in the same cluster were further grouped into sub-clusters and assembled into a consensus sequence. The starting gene is compared to the consensus sequence(s). Each consensus sequence is a potential splice variant for that gene. Even when a variant is identified that is not a full-length clone, that portion of the variant is very useful for antigen generation and for further cloning of the full-length splice variant, using techniques known in the art.


Moreover, computer programs are available in the art that identify splice variants based on genomic sequences. Genomic-based variant identification programs include FgenesH (A. Salamov and V. Solovyev, “Ab initio gene finding in Drosophila genomic DNA,” Genome Research. 2000 April; 10(4):516-22); Grail and GenScan. For a general discussion of splice variant identification protocols see., e.g., Southan C., “A genomic perspective on human proteases,” FEBS Lett. 2001 Jun. 8; 498(2-3):214-8; de Souza S J, et al., “Identification of human chromosome 22 transcribed sequences with ORF expressed sequence tags,” Proc. Natl. Acad Sci USA. 2000 Nov. 7; 97(23):12690-3.


To further confirm the parameters of a splice variant, a variety of techniques are available in the art, such as full-length cloning, proteomic validation, PCR-based validation, and 5′ RACE validation, etc. (see e.g., Proteomic Validation: Brennan S O, Fellowes A P, George P M.; “Albumin banks peninsula: a new termination variant characterised by electrospray mass spectrometry.” Biochim Biophys Acta. 1999 Aug. 17; 1433(1-2):321-6; Ferranti P, et al., “Differential splicing of pre-messenger RNA produces multiple forms of mature caprine alpha(s1)-casein.” Eur J. Biochem. 1997 Oct. 1; 249(1):1-7; PCR-based Validation: Wellmann S, et al., “Specific reverse transcription-PCR quantification of vascular endothelial growth factor (VEGF) splice variants by LightCycler technology.” Clin Chem. 2001 April; 47(4):654-60; Jia H P, et al., Discovery of new human beta-defensins using a genomics-based approach,” Gene. 2001 Jan. 24; 263(1-2):211-8; PCR-based and 5′ RACE Validation: Brigle K E, et al., “Organization of the murine reduced folate carrier gene and identification of variant splice forms,” Biochim Biophys Acta. 1997 Aug. 7; 1353(2): 191-8.


It is known in the art that genomic regions are modulated in cancers. When the genomic region to which the gene maps is modulated in a particular cancer, the splice variants are modulated as well. Disclosed herein is that 125P5C8 has a particular expression profile. Splice variants of 125P5C8 that are structurally and/or functionally similar to 125P5C8 share this expression pattern, thus serving as tumor-associated markers/antigens.


Using full-length cloning and EST data, no splice variant was identified for this gene.


Example 6
Single Nucleotide Polymorphisms of 125P5C8

Single Nucleotide Polymorphism (SNP) is a single base pair variation in nucleotide sequences. At a specific point of the genome, there are four possible nucleotide base pairs: A/T, C/G, G/C and T/A. Genotype refers to the base pair make-up of one or more spots in the genome of an individual, while haplotype refers to base pair make-up of more than one varied spots on the same DNA molecule (chromosome in higher organism). SNPs that occur on a cDNA are called cSNPs. These cSNPs may change amino acids of the protein encoded by the gene and thus change the functions of the protein. Some SNPs cause inherited diseases and some others contribute to quantitative variations in phenotype and reactions to environmental factors including diet and drugs among individuals. Therefore, SNPs and/or combinations of alleles (called haplotypes) have many applications including diagnosis of inherited diseases, determination of drug reactions and dosage, identification of genes responsible for diseases and discovery of genetic relationship between individuals (P. Nowotny, J. M. Kwon and A. M. Goate, “SNP analysis to dissect human traits,” Curr. Opin. Neurobiol. 2001 October; 11(5):637-641; M. Pirmohamed and B. K. Park, “Genetic susceptibility to adverse drug reactions,” Trends Pharmacol. Sci. 2001 June; 22(6):298-305; J. H. Riley, C. J. Allan, E. Lai and A. Roses, “The use of single nucleotide polymorphisms in the isolation of common disease genes,” Pharmacogenomics. 2000 February; 1(1):39-47; R. Judson, J. C. Stephens and A. Windemuth, “The predictive power of haplotypes in clinical response,” Pharmacogenomics. 2000 February; 1(1): 15-26).


SNPs are identified by a variety of art-accepted methods (P. Bean, “The promising voyage of SNP target discovery,” Am. Clin. Lab. 2001 October-November; 20(9):18-20; K. M. Weiss, “In search of human variation,” Genome Res. 1998 July; 8(7):691-697; M. M. She, “Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies,” Clin. Chem. 2001 February; 47(2):164-172). For example, SNPs are identified by sequencing DNA fragments that show polymorphism by gel-based methods such as restriction fragment length polymorphism (RFLP) and denaturing gradient gel electrophoresis (DGGE). They can also be discovered by direct sequencing of DNA samples pooled from different individuals or by comparing sequences from different DNA samples. With the rapid accumulation of sequence data in public and private databases, one can discover SNPs by comparing sequences using computer programs (Z. Gu, L. Hillier and P. Y. Kwok, “Single nucleotide polymorphism hunting in cyberspace,” Hum. Mutat. 1998; 12(4):221-225). SNPs can be verified and genotype or haplotype of an individual can be determined by a variety of methods including direct sequencing and high throughput microarrays (P. Y. Kwok, “Methods for genotyping single nucleotide polymorphisms,” Annu. Rev. Genomics Hum. Genet. 2001; 2:235-258; M. Kokoris, K. Dix, K. Moynihan, J. Mathis, B. Erwin, P. Grass, B. Hines and A. Duesterhoeft, “High-throughput SNP genotyping with the Masscode system,” Mol. Diagn. 2000 December; 5(4):329-340).


In addition, SNPs are identified by directly sequencing cDNA clones of the invention and by comparing the sequences with public and proprietary sequences. By sequencing these cDNA clones, one SNP was identified and the transcript, i.e. protein, with the alternative allele was designated as variant 5. By comparing the variant 1 sequence with high quality proprietary or public sequences, four SNPs were identified and the transcripts or proteins with alternative alleles were designated as variants 2, 3, 4, and 6. FIG. 10 shows the schematic alignment of the nucleotide variants. FIG. 11 shows the schematic alignment of protein variants, with variant nomenclature corresponding to nucleotide variants. Nucleotide variant 125P5C8 v.6 (FIG. 10) codes for the same protein as 125P5C8 v.1 (see FIG. 11). These alleles of the SNPs, though shown separately here, can occur in different combinations (haplotypes).


Example 7
Production of Recombinant 125P5C8 in Prokaryotic Systems

To express recombinant 125P5C8 in prokaryotic cells, the full or partial length 125P5C8 cDNA sequences can be cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 125P5C8 are expressed in these constructs, amino acids 1 to 699; or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 125P5C8, variants, or analogs thereof.


A. In Vitro Transcription and Translation Constructs:


pCRII: To generate 125P5C8 sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad Calif.) are generated encoding either all or fragments of the 125P5C8 cDNA. The pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of 125P5C8 RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of 125P5C8 at the RNA level. Transcribed 125P5C8 RNA representing the cDNA amino acid coding region of the 125P5C8 gene is used in in vitro translation systems such as the TnT™ Coupled Reticulolysate System (Promega, Corp., Madison, Wis.) to synthesize 125P5C8 protein.


B. Bacterial Constructs:


pGEX Constructs: To generate recombinant 125P5C8 proteins in bacteria that are fused to the Glutathione S-transferase (GST) protein, all or parts of the 125P5C8 cDNA protein coding sequence are fused to the GST gene by cloning into pGEX-6P-1 or any other GST-fusion vector of the pGEX family (Amersham Pharmacia Biotech, Piscataway, N.J.). These constructs allow controlled expression of recombinant 125P5C8 protein sequences with GST fused at the amino-terminus and a six histidine epitope (6×His) at the carboxyl-terminus. The GST and 6×His tags permit purification of the recombinant fusion protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-GST and anti-His antibodies. The 6×His tag is generated by adding 6 histidine codons to the cloning primer at the 3′ end, e.g., of the open reading frame (ORF). A proteolytic cleavage site, such as the PreScission™ recognition site in pGEX-6P-1, may be employed such that it permits cleavage of the GST tag from 125P5C8-related protein. The ampicillin resistance gene and pBR322 origin permits selection and maintenance of the pGEX plasmids in E. coli.


pMAL Constructs: To generate, in bacteria, recombinant 125P5C8 proteins that are fused to maltose-binding protein (MBP), all or parts of the 125P5C8 cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, Mass.). These constructs allow controlled expression of recombinant 125P5C8 protein sequences with MBP fused at the amino-terminus and a 6×His epitope tag at the carboxyl-terminus. The MBP and 6×His tags permit purification of the recombinant protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-MBP and anti-His antibodies. The 6×His epitope tag is generated by adding 6 histidine codons to the 3′ cloning primer. A Factor Xa recognition site permits cleavage of the pMAL tag from 125P5C8. The pMAL-c2X and pMAL-p2X vectors are optimized to express the recombinant protein in the cytoplasm or periplasm respectively. Periplasm expression enhances folding of proteins with disulfide bonds.


pET Constructs: To express 125P5C8 in bacterial cells, all or parts of the 125P5C8 cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, Wis.). These vectors allow tightly controlled expression of recombinant 125P5C8 protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6×His and S-Tag™ that aid purification and detection of the recombinant protein. For example, constructs are made utilizing pET NusA fusion system 43.1 such that regions of the 125P5C8 protein are expressed as amino-terminal fusions to NusA. In one embodiment, a NusA-fusion protein encompassing amino acids 412-699 of 125P5C8 with a C-terminal 6×His tag was expressed in E. Coli, purified by metal chelate affinity chromatography, and used as an immunogen for generation of antibodies.


C. Yeast Constructs:


pESC Constructs: To express 125P5C8 in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the 125P5C8 cDNA protein coding sequence are cloned into the pESC family of vectors each of which contain 1 of 4 selectable markers, HIS3, TRP1, LEU2, and URA3 (Stratagene, La Jolla, Calif.). These vectors allow controlled expression from the same plasmid of up to 2 different genes or cloned sequences containing either Flag™ or Myc epitope tags in the same yeast cell. This system is useful to confirm protein-protein interactions of 125P5C8. In addition, expression in yeast yields similar post-translational modifications, such as glycosylations and phosphorylations, that are found when expressed in eukaryotic cells.


pESP Constructs: To express 125P5C8 in the yeast species Saccharomyces pombe, all or parts of the 125P5C8 cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a 125P5C8 protein sequence that is fused at either the amino terminus or at the carboxyl terminus to GST which aids purification of the recombinant protein. A Flag™ epitope tag allows detection of the recombinant protein with anti-Flag™ antibody.


Example 8
Production of Recombinant 125P5C8 in Eukaryotic Systems

A. Mammalian Constructs:


To express recombinant 125P5C8 in eukaryotic cells, the full or partial length 125P5C8 cDNA sequences were cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 125P5C8 were expressed in these constructs, amino acids 1 to 699, or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 125P5C8, variants, or analogs thereof. In certain embodiments a region of 125P5C8 was expressed that encodes an amino acid not shared amongst at least variants.


The constructs were transfected into any one of a wide variety of mammalian cells such as 293T cells. Transfected 293T cell lysates can be probed with the anti-125P5C8 polyclonal serum, described herein.


pcDNA4/HisMax Constructs: To express 125P5C8 in mammalian cells, a 125P5C8 ORF, or portions thereof, of 125P5C8 are cloned into pcDNA4/HisMax Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter and the SP16 translational enhancer. The recombinant protein has Xpress™ and six histidine (6×His) epitopes fused to the amino-terminus. The pcDNA4/HisMax vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Zeocin resistance gene allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.


pcDNA3.1/MycHis Constructs: To express 125P5C8 in mammalian cells, a 125P5C8 ORF, or portions thereof, of 125P5C8 with a consensus Kozak translation initiation site was cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the myc epitope and 6×His epitope fused to the carboxyl-terminus. The pcDNA3.1/MycHis vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability, along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Neomycin resistance gene can be used, as it allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli. FIG. 17 shows expression of 125P5C8.pcDNA3.1/mychis in transiently transfected 293T cells.


pcDNA3.1/CT-GFP-TOPO Construct: To express 125P5C8 in mammalian cells and to allow detection of the recombinant proteins using fluorescence, a 125P5C8 ORF, or portions thereof, with a consensus Kozak translation initiation site are cloned into pcDNA3.1/CT-GFP-TOPO (Invitrogen, CA). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the Green Fluorescent Protein (GFP) fused to the carboxyl-terminus facilitating non-invasive, in vivo detection and cell biology studies. The pcDNA3.1CT-GFP-TOPO vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Neomycin resistance gene allows for selection of mammalian cells that express the protein, and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli. Additional constructs with an amino-terminal GFP fusion are made in pcDNA3.1/NT-GFP-TOPO spanning the entire length of a 125P5C8 protein.


PAPtag: A 125P5C8 ORF, or portions thereof, is cloned into pAPtag-5 (GenHunter Corp. Nashville, Tenn.). This construct generates an alkaline phosphatase fusion at the carboxyl-terminus of a 125P5C8 protein while fusing the IgGK signal sequence to the amino-terminus. Constructs are also generated in which alkaline phosphatase with an amino-terminal IgGK signal sequence is fused to the amino-terminus of a 125P5C8 protein. The resulting recombinant 125P5C8 proteins are optimized for secretion into the media of transfected mammalian cells and can be used to identify proteins such as ligands or receptors that interact with 125P5C8 proteins. Protein expression is driven from the CMV promoter and the recombinant proteins also contain myc and 6×His epitopes fused at the carboxyl-terminus that facilitates detection and purification. The Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the recombinant protein and the ampicillin resistance gene permits selection of the plasmid in E. coli.


ptag5: A 125P5C8 ORF, or portions thereof, is cloned into pTag-5. This vector is similar to pAPtag but without the alkaline phosphatase fusion. This construct generates 125P5C8 protein with an amino-terminal IgGK signal sequence and myc and 6×His epitope tags at the carboxyl-terminus that facilitate detection and affinity purification. The resulting recombinant 125P5C8 protein is optimized for secretion into the media of transfected mammalian cells, and is used as immunogen or ligand to identify proteins such as ligands or receptors that interact with the 125P5C8 proteins. Protein expression is driven from the CMV promoter. The Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.


PsecFc: A 125P5C8 ORF, or portions thereof, is also cloned into psecFc. The psecFc vector was assembled by cloning the human immunoglobulin G1 (IgG) Fc (hinge, CH2, CH3 regions) into pSecTag2 (Invitrogen, California). This construct generates an IgG1 Fc fusion at the carboxyl-terminus of the 125P5C8 proteins, while fusing the IgGK signal sequence to N-terminus. 125P5C8 fusions utilizing the murine IgG1 Fc region are also used. The resulting recombinant 125P5C8 proteins are optimized for secretion into the media of transfected mammalian cells, and can be used as immunogens or to identify proteins such as ligands or receptors that interact with 125P5C8 protein. Protein expression is driven from the CMV promoter. The hygromycin resistance gene present in the vector allows for selection of mammalian cells that express the recombinant protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.


pSRα Constructs: To generate mammalian cell lines that express 125P5C8 constitutively, 125P5C8 ORF, or portions thereof, of 125P5C8 were cloned into pSRα constructs. Amphotropic and ecotropic retroviruses were generated by transfection of pSRα constructs into the 293T-10A1 packaging line or co-transfection of pSRα and a helper plasmid (containing deleted packaging sequences) into the 293 cells, respectively. The retrovirus was used to infect a variety of mammalian cell lines, resulting in the integration of the cloned gene, 125P5C8, into the host cell-lines. Protein expression was driven from a long terminal repeat (LTR). The Neomycin resistance gene present in the vector allows for selection of mammalian cells that express the protein, and the ampicillin resistance gene and ColE1 origin permit selection and maintenance of the plasmid in E. coli. The retroviral vectors were thereafter used for infection and generation of various cell lines using, for example, PC3, NIH 3T3, TsuPr1, 293 or rat-1 cells.


Additional pSRα constructs are made that fuse an epitope tag such as the FLAG™ tag to the carboxyl-terminus of 125P5C8 sequences to allow detection using anti-Flag antibodies. For example, the FLAG™ sequence 5′ gat tac aag gat gac gac gat aag 3′ (SEQ ID NO: 33) is added to cloning primer at the 3′ end of the ORF. Additional pSRα constructs are made to produce both amino-terminal and carboxyl-terminal GFP and myc/6×His fusion proteins of the full-length 125P5C8 proteins.


Additional Viral Vectors: Additional constructs are made for viral-mediated delivery and expression of 125P5C8. High virus titer leading to high level expression of 125P5C8 is achieved in viral delivery systems such as adenoviral vectors and herpes amplicon vectors. A 125P5C8 coding sequences or fragments thereof are amplified by PCR and subcloned into the AdEasy shuttle vector (Stratagene). Recombination and virus packaging are performed according to the manufacturer's instructions to generate adenoviral vectors. Alternatively, 125P5C8 coding sequences or fragments thereof are cloned into the HSV-1 vector (Imgenex) to generate herpes viral vectors. The viral vectors are thereafter used for infection of various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.


Regulated Expression Systems: To control expression of 125P5C8 in mammalian cells, coding sequences of 125P5C8, or portions thereof, are cloned into regulated mammalian expression systems such as the T-Rex System (Invitrogen), the GeneSwitch System (Invitrogen) and the tightly-regulated Ecdysone System (Sratagene). These systems allow the study of the temporal and concentration dependent effects of recombinant 125P5C8. These vectors are thereafter used to control expression of 125P5C8 in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.


B. Baculovirus Expression Systems


To generate recombinant 125P5C8 proteins in a baculovirus expression system, 125P5C8 ORF, or portions thereof, are cloned into the baculovirus transfer vector pBlueBac 4.5 (Invitrogen), which provides a His-tag at the N-terminus. Specifically, pBlueBac-125P5C8 is co-transfected with helper plasmid pBac-N-Blue (Invitrogen) into SF9 (Spodoptera frugiperda) insect cells to generate recombinant baculovirus (see Invitrogen instruction manual for details). Baculovirus is then collected from cell supernatant and purified by plaque assay.


Recombinant 125P5C8 protein is then generated by infection of HighFive insect cells (Invitrogen) with purified baculovirus. Recombinant 125P5C8 protein can be detected using anti-125P5C8 or anti-His-tag antibody. 125P5C8 protein can be purified and used in various cell-based assays or as immunogen to generate polyclonal and monoclonal antibodies specific for 125P5C8. xx


Example 9
Antigenicity Profiles and Secondary Structure


FIG. 5, FIG. 6, FIG. 7, FIG. 8, and FIG. 9 depict graphically five amino acid profiles of the 125P5C8 var1 amino acid sequence, each assessment available by accessing the ProtScale website on the ExPasy molecular biology server; since variants 2-5 are SNPs of variant 1, these profiles would be the same for all variants.


These profiles: FIG. 5, Hydrophilicity, (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828); FIG. 6, Hydropathicity, (Kyte J., Doolittle R. F., 1982. J. Mol. Biol. 157:105-132); FIG. 7, Percentage Accessible Residues (Janin J., 1979 Nature 277:491-492); FIG. 8, Average Flexibility, (Bhaskaran R., and Ponnuswamy P. K., 1988. Int. J. Pept. Protein Res. 32:242-255); FIG. 9, Beta-turn (Deleage, G., Roux B. 1987 Protein Engineering 1:289-294); and optionally others available in the art, such as on the ProtScale website, were used to identify antigenic regions of the 125P5C8 protein. Each of the above amino acid profiles of 125P5C8 were generated using the following ProtScale parameters for analysis: 1) A window size of 9; 2) 100% weight of the window edges compared to the window center; and, 3) amino acid profile values normalized to lie between 0 and 1.


Hydrophilicity (FIG. 5), Hydropathicity (FIG. 6) and Percentage Accessible Residues (FIG. 7) profiles were used to determine stretches of hydrophilic amino acids (i.e., values greater than 0.5 on the Hydrophilicity and Percentage Accessible Residues profile, and values less than 0.5 on the Hydropathicity profile). Such regions are likely to be exposed to the aqueous environment, be present on the surface of the protein, and thus available for immune recognition, such as by antibodies.


Average Flexibility (FIG. 8) and Beta-turn (FIG. 9) profiles determine stretches of amino acids (i.e., values greater than 0.5 on the Beta-turn profile and the Average Flexibility profile) that are not constrained in secondary structures such as beta sheets and alpha helices. Such regions are also more likely to be exposed on the protein and thus accessible to immune recognition, such as by antibodies.


Antigenic sequences of the 125P5C8 protein indicated, e.g., by the profiles set forth in FIG. 5, FIG. 6, FIG. 7, FIG. 8, and/or FIG. 9 are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-125P5C8 antibodies. The immunogen can be any 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more than 50 contiguous amino acids, or the corresponding nucleic acids that encode them, from the 125P5C8 protein. In particular, peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of FIG. 2 in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5; a peptide region of at least 5 amino acids of FIG. 2 in any whole number increment up to 699 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of FIG. 6; a peptide region of at least 5 amino acids of FIG. 2 in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7; a peptide region of at least 5 amino acids of FIG. 2 in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile on FIG. 8; and, a peptide region of at least 5 amino acids of FIG. 2 in any whole number increment up to 699 that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of FIG. 9. Peptide immunogens of the invention can also comprise nucleic acids that encode any of the forgoing.


All immunogens of the invention, peptide or nucleic acid, can be embodied in human unit dose form, or comprised by a composition that includes a pharmaceutical excipient compatible with human physiology.


The secondary structure of 125P5C8, namely the predicted presence and location of alpha helices, extended strands, and random coils, is predicted from the primary amino acid sequence using the HNN—Hierarchical Neural Network method (Guermeur, 1997) accessed from the ExPasy molecular biology server. The analysis indicates that 125P5C8 is composed 40.49% alpha helix, 17.31% extended strand, and 42.20% random coil (FIG. 20A).


Analysis for the potential presence of transmembrane domains in 125P5C8 was carried out using a variety of transmembrane prediction algorithms accessed from the ExPasy molecular biology server. The programs predict the presence of multiple transmembrane domain in 125P5C8. The highest probability of topology is that of a 10 transmembrane cell surface protein with the amino and carboxyl termini being intracellular. Shown graphically in FIGS. 20B and 20C are the results of analysis using the TMpred (FIG. 20B) and TMHMM (FIG. 20C) prediction programs depicting the location of the transmembrane domain.


Example 10
Generation of 125P5C8 Polyclonal Antibodies

Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. In addition to immunizing with the full length 125P5C8 protein, computer algorithms are employed in design of immunogens that, based on amino acid sequence analysis contain characteristics of being antigenic and available for recognition by the immune system of the immunized host (see Example 9). Such regions would be predicted to be hydrophilic, flexible, in beta-turn conformations, and be exposed on the surface of the protein (see, e.g., FIG. 5, FIG. 6, FIG. 7, FIG. 8, or FIG. 9 for amino acid profiles that indicate such regions of 125P5C8).


For example, 125P5C8 recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta-turn regions of the 125P5C8, generally found in regions between transmembrane domains and at the amino and carboxyl termini, are used as antigens to generate polyclonal antibodies in New Zealand White rabbits. For example, such regions include, but are not limited to, amino acids 163-190 and amino acids 412-699 encoding regions predicted to be intracellular or amino acids 32-45, amino acids 112-122, amino acids 214-236, amino acids 295-317, and amino acids 368-391, regions predicted to be extracellular. It is useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include, but are not limited to, keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. In one embodiment, a peptide encoding amino acids 295-317 of 125P5C8 is conjugated to KLH and used to immunize the rabbit. Alternatively the immunizing agent may include all or portions of the 125P5C8 protein, analogs or fusion proteins thereof. For example, the 125P5C8 amino acid sequence can be fused using recombinant DNA techniques to any one of a variety of fusion protein partners that are well known in the art, such as glutathione-S-transferase (GST) and HIS tagged fusion proteins. Such fusion proteins are purified from induced bacteria using the appropriate affinity matrix.


In one embodiment, a NusA-fusion protein encoding amino acids 412-699 is produced and purified and used as immunogen. Other recombinant bacterial fusion proteins that may be employed include maltose binding protein, LacZ, thioredoxin, GST, or an immunoglobulin constant region (see the section entitled “Production of 125P5C8 in Prokaryotic Systems” and Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubul et al. eds., 1995; Linsley, P.S., Brady, W., Urnes, M., Grosmaire, L., Damle, N., and Ledbetter, L. (1991) J. Exp. Med. 174, 561-566).


In addition to bacterial derived fusion proteins, mammalian expressed protein antigens are also used. These antigens are expressed from mammalian expression vectors such as the Tag5 and Fc-fusion vectors (see the section entitled “Production of Recombinant 125P5C8 in Eukaryotic Systems”), and retain post-translational modifications such as glycosylations found in native protein. In one embodiment, amino acids 368-391 is cloned into the Tag5 mammalian secretion vector. The recombinant protein is purified by metal chelate chromatography from tissue culture supernatants of 293T cells stably expressing the recombinant vector. The purified Tag5 125P5C8 protein is then used as immunogen.


During the immunization protocol, it is useful to mix or emulsify the antigen in adjuvants that enhance the immune response of the host animal. Examples of adjuvants include, but are not limited to, complete Freund's adjuvant (CFA) and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).


In a typical protocol, rabbits are initially immunized subcutaneously with up to 200 μg, typically 100-200 μg, of fusion protein or peptide conjugated to KLH mixed in complete Freund's adjuvant (CFA). Rabbits are then injected subcutaneously every two weeks with up to 200 μg, typically 100-200 μg, of the immunogen in incomplete Freund's adjuvant (IFA). Test bleeds are taken approximately 7-10 days following each immunization and used to monitor the titer of the antiserum by ELISA.


To test reactivity and specificity of immune serum, such as the rabbit serum derived from immunization with Tag5 125P5C8 encoding amino acids 368-391, the full-length 125P5C8 cDNA is cloned into pCDNA 3.1 myc-his expression vector (Invitrogen, see Example 8). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-125P5C8 serum and with anti-His antibody (Santa Cruz Biotechnologies, Santa Cruz, Calif.) to determine specific reactivity to denatured 125P5C8 protein using the Western blot technique. Shown in FIG. 18 is expression of Myc His tagged 125P5C8 protein in 293T cells as detected by Western blot with anti-His antibody. FIG. 19 shows cell surface expression as detected by fluorescence microscopy and flow cytometry. Moreover, immunoprecipitation and flow cytometric analyses of 293T and other recombinant 125P5C8-expressing cells determine recognition of native protein by the antiserum. In addition, Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express 125P5C8 are carried out to test specificity.


The anti-serum from the NusA 125P5C8 immunized rabbit is affinity purified by passage over a column composed of the a bacterially MBP-fusion protein encoding amino acids 412-699 covalently coupled to Affigel matrix (BioRad, Hercules, Calif.). The serum is then further purified by protein G affinity chromatography to isolate the IgG fraction. Serum from rabbits immunized with fusion proteins, such as GST and MBP fusion proteins, are purified by depletion of antibodies reactive to the fusion partner sequence by passage over an affinity column containing the fusion partner either alone or in the context of an irrelevant fusion protein. Sera from other His-tagged antigens and peptide immunized rabbits as well as fusion partner depleted sera are affinity purified by passage over a column matrix composed of the original protein immunogen or free peptide.


Example 11
Generation of 125P5C8 Monoclonal Antibodies (mAbs)

In one embodiment, therapeutic mAbs to 125P5C8 comprise those that react with epitopes of the protein that would disrupt or modulate the biological function of 125P5C8, for example those that would disrupt its biological activity such as antibodies that disrupt its interaction with ligands and binding partners. Therapeutic mAbs also comprise those that specifically bind epitopes of 125P5C8 exposed on the cell surface and thus are useful in targeting mAb-toxin conjugates. Immunogens for generation of such mAbs include those designed to encode or contain the entire 125P5C8 protein, regions of the 125P5C8 protein predicted to be antigenic from computer analysis of the amino acid sequence (see, e.g., FIG. 5, FIG. 6, FIG. 7, FIG. 8, or FIG. 9, and Example 9) such as predicted extracellular domains. Immunogens include peptides, recombinant bacterial proteins, and mammalian expressed Tag 5 proteins and human and murine IgG FC fusion proteins. In addition, cells expressing high levels of 125P5C8, such as 293T-125P5C8 or 300.19-125P5C8 murine Pre-B cells, are used to immunize mice.


To generate mAbs to 125P5C8, mice are first immunized intraperitoneally (IP) with, typically, 10-50 μg of protein immunogen or 107 125P5C8-expressing cells mixed in complete Freund's adjuvant. Mice are then subsequently immunized IP every 2-4 weeks with, typically, 10-50 μg of protein immunogen or 107 cells mixed in incomplete Freund's adjuvant. Alternatively, MPL-TDM adjuvant is used in immunizations. In addition to the above protein and cell-based immunization strategies, a DNA-based immunization protocol is employed in which a mammalian expression vector encoding 125P5C8 sequence is used to immunize mice by direct injection of the plasmid DNA. For example, amino acids 368-391 is cloned into the Tag5 mammalian secretion vector and the recombinant vector is used as immunogen. In another example the same amino acids are cloned into an Fc-fusion secretion vector in which the 125P5C8 sequence is fused at the amino-terminus to an IgK leader sequence and at the carboxyl-terminus to the coding sequence of the human or murine IgG Fc region. This recombinant vector is then used as immunogen. The plasmid immunization protocols are used in combination with purified proteins expressed from the same vector and with cells expressing 125P5C8.


During the immunization protocol, test bleeds are taken 7-10 days following an injection to monitor titer and specificity of the immune response. Once appropriate reactivity and specificity is obtained as determined by ELISA, Western blotting, immunoprecipitation, fluorescence microscopy, and flow cytometric analyses, fusion and hybridoma generation is then carried out with established procedures well known in the art (see, e.g., Harlow and Lane, 1988).


In one embodiment for generating 125P5C8 monoclonal antibodies, a Tag5-125P5C8 antigen encoding amino acids 214-236, a predicted extracellular domain, is expressed and purified from stably transfected 293T cells.


In another embodiment, Balb/c mice were immunized with 125P5C8-NusA fusion protein “the immunogen”. This fusion protein encompasses the complete protein encoded by the gene 125P5C8 fused with NusA, a commonly used recombinant protein tag. Injections of the immunogen were given at two week intervals intra-peritoneally. A total of 3 injections were given the first in Complete Freund's Adjuvant (CFA) and the subsequent two in Incomplete Freund's Adjuvant (IFA). Mice were bled 10 days after the final injection and the sera were tested by ELISA for reactivity to both 125P5C8-NusA (the immunogen) and 125P5C8-MBP (protein encoded by the gene 125P5C8 fused with MBP which is also a tag protein. It has no similarity with NusA). Thus reaction to the MBP fusion protein indicates a humoral reaction exclusively to the 125P5C8 portion of the immunogen. Specific titers for the individual mice are listed in the following chart.
















Mouse number
Titer in 125P5C8-MBP









1
3 × 10−4



2
2 × 10−4



3
1.5 × 10−4  



4
5 × 10−4










Fusions were performed using donor cells from mouse 1 and mouse 4, by the method of Kohler et. al, J. Immunol. 6, 511 (1976). When screened by ELISA on 125P5C8-MBP these yielded a total of 7 positive hybridoma clones; X12(1)4, X12(1)7, X12(1)8, X12(1)9, X12(5)6, X12(5)23 and X12(5)37. Immunocytochemistry was performed on cytosine preparations of 125P5C8 expressing 293T cells using supernatants from these hybridomas. This immunohistochemistry showed that three hybridomas X12(1)4, X12(1)9 and X12(5)37 secreted monoclonal antibodies which recognize 125P5C8 on the surface of cells.


The binding affinity of a 125P5C8 monoclonal antibody is determined using standard technologies. Affinity measurements quantify the strength of antibody to epitope binding and are used to help define which 125P5C8 monoclonal antibodies preferred for diagnostic or therapeutic use, as appreciated by one of skill in the art. The BIAcore system (Uppsala, Sweden) is one preferred method for determining binding affinity. The BIAcore system uses surface plasmon resonance (SPR, Welford K. 1991, Opt. Quant. Elect. 23:1; Morton and Myszka, 1998, Methods in Enzymology 295: 268) to monitor biomolecular interactions in real time. BIAcore analysis conveniently generates association rate constants, dissociation rate constants, equilibrium dissociation constants, and affinity constants.


Example 12
HLA Class I and Class II Binding Assays

HLA class I and class II binding assays using purified HLA molecules are performed in accordance with disclosed protocols (e.g., PCT publications WO 94/20127 and WO 94/03205; Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et al., J. Immunol. 154:247 (1995); Sette, et al., Mol. Immunol. 31:813 (1994)). Briefly, purified MHC molecules (5 to 500 nM) are incubated with various unlabeled peptide inhibitors and 1-10 nM 125I-radiolabeled probe peptides as described. Following incubation, MHC-peptide complexes are separated from free peptide by gel filtration and the fraction of peptide bound is determined. Typically, in preliminary experiments, each MHC preparation is titered in the presence of fixed amounts of radiolabeled peptides to determine the concentration of HLA molecules necessary to bind 10-20% of the total radioactivity. All subsequent inhibition and direct binding assays are performed using these HLA concentrations.


Since under these conditions [label]<[HLA] and IC50≧[HLA], the measured IC50 values are reasonable approximations of the true KD values. Peptide inhibitors are typically tested at concentrations ranging from 120 μg/ml to 1.2 ng/ml, and are tested in two to four completely independent experiments. To allow comparison of the data obtained in different experiments, a relative binding figure is calculated for each peptide by dividing the IC50 of a positive control for inhibition by the IC50 for each tested peptide (typically unlabeled versions of the radiolabeled probe peptide). For database purposes, and inter-experiment comparisons, relative binding values are compiled. These values can subsequently be converted back into IC50 nM values by dividing the IC50 nM of the positive controls for inhibition by the relative binding of the peptide of interest. This method of data compilation is accurate and consistent for comparing peptides that have been tested on different days, or with different lots of purified MHC.


Binding assays as outlined above may be used to analyze HLA supermotif and/or HLA motif-bearing peptides.


Example 13
Identification of HLA Supermotif- and Motif-Bearing CTL Candidate Epitopes

HLA vaccine compositions of the invention can include multiple epitopes. The multiple epitopes can comprise multiple HLA supermotifs or motifs to achieve broad population coverage. This example illustrates the identification and confirmation of supermotif- and motif-bearing epitopes for the inclusion in such a vaccine composition. Calculation of population coverage is performed using the strategy described below.


Computer Searches and Algorithms for Identification of Supermotif and/or Motif-Bearing Epitopes


The searches performed to identify the motif-bearing peptide sequences in Example 9 and Tables V-XVIII employ the protein sequence data from the gene product of 125P5C8 set forth in FIGS. 2 and 3.


Computer searches for epitopes bearing HLA Class I or Class II supermotifs or motifs are performed as follows. All translated 125P5C8 protein sequences are analyzed using a text string search software program to identify potential peptide sequences containing appropriate HLA binding motifs; such programs are readily produced in accordance with information in the art in view of known motif/supermotif disclosures. Furthermore, such calculations can be made mentally.


Identified A2-, A3-, and DR-supermotif sequences are scored using polynomial algorithms to predict their capacity to bind to specific HLA-Class I or Class II molecules. These polynomial algorithms account for the impact of different amino acids at different positions, and are essentially based on the premise that the overall affinity (or ΔG) of peptide-HLA molecule interactions can be approximated as a linear polynomial function of the type:





“ΔG”=a1i×a2i×a3i . . . ×ani


where aji is a coefficient which represents the effect of the presence of a given amino acid (j) at a given position (i) along the sequence of a peptide of n amino acids. The crucial assumption of this method is that the effects at each position are essentially independent of each other (i.e., independent binding of individual side-chains). When residue j occurs at position i in the peptide, it is assumed to contribute a constant amount ji to the free energy of binding of the peptide irrespective of the sequence of the rest of the peptide.


The method of derivation of specific algorithm coefficients has been described in Gulukota et al., J. Mol. Biol. 267:1258-126, 1997; (see also Sidney et al., Human Immunol. 45:79-93, 1996; and Southwood et al., J. Immunol. 160:3363-3373, 1998). Briefly, for all i positions, anchor and non-anchor alike, the geometric mean of the average relative binding (ARB) of all peptides carrying j is calculated relative to the remainder of the group, and used as the estimate of ji. For Class II peptides, if multiple alignments are possible, only the highest scoring alignment is utilized, following an iterative procedure. To calculate an algorithm score of a given peptide in a test set, the ARB values corresponding to the sequence of the peptide are multiplied. If this product exceeds a chosen threshold, the peptide is predicted to bind. Appropriate thresholds are chosen as a function of the degree of stringency of prediction desired.


Selection of HLA-A2 Supertype Cross-Reactive Peptides


Protein sequences from 125P5C8 are scanned utilizing motif identification software, to identify 8-, 9-10- and 11-mer sequences containing the HLA-A2-supermotif main anchor specificity. Typically, these sequences are then scored using the protocol described above and the peptides corresponding to the positive-scoring sequences are synthesized and tested for their capacity to bind purified HLA-A*0201 molecules in vitro (HLA-A*0201 is considered a prototype A2 supertype molecule).


These peptides are then tested for the capacity to bind to additional A2-supertype molecules (A*0202, A*0203, A*0206, and A*6802). Peptides that bind to at least three of the five A2-supertype alleles tested are typically deemed A2-supertype cross-reactive binders. Preferred peptides bind at an affinity equal to or less than 500 nM to three or more HLA-A2 supertype molecules.


Selection of HLA-A3 Supermotif-Bearing Epitopes


The 125P5C8 protein sequence(s) scanned above is also examined for the presence of peptides with the HLA-A3-supermotif primary anchors. Peptides corresponding to the HLA A3 supermotif-bearing sequences are then synthesized and tested for binding to HLA-A*0301 and HLA-A*1101 molecules, the molecules encoded by the two most prevalent A3-supertype alleles. The peptides that bind at least one of the two alleles with binding affinities of ≦500 nM, often ≦200 nM, are then tested for binding cross-reactivity to the other common A3-supertype alleles (e.g., A*3101, A*3301, and A*6801) to identify those that can bind at least three of the five HLA-A3-supertype molecules tested.


Selection of HLA-B7 Supermotif Bearing Epitopes

The 125P5C8 protein(s) scanned above is also analyzed for the presence of 8-, 9-10-, or 11-mer peptides with the HLA-B7-supermotif. Corresponding peptides are synthesized and tested for binding to HLA-B*0702, the molecule encoded by the most common B7-supertype allele (i.e., the prototype B7 supertype allele). Peptides binding B*0702 with IC50 of ≦500 nM are identified using standard methods. These peptides are then tested for binding to other common B7-supertype molecules (e.g., B*3501, B*5101, B*5301, and B*5401). Peptides capable of binding to three or more of the five B7-supertype alleles tested are thereby identified.


Selection of A1 and A24 Motif-Bearing Epitopes


To further increase population coverage, HLA-A1 and -A24 epitopes can also be incorporated into vaccine compositions. An analysis of the 125P5C8 protein can also be performed to identify HLA-A1- and A24-motif-containing sequences.


High affinity and/or cross-reactive binding epitopes that bear other motif and/or supermotifs are identified using analogous methodology.


Example 14
Confirmation of Immunogenicity

Cross-reactive candidate CTL A2-supermotif-bearing peptides that are identified as described herein are selected to confirm in vitro immunogenicity. Confirmation is performed using the following methodology:


Target Cell Lines for Cellular Screening:


The 0.221A2.1 cell line, produced by transferring the HLA-A2.1 gene into the HLA-A, -B, -C null mutant human B-lymphoblastoid cell line 721.221, is used as the peptide-loaded target to measure activity of HLA-A2.1-restricted CTL. This cell line is grown in RPMI-1640 medium supplemented with antibiotics, sodium pyruvate, nonessential amino acids and 10% (v/v) heat inactivated FCS. Cells that express an antigen of interest, or transfectants comprising the gene encoding the antigen of interest, can be used as target cells to confirm the ability of peptide-specific CTLs to recognize endogenous antigen.


Primary CTL Induction Cultures:


Generation of Dendritic Cells (DC): PBMCs are thawed in RPMI with 30 μg/ml DNAse, washed twice and resuspended in complete medium (RPMI-1640 plus 5% AB human serum, non-essential amino acids, sodium pyruvate, L-glutamine and penicillin/streptomycin). The monocytes are purified by plating 10×106 PBMC/well in a 6-well plate. After 2 hours at 37° C., the non-adherent cells are removed by gently shaking the plates and aspirating the supernatants. The wells are washed a total of three times with 3 ml RPMI to remove most of the non-adherent and loosely adherent cells. Three ml of complete medium containing 50 ng/ml of GM-CSF and 1,000 U/ml of IL-4 are then added to each well. TNFα is added to the DCs on day 6 at 75 ng/ml and the cells are used for CTL induction cultures on day 7.


Induction of CTL with DC and Peptide: CD8+ T-cells are isolated by positive selection with Dynal immunomagnetic beads (Dynabeads® M-450) and the Detacha-Bead® reagent. Typically about 200−250×106 PBMC are processed to obtain 24×106 CD8+ T-cells (enough for a 48-well plate culture). Briefly, the PBMCs are thawed in RPMI with 30 μg/ml DNAse, washed once with PBS containing 1% human AB serum and resuspended in PBS/1% AB serum at a concentration of 20×106 cells/ml. The magnetic beads are washed 3 times with PBS/AB serum, added to the cells (140 μl beads/20×106 cells) and incubated for 1 hour at 4° C. with continuous mixing. The beads and cells are washed 4× with PBS/AB serum to remove the nonadherent cells and resuspended at 100×106 cells/ml (based on the original cell number) in PBS/AB serum containing 100 μl/ml Detacha-Bead® reagent and 30 μg/ml DNAse. The mixture is incubated for 1 hour at room temperature with continuous mixing. The beads are washed again with PBS/AB/DNAse to collect the CD8+ T-cells. The DC are collected and centrifuged at 1300 rpm for 5-7 minutes, washed once with PBS with 1% BSA, counted and pulsed with 40 μg/ml of peptide at a cell concentration of 1−2×106/ml in the presence of 3 μg/ml β2-microglobulin for 4 hours at 20° C. The DC are then irradiated (4,200 rads), washed 1 time with medium and counted again.


Setting up induction cultures: 0.25 ml cytokine-generated DC (at 1×105 cells/ml) are co-cultured with 0.25 ml of CD8+ T-cells (at 2×106 cell/ml) in each well of a 48-well plate in the presence of 10 ng/ml of IL-7. Recombinant human IL-10 is added the next day at a final concentration of 10 ng/ml and Rhuman IL-2 is added 48 hours later at 10 IU/ml.


Restimulation of the induction cultures with peptide-pulsed adherent cells: Seven and fourteen days after the primary induction, the cells are restimulated with peptide-pulsed adherent cells. The PBMCs are thawed and washed twice with RPMI and DNAse. The cells are resuspended at 5×106 cells/ml and irradiated at ˜4200 rads. The PBMCs are plated at 2×106 in 0.5 ml complete medium per well and incubated for 2 hours at 37° C. The plates are washed twice with RPMI by tapping the plate gently to remove the nonadherent cells and the adherent cells pulsed with 10 μg/ml of peptide in the presence of 3 μg/ml β2 microglobulin in 0.25 ml RPMI/5% AB per well for 2 hours at 37° C. Peptide solution from each well is aspirated and the wells are washed once with RPMI. Most of the media is aspirated from the induction cultures (CD8+ cells) and brought to 0.5 ml with fresh media. The cells are then transferred to the wells containing the peptide-pulsed adherent cells. Twenty four hours later recombinant human IL-10 is added at a final concentration of 10 ng/ml and recombinant human IL2 is added the next day and again 2-3 days later at 501 U/ml (Tsai et al., Critical Reviews in Immunology 18(1-2):65-75, 1998). Seven days later, the cultures are assayed for CTL activity in a 51Cr release assay. In some experiments the cultures are assayed for peptide-specific recognition in the in situ IFNγ ELISA at the time of the second restimulation followed by assay of endogenous recognition 7 days later. After expansion, activity is measured in both assays for a side-by-side comparison.


Measurement of CTL Lytic Activity by 51Cr Release.


Seven days after the second restimulation, cytotoxicity is determined in a standard (5 hr) 51Cr release assay by assaying individual wells at a single E:T. Peptide-pulsed targets are prepared by incubating the cells with 10 μg/ml peptide overnight at 37° C.


Adherent target cells are removed from culture flasks with trypsin-EDTA. Target cells are labeled with 200 μCi of 51Cr sodium chromate (Dupont, Wilmington, Del.) for 1 hour at 37° C. Labeled target cells are resuspended at 106 per ml and diluted 1:10 with K562 cells at a concentration of 3.3×106/ml (an NK-sensitive erythroblastoma cell line used to reduce non-specific lysis). Target cells (100 μl) and effectors (100 μl) are plated in 96 well round-bottom plates and incubated for 5 hours at 37° C. At that time, 100 μl of supernatant are collected from each well and percent lysis is determined according to the formula:





[(cpm of the test sample-cpm of the spontaneous 51Cr release sample)/(cpm of the maximal 51Cr release sample-cpm of the spontaneous 51Cr release sample)]×100.


Maximum and spontaneous release are determined by incubating the labeled targets with 1% Triton X-100 and media alone, respectively. A positive culture is defined as one in which the specific lysis (sample-background) is 10% or higher in the case of individual wells and is 15% or more at the two highest E:T ratios when expanded cultures are assayed.


In Situ Measurement of Human IFNγ Production as an Indicator of Peptide-Specific and Endogenous Recognition


Immulon 2 plates are coated with mouse anti-human IFNγ monoclonal antibody (4 μg/ml 0.1M NaHCO3, pH8.2) overnight at 4° C. The plates are washed with Ca2+, Mg2+-free PBS/0.05% Tween 20 and blocked with PBS/10% FCS for two hours, after which the CTLs (100 μl/well) and targets (100 μl/well) are added to each well, leaving empty wells for the standards and blanks (which received media only). The target cells, either peptide-pulsed or endogenous targets, are used at a concentration of 1×106 cells/ml. The plates are incubated for 48 hours at 37° C. with 5% CO2.


Recombinant human IFN-gamma is added to the standard wells starting at 400 pg or 1200 pg/100 microliter/well and the plate incubated for two hours at 37° C. The plates are washed and 100 μl of biotinylated mouse anti-human IFN-gamma monoclonal antibody (2 microgram/ml in PBS/3% FCS/0.05% Tween 20) are added and incubated for 2 hours at room temperature. After washing again, 100 microliter HRP-streptavidin (1:4000) are added and the plates incubated for one hour at room temperature. The plates are then washed 6× with wash buffer, 100 microliter/well developing solution (TMB 1:1) are added, and the plates allowed to develop for 5-15 minutes. The reaction is stopped with 50 microliter/well 1M H3PO4 and read at OD450. A culture is considered positive if it measured at least 50 pg of IFN-gamma/well above background and is twice the background level of expression.


CTL Expansion.


Those cultures that demonstrate specific lytic activity against peptide-pulsed targets and/or tumor targets are expanded over a two week period with anti-CD3. Briefly, 5×104 CD8+ cells are added to a T25 flask containing the following: 1×106 irradiated (4,200 rad) PBMC (autologous or allogeneic) per mil, 2×105 irradiated (8,000 rad) EBV-transformed cells per mil, and OKT3 (anti-CD3) at 30 ng per mil in RPMI-1640 containing 10% (v/v) human AB serum, non-essential amino acids, sodium pyruvate, 25 μM 2-mercaptoethanol, L-glutamine and penicillin/streptomycin. Recombinant human IL2 is added 24 hours later at a final concentration of 200 IU/ml and every three days thereafter with fresh media at 50 IU/ml. The cells are split if the cell concentration exceeds 1×106/ml and the cultures are assayed between days 13 and 15 at E:T ratios of 30, 10, 3 and 1:1 in the 51Cr release assay or at 1×106/ml in the in situ IFNγ assay using the same targets as before the expansion.


Cultures are expanded in the absence of anti-CD3+ as follows. Those cultures that demonstrate specific lytic activity against peptide and endogenous targets are selected and 5×104 CD8+ cells are added to a T25 flask containing the following: 1×106 autologous PBMC per ml which have been peptide-pulsed with 10 μg/ml peptide for two hours at 37° C. and irradiated (4,200 rad); 2×105 irradiated (8,000 rad) EBV-transformed cells per ml RPMI-1640 containing 10% (v/v) human AB serum, non-essential AA, sodium pyruvate, 25 mM 2-ME, L-glutamine and gentamicin.


Immunogenicity of A2 supermotif-bearing peptides


A2-supermotif cross-reactive binding peptides are tested in the cellular assay for the ability to induce peptide-specific CTL in normal individuals. In this analysis, a peptide is typically considered to be an epitope if it induces peptide-specific CTLs in at least individuals, and preferably, also recognizes the endogenously expressed peptide.


Immunogenicity can also be confirmed using PBMCs isolated from patients bearing a tumor that expresses 125P5C8. Briefly, PBMCs are isolated from patients, re-stimulated with peptide-pulsed monocytes and assayed for the ability to recognize peptide-pulsed target cells as well as transfected cells endogenously expressing the antigen.


Evaluation of A*03/A11 Immunogenicity


HLA-A3 supermotif-bearing cross-reactive binding peptides are also evaluated for immunogenicity using methodology analogous for that used to evaluate the immunogenicity of the HLA-A2 supermotif peptides.


Evaluation of B7 Immunogenicity


Immunogenicity screening of the B7-supertype cross-reactive binding peptides identified as set forth herein are confirmed in a manner analogous to the confirmation of A2- and A3-supermotif-bearing peptides.


Peptides bearing other supermotifs/motifs, e.g., HLA-A1, HLA-A24 etc. are also confirmed using similar methodology


Example 15
Implementation of the Extended Supermotif to Improve the Binding Capacity of Native Epitopes by Creating Analogs

HLA motifs and supermotifs (comprising primary and/or secondary residues) are useful in the identification and preparation of highly cross-reactive native peptides, as demonstrated herein. Moreover, the definition of HLA motifs and supermotifs also allows one to engineer highly cross-reactive epitopes by identifying residues within a native peptide sequence which can be analoged to confer upon the peptide certain characteristics, e.g. greater cross-reactivity within the group of HLA molecules that comprise a supertype, and/or greater binding affinity for some or all of those HLA molecules. Examples of analoging peptides to exhibit modulated binding affinity are set forth in this example.


Analoging at Primary Anchor Residues


Peptide engineering strategies are implemented to further increase the cross-reactivity of the epitopes. For example, the main anchors of A2-supermotif-bearing peptides are altered, for example, to introduce a preferred L, I, V, or M at position 2, and I or V at the C-terminus.


To analyze the cross-reactivity of the analog peptides, each engineered analog is initially tested for binding to the prototype A2 supertype allele A*0201, then, if A*0201 binding capacity is maintained, for A2-supertype cross-reactivity.


Alternatively, a peptide is confirmed as binding one or all supertype members and then analoged to modulate binding affinity to any one (or more) of the supertype members to add population coverage.


The selection of analogs for immunogenicity in a cellular screening analysis is typically further restricted by the capacity of the parent wild type (WT) peptide to bind at least weakly, i.e., bind at an IC50 of 500 nM or less, to three of more A2 supertype alleles. The rationale for this requirement is that the WT peptides must be present endogenously in sufficient quantity to be biologically relevant. Analoged peptides have been shown to have increased immunogenicity and cross-reactivity by T cells specific for the parent epitope (see, e.g., Parkhurst et al., J. Immunol. 157:2539, 1996; and Pogue et al., Proc. Natl. Acad. Sci. USA 92:8166, 1995).


In the cellular screening of these peptide analogs, it is important to confirm that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, target cells that endogenously express the epitope.


Analoging of HLA-A3 and B7-Supermotif-Bearing Peptides


Analogs of HLA-A3 supermotif-bearing epitopes are generated using strategies similar to those employed in analoging HLA-A2 supermotif-bearing peptides. For example, peptides binding to ⅗ of the A3-supertype molecules are engineered at primary anchor residues to possess a preferred residue (V, S, M, or A) at position 2.


The analog peptides are then tested for the ability to bind A*03 and A*11 (prototype A3 supertype alleles). Those peptides that demonstrate ≦500 nM binding capacity are then confirmed as having A3-supertype cross-reactivity.


Similarly to the A2- and A3-motif bearing peptides, peptides binding 3 or more B7-supertype alleles can be improved, where possible, to achieve increased cross-reactive binding or greater binding affinity or binding half life. B7 supermotif-bearing peptides are, for example, engineered to possess a preferred residue (V, I, L, or F) at the C-terminal primary anchor position, as demonstrated by Sidney et al. (J. Immunol. 157:3480-3490, 1996).


Analoging at primary anchor residues of other motif and/or supermotif-bearing epitopes is performed in a like manner.


The analog peptides are then be confirmed for immunogenicity, typically in a cellular screening assay. Again, it is generally important to demonstrate that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, targets that endogenously express the epitope.


Analoging at Secondary Anchor Residues


Moreover, HLA supermotifs are of value in engineering highly cross-reactive peptides and/or peptides that bind HLA molecules with increased affinity by identifying particular residues at secondary anchor positions that are associated with such properties. For example, the binding capacity of a B7 supermotif-bearing peptide with an F residue at position 1 is analyzed. The peptide is then analoged to, for example, substitute L for F at position 1. The analoged peptide is evaluated for increased binding affinity, binding half life and/or increased cross-reactivity. Such a procedure identifies analoged peptides with enhanced properties.


Engineered analogs with sufficiently improved binding capacity or cross-reactivity can also be tested for immunogenicity in HLA-B7-transgenic mice, following for example, IFA immunization or lipopeptide immunization. Analoged peptides are additionally tested for the ability to stimulate a recall response using PBMC from patients with 125P5C8-expressing tumors.


Other Analoging Strategies


Another form of peptide analoging, unrelated to anchor positions, involves the substitution of a cysteine with α-amino butyric acid. Due to its chemical nature, cysteine has the propensity to form disulfide bridges and sufficiently alter the peptide structurally so as to reduce binding capacity. Substitution of α-amino butyric acid for cysteine not only alleviates this problem, but has been shown to improve binding and crossbinding capabilities in some instances (see, e.g., the review by Sette et al., In: Persistent Viral Infections, Eds. R. Ahmed and I. Chen, John Wiley & Sons, England, 1999).


Thus, by the use of single amino acid substitutions, the binding properties and/or cross-reactivity of peptide ligands for HLA supertype molecules can be modulated.


Example 16
Identification and Confirmation of 125P5C8-Derived Sequences with HLA-DR Binding Motifs

Peptide epitopes bearing an HLA class II supermotif or motif are identified and confirmed as outlined below using methodology similar to that described for HLA Class I peptides.


Selection of HLA-DR-Supermotif-Bearing Epitopes.


To identify 125P5C8-derived, HLA class II HTL epitopes, a 125P5C8 antigen is analyzed for the presence of sequences bearing an HLA-DR-motif or supermotif. Specifically, 15-mer sequences are selected comprising a DR-supermotif, comprising a 9-mer core, and three-residue N- and C-terminal flanking regions (15 amino acids total).


Protocols for predicting peptide binding to DR molecules have been developed (Southwood et al., J. Immunol. 160:3363-3373, 1998). These protocols, specific for individual DR molecules, allow the scoring, and ranking, of 9-mer core regions. Each protocol not only scores peptide sequences for the presence of DR-supermotif primary anchors (i.e., at position 1 and position 6) within a 9-mer core, but additionally evaluates sequences for the presence of secondary anchors. Using allele-specific selection tables (see, e.g., Southwood et al., ibid.), it has been found that these protocols efficiently select peptide sequences with a high probability of binding a particular DR molecule. Additionally, it has been found that performing these protocols in tandem, specifically those for DR1, DR4w4, and DR7, can efficiently select DR cross-reactive peptides.


The 125P5C8-derived peptides identified above are tested for their binding capacity for various common HLA-DR molecules. All peptides are initially tested for binding to the DR molecules in the primary panel: DR1, DR4w4, and DR7. Peptides binding at least two of these three DR molecules are then tested for binding to DR2w2 β1, DR2w2 β2, DR6w19, and DR9 molecules in secondary assays. Finally, peptides binding at least two of the four secondary panel DR molecules, and thus cumulatively at least four of seven different DR molecules, are screened for binding to DR4w15, DR5w11, and DR8w2 molecules in tertiary assays. Peptides binding at least seven of the ten DR molecules comprising the primary, secondary, and tertiary screening assays are considered cross-reactive DR binders. 125P5C8-derived peptides found to bind common HLA-DR alleles are of particular interest.


Selection of DR3 Motif Peptides


Because HLA-DR3 is an allele that is prevalent in Caucasian, Black, and Hispanic populations, DR3 binding capacity is a relevant criterion in the selection of HTL epitopes. Thus, peptides shown to be candidates may also be assayed for their DR3 binding capacity. However, in view of the binding specificity of the DR3 motif, peptides binding only to DR3 can also be considered as candidates for inclusion in a vaccine formulation.


To efficiently identify peptides that bind DR3, target 125P5C8 antigens are analyzed for sequences carrying one of the two DR3-specific binding motifs reported by Geluk et al. (J. Immunol. 152:5742-5748, 1994). The corresponding peptides are then synthesized and confirmed as having the ability to bind DR3 with an affinity of 1 μM or better, i.e., less than 1 μM. Peptides are found that meet this binding criterion and qualify as HLA class II high affinity binders.


DR3 binding epitopes identified in this manner are included in vaccine compositions with DR supermotif-bearing peptide epitopes.


Similarly to the case of HLA class I motif-bearing peptides, the class II motif-bearing peptides are analoged to improve affinity or cross-reactivity. For example, aspartic acid at position 4 of the 9-mer core sequence is an optimal residue for DR3 binding, and substitution for that residue often improves DR 3 binding.


Example 17
Immunogenicity of 125P5C8-Derived HTL Epitopes

This example determines immunogenic DR supermotif- and DR3 motif-bearing epitopes among those identified using the methodology set forth herein.


Immunogenicity of HTL epitopes are confirmed in a manner analogous to the determination of immunogenicity of CTL epitopes, by assessing the ability to stimulate HTL responses and/or by using appropriate transgenic mouse models. Immunogenicity is determined by screening for: 1.) in vitro primary induction using normal PBMC or 2.) recall responses from patients who have 125P5C8-expressing tumors.


Example 18
Calculation of Phenotypic Frequencies of HLA-Supertypes in Various Ethnic Backgrounds to Determine Breadth of Population Coverage

This example illustrates the assessment of the breadth of population coverage of a vaccine composition comprised of multiple epitopes comprising multiple supermotifs and/or motifs.


In order to analyze population coverage, gene frequencies of HLA alleles are determined. Gene frequencies for each HLA allele are calculated from antigen or allele frequencies utilizing the binomial distribution formulae gf=1-(SQRT(1-af)) (see, e.g., Sidney et al., Human Immunol. 45:79-93, 1996). To obtain overall phenotypic frequencies, cumulative gene frequencies are calculated, and the cumulative antigen frequencies derived by the use of the inverse formula [af=1−(1−Cgf)2].


Where frequency data is not available at the level of DNA typing, correspondence to the serologically defined antigen frequencies is assumed. To obtain total potential supertype population coverage no linkage disequilibrium is assumed, and only alleles confirmed to belong to each of the supertypes are included (minimal estimates). Estimates of total potential coverage achieved by inter-loci combinations are made by adding to the A coverage the proportion of the non-A covered population that could be expected to be covered by the B alleles considered (e.g., total=A+B*(1−A)). Confirmed members of the A3-like supertype are A3, A11, A31, A*3301, and A*6801. Although the A3-like supertype may also include A34, A66, and A*7401, these alleles were not included in overall frequency calculations. Likewise, confirmed members of the A2-like supertype family are A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*0207, A*6802, and A*6901. Finally, the B7-like supertype-confirmed alleles are: B7, B*3501-03, B51, B*5301, B*5401, B*5501-2, B*5601, B*6701, and B*7801 (potentially also B*1401, B*3504-06, B*4201, and B*5602).


Population coverage achieved by combining the A2-, A3- and B7-supertypes is approximately 86% in five major ethnic groups. Coverage may be extended by including peptides bearing the A1 and A24 motifs. On average, A1 is present in 12% and A24 in 29% of the population across five different major ethnic groups (Caucasian, North American Black, Chinese, Japanese, and Hispanic). Together, these alleles are represented with an average frequency of 39% in these same ethnic populations. The total coverage across the major ethnicities when A1 and A24 are combined with the coverage of the A2-, A3- and B7-supertype alleles is >95%. An analogous approach can be used to estimate population coverage achieved with combinations of class II motif-bearing epitopes.


Immunogenicity studies in humans (e.g., Bertoni et al., J. Clin. Invest. 100:503, 1997; Doolan et al., Immunity 7:97, 1997; and Threlkeld et al., J. Immunol. 159:1648, 1997) have shown that highly cross-reactive binding peptides are almost always recognized as epitopes. The use of highly cross-reactive binding peptides is an important selection criterion in identifying candidate epitopes for inclusion in a vaccine that is immunogenic in a diverse population.


With a sufficient number of epitopes (as disclosed herein and from the art), an average population coverage is predicted to be greater than 95% in each of five major ethnic populations. The game theory Monte Carlo simulation analysis, which is known in the art (see e.g., Osborne, M. J. and Rubinstein, A. “A course in game theory” MIT Press, 1994), can be used to estimate what percentage of the individuals in a population comprised of the Caucasian, North American Black, Japanese, Chinese, and Hispanic ethnic groups would recognize the vaccine epitopes described herein. A preferred percentage is 90%. A more preferred percentage is 95%.


Example 19
CTL Recognition of Endogenously Processed Antigens After Priming

This example confirms that CTL induced by native or analoged peptide epitopes identified and selected as described herein recognize endogenously synthesized, i.e., native antigens.


Effector cells isolated from transgenic mice that are immunized with peptide epitopes, for example HLA-A2 supermotif-bearing epitopes, are re-stimulated in vitro using peptide-coated stimulator cells. Six days later, effector cells are assayed for cytotoxicity and the cell lines that contain peptide-specific cytotoxic activity are further re-stimulated. An additional six days later, these cell lines are tested for cytotoxic activity on 51Cr labeled Jurkat-A2.1/Kb target cells in the absence or presence of peptide, and also tested on 51Cr labeled target cells bearing the endogenously synthesized antigen, i.e. cells that are stably transfected with 125P5C8 expression vectors.


The results demonstrate that CTL lines obtained from animals primed with peptide epitope recognize endogenously synthesized 125P5C8 antigen. The choice of transgenic mouse model to be used for such an analysis depends upon the epitope(s) that are being evaluated. In addition to HLA-A*0201/Kb transgenic mice, several other transgenic mouse models including mice with human A11, which may also be used to evaluate A3 epitopes, and B7 alleles have been characterized and others (e.g., transgenic mice for HLA-A1 and A24) are being developed. HLA-DR1 and HLA-DR3 mouse models have also been developed, which may be used to evaluate HTL epitopes.


Example 20
Activity of CTL-HTL Conjugated Epitopes in Transgenic Mice

This example illustrates the induction of CTLs and HTLs in transgenic mice, by use of a 125P5C8-derived CTL and HTL peptide vaccine compositions. The vaccine composition used herein comprise peptides to be administered to a patient with a 125P5C8-expressing tumor. The peptide composition can comprise multiple CTL and/or HTL epitopes. The epitopes are identified using methodology as described herein. This example also illustrates that enhanced immunogenicity can be achieved by inclusion of one or more HTL epitopes in a CTL vaccine composition; such a peptide composition can comprise an HTL epitope conjugated to a CTL epitope. The CTL epitope can be one that binds to multiple HLA family members at an affinity of 500 nM or less, or analogs of that epitope. The peptides may be lipidated, if desired.


Immunization procedures: Immunization of transgenic mice is performed as described (Alexander et al., J. Immunol. 159:4753-4761, 1997). For example, A2/Kb mice, which are transgenic for the human HLA A2.1 allele and are used to confirm the immunogenicity of HLA-A*0201 motif- or HLA-A2 supermotif-bearing epitopes, and are primed subcutaneously (base of the tail) with a 0.1 ml of peptide in Incomplete Freund's Adjuvant, or if the peptide composition is a lipidated CTL/HTL conjugate, in DMSO/saline, or if the peptide composition is a polypeptide, in PBS or Incomplete Freund's Adjuvant. Seven days after priming, splenocytes obtained from these animals are restimulated with syngenic irradiated LPS-activated lymphoblasts coated with peptide.


Cell lines: Target cells for peptide-specific cytotoxicity assays are Jurkat cells transfected with the HLA-A2.1/Kb chimeric gene (e.g., Vitiello et al., J. Exp. Med. 173:1007, 1991)


In vitro CTL activation: One week after priming, spleen cells (30×106 cells/flask) are co-cultured at 37° C. with syngeneic, irradiated (3000 rads), peptide coated lymphoblasts (10×106 cells/flask) in 10 ml of culture medium/T25 flask. After six days, effector cells are harvested and assayed for cytotoxic activity.


Assay for cytotoxic activity: Target cells (1.0 to 1.5×106) are incubated at 37° C. in the presence of 200 μl of 51Cr. After 60 minutes, cells are washed three times and resuspended in R10 medium. Peptide is added where required at a concentration of 1 μg/ml. For the assay, 104 51Cr-labeled target cells are added to different concentrations of effector cells (final volume of 200 μl) in U-bottom 96-well plates. After a six hour incubation period at 37° C., a 0.1 ml aliquot of supernatant is removed from each well and radioactivity is determined in a Micromedic automatic gamma counter. The percent specific lysis is determined by the formula: percent specific release=100×(experimental release−spontaneous release)/(maximum release−spontaneous release). To facilitate comparison between separate CTL assays run under the same conditions, % 51Cr release data is expressed as lytic units/106 cells. One lytic unit is arbitrarily defined as the number of effector cells required to achieve 30% lysis of 10,000 target cells in a six hour 51Cr release assay. To obtain specific lytic units/106, the lytic units/106 obtained in the absence of peptide is subtracted from the lytic units/106 obtained in the presence of peptide. For example, if 30% 51Cr release is obtained at the effector (E): target (T) ratio of 50:1 (i.e., 5×105 effector cells for 10,000 targets) in the absence of peptide and 5:1 (i.e., 5×104 effector cells for 10,000 targets) in the presence of peptide, the specific lytic units would be: [( 1/50,000)-( 1/500,000)]×106=18 LU.


The results are analyzed to assess the magnitude of the CTL responses of animals injected with the immunogenic CTL/HTL conjugate vaccine preparation and are compared to the magnitude of the CTL response achieved using, for example, CTL epitopes as outlined above in Example 14. Analyses similar to this may be performed to confirm the immunogenicity of peptide conjugates containing multiple CTL epitopes and/or multiple HTL epitopes. In accordance with these procedures, it is found that a CTL response is induced, and concomitantly that an HTL response is induced upon administration of such compositions.


Example 21
Selection of CTL and HTL Epitopes for Inclusion in a 125P5C8-Specific Vaccine

This example illustrates a procedure for selecting peptide epitopes for vaccine compositions of the invention. The peptides in the composition can be in the form of a nucleic acid sequence, either single or one or more sequences (i.e., minigene) that encodes peptide(s), or can be single and/or polyepitopic peptides.


The following principles are utilized when selecting a plurality of epitopes for inclusion in a vaccine composition. Each of the following principles is balanced in order to make the selection.


Epitopes are selected which, upon administration, mimic immune responses that are correlated with 125P5C8 clearance. The number of epitopes used depends on observations of patients who spontaneously clear 125P5C8. For example, if it has been observed that patients who spontaneously clear 125P5C8 generate an immune response to at least three (3) from 125P5C8 antigen, then three or four (3-4) epitopes should be included for HLA class I. A similar rationale is used to determine HLA class II epitopes.


Epitopes are often selected that have a binding affinity of an IC50 of 500 nM or less for an HLA class I molecule, or for class II, an IC50 of 1000 nM or less; or HLA Class I peptides with high binding scores from the BIMAS web site, at URL bimas.dcrt.nih.gov/.


In order to achieve broad coverage of the vaccine through out a diverse population, sufficient supermotif bearing peptides, or a sufficient array of allele-specific motif bearing peptides, are selected to give broad population coverage. In one embodiment, epitopes are selected to provide at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess breadth, or redundancy, of population coverage.


When creating polyepitopic compositions, or a minigene that encodes same, it is typically desirable to generate the smallest peptide possible that encompasses the epitopes of interest. The principles employed are similar, if not the same, as those employed when selecting a peptide comprising nested epitopes. For example, a protein sequence for the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, i.e., it has a high concentration of epitopes. Epitopes may be nested or overlapping (i.e., frame shifted relative to one another). For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a 10 amino acid peptide. Each epitope can be exposed and bound by an HLA molecule upon administration of such a peptide. A multi-epitopic, peptide can be generated synthetically, recombinantly, or via cleavage from the native source. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes. This embodiment provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally such an embodiment provides for the possibility of motif-bearing epitopes for an HLA makeup that is presently unknown. Furthermore, this embodiment (absent the creating of any analogs) directs the immune response to multiple peptide sequences that are actually present in 125P5C8, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing nucleic acid vaccine compositions. Related to this embodiment, computer programs can be derived in accordance with principles in the art, which identify in a target sequence, the greatest number of epitopes per sequence length.


A vaccine composition comprised of selected peptides, when administered, is safe, efficacious, and elicits an immune response similar in magnitude to an immune response that controls or clears cells that bear or overexpress 125P5C8.


Example 22
Construction of “Minigene” Multi-Epitope DNA Plasmids

This example discusses the construction of a minigene expression plasmid. Minigene plasmids may, of course, contain various configurations of B cell, CTL and/or HTL epitopes or epitope analogs as described herein.


A minigene expression plasmid typically includes multiple CTL and HTL peptide epitopes. In the present example, HLA-A2, -A3, -B7 supermotif-bearing peptide epitopes and HLA-A1 and -A24 motif-bearing peptide epitopes are used in conjunction with DR supermotif-bearing epitopes and/or DR3 epitopes. HLA class I supermotif or motif-bearing peptide epitopes derived 125P5C8, are selected such that multiple supermotifs/motifs are represented to ensure broad population coverage. Similarly, HLA class II epitopes are selected from 125P5C8 to provide broad population coverage, i.e. both HLA DR-1-4-7 supermotif-bearing epitopes and HLA DR-3 motif-bearing epitopes are selected for inclusion in the minigene construct. The selected CTL and HTL epitopes are then incorporated into a minigene for expression in an expression vector.


Such a construct may additionally include sequences that direct the HTL epitopes to the endoplasmic reticulum. For example, the Ii protein may be fused to one or more HTL epitopes as described in the art, wherein the CLIP sequence of the Ii protein is removed and replaced with an HLA class II epitope sequence so that HLA class II epitope is directed to the endoplasmic reticulum, where the epitope binds to an HLA class II molecules.


This example illustrates the methods to be used for construction of a minigene-bearing expression plasmid. Other expression vectors that may be used for minigene compositions are available and known to those of skill in the art.


The minigene DNA plasmid of this example contains a consensus Kozak sequence and a consensus murine kappa Ig-light chain signal sequence followed by CTL and/or HTL epitopes selected in accordance with principles disclosed herein. The sequence encodes an open reading frame fused to the Myc and His antibody epitope tag coded for by the pcDNA 3.1 Myc-His vector.


Overlapping oligonucleotides that can, for example, average about 70 nucleotides in length with 15 nucleotide overlaps, are synthesized and HPLC-purified. The oligonucleotides encode the selected peptide epitopes as well as appropriate linker nucleotides, Kozak sequence, and signal sequence. The final multiepitope minigene is assembled by extending the overlapping oligonucleotides in three sets of reactions using PCR. A Perkin/Elmer 9600 PCR machine is used and a total of 30 cycles are performed using the following conditions: 95° C. for 15 sec, annealing temperature (5° below the lowest calculated Tm of each primer pair) for 30 sec, and 72° C. for 1 min.


For example, a minigene is prepared as follows. For a first PCR reaction, 5 μg of each of two oligonucleotides are annealed and extended: In an example using eight oligonucleotides, i.e., four pairs of primers, oligonucleotides 1+2, 3+4, 5+6, and 7+8 are combined in 1001 reactions containing Pfu polymerase buffer (1×=10 mM KCL, 10 mM (NH4)2SO4, 20 mM Tris-chloride, pH 8.75, 2 mM MgSO4, 0.1% Triton X-100, 100 μg/ml BSA), 0.25 mM each dNTP, and 2.5 U of Pfu polymerase. The full-length dimer products are gel-purified, and two reactions containing the product of 1+2 and 3+4, and the product of 5+6 and 7+8 are mixed, annealed, and extended for 10 cycles. Half of the two reactions are then mixed, and 5 cycles of annealing and extension carried out before flanking primers are added to amplify the full length product. The full-length product is gel-purified and cloned into pCR-blunt (Invitrogen) and individual clones are screened by sequencing.


Example 23
The Plasmid Construct and the Degree to which it Induces Immunogenicity

The degree to which a plasmid construct, for example a plasmid constructed in accordance with the previous Example, is able to induce immunogenicity is confirmed in vitro by determining epitope presentation by APC following transduction or transfection of the APC with an epitope-expressing nucleic acid construct. Such a study determines “antigenicity” and allows the use of human APC. The assay determines the ability of the epitope to be presented by the APC in a context that is recognized by a T cell by quantifying the density of epitope-HLA class I complexes on the cell surface. Quantitation can be performed by directly measuring the amount of peptide eluted from the APC (see, e.g., Sijts et al., J. Immunol. 156:683-692, 1996; Demotz et al., Nature 342:682-684, 1989); or the number of peptide-HLA class I complexes can be estimated by measuring the amount of lysis or lymphokine release induced by diseased or transfected target cells, and then determining the concentration of peptide necessary to obtain equivalent levels of lysis or lymphokine release (see, e.g., Kageyama et al., J. Immunol. 154:567-576, 1995).


Alternatively, immunogenicity is confirmed through in vivo injections into mice and subsequent in vitro assessment of CTL and HTL activity, which are analyzed using cytotoxicity and proliferation assays, respectively, as detailed e.g., in Alexander et al., Immunity 1:751-761, 1994.


For example, to confirm the capacity of a DNA minigene construct containing at least one HLA-A2 supermotif peptide to induce CTLs in vivo, HLA-A2.1/Kb transgenic mice, for example, are immunized intramuscularly with 100 μg of naked cDNA. As a means of comparing the level of CTLs induced by cDNA immunization, a control group of animals is also immunized with an actual peptide composition that comprises multiple epitopes synthesized as a single polypeptide as they would be encoded by the minigene.


Splenocytes from immunized animals are stimulated twice with each of the respective compositions (peptide epitopes encoded in the minigene or the polyepitopic peptide), then assayed for peptide-specific cytotoxic activity in a 51Cr release assay. The results indicate the magnitude of the CTL response directed against the A2-restricted epitope, thus indicating the in vivo immunogenicity of the minigene vaccine and polyepitopic vaccine.


It is, therefore, found that the minigene elicits immune responses directed toward the HLA-A2 supermotif peptide epitopes as does the polyepitopic peptide vaccine. A similar analysis is also performed using other HLA-A3 and HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 and HLA-B7 motif or supermotif epitopes, whereby it is also found that the minigene elicits appropriate immune responses directed toward the provided epitopes.


To confirm the capacity of a class II epitope-encoding minigene to induce HTLs in vivo, DR transgenic mice, or for those epitopes that cross react with the appropriate mouse MHC molecule, I-Ab-restricted mice, for example, are immunized intramuscularly with 100 μg of plasmid DNA. As a means of comparing the level of HTLs induced by DNA immunization, a group of control animals is also immunized with an actual peptide composition emulsified in complete Freund's adjuvant. CD4+ T cells, i.e. HTLs, are purified from splenocytes of immunized animals and stimulated with each of the respective compositions (peptides encoded in the minigene). The HTL response is measured using a 3H-thymidine incorporation proliferation assay, (see, e.g., Alexander et al. Immunity 1:751-761, 1994). The results indicate the magnitude of the HTL response, thus demonstrating the in vivo immunogenicity of the minigene.


DNA minigenes, constructed as described in the previous Example, can also be confirmed as a vaccine in combination with a boosting agent using a prime boost protocol. The boosting agent can consist of recombinant protein (e.g., Barnett et al., Aids Res. and Human Retroviruses 14, Supplement 3:S299-S309, 1998) or recombinant vaccinia, for example, expressing a minigene or DNA encoding the complete protein of interest (see, e.g., Hanke et al., Vaccine 16:439-445, 1998; Sedegah et al., Proc. Natl. Acad. Sci USA 95:7648-53, 1998; Hanke and McMichael, Immunol. Letters 66:177-181, 1999; and Robinson et al., Nature Med. 5:526-34, 1999).


For example, the efficacy of the DNA minigene used in a prime boost protocol is initially evaluated in transgenic mice. In this example, A2.1/Kb transgenic mice are immunized IM with 100 μg of a DNA minigene encoding the immunogenic peptides including at least one HLA-A2 supermotif-bearing peptide. After an incubation period (ranging from 3-9 weeks), the mice are boosted IP with 107 pfu/mouse of a recombinant vaccinia virus expressing the same sequence encoded by the DNA minigene. Control mice are immunized with 100 μg of DNA or recombinant vaccinia without the minigene sequence, or with DNA encoding the minigene, but without the vaccinia boost. After an additional incubation period of two weeks, splenocytes from the mice are immediately assayed for peptide-specific activity in an ELISPOT assay. Additionally, splenocytes are stimulated in vitro with the A2-restricted peptide epitopes encoded in the minigene and recombinant vaccinia, then assayed for peptide-specific activity in an alpha, beta and/or gamma IFN ELISA.


It is found that the minigene utilized in a prime-boost protocol elicits greater immune responses toward the HLA-A2 supermotif peptides than with DNA alone. Such an analysis can also be performed using HLA-A11 or HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 or HLA-B7 motif or supermotif epitopes. The use of prime boost protocols in humans is described below in Example 31.


Example 24
Peptide Compositions for Prophylactic Uses

Vaccine compositions of the present invention can be used to prevent 125P5C8 expression in persons who are at risk for tumors that bear this antigen. For example, a polyepitopic peptide epitope composition (or a nucleic acid comprising the same) containing multiple CTL and HTL epitopes such as those selected in the above Examples, which are also selected to target greater than 80% of the population, is administered to individuals at risk for a 125P5C8-associated tumor.


For example, a peptide-based composition is provided as a single polypeptide that encompasses multiple epitopes. The vaccine is typically administered in a physiological solution that comprises an adjuvant, such as Incomplete Freunds Adjuvant. The dose of peptide for the initial immunization is from about 1 to about 50,000 μg, generally 100-5,000 μg, for a 70 kg patient. The initial administration of vaccine is followed by booster dosages at 4 weeks followed by evaluation of the magnitude of the immune response in the patient, by techniques that determine the presence of epitope-specific CTL populations in a PBMC sample. Additional booster doses are administered as required. The composition is found to be both safe and efficacious as a prophylaxis against 125P5C8-associated disease.


Alternatively, a composition typically comprising transfecting agents is used for the administration of a nucleic acid-based vaccine in accordance with methodologies known in the art and disclosed herein.


Example 25
Polyepitopic Vaccine Compositions Derived from Native 125P5C8 Sequences

A native 125P5C8 polyprotein sequence is analyzed, preferably using computer algorithms defined for each class I and/or class II supermotif or motif, to identify “relatively short” regions of the polyprotein that comprise multiple epitopes. The “relatively short” regions are preferably less in length than an entire native antigen. This relatively short sequence that contains multiple distinct or overlapping, “nested” epitopes is selected; it can be used to generate a minigene construct. The construct is engineered to express the peptide, which corresponds to the native protein sequence. The “relatively short” peptide is generally less than 250 amino acids in length, often less than 100 amino acids in length, preferably less than 75 amino acids in length, and more preferably less than 50 amino acids in length. The protein sequence of the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, i.e., it has a high concentration of epitopes. As noted herein, epitope motifs may be nested or overlapping (i.e., frame shifted relative to one another). For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a 10 amino acid peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes.


The vaccine composition will include, for example, multiple CTL epitopes from 125P5C8 antigen and at least one HTL epitope. This polyepitopic native sequence is administered either as a peptide or as a nucleic acid sequence which encodes the peptide. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide.


The embodiment of this example provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally such an embodiment provides for the possibility of motif-bearing epitopes for an HLA makeup that is presently unknown. Furthermore, this embodiment (excluding an analoged embodiment) directs the immune response to multiple peptide sequences that are actually present in native 125P5C8, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing peptide or nucleic acid vaccine compositions.


Related to this embodiment, computer programs are available in the art which can be used to identify in a target sequence, the greatest number of epitopes per sequence length.


Example 26
Polyepitopic Vaccine Compositions from Multiple Antigens

The 125P5C8 peptide epitopes of the present invention are used in conjunction with epitopes from other target tumor-associated antigens, to create a vaccine composition that is useful for the prevention or treatment of cancer that expresses 125P5C8 and such other antigens. For example, a vaccine composition can be provided as a single polypeptide that incorporates multiple epitopes from 125P5C8 as well as tumor-associated antigens that are often expressed with a target cancer associated with 125P5C8 expression, or can be administered as a composition comprising a cocktail of one or more discrete epitopes. Alternatively, the vaccine can be administered as a minigene construct or as dendritic cells which have been loaded with the peptide epitopes in vitro.


Example 27
Use of Peptides to Evaluate an Immune Response

Peptides of the invention may be used to analyze an immune response for the presence of specific antibodies, CTL or HTL directed to 125P5C8. Such an analysis can be performed in a manner described by Ogg et al., Science 279:2103-2106, 1998. In this Example, peptides in accordance with the invention are used as a reagent for diagnostic or prognostic purposes, not as an immunogen.


In this example highly sensitive human leukocyte antigen tetrameric complexes (“tetramers”) are used for a cross-sectional analysis of, for example, 125P5C8 HLA-A*0201-specific CTL frequencies from HLA A*0201-positive individuals at different stages of disease or following immunization comprising a 125P5C8 peptide containing an A*0201 motif. Tetrameric complexes are synthesized as described (Musey et al., N. Engl. J. Med. 337:1267, 1997). Briefly, purified HLA heavy chain (A*0201 in this example) and β2-microglobulin are synthesized by means of a prokaryotic expression system. The heavy chain is modified by deletion of the transmembrane-cytosolic tail and COOH-terminal addition of a sequence containing a BirA enzymatic biotinylation site. The heavy chain, β2-microglobulin, and peptide are refolded by dilution. The 45-kD refolded product is isolated by fast protein liquid chromatography and then biotinylated by BirA in the presence of biotin (Sigma, St. Louis, Mo.), adenosine 5′ triphosphate and magnesium. Streptavidin-phycoerythrin conjugate is added in a 1:4 molar ratio, and the tetrameric product is concentrated to 1 mg/ml. The resulting product is referred to as tetramer-phycoerythrin.


For the analysis of patient blood samples, approximately one million PBMCs are centrifuged at 300 g for 5 minutes and resuspended in 501 of cold phosphate-buffered saline. Tri-color analysis is performed with the tetramer-phycoerythrin, along with anti-CD8-Tricolor, and anti-CD38. The PBMCs are incubated with tetramer and antibodies on ice for 30 to 60 min and then washed twice before formaldehyde fixation. Gates are applied to contain >99.98% of control samples. Controls for the tetramers include both A*0201-negative individuals and A*0201-positive non-diseased donors. The percentage of cells stained with the tetramer is then determined by flow cytometry. The results indicate the number of cells in the PBMC sample that contain epitope-restricted CTLs, thereby readily indicating the extent of immune response to the 125P5C8 epitope, and thus the status of exposure to 125P5C8, or exposure to a vaccine that elicits a protective or therapeutic response.


Example 28
Use of Peptide Epitopes to Evaluate Recall Responses

The peptide epitopes of the invention are used as reagents to evaluate T cell responses, such as acute or recall responses, in patients. Such an analysis may be performed on patients who have recovered from 125P5C8-associated disease or who have been vaccinated with a 125P5C8 vaccine.


For example, the class I restricted CTL response of persons who have been vaccinated may be analyzed. The vaccine may be any 125P5C8 vaccine. PBMC are collected from vaccinated individuals and HLA typed. Appropriate peptide epitopes of the invention that, optimally, bear supermotifs to provide cross-reactivity with multiple HLA supertype family members, are then used for analysis of samples derived from individuals who bear that HLA type.


PBMC from vaccinated individuals are separated on Ficoll-Histopaque density gradients (Sigma Chemical Co., St. Louis, Mo.), washed three times in HBSS (GIBCO Laboratories), resuspended in RPMI-1640 (GIBCO Laboratories) supplemented with L-glutamine (2 mM), penicillin (50 U/ml), streptomycin (50 μg/ml), and Hepes (10 mM) containing 10% heat-inactivated human AB serum (complete RPMI) and plated using microculture formats. A synthetic peptide comprising an epitope of the invention is added at 10 μg/ml to each well and HBV core 128-140 epitope is added at 1 μg/ml to each well as a source of T cell help during the first week of stimulation.


In the microculture format, 4×105 PBMC are stimulated with peptide in 8 replicate cultures in 96-well round bottom plate in 100 μl/well of complete RPMI. On days 3 and 10, 100 μl of complete RPMI and 20 U/ml final concentration of rIL-2 are added to each well. On day 7 the cultures are transferred into a 96-well flat-bottom plate and restimulated with peptide, rIL-2 and 105 irradiated (3,000 rad) autologous feeder cells. The cultures are tested for cytotoxic activity on day 14. A positive CTL response requires two or more of the eight replicate cultures to display greater than 10% specific 51Cr release, based on comparison with non-diseased control subjects as previously described (Rehermann, et al., Nature Med. 2:1104, 1108, 1996; Rehermann et al., J. Clin. Invest. 97:1655-1665, 1996; and Rehermann et al. J. Clin. Invest. 98:1432-1440, 1996).


Target cell lines are autologous and allogeneic EBV-transformed B-LCL that are either purchased from the American Society for Histocompatibility and Immunogenetics (ASHI, Boston, Mass.) or established from the pool of patients as described (Guilhot, et al. J. Virol. 66:2670-2678, 1992).


Cytotoxicity assays are performed in the following manner. Target cells consist of either allogeneic HLA-matched or autologous EBV-transformed B lymphoblastoid cell line that are incubated overnight with the synthetic peptide epitope of the invention at 10 μM, and labeled with 100 μCi of 51Cr (Amersham Corp., Arlington Heights, Ill.) for 1 hour after which they are washed four times with HBSS.


Cytolytic activity is determined in a standard 4-h, split well 51Cr release assay using U-bottomed 96 well plates containing 3,000 targets/well. Stimulated PBMC are tested at effector/target (E/T) ratios of 20-50:1 on day 14. Percent cytotoxicity is determined from the formula: 100×[(experimental release-spontaneous release)/maximum release-spontaneous release)]. Maximum release is determined by lysis of targets by detergent (2% Triton X-100; Sigma Chemical Co., St. Louis, Mo.). Spontaneous release is <25% of maximum release for all experiments.


The results of such an analysis indicate the extent to which HLA-restricted CTL populations have been stimulated by previous exposure to 125P5C8 or a 125P5C8 vaccine.


Similarly, Class II restricted HTL responses may also be analyzed. Purified PBMC are cultured in a 96-well flat bottom plate at a density of 1.5×105 cells/well and are stimulated with 10 μg/ml synthetic peptide of the invention, whole 125P5C8 antigen, or PHA. Cells are routinely plated in replicates of 4-6 wells for each condition. After seven days of culture, the medium is removed and replaced with fresh medium containing 10 U/ml IL-2. Two days later, 1 μCi 3H-thymidine is added to each well and incubation is continued for an additional 18 hours. Cellular DNA is then harvested on glass fiber mats and analyzed for 3H-thymidine incorporation. Antigen-specific T cell proliferation is calculated as the ratio of 3H-thymidine incorporation in the presence of antigen divided by the 3H-thymidine incorporation in the absence of antigen.


Example 29
Induction of Specific CTL Response in Humans

A human clinical trial for an immunogenic composition comprising CTL and HTL epitopes of the invention is set up as an IND Phase I, dose escalation study and carried out as a randomized, double-blind, placebo-controlled trial. Such a trial is designed, for example, as follows:


A total of about 27 individuals are enrolled and divided into 3 groups:


Group I: 3 subjects are injected with placebo and 6 subjects are injected with 5 μg of peptide composition;


Group II: 3 subjects are injected with placebo and 6 subjects are injected with 50 μg peptide composition;


Group III: 3 subjects are injected with placebo and 6 subjects are injected with 500 μg of peptide composition.


After 4 weeks following the first injection, all subjects receive a booster inoculation at the same dosage.


The endpoints measured in this study relate to the safety and tolerability of the peptide composition as well as its immunogenicity. Cellular immune responses to the peptide composition are an index of the intrinsic activity of this the peptide composition, and can therefore be viewed as a measure of biological efficacy. The following summarize the clinical and laboratory data that relate to safety and efficacy endpoints.


Safety: The incidence of adverse events is monitored in the placebo and drug treatment group and assessed in terms of degree and reversibility.


Evaluation of Vaccine Efficacy: For evaluation of vaccine efficacy, subjects are bled before and after injection. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.


The vaccine is found to be both safe and efficacious.


Example 30
Phase II Trials In Patients Expressing 125P5C8

Phase II trials are performed to study the effect of administering the CTL-HTL peptide compositions to patients having cancer that expresses 125P5C8. The main objectives of the trial are to determine an effective dose and regimen for inducing CTLs in cancer patients that express 125P5C8, to establish the safety of inducing a CTL and HTL response in these patients, and to see to what extent activation of CTLs improves the clinical picture of these patients, as manifested, e.g., by the reduction and/or shrinking of lesions. Such a study is designed, for example, as follows:


The studies are performed in multiple centers. The trial design is an open-label, uncontrolled, dose escalation protocol wherein the peptide composition is administered as a single dose followed six weeks later by a single booster shot of the same dose. The dosages are 50,500 and 5,000 micrograms per injection. Drug-associated adverse effects (severity and reversibility) are recorded.


There are three patient groupings. The first group is injected with 50 micrograms of the peptide composition and the second and third groups with 500 and 5,000 micrograms of peptide composition, respectively. The patients within each group range in age from 21-65 and represent diverse ethnic backgrounds. All of them have a tumor that expresses 125P5C8.


Clinical manifestations or antigen-specific T-cell responses are monitored to assess the effects of administering the peptide compositions. The vaccine composition is found to be both safe and efficacious in the treatment of 125P5C8-associated disease.


Example 31
Induction of CTL Responses Using a Prime Boost Protocol

A prime boost protocol similar in its underlying principle to that used to confirm the efficacy of a DNA vaccine in transgenic mice, such as described above in Example 23, can also be used for the administration of the vaccine to humans. Such a vaccine regimen can include an initial administration of, for example, naked DNA followed by a boost using recombinant virus encoding the vaccine, or recombinant protein/polypeptide or a peptide mixture administered in an adjuvant.


For example, the initial immunization may be performed using an expression vector, such as that constructed in Example 22 in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 μg) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5×109 pfu. An alternative recombinant virus, such as an MVA, canarypox, adenovirus, or adeno-associated virus, can also be used for the booster, or the polyepitopic protein or a mixture of the peptides can be administered. For evaluation of vaccine efficacy, patient blood samples are obtained before immunization as well as at intervals following administration of the initial vaccine and booster doses of the vaccine. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.


Analysis of the results indicates that a magnitude of response sufficient to achieve a therapeutic or protective immunity against 125P5C8 is generated.


Example 32
Administration of Vaccine Compositions Using Dendritic Cells (DC)

Vaccines comprising peptide epitopes of the invention can be administered using APCs, or “professional” APCs such as DC. In this example, peptide-pulsed DC are administered to a patient to stimulate a CTL response in vivo. In this method, dendritic cells are isolated, expanded, and pulsed with a vaccine comprising peptide CTL and HTL epitopes of the invention. The dendritic cells are infused back into the patient to elicit CTL and HTL responses in vivo. The induced CTL and HTL then destroy or facilitate destruction, respectively, of the target cells that bear the 125P5C8 protein from which the epitopes in the vaccine are derived.


For example, a cocktail of epitope-comprising peptides is administered ex vivo to PBMC, or isolated DC therefrom. A pharmaceutical to facilitate harvesting of DC can be used, such as Progenipoietin™ (Monsanto, St. Louis, Mo.) or GM-CSF/IL-4. After pulsing the DC with peptides, and prior to reinfusion into patients, the DC are washed to remove unbound peptides.


As appreciated clinically, and readily determined by one of skill based on clinical outcomes, the number of DC reinfused into the patient can vary (see, e.g., Nature Med. 4:328, 1998; Nature Med. 2:52, 1996 and Prostate 32:272, 1997). Although 2−50×106 DC per patient are typically administered, larger number of DC, such as 107 or 108 can also be provided. Such cell populations typically contain between 50-90% DC.


In some embodiments, peptide-loaded PBMC are injected into patients without purification of the DC. For example, PBMC generated after treatment with an agent such as Progenipoietin™ are injected into patients without purification of the DC. The total number of PBMC that are administered often ranges from 108 to 1010. Generally, the cell doses injected into patients is based on the percentage of DC in the blood of each patient, as determined, for example, by immunofluorescence analysis with specific anti-DC antibodies. Thus, for example, if Progenipoietin™ mobilizes 2% DC in the peripheral blood of a given patient, and that patient is to receive 5×106 DC, then the patient will be injected with a total of 2.5×108 peptide-loaded PBMC. The percent DC mobilized by an agent such as Progenipoietin™ is typically estimated to be between 2-10%, but can vary as appreciated by one of skill in the art.


Ex Vivo Activation of CTL/HTL Responses


Alternatively, ex vivo CTL or HTL responses to 125P5C8 antigens can be induced by incubating, in tissue culture, the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of APC, such as DC, and immunogenic peptides. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cells, i.e., tumor cells.


Example 33
An Alternative Method of Identifying and Confirming Motif-Bearing Peptides

Another method of identifying and confirming motif-bearing peptides is to elute them from cells bearing defined MHC molecules. For example, EBV transformed B cell lines used for tissue typing have been extensively characterized to determine which HLA molecules they express. In certain cases these cells express only a single type of HLA molecule. These cells can be transfected with nucleic acids that express the antigen of interest, e.g. 125P5C8. Peptides produced by endogenous antigen processing of peptides produced as a result of transfection will then bind to HLA molecules within the cell and be transported and displayed on the cell's surface. Peptides are then eluted from the HLA molecules by exposure to mild acid conditions and their amino acid sequence determined, e.g., by mass spectral analysis (e.g., Kubo et al., J. Immunol. 152:3913, 1994). Because the majority of peptides that bind a particular HLA molecule are motif-bearing, this is an alternative modality for obtaining the motif-bearing peptides correlated with the particular HLA molecule expressed on the cell.


Alternatively, cell lines that do not express endogenous HLA molecules can be transfected with an expression construct encoding a single HLA allele. These cells can then be used as described, i.e., they can then be transfected with nucleic acids that encode 125P5C8 to isolate peptides corresponding to 125P5C8 that have been presented on the cell surface. Peptides obtained from such an analysis will bear motif(s) that correspond to binding to the single HLA allele that is expressed in the cell.


As appreciated by one in the art, one can perform a similar analysis on a cell bearing more than one HLA allele and subsequently determine peptides specific for each HLA allele expressed. Moreover, one of skill would also recognize that means other than transfection, such as loading with a protein antigen, can be used to provide a source of antigen to the cell.


Example 34
Complementary Polynucleotides

Sequences complementary to the 125P5C8-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring 125P5C8. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using, e.g., OLIGO 4.06 software (National Biosciences) and the coding sequence of 125P5C8. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5′ sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to a 125P5C8-encoding transcript.


Example 35
Purification of Naturally-Occurring or Recombinant 125P5C8 Using 125P5C8-Specific Antibodies

Naturally occurring or recombinant 125P5C8 is substantially purified by immunoaffinity chromatography using antibodies specific for 125P5C8. An immunoaffinity column is constructed by covalently coupling anti-125P5C8 antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.


Media containing 125P5C8 are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of 125P5C8 (e.g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/125P5C8 binding (e.g., a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and GCR.P is collected.


Example 36
Identification of Molecules which Interact with 125P5C8

125P5C8, or biologically active fragments thereof, are labeled with 121 1 Bolton-Hunter reagent. (See, e.g., Bolton et al. (1973) Biochem. J. 133:529.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled 125P5C8, washed, and any wells with labeled 125P5C8 complex are assayed. Data obtained using different concentrations of 125P5C8 are used to calculate values for the number, affinity, and association of 125P5C8 with the candidate molecules.


Example 37
In Vivo Assay for 125P5C8 Tumor Growth Promotion

The effect of the 125P5C8 protein on tumor cell growth is evaluated in vivo by evaluating tumor development and growth of cells expressing or lacking 125P5C8. For example, SCID mice are injected subcutaneously on each flank with 1×106 of either 3T3, prostate, kidney or breast cancer cell lines (e.g. PC3, DU145, CaKi, SW 839, MCF7 cells) containing tkNeo empty vector or 125P5C8. At least two strategies can be used: (1) Constitutive 125P5C8 expression under regulation of a promoter, such as a constitutive promoter obtained from the genomes of viruses such as polyoma virus, fowlpox virus (see UK 2,211,504, published 5 Jul. 1989), adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), or from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, provided such promoters are compatible with the host cell systems, and (2) Regulated expression under control of an inducible vector system, such as ecdysone, tetracycline, etc., provided such promoters are compatible with the host cell systems. Tumor volume is then monitored by caliper measurement at the appearance of palpable tumors and followed over time to determine if 125P5C8-expressing cells grow at a faster rate and whether tumors produced by 125P5C8-expressing cells demonstrate characteristics of altered aggressiveness (e.g. enhanced metastasis, vascularization, reduced responsiveness to chemotherapeutic drugs).


Additionally, mice can be implanted with 1×105 of the same cells orthotopically to determine if 125P5C8 has an effect on local growth in the prostate, kidney or mammary gland, and whether 125P5C8 affects the ability of the cells to metastasize, specifically to lungs, lymph nodes, and bone marrow.


The assay is also useful to determine the 125P5C8 inhibitory effect of candidate therapeutic compositions, such as for example, 125P5C8 intrabodies, 125P5C8 antisense molecules and ribozymes.


Example 38
125P5C8 Monoclonal Antibody-Mediated Inhibition of Prostate and Kidney Tumors In Vivo

The significant expression of 125P5C8 in cancer tissues, together with its restrictive expression in normal tissues and its surface expression make 125P5C8 a good target for antibody therapy. Similarly, 125P5C8 is a target for T cell-based immunotherapy. Thus, the therapeutic efficacy of anti-125P5C8 mAbs in human prostate cancer xenograft mouse models is evaluated by using androgen-independent LAPC-4 and LAPC-9 xenografts (Craft, N., et al., Cancer Res, 1999. 59(19): p. 5030-6) the androgen independent recombinant cell line PC3-125P5C8 and 3T3-125P5C8 (see, e.g., Kaighn, M. E., et al., Invest Urol, 1979. 17(1): p. 16-23). Similarly, anti-125P5C8 mAbs are evaluated in human kidney cancer xenograft models such as AGS-K3 and AGS-K6 and in recombinant kidney cell lines such as CaKi-125P5C8.


Antibody efficacy on tumor growth and metastasis formation is studied, e.g., in a mouse orthotopic prostate cancer xenograft models and mouse kidney xenograft models. The antibodies can be unconjugated, as discussed in this Example, or can be conjugated to a therapeutic modality, as appreciated in the art. Anti-125P5C8 mAbs inhibit formation of both the androgen-dependent LAPC-9 and androgen-independent PC3-125P5C8 tumor xenografts. Anti-125P5C8 mAbs also retard the growth of established orthotopic tumors and prolonged survival of tumor-bearing mice. These results indicate the utility of anti-125P5C8 mAbs in the treatment of local and advanced stages of prostate cancer. (See, e.g., Saffran, D., et al., PNAS 10:1073-1078). Similarly, anti-125P5C8 mAbs can inhibit formation of AGS-K3 and AGS-K6 tumors in SCID mice, and prevent or retard the growth CaKi-125P5C8 tumor xenografts. These results indicate the use of anti-125P5C8 mAbs in the treatment of kidney cancer.


Administration of the anti-125P5C8 mAbs led to retardation of established orthotopic tumor growth and inhibition of metastasis to distant sites, resulting in a significant prolongation in the survival of tumor-bearing mice. These studies indicate that 125P5C8 as an attractive target for immunotherapy and demonstrate the therapeutic potential of anti-125P5C8 mAbs for the treatment of local and metastatic prostate cancer. This example demonstrates that unconjugated 125P5C8 monoclonal antibodies are effective to inhibit the growth of human prostate tumor xenografts and human kidney xenografts grown in SCID mice; accordingly a combination of such efficacious monoclonal antibodies is also effective.


Tumor Inhibition Using Multiple Unconjugated 125P5C8 Mabs


Materials and Methods


125P5C8 Monoclonal Antibodies:


Monoclonal antibodies are raised against 125P5C8 as described in Example 11. The antibodies are characterized by ELISA, Western blot, FACS, and immunoprecipitation for their capacity to bind 125P5C8. Epitope mapping data for the anti-125P5C8 mAbs, as determined by ELISA and Western analysis, recognize epitopes on the 125P5C8 protein. Immunohistochemical analysis of prostate cancer tissues and cells with these antibodies is performed.


The monoclonal antibodies are purified from ascites or hybridoma tissue culture supernatants by Protein-G Sepharose chromatography, dialyzed against PBS, filter sterilized, and stored at −20° C. Protein determinations are performed by a Bradford assay (Bio-Rad, Hercules, Calif.). A therapeutic monoclonal antibody or a cocktail comprising a mixture of individual monoclonal antibodies is prepared and used for the treatment of mice receiving subcutaneous or orthotopic injections of LAPC-9 prostate tumor xenografts.


Cancer Xenografts and Cell Lines


The LAPC-9 xenograft, which expresses a wild-type androgen receptor and produces prostate-specific antigen (PSA), is passaged in 6- to 8-week-old male ICR-severe combined immunodeficient (SCID) mice (Taconic Farms) by s.c. trocar implant (Craft, N., et al., supra). The AGS-K3 and AGS-K6 kidney xenografts are also passaged by subcutaneous implants in 6- to 8-week old SCID mice. Single-cell suspensions of tumor cells are prepared as described in Craft, et al. The prostate carcinoma cell line PC3 (American Type Culture Collection) is maintained in RPMI supplemented with L-glutamine and 10% FBS, and the kidney carcinoma line CaKi as well as NIH-3T3 cells (American Type Culture Collection) are maintained in DMEM supplemented with L-glutamine and 10% FBS.


A PC3-125P5C8, CaKi-125P5C8 and 3T3-125P5C8 cell populations are generated by retroviral gene transfer as described in Hubert, R. S., et al., STEAP: a prostate-specific cell-surface antigen highly expressed in human prostate tumors. Proc Natl Acad Sci USA, 1999. 96(25): p. 14523-8.


Xenograft Mouse Models.


Subcutaneous (s.c.) tumors are generated by injection of 1×106 LAPC-9, AGS-K3, AGS-K6, PC3, PC3-125P5C8, CaKi or CaKi-125P5C8 cells mixed at a 1:1 dilution with Matrigel (Collaborative Research) in the right flank of male SCID mice. To test antibody efficacy on tumor formation, i.p. antibody injections are started on the same day as tumor-cell injections. As a control, mice are injected with either purified mouse IgG (ICN) or PBS; or a purified monoclonal antibody that recognizes an irrelevant antigen not expressed in human cells. In preliminary studies, no difference is found between mouse IgG or PBS on tumor growth. Tumor sizes are determined by vernier caliper measurements, and the tumor volume is calculated as length×width×height. Mice with s.c. tumors greater than 1.5 cm in diameter are sacrificed. PSA levels are determined by using a PSA ELISA kit (Anogen, Mississauga, Ontario). Circulating levels of anti-125P5C8 mAbs are determined by a capture ELISA kit (Bethyl Laboratories, Montgomery, Tex.). (See, e.g., (Saffran, D., et al., PNAS 10: 1073-1078)


Orthotopic injections are performed under anesthesia by using ketamine/xylazine. For prostate orthotopic studies, an incision is made through the abdominal muscles to expose the bladder and seminal vesicles, which then are delivered through the incision to expose the dorsal prostate. LAPC-9 cells (5×105) mixed with Matrigel are injected into each dorsal lobe in a 10-μl volume. To monitor tumor growth, mice are bled on a weekly basis for determination of PSA levels. For kidney orthopedic models, an incision is made through the abdominal muscles to expose the kidney. AGS-K3 or AGS-K6 cells mixed with Matrigel are injected under the kidney capsule. The mice are segregated into groups for the appropriate treatments, with anti-125P5C8 or control mAbs being injected i.p.


Anti-125P5C8 mAbs Inhibit Growth of 125P5C8-Expressing Xenograft-Cancer Tumors


The effect of anti-125P5C8 mAbs on tumor formation is tested by using LAPC-9 and AGS-K3 orthotopic models. As compared with the s.c. tumor model, the orthotopic model, which requires injection of tumor cells directly in the mouse prostate or kidney, respectively, results in a local tumor growth, development of metastasis in distal sites, deterioration of mouse health, and subsequent death (Saffran, D., et al., PNAS supra; Fu, X., et al., Int J Cancer, 1992. 52(6): p. 987-90; Kubota, T., J Cell Biochem, 1994. 56(1): p. 4-8). The features make the orthotopic model more representative of human disease progression and allowed us to follow the therapeutic effect of mAbs on clinically relevant end points.


Accordingly, tumor cells are injected into the mouse prostate or kidney, and 2 days later, the mice are segregated into two groups and treated with either: a) 200-500 μg, of anti-125P5C8 Ab, or b) PBS three times per week for two to five weeks.


A major advantage of the orthotopic prostate-cancer model is the ability to study the development of metastases. Formation of metastasis in mice bearing established orthotopic tumors is studies by IHC analysis on lung sections using an antibody against a prostate-specific cell-surface protein STEAP expressed at high levels in LAPC-9 xenografts (Hubert, R. S., et al., Proc Natl Acad Sci USA, 1999. 96(25): p. 14523-8) or anti-G250 antibody for kidney cancer models.


Mice bearing established orthotopic LAPC-9 tumors are administered 1000 μg injections of either anti-125P5C8 mAb or PBS over a 4-week period. Mice in both groups are allowed to establish a high tumor burden (PSA levels greater than 300 ng/ml), to ensure a high frequency of metastasis formation in mouse lungs. Mice then are killed and their prostate/kidney and lungs are analyzed for the presence of tumor cells by IHC analysis.


These studies demonstrate a broad anti-tumor efficacy of anti-125P5C8 antibodies on initiation and progression of prostate and kidney cancer in xenograft mouse models. Anti-125P5C8 antibodies inhibit tumor formation of both androgen-dependent and androgen-independent tumors as well as retarding the growth of already established tumors and prolong the survival of treated mice. Moreover, anti-125P5C8 mAbs demonstrate a dramatic inhibitory effect on the spread of local prostate tumor to distal sites, even in the presence of a large tumor burden. Thus, anti-125P5C8 mAbs are efficacious on major clinically relevant end points (tumor growth), prolongation of survival, and health.


Example 39
Therapeutic and Diagnostic Use of Anti-125P5C8 Antibodies in Humans

Anti-125P5C8 monoclonal antibodies are safely and effectively used for diagnostic, prophylactic, prognostic and/or therapeutic purposes in humans. Western blot and immunohistochemical analysis of cancer tissues and cancer xenografts with anti-125P5C8 mAb show strong extensive staining in carcinoma but significantly lower or undetectable levels in normal tissues. Detection of 125P5C8 in carcinoma and in metastatic disease demonstrates the usefulness of the mAb as a diagnostic and/or prognostic indicator. Anti-125P5C8 antibodies are therefore used in diagnostic applications such as immunohistochemistry of kidney biopsy specimens to detect cancer from suspect patients.


As determined by flow cytometry, anti-125P5C8 mAb specifically binds to carcinoma cells. Thus, anti-125P5C8 antibodies are used in diagnostic whole body imaging applications, such as radioimmunoscintigraphy and radioimmunotherapy, (see, e.g., Potamianos S., et. al. Anticancer Res 20(2A):925-948 (2000)) for the detection of localized and metastatic cancers that exhibit expression of 125P5C8. Shedding or release of an extracellular domain of 125P5C8 into the extracellular milieu, such as that seen for alkaline phosphodiesterase B10 (Meerson, N. R., Hepatology 27:563-568 (1998)), allows diagnostic detection of 125P5C8 by anti-125P5C8 antibodies in serum and/or urine samples from suspect patients.


Anti-125P5C8 antibodies that specifically bind 125P5C8 are used in therapeutic applications for the treatment of cancers that express 125P5C8. Anti-125P5C8 antibodies are used as an unconjugated modality and as conjugated form in which the antibodies are attached to one of various therapeutic or imaging modalities well known in the art, such as a prodrugs, enzymes or radioisotopes. In preclinical studies, unconjugated and conjugated anti-125P5C8 antibodies are tested for efficacy of tumor prevention and growth inhibition in the SCID mouse cancer xenograft models, e.g., kidney cancer models AGS-K3 and AGS-K6, (see, e.g., Example 38). Conjugated and unconjugated anti-125P5C8 antibodies are used as a therapeutic modality in human clinical trials either alone or in combination with other treatments as described in following Examples.


Example 40
Human Clinical Trials for the Treatment and Diagnosis of Human Carcinomas Through Use of Human Anti-125P5C8 Antibodies In vivo

Antibodies are used in accordance with the present invention which recognize an epitope on 125P5C8, and are used in the treatment of certain tumors such as those listedin Table I. Based upon a number of factors, including 125P5C8 expression levels, tumors such as those listed in Table I are presently preferred indications. In connection with each of these indications, three clinical approaches are successfully pursued.


I.) Adjunctive therapy: In adjunctive therapy, patients are treated with anti-125P5C8 antibodies in combination with a chemotherapeutic or antineoplastic agent and/or radiation therapy. Primary cancer targets, such as those listed in Table I, are treated under standard protocols by the addition anti-125P5C8 antibodies to standard first and second line therapy. Protocol designs address effectiveness as assessed by reduction in tumor mass as well as the ability to reduce usual doses of standard chemotherapy. These dosage reductions allow additional and/or prolonged therapy by reducing dose-related toxicity of the chemotherapeutic agent. Anti-125P5C8 antibodies are utilized in several adjunctive clinical trials in combination with the chemotherapeutic or antineoplastic agents adriamycin (advanced prostrate carcinoma), cisplatin (advanced head and neck and lung carcinomas), taxol (breast cancer), and doxorubicin (preclinical).


II.) Monotherapy: In connection with the use of the anti-125P5C8 antibodies in monotherapy of tumors, the antibodies are administered to patients without a chemotherapeutic or antineoplastic agent. In one embodiment, monotherapy is conducted clinically in end stage cancer patients with extensive metastatic disease. Patients show some disease stabilization. Trials demonstrate an effect in refractory patients with cancerous tumors.


III.) Imaging Agent: Through binding a radionuclide (e.g., iodine or yttrium (I131, Y90) to anti-125P5C8 antibodies, the radiolabeled antibodies are utilized as a diagnostic and/or imaging agent. In such a role, the labeled antibodies localize to both solid tumors, as well as, metastatic lesions of cells expressing 125P5C8. In connection with the use of the anti-125P5C8 antibodies as imaging agents, the antibodies are used as an adjunct to surgical treatment of solid tumors, as both a pre-surgical screen as well as a post-operative follow-up to determine what tumor remains and/or returns. In one embodiment, a (111In)-125P5C8 antibody is used as an imaging agent in a Phase I human clinical trial in patients having a carcinoma that expresses 125P5C8 (by analogy see, e.g., Divgi et al. J. Natl. Cancer Inst. 83:97-104 (1991)). Patients are followed with standard anterior and posterior gamma camera. The results indicate that primary lesions and metastatic lesions are identified.


Dose and Route of Administration


As appreciated by those of ordinary skill in the art, dosing considerations can be determined through comparison with the analogous products that are in the clinic. Thus, anti-125P5C8 antibodies can be administered with doses in the range of 5 to 400 mg/m2, with the lower doses used, e.g., in connection with safety studies. The affinity of anti-125P5C8 antibodies relative to the affinity of a known antibody for its target is one parameter used by those of skill in the art for determining analogous dose regimens. Further, anti-125P5C8 antibodies that are fully human antibodies, as compared to the chimeric antibody, have slower clearance; accordingly, dosing in patients with such fully human anti-125P5C8 antibodies can be lower, perhaps in the range of 50 to 300 mg/m2, and still remain efficacious. Dosing in mg/m2, as opposed to the conventional measurement of dose in mg/kg, is a measurement based on surface area and is a convenient dosing measurement that is designed to include patients of all sizes from infants to adults.


Three distinct delivery approaches are useful for delivery of anti-125P5C8 antibodies. Conventional intravenous delivery is one standard delivery technique for many tumors. However, in connection with tumors in the peritoneal cavity, such as tumors of the ovaries, biliary duct, other ducts, and the like, intraperitoneal administration may prove favorable for obtaining high dose of antibody at the tumor and to also minimize antibody clearance. In a similar manner, certain solid tumors possess vasculature that is appropriate for regional perfusion. Regional perfusion allows for a high dose of antibody at the site of a tumor and minimizes short term clearance of the antibody.


Clinical Development Plan (CDP)


Overview: The CDP follows and develops treatments of anti-125P5C8 antibodies in connection with adjunctive therapy, monotherapy, and as an imaging agent. Trials initially demonstrate safety and thereafter confirm efficacy in repeat doses. Trails are open label comparing standard chemotherapy with standard therapy plus anti-125P5C8 antibodies. As will be appreciated, one criteria that can be utilized in connection with enrollment of patients is 125P5C8 expression levels in their tumors as determined by biopsy.


As with any protein or antibody infusion-based therapeutic, safety concerns are related primarily to (i) cytokine release syndrome, i.e., hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material (i.e., development of human antibodies by the patient to the antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express 125P5C8. Standard tests and follow-up are utilized to monitor each of these safety concerns. Anti-125P5C8 antibodies are found to be safe upon human administration.


Example 41
Human Clinical Trial Adjunctive Therapy with Human Anti-125P5C8 Antibody and Chemotherapeutic Agent

A phase I human clinical trial is initiated to assess the safety of six intravenous doses of a human anti-125P5C8 antibody in connection with the treatment of a solid tumor, e.g., a cancer of a tissue listed in Table I. In the study, the safety of single doses of anti-125P5C8 antibodies when utilized as an adjunctive therapy to an antineoplastic or chemotherapeutic agent, such as cisplatin, topotecan, doxorubicin, adriamycin, taxol, or the like, is assessed. The trial design includes delivery of six single doses of an anti-125P5C8 antibody with dosage of antibody escalating from approximately about 25 mg/m2 to about 275 mg/m2 over the course of the treatment in accordance with the following schedule:




















Day 0
Day 7
Day 14
Day 21
Day 28
Day 35






















mAb Dose
25
75
125
175
225
275



mg/m2
mg/m2
mg/m2
mg/m2
mg/m2
mg/m2


Chemotherapy
+
+
+
+
+
+


(standard dose)









Patients are closely followed for one-week following each administration of antibody and chemotherapy. In particular, patients are assessed for the safety concerns mentioned above: (i) cytokine release syndrome, i.e., hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material (i.e., development of human antibodies by the patient to the human antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express 125P5C8. Standard tests and follow-up are utilized to monitor each of these safety concerns. Patients are also assessed for clinical outcome, and particularly reduction in tumor mass as evidenced by MRI or other imaging.


The anti-125P5C8 antibodies are demonstrated to be safe and efficacious, Phase II trials confirm the efficacy and refine optimum dosing.


Example 42
Human Clinical Trial
Monotherapy with Human Anti-125P5C8 Antibody

Anti-125P5C8 antibodies are safe in connection with the above-discussed adjunctive trial, a Phase II human clinical trial confirms the efficacy and optimum dosing for monotherapy. Such trial is accomplished, and entails the same safety and outcome analyses, to the above-described adjunctive trial with the exception being that patients do not receive chemotherapy concurrently with the receipt of doses of anti-125P5C8 antibodies.


Example 43
Human Clinical Trial
Diagnostic Imaging with Anti-125P5C8 Antibody

Once again, as the adjunctive therapy discussed above is safe within the safety criteria discussed above, a human clinical trial is conducted concerning the use of anti-125P5C8 antibodies as a diagnostic imaging agent. The protocol is designed in a substantially similar manner to those described in the art, such as in Divgi et al. J. Natl. Cancer Inst. 83:97-104 (1991). The antibodies are found to be both safe and efficacious when used as a diagnostic modality.


Example 44
Homology Comparison of 125P5C8 to Known Sequences

The 125P5C8 gene is identical to a recently sequenced gene, namely human hypothetical protein FLJ21511 (gi113376644) showing 100% identity that protein. The closest homolog to the 125P5C8 protein is a mouse hypothetical protein (gil116741400), displaying 88% homology with that protein. However, neither the human nor the mouse homolog of 125P5C8 have a known function. The 125P5C8 protein consists of 699 amino acids, with calculated molecular weight of 78.5 kDa, and pI of 8.75. Based on structural analysis, 125P5C8 contains 10-11 transmembrane domains, with the C-terminus oriented intracellularly. The multiple transmembrane topology was also confirmed by Prints analysis which revealed the presence of a Srg-like motif (Table XXII). The 125P5C8 protein localizes to the cell surface, with low likelihood of mitochondrial localization. Motif analysis revealed the presence of a Phosphoribulokinase/Uridine kinase family at aa 296-322.


Uridine kinase is a rate limiting enzyme of the UMP salvage pathway. It phosphorylates nucleosides into nucleotides and regulates the availability of pyrimidine nucleotides for cellular nucleic acid synthesis (Cihak A and Rada B. Neoplasma. 1976, 23:233). In addition to participating in RNA synthesis, uridine nucleotides play an important role in hexose metabolism, polymerization of sugars into starch and assembly of oligosaccharide moieties of glycoproteins (Striepen B et al. Biochemistry. 1999, 38:1478; O'Regan M, et al. Int J Biol Macromol. 1994, 16:283). Uridine kinase activity has been associated with several cancers. Specifically, enhanced uridine kinase activity has been observed in human hepatomas, colon carcinoma and mesothelioma (Luccioni C et al, Int. J. Cancer 1994, 58:517; Otal-Brun M and Webb T E. Cancer Lett. 1979, 6:39; Greengard O et al, J Natl Cancer Inst. 1987, 78:617). In addition, enhanced uridine kinase activity correlates with enhanced proliferation (Cheng N, Traut T W. J Cell Biochem. 1987, 35:217).


The presence of 10-11 transmembrane domains in 125P5C8 is indicative of transporter function. Several transporters containing 10 transmembrane domains have been previously described, including glucose transporters, ion transporters, water pumps and small molecule efflux (Rogers S et al, Am J Physiol Endocrinol Metab. 2002, 282:E733; Gerelsaikhan T, Turner R J. J Biol. Chem. 2000, 275:0471; Kage K, et al, Int J. Cancer. 2002, 97:626)


The presence of uridine kinase motif, the multiple transmembrane structure along with its cell surface localization indicate that 125P5C8 plays a role in sugar metabolism, RNA synthesis, ion, small molecule and sugar transport as well as regulating gene transcription in mammalian cells. These biological function thereby regulates cellular proliferation, transformation, differentiation and apoptosis. These biological functions have a direct effect on transformation, tumor growth and progression.


Accordingly, when 125P5C8 functions as a regulator of cellular growth, cell transformation, tumor formation, as a transporter or as a modulator of transcription, 125P5C8 is used for therapeutic, diagnostic, prognostic and/or preventative purposes. In addition, when a molecule, such as a variant or SNP of 125P5C8 is expressed in cancerous tissues, such as those listed in Table I, they are used for therapeutic, diagnostic, prognostic and/or preventative purposes.


Several SNP of 125P5C8 have been identified, including the 5 variants shown in FIG. 11. While all 5 SNPs encode for 1 amino acid change relative to 125P5C8v1, the protein characteristics of the 5 SNPs are highly consistent with those of 125P5C8v1.


Example 45
Regulation of Transcription

The presence of a uridine kinase domain at aa 296 indicates that 125P5C8 participates in RNA synthesis. In addition, its surface location suggests that 125P5C8 can mediate signaling from the cell surface to the nucleus, thereby regulating the expression of eukaryotic genes. Regulation of gene expression is confirmed, e.g., by studying gene expression in cells expressing or lacking 125P5C8. For this purpose, two types of experiments are performed.


In the first set of experiments, RNA from parental and 125P5C8-expressing cells are extracted and hybridized to commercially available gene arrays (Clontech) (Smid-Koopman E et al. Br J Cancer. 2000. 83:246). Resting cells as well as cells treated with FBS, androgen or growth factors are compared. Differentially expressed genes are identified in accordance with procedures known in the art. The differentially expressed genes are then mapped to biological pathways (Chen K et al. Thyroid. 2001. 11:41.).


In the second set of experiments, specific transcriptional pathway activation is evaluated using commercially available (Stratagene) luciferase reporter constructs including: NFkB-luc, SRE-luc, ELK1-luc, ARE-luc, p53-luc, and CRE-luc. In addition, a Myc/Max specific response element, namely E-box hexamer CACGTG reporter is also evaluated (Ben-Porath I et al, Mol Cell Biol 1999; 19:3529). These transcriptional reporters contain consensus binding sites for known transcription factors that lie downstream of well-characterized signal transduction pathways, and represent a good tool to ascertain pathway activation and screen for positive and negative modulators of pathway activation.


Thus, 125P5C8 plays a role in gene regulation, and it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes.


Example 46
Identification and Confirmation of Potential Signal Transduction Pathways

Many mammalian proteins have been reported to interact with signaling molecules and to participate in regulating signaling pathways. (J Neurochem. 2001; 76:217-223). In particular, multi-transmembrane proteins and ion channels induce ion accumulation that, in turn, mediate activation of signaling cascades such as PI3K (Putney J. Nature. 2001, 410:648). In addition, multi-transmembrane proteins interact with a variety of signaling molecules, such as kinases, adaptor proteins, etc, thereby regulating downstream events (Hruska-Hageman A M et al, Biochem J. 2002, 361:443; Cook K K, Fadool D A., J Biol Chem. 2002). Using immunoprecipitation and Western blotting techniques, proteins are identified that associate with 125P5C8 and mediate signaling events. Several pathways known to play a role in cancer biology can be regulated by 125P5C8, including phospholipid pathways such as PI3K, AKT, etc, adhesion and migration pathways, including FAK, Rho, Rac-1, etc, as well as mitogenic/survival cascades such as ERK, p38, etc (Cell Growth Differ. 2000, 11:279; J Biol. Chem. 1999, 274:801; Oncogene. 2000, 19:3003, J. Cell Biol. 1997, 138:913.).


To confirm that 125P5C8 directly or indirectly activates known signal transduction pathways in cells, luciferase (luc) based transcriptional reporter assays are carried out in cells expressing individual genes. These transcriptional reporters contain consensus-binding sites for known transcription factors that lie downstream of well-characterized signal transduction pathways. The reporters and examples of these associated transcription factors, signal transduction pathways, and activation stimuli are listed below.


1. NFkB-luc, NFkB/Rel; Ik-kinase/SAPK; growth/apoptosis/stress


2. SRE-luc, SRF/TCF/ELK1; MAPK/SAPK; growth/differentiation


3. AP-1-luc, FOS/JUN; MAPK/SAPK/PKC; growth/apoptosis/stress


4. ARE-luc, androgen receptor; steroids/MAPK; growth/differentiation/apoptosis


5. p53-luc, p53; SAPK; growth/differentiation/apoptosis


6. CRE-luc, CREB/ATF2; PKA/p38; growth/apoptosis/stress


Gene-mediated effects can be assayed in cells showing mRNA expression. Luciferase reporter plasmids can be introduced by lipid-mediated transfection (TFX-50, Promega). Luciferase activity, an indicator of relative transcriptional activity, is measured by incubation of cell extracts with luciferin substrate and luminescence of the reaction is monitored in a luminometer.


Signaling pathways activated by 125P5C8 are mapped and used for the identification and validation of therapeutic targets. When 125P5C8 is involved in cell signaling, it is used as target for diagnostic, prognostic, preventative and/or therapeutic purposes.


Example 47
Involvement in Tumor Progression

Based on the documented role the uridine kinase pathway in cell growth and proliferation (Cheng N above), the 125P5C8 gene can contribute to the growth of cancer cells. The role of 125P5C8 in tumor growth is confirmed in a variety of primary and transfected cell lines including prostate, breast and kidney cell lines, as well as NIH 3T3 cells engineered to stably express 125P5C8. Parental cells lacking 125P5C8 and cells expressing 125P5C8 are evaluated for cell growth using a well-documented proliferation assay (Fraser S P, Grimes J A, Djamgoz M B. Prostate. 2000; 44:61, Johnson D E, Ochieng J, Evans SL. Anticancer Drugs. 1996, 7:288).


To confirm the role of 125P5C8 in the transformation process, its effect in colony forming assays is investigated. Parental NIH-3T3 cells lacking 125P5C8 are compared to NIH-3T3 cells expressing 125P5C8, using a soft agar assay under stringent and more permissive conditions (Song Z. et al. Cancer Res. 2000; 60:6730).


To confirm the role of 125P5C8 in invasion and metastasis of cancer cells, a well-established assay is used, e.g., a Transwell Insert System assay (Becton Dickinson) (Cancer Res. 1999; 59:6010). Control cells, including prostate, breast and kidney cell lines lacking 125P5C8 are compared to cells expressing 125P5C8. Cells are loaded with the fluorescent dye, calcein, and plated in the top well of the Transwell insert coated with a basement membrane analog. Invasion is determined by fluorescence of cells in the lower chamber relative to the fluorescence of the entire cell population.


125P5C8 can also play a role in cell cycle and apoptosis. Parental cells and cells expressing 125P5C8 are compared for differences in cell cycle regulation using a well-established BrdU assay (Abdel-Malek Z A. J Cell Physiol. 1988, 136:247). In short, cells are grown under both optimal (full serum) and limiting (low serum) conditions are labeled with BrdU and stained with anti-BrdU Ab and propidium iodide. Cells are analyzed for entry into the G1, S, and G2M phases of the cell cycle. Alternatively, the effect of stress on apoptosis is evaluated in control parental cells and cells expressing 125P5C8, including normal and tumor prostate, colon and lung cells. Engineered and parental cells are treated with various chemotherapeutic agents, such as etoposide, flutamide, etc, and protein synthesis inhibitors, such as cycloheximide. Cells are stained with annexin V-FITC and cell death is measured by FACS analysis. The modulation of cell death by 125P5C8 can play a critical role in regulating tumor progression and tumor load.


When 125P5C8 plays a role in cell growth, transformation, invasion or apoptosis, it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes.


Example 48
Involvement in Angiogenesis

Angiogenesis or new capillary blood vessel formation is necessary for tumor growth (Hanahan D, Folkman J. Cell. 1996, 86:353; Folkman J. Endocrinology. 1998 139:441). Based on the effect of phsophodieseterase inhibitors on endothelial cells, 125P5C8 plays a role in angiogenesis (DeFouw L et al, Microvasc Res 2001, 62:263). Several assays have been developed to measure angiogenesis in vitro and in vivo, such as the tissue culture assays endothelial cell tube formation and endothelial cell proliferation. Using these assays as well as in vitro neo-vascularization, the role of 125P5C8 in angiogenesis, enhancement or inhibition, is confirmed.


For example, endothelial cells engineered to express 125P5C8 are evaluated using tube formation and proliferation assays. The effect of 125P5C8 is also confirmed in animal models in vivo. For example, cells either expressing or lacking 125P5C8 are implanted subcutaneously in immunocompromised mice. Endothelial cell migration and angiogenesis are evaluated 5-15 days later using immunohistochemistry techniques. 125P5C8 affects angiogenesis, and it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes


Example 49
Involvement in Protein-Protein Interactions

Multi-transmembrane proteins have been shown to interact with other proteins, such as signaling molecules, structural proteins e.g. syntaxin, co-enzymes, etc (Peters K W et al, Pflugers Arch. 2001; 443:S65; Yue G et al, J Biol. Chem. 2002; Liu S Q et al, J Biol. Chem. 2001, 276:11812), and to multimerize forming hetero- and homo-multimers (Ludwig M et al, Hum Genet. 1998, 102:576). Using immunoprecipitation techniques as well as two yeast hybrid systems, proteins are identified that associate with 125P5C8. Immunoprecipitates from cells expressing 125P5C8 and cells lacking 125P5C8 are compared for specific protein-protein associations.


Studies are performed to confirm the extent of association of 125P5C8 with effector molecules, such as cell surface protein, signaling intermediates, nuclear proteins, transcription factors, kinases, phosphates etc. Studies comparing 125P5C8 positive and 125P5C8 negative cells as well as studies comparing unstimulated/resting cells and cells treated with epithelial cell activators, such as cytokines, growth factors, androgen and anti-integrin Ab reveal unique interactions.


In addition, protein-protein interactions are confirmed using two yeast hybrid methodology (Curr Opin Chem. Biol. 1999, 3:64). A vector carrying a library of proteins fused to the activation domain of a transcription factor is introduced into yeast expressing a 125P5C8-DNA-binding domain fusion protein and a reporter construct. Protein-protein interaction is detected by colorimetric reporter activity. Specific association with effector molecules and transcription factors directs one of skill to the mode of action of 125P5C8, and thus identifies therapeutic, prognostic, preventative and/or diagnostic targets for cancer. This and similar assays are also used to identify and screen for small molecules that interact with 125P5C8.


Thus it is found that 125P5C8 associates with proteins and small molecules. Accordingly, 125P5C8 and these proteins and small molecules are used for diagnostic, prognostic, preventative and/or therapeutic purposes.


Example 50
Protein and Ion Transporter Function

Using a modified rhodamine retention assay (Davies J et al. Science 2000, 290:2295; Leith C et al. Blood 1995, 86:2329) we can confirm that 125P5CD8 functions as a protein transporter. Cell lines, such as prostate, and kidney cancer cells as well as NIH-3T3 cells, expressing or lacking 125P5C8 are loaded with Calcein AM (Molecular Probes). Cells are examined over time for dye transport using a fluorescent microscope or fluorometer. Quantitation is performed using a fluorometer (Hollo Z. et al. 1994. 1191:384). Information obtained from such experiments is used in determining whether 125P5C8 mediates the efflux of small molecules and chemotherapeutic drugs, such as doxorubicin, paclitaxel, etoposide, etc, from tumor cells, thereby lowering drug content and reducing tumor responsiveness to treatment. Such a system is also used to determine whether 125P5C8 functions in transporting ions and other small molecules. When 125P5C8 functions as a transporter, it is used as a target for preventative and therapeutic purposes as well as drug sensitivity/resistance.


To determine whether 125P5C8 functions as an ion channel, FACS analysis and electrophysiology techniques are used (Gergely L, Cook L, Agnello V. Clin Diagn Lab Immunol. 1997; 4:70; Skryma R, et al. J. Physiol. 2000, 527: 71). Using FACS analysis and commercially available indicators (Molecular Probes), parental cells and cells expressing genes under consideration are compared for their ability to transport calcium, sodium and potassium. Prostate, colon, bladder and kidney normal and tumor cell lines are used in these studies. For example cells loaded with calcium responsive indicators such as Fluo4 and Fura red are incubated in the presence or absence of ions and analyzed by flow cytometry. Information derived from these experiments provides a mechanism by which cancer cells are regulated. This is particularly true in the case of calcium, as calcium channel inhibitors have been reported to induce the death of certain cancer cells, including prostate cancer cell lines (Batra S, Popper L D, Hartley-Asp B. Prostate. 1991, 19: 299).


Using electrophysiology, uninjected oocytes and oocytes injected with 125P5C8 cRNA are compared for ion channel activity. Patch/voltage clamp assays are performed on oocytes in the presence or absence of selected ions, including calcium, potassium, sodium, etc as well as sugar. Ion channel activators (such as cAMP/GMP, forskolin, TPA, etc) and inhibitors (such as calcicludine, conotoxin, TEA, tetrodotoxin, etc) are used to confirm the function of 125P5C8 as ion channels (Schweitz H. et al. Proc. Natl. Acad. Sci. 1994. 91:878; Skryma R. et al. Prostate. 1997. 33:112).


When 125P5C8 functions as an ion channel, it is used as a target for diagnostic, preventative and therapeutic purposes.


Example 51
Involvement in Cell-Cell Communication

Cell-cell communication is essential in maintaining organ integrity and homeostasis, both of which become deregulated during tumor formation and progression. The presence of 125P5C8 on the cell surface suggests that 125P5C8 can mediate cell-cell communication. Intercellular communications can be measured using two types of assays (J. Biol. Chem. 2000, 275:25207). Cells loaded with a fluorescent dye are incubated in the presence of unlabeled recipient cells and the cell populations are examined under fluorescent microscopy. This qualitative assay measures the exchange of dye between adjacent cells. Using this assay system, we can compare cell expressing or lacking 125P5C8 and determine whether 125P5C8 can enhance or suppress cell communications. This assay can be used to identify small molecules and/or specific antibodies that modulate cell-cell communication.


When 125P5C8 functions in cell-cell communication, it is used as a target for diagnostic, preventative and therapeutic purposes.


Throughout this application, various website data content, publications, patent applications and patents are referenced. (Websites are referenced by their Uniform Resource Locator, or URL, addresses on the World Wide Web.) The disclosures of each of these references are hereby incorporated by reference herein in their entireties.


The present invention is not to be limited in scope by the embodiments disclosed herein, which are intended as single illustrations of individual aspects of the invention, and any that are functionally equivalent are within the scope of the invention. Various modifications to the models and methods of the invention, in addition to those described herein, will become apparent to those skilled in the art from the foregoing description and teachings, and are similarly intended to fall within the scope of the invention. Such modifications or other embodiments can be practiced without departing from the true scope and spirit of the invention.









TABLE I





Tissues that Express 125P5C8 When Malignant

















Prostate



Bladder



Kidney



Colon



Ovary



Breast

















TABLE II







Amino Acid Abbreviations











SINGLE LETTER
THREE LETTER
FULL NAME







F
Phe
phenylalanine



L
Leu
leucine



S
Ser
serine



Y
Tyr
tyrosine



C
Cys
cysteine



W
Trp
tryptophan



P
Pro
proline



H
His
histidine



Q
Gln
glutamine



R
Arg
arginine



I
Ile
isoleucine



M
Met
methionine



T
Thr
threonine



N
Asn
asparagine



K
Lys
lysine



V
Val
valine



A
Ala
alanine



D
Asp
aspartic acid



E
Glu
glutamic acid



G
Gly
glycine

















TABLE III







Amino Acid Substitution Matrix



























A
C
D
E
F
G
H
I
K
L
M
N
P
Q
R
S
T
V
W
Y
.






























4
0
−2
−1
−2
0
−2
−1
−1
−1
−1
−2
−1
−1
−1
1
0
0
−3
−2
A



9
−3
−4
−2
−3
−3
−1
−3
−1
−1
−3
−3
−3
−3
−1
−1
−1
−2
−2
C




6
2
−3
−1
−1
−3
−1
−4
−3
1
−1
0
−2
0
−1
−3
−4
−3
D





5
−3
−2
0
−3
1
−3
−2
0
−1
2
0
0
−1
−2
−3
−2
E






6
−3
−1
0
−3
0
0
−3
−4
−3
−3
−2
−2
−1
1
3
F







6
−2
−4
−2
−4
−3
0
−2
−2
−2
0
−2
−3
−2
−3
G








8
−3
−1
−3
−2
1
−2
0
0
−1
−2
−3
−2
2
H









4
−3
2
1
−3
−3
−3
−3
−2
−1
3
−3
−1
I










5
−2
−1
0
−1
1
2
0
−1
−2
−3
−2
K











4
2
−3
−3
−2
−2
−2
−1
1
−2
−1
L












5
−2
−2
0
−1
−1
−1
1
−1
−1
M













6
−2
0
0
1
0
−3
−4
−2
N














7
−1
−2
−1
−1
−2
−4
−3
P















5
1
0
−1
−2
−2
−1
Q
















5
−1
−1
−3
−3
−2
R

















4
1
−2
−3
−2
S


















5
0
−2
−2
T



















4
−3
−1
V




















11
2
W





















7
Y





Adapted from the GCG Software 9.0 BLOSUM62 amino acid substitution matrix (block substitution matrix). The higher the value, the more likely a substitution is found in related, natural proteins.













TABLE IV (A)







HLA Class I Supermotifs/Motifs












POSITION
POSITION
POSITION




2 (Primary Anchor)
3 (Primary Anchor)
C Terminus (Primary Anchor)














SUPERMOTIFS





A1

TI
LVMS (SEQ ID NO: 56)



FWY






A2

LIVM
ATQ (SEQ ID NO: 57)



IV
MATL (SEQ ID NO: 58)






A3

VSMA
TLI (SEQ ID NO: 59)



RK






A24

YF
WIVLMT (SEQ ID NO: 60)



FI
YWLM (SEQ ID NO: 61)






B7

P



VILF
MWYA (SEQ ID NO: 62)






B27

RHK



FYL
WMIVA (SEQ ID NO: 63)






B44

E
D



FWYLIMVA (SEQ ID NO: 64)






B58

ATS



FWY
LIVMA (SEQ ID NO: 65)






B62

QL
IVMP (SEQ ID NO: 66)



FWYMIVLA (SEQ ID NO: 67)






MOTIFS


A1

TSM



Y






A1


DE
AS


Y





(SEQ ID NO: 68)





A2.1

LM
VQIAT (SEQ ID NO: 69)



V
LIMAT (SEQ ID NO: 70)






A3

LMVISATF
CGD (SEQ ID



KYR
HFA (SEQ ID NO: 72)




NO: 71)





A11

VTMLISAGN
CDF (SEQ ID



K
RYH (SEQ ID NO: 74)




NO: 73)





A24

YFWM (SEQ ID NO: 75)



FLIW (SEQ ID NO: 76)






A*3101

MVT
ALIS (SEQ ID NO: 77)



R
K






A*3301

MVALF
IST (SEQ ID NO: 78)



RK






A*6801

AVT
MSLI (SEQ ID NO: 79)



RK






B*0702

P



LMF
WYAIV (SEQ ID NO: 80)






B*3501

P



LMFWY
IVA (SEQ ID NO: 81)






B51

P



LIVF
WYAM (SEQ ID NO: 82)






B*5301

P



IMFWY
ALV (SEQ ID NO: 83)






B*5401

P



ATIV
LMFWY (SEQ ID NO: 84)






Bolded residues are preferred, italicized residues are less preferred: A peptide is considered motif-bearing if it has primary anchors at each primary anchor position for a motif or supermotif as specified in the above table.













TABLE IV (B)







HLA Class II Supermotif









1
6
9





W, F, Y, V, .I, L
A, V, I, L, P, C, S, T
A, V, I, L, C, S, T, M, Y
















TABLE IV (C)





HLA Class II Motifs

























MOTIFS

1° anchor 1
2
3
4
5
1° anchor 6
7
8
9





DR4
preferred
FMYLIVW
M
T

I
VSTCPALIM
MH

MH



deleterious



W


R

WDE





DR1
preferred
MFLIVWY


PAMQ

VMATSPLIC
M

AVM



deleterious

C
CH
FD
CWD

GDE
D





DR7
preferred
MFLIVWY
M
W
A

IVMSACTPL
M

IV



deleterious

C

G


GRD
N
G

















DR3
MOTIFS
1° anchor 1
2
3
1° anchor 4
5
1° anchor 6





motif a preferred

LIVMFY


D


motif b preferred

LIVMFAY


DNQEST

KRH





DR Supermotif

MFLIVWY




VMSTACPLI





Italicized residues indicate less preferred or “tolerated” residues.













TABLE IV (D)







HLA Class I Supermotifs








SUPER-
POSITION:
















MOTIFS
1
2
3
4
5
6
7
8
C-terminus





















A1



1° Anchor








1° Anchor







TILVMS






FWY





A2



1° Anchor








1° Anchor






LIVMATQ






LIVMAT





A3
preferred


1° Anchor

YFW


YFW
YFW
P

1° Anchor






VSMATLI
(4/5)


(3/5)
(4/5)
(4/5)
RK



deleterious
DE (3/5);

DE




P (5/5)

(4/5)





A24



1° Anchor








1° Anchor






YFWIVLMT






FIYWLM





B7
preferred
FWY (5/5)

1° Anchor

FWY




FWY

1° Anchor





LIVM
P
(4/5)




(3/5)
VILFMWYA




(3/5)



deleterious
DE (3/5);



DE
G
QN
DE




P(5/5);



(3/
(4/5)
(4/5)
(4/5)




G(4/5);



5)




A(3/5);




QN(3/5)





B27



1° Anchor








1° Anchor






RHK






FYLWMIVA





B44



1° Anchor








1° Anchor






ED






FWYLIMVA





B58



1° Anchor








1° Anchor






ATS






FWYLIVMA





B62



1° Anchor








1° Anchor






QLIVMP






FWYMIVLA
















TABLE IV (E)







HLA Class I Motifs









POSITION:



























9













or



1
2
3
4
5
6
7
8
C-terminus
C-terminus























A1
preferred
GFYW

1°Anchor

DEA
YFW

P
DEQN
YFW

1°Anchor





9-mer


STM






Y



deleterious
DE

RHKLIVMP
A
G
A





A1
preferred
GRHK
ASTCLIVM

1°Anchor

GSTC

ASTC
LIVM
DE

1°Anchor



9-mer



DEAS





Y



deleterious
A
RHKDEPYF

DE
PQN
RHK
PG
GP





W





A1
preferred
YFW

1°Anchor

DEAQN
A
YFWQN

PASTC
GDE
P

1°Anchor



10-mer


STM







Y



deleterious
GP

RHKGLIVM
DE
RHK
QNA
RHKYFW
RHK
A





A1
preferred
YFW
STCLIVM

1°Anchor

A
YFW

PG
G
YFW

1°Anchor



10-mer



DEAS






Y



deleterious
RHK
RHKDEPY


P
G

PRHK
QN





FW





A2.1
preferred
YFW

1°Anchor

YFW
STC
YFW

A
P

1°Anchor



9-mer


LMIVQAT






VLIMAT



deleterious
DEP

DERKH


RKH
DERKH





A2.1
preferred
AYFW

1°Anchor

LVIM
G

G

FYWL


1°Anchor



10-mer


LMIVQAT





VIM

VLIMAT



deleterious
DEP

DE
RKHA
P

RKH
DERKH
RKH





A3
preferred
RHK

1°Anchor

YFW
PRHKYFW
A
YFW

P

1°Anchor






LMVISATF






KYRHFA





CGD



deleterious
DEP

DE





A11
preferred
A

1°Anchor

YFW
YFW
A
YFW
YFW
P

1°Anchor






VTLMISAG






KRYH





NCDF



deleterious
DEP





A
G





A24
preferred
YFWRHK

1°Anchor


STC


YFW
YFW

1°Anchor



9-mer


YFWM






FLIW



deleterious
DEG

DE
G
QNP
DERHK
G
AQN





A24
preferred


1°Anchor


P
YFWP

P



1°Anchor



10-mer


YFWM







FLIW



deleterious


GDE
QN
RHK
DE
A
QN
DEA





A3101
preferred
RHK

1°Anchor

YFW
P

YFW
YFW
AP

1°Anchor






MVTALIS






RK



deleterious
DEP

DE

ADE
DE
DE
DE





A3301
preferred


1°Anchor

YFW



AYFW


1°Anchor






MVALFIST






RK



deleterious
GP

DE





A6801
preferred
YFWSTC

1°Anchor



YFWLIVM

YFW
P

1°Anchor






AVTMSLI






RK



deleterious
GP

DEG

RHK


A





B0702
preferred
RHKFWY

1°Anchor

RHK

RHK
RHK
RHK
PA

1°Anchor






P






LMFWYAIV



deleterious
DEQNP

DEP
DE
DE
GDE
QN
DE





B3501
preferred
FWYLIVM

1°Anchor

FWY



FWY


1°Anchor






P






LMFWYIVA



deleterious
AGP



G
G





B51
preferred
LIVMFWY

1°Anchor

FWY
STC
FWY

G
FWY

1°Anchor






P






LIVFWYAM



deleterious
AGPDERH



DE
G
DEQN
GDE




KSTC





B5301
preferred
LIVMFWY

1°Anchor

FWY
STC
FWY

LIVMFWY
FWY

1°Anchor






P






IMFWYALV



deleterious
AGPQN




G
RHKQN
DE





B5401
preferred
FWY

1°Anchor

FWYL

LIVM

ALIVM
FWYAP

1°Anchor






P
IVM





ATIVLMFWY



deleterious
GPQNDE

GDES

RHKDE
DE
QNDGE
DE






TC





Italicized residues indicate less preferred or “tolerated” residues. The information in this Table is specific for 9-mers unless otherwise specified.













TABLE V







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A1, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
41
GLEGFSIAF
45.000
Portion



2
635
DSEIQMAKF
27.000
of SEQ ID


3
490
YTDFGPSTR
12.500
NO: 3;


4
371
NLDLLLQTK
10.000
each


5
583
TSAPGSRDY
7.500
start


6
514
KSEHHLLPS
6.750
position


7
231
GPDPNPFGG
6.250
is


8
22
YHDLGPMIY
6.250
specified,


9
602
DIDSTDHDR
5.000
the


10
541
LVDFVVTHF
5.000
length of


11
213
FGEVSLVSR
4.500
each


12
36
TLELTGLEG
4.500
peptide


13
249
LMLPSCLWF
2.500
is 9


14
269
TASAAGLLY
2.500
amino


15
132
VLVVLRIWY
2.500
acids,


16
431
AIWPFRFGY
2.500
the end


17
24
DLGPMIYYF
2.000
position


18
611
WCEYIMYRG
1.800
for each


19
466
ESDASKPYM
1.500
peptide


20
388
KSEKYMKLF
1.350
is the


21
315
TMTIAMIFY
1.250
start


22
314
KTMTIAMIF
1.250
position


23
645
IPDDPTNYR
1.250
plus


24
562
AIAVSKLLK
1.000
eight


25
413
KAYERKLGK
1.000


26
54
FLTITPFWK
1.000


27
9
LLESLLGCV
0.900


28
324
LLEIFFCAW
0.900


29
551
NHEDDLDRK
0.900


30
630
HAELSDSEI
0.900


31
159
LSAIATLDR
0.750


32
141
TSLNPIWSY
0.750


33
348
RSDVLLGTM
0.750


34
112
WSGSHLQRY
0.750


35
633
LSDSEIQMA
0.750


36
573
SNQVIFLGY
0.625


37
358
LIIGLNMLF
0.500


38
49
FLSPIFLTI
0.500


39
429
SAAIWPFRF
0.500


40
644
RIPDDPTNY
0.500


41
407
GLGLRHKAY
0.500


42
482
WLGEKLGFY
0.500


43
614
YIMYRGLIR
0.500


44
76
ITIGSIASF
0.500


45
199
GAAFGSLVF
0.500


46
594
LTEHGNVKD
0.450


47
524
EGEIAPAIT
0.450


48
522
SPEGEIAPA
0.450


49
559
KLQAIAVSK
0.400


50
463
TILESDASK
0.400
















TABLE VI







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A1, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
633
LSDSEIQMAK
75.000
Portion



2
605
STDHDRWCEY
62.500
of SEQ ID


3
490
YTDFGPSTRY
62.500
NO: 3;


4
464
ILESDASKPY
45.000
each


5
635
DSEIQMAKFR
13.500
start


6
440
DNEGWSSLER
11.250
position


7
659
VIDHREVSEK
10.000
is


8
36
TLELTGLEGF
9.000
specified,


9
22
YHDLGPMIYY
6.250
the


10
268
GTASAAGLLY
6.250
length of


11
314
KTMTIAMIFY
6.250
each


12
572
SSNQVIFLGY
3.750
peptide


13
171
DGDCSKPEEK
2.500
is 10


14
430
AAIWPFRFGY
2.500
amino


15
131
IVLVVLRIWY
2.500
acids,


16
458
GADFITILES
2.500
the end


17
662
HREVSEKIHF
2.250
position


18
594
LTEHGNVKDI
2.250
for each


19
41
GLEGFSIAFL
1.800
peptide


20
324
LLEIFFCAWC
1.800
is the


21
466
ESDASKPYMG
1.500
start


22
665
VSEKIHFNPR
1.350
position


23
140
YTSLNPIWSY
1.250
plus nine


24
309
GTNPGKTMTI
1.250


25
582
ITSAPGSRDY
1.250


26
231
GPDPNPFGGA
1.250


27
524
EGEIAPAITL
1.125


28
182
TGEVATGMAS
1.125


29
454
LNETGADFIT
1.125


30
57
ITPFWKLVNK
1.000


31
505
MALSRYPIVK
1.000


32
561
QAIAVSKLLK
1.000


33
462
ITILESDASK
1.000


34
9
LLESLLGCVS
0.900


35
630
HAELSDSEIQ
0.900


36
611
WCEYIMYRGL
0.900


37
428
VSAAIWPFRF
0.750


38
348
RSDVLLGTMM
0.750


39
388
KSEKYMKLFL
0.675


40
329
FCAWCTAFKF
0.500


41
541
LVDFVVTHFG
0.500


42
158
TLSAIATLDR
0.500


43
531
ITLTVNISGK
0.500


44
320
MIFYLLEIFF
0.500


45
13
LLGCVSWSLY
0.500


46
371
NLDLLLQTKN
0.500


47
56
TITPFWKLVN
0.500


48
526
EIAPAITLTV
0.500


49
383
KVLFRKSEKY
0.500


50
357
MLIIGLNMLF
0.500
















TABLE VII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A2, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
323
YLLEIFFCA
3820.380
Portion



2
62
KLVNKKWML
560.763
of SEQ ID


3
204
SLVFLTHWV
382.536
NO: 3;


4
126
FILGQIVLV
374.369
each


5
277
YLHTWAAAV
319.939
start


6
8
ILLESLLGC
294.675
position


7
13
LLGCVSWSL
272.371
is


8
92
RLMVLALGV
257.342
specified,


9
211
WVFGEVSLV
238.235
the


10
275
LLYLHTWAA
202.694
length of


11
615
IMYRGLIRL
193.040
each


12
254
CLWFRGTGL
177.308
peptide


13
392
YMKLFLWLL
162.824
is 9


14
351
VLLGTMMLI
150.931
amino


15
364
MLFGPKKNL
134.369
acids,


16
241
VLLCLASGL
134.369
the end


17
127
ILGQIVLVV
111.499
position


18
398
WLLVGVGLL
108.713
for each


19
133
LVVLRIWYT
105.168
peptide


20
274
GLLYLHTWA
101.099
is the


21
49
FLSPIFLTI
91.183
start


22
188
GMASRPNWL
84.856
position


23
357
MLIIGLNML
83.527
plus


24
56
TITPFWKLV
61.780
eight


25
258
RGTGLIWWV
43.075


26
316
MTIAMIFYL
37.007


27
68
WMLTLLRII
24.186


28
356
MMLIIGLNM
22.569


29
216
VSLVSRWAV
21.418


30
28
MIYYFPLQT
21.182


31
120
YLRIWGFIL
17.760


32
319
AMIFYLLEI
17.330


33
261
GLIWWVTGT
17.140


34
352
LLGTMMLII
16.725


35
473
YMGNNDLTM
16.505


36
149
YQMSNKVIL
15.114


37
200
AAFGSLVFL
13.887


38
90
KLRLMVLAL
13.070


39
504
IMALSRYPI
12.809


40
156
ILTLSAIAT
12.668


41
150
QMSNKVILT
12.379


42
284
AVSGCVFAI
12.178


43
376
LQTKNSSKV
11.988


44
97
ALGVSSSLI
10.433


45
47
IAFLSPIFL
10.264


46
540
KLVDFVVTH
9.346


47
42
LEGFSIAFL
8.933


48
560
LQAIAVSKL
8.469


49
34
LQTLELTGL
8.469


50
154
KVILTLSAI
7.349
















TABLE VIII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A2, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
482
WLGEKLGFYT
4483.377
Portion



2
394
KLFLWLLVGV
2071.606
of SEQ ID


3
54
FLTITPFWKL
1400.305
NO: 3;


4
132
VLVVLRIWYT
1201.914
each


5
315
TMTIAMIFYL
1131.982
start


6
567
KLLKSSSNQV
900.698
position


7
396
FLWLLVGVGL
815.616
is


8
12
SLLGCVSWSL
592.807
specified,


9
8
ILLESLLGCV
536.309
the


10
356
MMLIIGLNML
223.203
length of


11
453
LLNETGADFI
195.971
each


12
559
KLQAIAVSKL
171.967
peptide


13
384
VLFRKSEKYM
171.868
is 10


14
126
FILGQIVLVV
153.491
amino


15
274
GLLYLHTWAA
137.862
acids,


16
49
FLSPIFLTIT
122.836
the end


17
375
LLQTKNSSKV
118.238
position


18
188
GMASRPNWLL
115.713
for each


19
614
YIMYRGLIRL
114.985
peptide


20
330
CAWCTAFKFV
83.786
is the


21
399
LLVGVGLLGL
83.527
start


22
156
ILTLSAIATL
83.527
position


23
207
FLTHWVFGEV
79.025
plus nine


24
351
VLLGTMMLII
61.882


25
536
NISGKLVDFV
59.279


26
363
NMLFGPKKNL
57.085


27
504
IMALSRYPIV
52.518


28
275
LLYLHTWAAA
45.944


29
62
KLVNKKWMLT
44.339


30
591
YLQLTEHGNV
41.592


31
69
MLTLLRIITI
40.792


32
296
SMWPQTLGHL
38.289


33
68
WMLTLLRIIT
37.557


34
323
YLLEIFFCAW
37.545


35
28
MIYYFPLQTL
36.752


36
242
LLCLASGLML
36.316


37
95
VLALGVSSSL
36.316


38
150
QMSNKVILTL
35.485


39
127
ILGQIVLVVL
34.246


40
20
SLYHDLGPMI
33.385


41
149
YQMSNKVILT
29.577


42
97
ALGVSSSLIV
28.516


43
137
RIWYTSLNPI
27.385


44
342
GVYARERSDV
19.475


45
134
VVLRIWYTSL
17.636


46
41
GLEGFSIAFL
17.295


47
46
SIAFLSPIFL
16.155


48
619
GLIRLGYARI
15.649


49
392
YMKLFLWLLV
13.748


50
355
TMMLIIGLNM
13.276
















TABLE IX







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A3, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
506
ALSRYPIVK
120.000
Portion



2
418
KLGKVAPTK
90.000
of SEQ ID


3
559
KLQAIAVSK
90.000
NO: 3;


4
54
FLTITPFWK
60.000
each


5
619
GLIRLGYAR
54.000
start


6
361
GLNMLFGPK
54.000
position


7
41
GLEGFSIAF
54.000
is


8
409
GLRHKAYER
36.000
specified,


9
371
NLDLLLQTK
30.000
the


10
532
TLTVNISGK
30.000
length of


11
593
QLTEHGNVK
30.000
each


12
431
AIWPFRFGY
27.000
peptide


13
384
VLFRKSEKY
20.000
is 9


14
375
LLQTKNSSK
20.000
amino


15
250
MLPSCLWFR
18.000
acids,


16
315
TMTIAMIFY
12.000
the end


17
132
VLVVLRIWY
12.000
position


18
90
KLRLMVLAL
10.800
for each


19
413
KAYERKLGK
9.000
peptide


20
615
IMYRGLIRL
9.000
is the


21
249
LMLPSCLWF
9.000
start


22
383
KVLFRKSEK
9.000
position


23
392
YMKLFLWLL
8.100
plus


24
319
AMIFYLLEI
8.100
eight


25
540
KLVDFVVTH
8.100


26
478
DLTMWLGEK
8.100


27
62
KLVNKKWML
8.100


28
49
FLSPIFLTI
8.100


29
323
YLLEIFFCA
6.075


30
407
GLGLRHKAY
6.000


31
338
FVPGGVYAR
5.400


32
120
YLRIWGFIL
5.400


33
463
TILESDASK
4.500


34
24
DLGPMIYYF
4.050


35
351
VLLGTMMLI
4.050


36
261
GLIWWVTGT
4.050


37
562
AIAVSKLLK
4.000


38
364
MLFGPKKNL
3.375


39
275
LLYLHTWAA
3.000


40
453
LLNETGADF
3.000


41
205
LVFLTHWVF
3.000


42
405
LLGLGLRHK
3.000


43
296
SMWPQTLGH
3.000


44
254
CLWFRGTGL
3.000


45
691
HMNTPKYFL
2.700


46
404
GLLGLGLRH
2.700


47
482
WLGEKLGFY
2.700


48
13
LLGCVSWSL
2.700


49
394
KLFLWLLVG
2.700


50
188
GMASRPNWL
1.800
















TABLE X







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A3, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
361
GLNMLFGPKK
180.000
Portion



2
409
GLRHKAYERK
60.000
of SEQ ID


3
540
KLVDFVVTHF
40.500
NO: 3;


4
249
LMLPSCLWFR
40.500
each


5
374
LLLQTKNSSK
30.000
start


6
404
GLLGLGLRHK
20.250
position


7
480
TMWLGEKLGF
20.000
is


8
248
GLMLPSCLWF
18.000
specified,


9
204
SLVFLTHWVF
9.000
the


10
188
GMASRPNWLL
8.100
length of


11
54
FLTITPFWKL
8.100
each


12
158
TLSAIATLDR
8.000
peptide


13
12
SLLGCVSWSL
6.075
is 10


14
659
VIDHREVSEK
6.000
amino


15
357
MLIIGLNMLF
6.000
acids,


16
559
KLQAIAVSKL
5.400
the end


17
394
KLFLWLLVGV
4.500
position


18
396
FLWLLVGVGL
4.500
for each


19
319
AMIFYLLEIF
4.500
peptide


20
351
VLLGTMMLII
4.050
is the


21
323
YLLEIFFCAW
4.050
start


22
399
LLVGVGLLGL
4.050
position


23
41
GLEGFSIAFL
4.050
plus nine


24
13
LLGCVSWSLY
4.000


25
20
SLYHDLGPMI
3.000


26
452
HLLNETGADF
3.000


27
500
HTWGIMALSR
3.000


28
36
TLELTGLEGF
3.000


29
58
TPFWKLVNKK
3.000


30
150
QMSNKVILTL
2.700


31
619
GLIRLGYARI
2.700


32
315
TMTIAMIFYL
2.700


33
142
SLNPIWSYQM
2.700


34
274
GLLYLHTWAA
2.700


35
314
KTMTIAMIFY
2.700


36
531
ITLTVNISGK
2.250


37
296
SMWPQTLGHL
2.025


38
167
RIGTDGDCSK
2.000


39
464
ILESDASKPY
2.000


40
320
MIFYLLEIFF
2.000


41
305
LINSGTNPGK
2.000


42
383
KVLFRKSEKY
1.800


43
505
MALSRYPIVK
1.800


44
69
MLTLLRIITI
1.800


45
145
PIWSYQMSNK
1.500


46
57
ITPFWKLVNK
1.500


47
462
ITILESDASK
1.500


48
127
ILGQIVLVVL
1.350


49
284
AVSGCVFAIF
1.350


50
140
YTSLNPIWSY
1.350
















TABLE XI







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A11, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
383
KVLFRKSEK
9.000
Portion



2
413
KAYERKLGK
2.400
of SEQ ID


3
54
FLTITPFWK
1.200
NO: 3;


4
418
KLGKVAPTK
1.200
each


5
361
GLNMLFGPK
1.200
start


6
559
KLQAIAVSK
1.200
position


7
506
ALSRYPIVK
0.800
is


8
562
AIAVSKLLK
0.800
specified,


9
338
FVPGGVYAR
0.800
the


10
619
GLIRLGYAR
0.720
length of


11
463
TILESDASK
0.600
each


12
129
GQIVLVVLR
0.540
peptide


13
409
GLRHKAYER
0.480
is 9


14
371
NLDLLLQTK
0.400
amino


15
532
TLTVNISGK
0.400
acids,


16
375
LLQTKNSSK
0.400
the end


17
58
TPFWKLVNK
0.400
position


18
593
QLTEHGNVK
0.400
for each


19
614
YIMYRGLIR
0.320
peptide


20
329
FCAWCTAFK
0.200
is the


21
490
YTDFGPSTR
0.200
start


22
250
MLPSCLWFR
0.160
position


23
314
KTMTIAMIF
0.120
plus


24
402
GVGLLGLGL
0.120
eight


25
655
NQKVVIDHR
0.120


26
478
DLTMWLGEK
0.120


27
354
GTMMLIIGL
0.120


28
184
EVATGMASR
0.120


29
649
PTNYRDNQK
0.100


30
154
KVILTLSAI
0.090


31
581
YITSAPGSR
0.080


32
362
LNMLFGPKK
0.080


33
205
LVFLTHWVF
0.080


34
380
NSSKVLFRK
0.060


35
172
GDCSKPEEK
0.060


36
284
AVSGCVFAI
0.060


37
84
FQAPNAKLR
0.060


38
173
DCSKPEEKK
0.060


39
550
GNHEDDLDR
0.048


40
66
KKWMLTLLR
0.048


41
379
KNSSKVLFR
0.048


42
92
RLMVLALGV
0.048


43
391
KYMKLFLWL
0.048


44
316
MTIAMIFYL
0.045


45
688
HHFHMNTPK
0.040


46
386
FRKSEKYMK
0.040


47
671
FNPRFGSYK
0.040


48
405
LLGLGLRHK
0.040


49
400
LVGVGLLGL
0.040


50
306
INSGTNPGK
0.040
















TABLE XII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A11, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
462
ITILESDASK
1.500
Portion



2
531
ITLTVNISGK
1.500
of SEQ ID


3
409
GLRHKAYERK
1.200
NO: 3;


4
361
GLNMLFGPKK
1.200
each


5
402
GVGLLGLGLR
1.200
start


6
167
RIGTDGDCSK
1.200
position


7
57
ITPFWKLVNK
1.000
is


8
53
IFLTITPFWK
0.900
specified,


9
592
LQLTEHGNVK
0.900
the


10
500
HTWGIMALSR
0.800
length of


11
374
LLLQTKNSSK
0.600
each


12
561
QAIAVSKLLK
0.600
peptide


13
505
MALSRYPIVK
0.600
is 10


14
305
LINSGTNPGK
0.400
amino


15
58
TPFWKLVNKK
0.400
acids,


16
659
VIDHREVSEK
0.400
the end


17
385
LFRKSEKYMK
0.400
position


18
379
KNSSKVLFRK
0.360
for each


19
337
KFVPGGVYAR
0.360
peptide


20
580
GYITSAPGSR
0.360
is the


21
249
LMLPSCLWFR
0.240
start


22
644
RIPDDPTNYR
0.240
position


23
670
HFNPRFGSYK
0.200
plus nine


24
328
FFCAWCTAFK
0.200


25
81
IASFQAPNAK
0.200


26
370
KNLDLLLQTK
0.180


27
404
GLLGLGLRHK
0.180


28
158
TLSAIATLDR
0.160


29
550
GNHEDDLDRK
0.120


30
342
GVYARERSDV
0.120


31
427
EVSAAIWPFR
0.120


32
314
KTMTIAMIFY
0.120


33
558
RKLQAIAVSK
0.090


34
417
RKLGKVAPTK
0.090


35
383
KVLFRKSEKY
0.090


36
154
KVILTLSAIA
0.090


37
145
PIWSYQMSNK
0.080


38
489
FYTDFGPSTR
0.080


39
613
EYIMYRGLIR
0.072


40
172
GDCSKPEEKK
0.060


41
268
GTASAAGLLY
0.060


42
421
KVAPTKEVSA
0.060


43
131
IVLVVLRIWY
0.060


44
648
DPTNYRDNQK
0.060


45
99
GVSSSLIVQA
0.060


46
309
GTNPGKTMTI
0.060


47
129
GQIVLVVLRI
0.054


48
119
RYLRIWGFIL
0.054


49
183
GEVATGMASR
0.054


50
248
GLMLPSCLWF
0.048
















TABLE XIII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A24, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
391
KYMKLFLWL
864.000
Portion



2
29
IYYFPLQTL
240.000
of SEQ ID


3
119
RYLRIWGFI
210.000
NO: 3;


4
148
SYQMSNKVI
75.000
each


5
613
EYIMYRGLI
75.000
start


6
21
LYHDLGPMI
72.000
position


7
31
YFPLQTLEL
33.000
is


8
83
SFQAPNAKL
33.000
specified,


9
125
GFILGQIVL
30.000
the


10
548
HFGNHEDDL
20.000
length of


11
62
KLVNKKWML
12.000
each


12
321
IFYLLEIFF
12.000
peptide


13
328
FFCAWCTAF
10.000
is 9


14
498
RYHTWGIMA
10.000
amino


15
471
KPYMGNNDL
9.600
acids,


16
533
LTVNISGKL
9.240
the end


17
475
GNNDLTMWL
8.640
position


18
314
KTMTIAMIF
8.400
for each


19
96
LALGVSSSL
8.400
peptide


20
151
MSNKVILTL
8.400
is the


21
397
LWLLVGVGL
8.400
start


22
561
QAIAVSKLL
8.400
position


23
128
LGQIVLVVL
8.400
plus


24
414
AYERKLGKV
8.250
eight


25
90
KLRLMVLAL
8.000


26
55
LTITPFWKL
7.920


27
479
LTMWLGEKL
7.920


28
276
LYLHTWAAA
7.500


29
580
GYITSAPGS
7.500


30
322
FYLLEIFFC
7.500


31
247
SGLMLPSCL
7.200


32
445
SSLERSAHL
7.200


33
357
MLIIGLNML
7.200


34
241
VLLCLASGL
7.200


35
354
GTMMLIIGL
7.200


36
317
TIAMIFYLL
6.720


37
85
QAPNAKLRL
6.000


38
388
KSEKYMKLF
6.000


39
584
SAPGSRDYL
6.000


40
243
LCLASGLML
6.000


41
149
YQMSNKVIL
6.000


42
26
GPMIYYFPL
6.000


43
297
MWPQTLGHL
6.000


44
366
FGPKKNLDL
6.000


45
157
LTLSAIATL
6.000


46
350
DVLLGTMML
6.000


47
489
FYTDFGPST
6.000


48
691
HMNTPKYFL
6.000


49
210
HWVFGEVSL
6.000


50
139
WYTSLNPIW
6.000
















TABLE XIV







HLA PEPTIDE SCORING RESULTS - 125P5C8 - A24, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
391
KYMKLFLWLL
600.000
Portion



2
119
RYLRIWGFIL
600.000
of SEQ ID


3
498
RYHTWGIMAL
400.000
NO: 3;


4
148
SYQMSNKVIL
300.000
each


5
30
YYFPLQTLEL
264.000
start


6
624
GYARISHAEL
220.000
position


7
343
VYARERSDVL
200.000
is


8
438
GYDNEGWSSL
200.000
specified,


9
651
NYRDNQKVVI
60.000
the


10
683
NYENNHHFHM
37.500
length of


11
365
LFGPKKNLDL
24.000
each


12
559
KLQAIAVSKL
13.200
peptide


13
322
FYLLEIFFCA
12.600
is 10


14
48
AFLSPIFLTI
12.600
amino


15
388
KSEKYMKLFL
12.000
acids,


16
316
MTIAMIFYLL
10.080
the end


17
540
KLVDFVVTHF
10.080
position


18
689
HFHMNTPKYF
10.000
for each


19
327
IFFCAWCTAF
10.000
peptide


20
414
AYERKLGKVA
9.000
is the


21
12
SLLGCVSWSL
8.400
start


22
570
KSSSNQVIFL
8.000
position


23
590
DYLQLTEHGN
7.500
plus nine


24
276
LYLHTWAAAV
7.500


25
401
VGVGLLGLGL
7.200


26
445
SSLERSAHLL
7.200


27
474
MGNNDLTMWL
7.200


28
187
TGMASRPNWL
7.200


29
233
DPNPFGGAVL
7.200


30
240
AVLLCLASGL
7.200


31
356
MMLIIGLNML
7.200


32
616
MYRGLIRLGY
7.000


33
677
SYKEGHNYEN
6.600


34
532
TLTVNISGKL
6.160


35
614
YIMYRGLIRL
6.000


36
611
WCEYIMYRGL
6.000


37
363
NMLFGPKKNL
6.000


38
510
YPIVKSEHHL
6.000


39
63
LVNKKWMLTL
6.000


40
21
LYHDLGPMIY
6.000


41
399
LLVGVGLLGL
6.000


42
397
LWLLVGVGLL
6.000


43
134
VVLRIWYTSL
6.000


44
366
FGPKKNLDLL
6.000


45
524
EGEIAPAITL
6.000


46
25
LGPMIYYFPL
6.000


47
253
SCLWFRGTGL
6.000


48
41
GLEGFSIAFL
6.000


49
4
LWREILLESL
5.760


50
127
ILGQIVLVVL
5.600
















TABLE XV







HLA PEPTIDE SCORING RESULTS - 125P5C8 - B7, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
26
GPMIYYFPL
240.000
Portion



2
344
YARERSDVL
120.000
of SEQ ID


3
625
YARISHAEL
120.000
NO: 3;


4
367
GPKKNLDLL
80.000
each


5
235
NPFGGAVLL
80.000
start


6
471
KPYMGNNDL
80.000
position


7
86
APNAKLRLM
60.000
is


8
90
KLRLMVLAL
40.000
specified,


9
120
YLRIWGFIL
40.000
the


10
135
VLRIWYTSL
40.000
length of


11
200
AAFGSLVFL
36.000
each


12
402
GVGLLGLGL
20.000
peptide


13
350
DVLLGTMML
20.000
is 9


14
400
LVGVGLLGL
20.000
amino


15
512
IVKSEHHLL
20.000
acids,


16
189
MASRPNWLL
18.000
the end


17
584
SAPGSRDYL
18.000
position


18
270
ASAAGLLYL
12.000
for each


19
85
QAPNAKLRL
12.000
peptide


20
197
LAGAAFGSL
12.000
is the


21
47
IAFLSPIFL
12.000
start


22
561
QAIAVSKLL
12.000
position


23
354
GTMMLIIGL
12.000
plus


24
294
TASMWPQTL
12.000
eight


25
149
YQMSNKVIL
12.000


26
479
LTMWLGEKL
12.000


27
96
LALGVSSSL
12.000


28
88
NAKLRLMVL
12.000


29
298
WPQTLGHLI
8.000


30
55
LTITPFWKL
6.000


31
691
HMNTPKYFL
6.000


32
1
MTSLWREIL
6.000


33
364
MLFGPKKNL
6.000


34
284
AVSGCVFAI
6.000


35
423
APTKEVSAA
6.000


36
64
VNKKWMLTL
4.000


37
392
YMKLFLWLL
4.000


38
254
CLWFRGTGL
4.000


39
2
TSLWREILL
4.000


40
366
FGPKKNLDL
4.000


41
571
SSSNQVIFL
4.000


42
151
MSNKVILTL
4.000


43
109
VTWWSGSHL
4.000


44
357
MLIIGLNML
4.000


45
620
LIRLGYARI
4.000


46
237
FGGAVLLCL
4.000


47
34
LQTLELTGL
4.000


48
128
LGQIVLVVL
4.000


49
268
GTASAAGLL
4.000


50
316
MTIAMIFYL
4.000
















TABLE XVI







HLA PEPTIDE SCORING RESULTS - 125P5C8 - B7, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
585
APGSRDYLQL
240.000
Portion



2
344
YARERSDVLL
120.000
of SEQ ID


3
510
YPIVKSEHHL
80.000
NO: 3;


4
367
GPKKNLDLLL
80.000
each


5
233
DPNPFGGAVL
80.000
start


6
108
AVTWWSGSHL
60.000
position


7
240
AVLLCLASGL
60.000
is


8
528
APAITLTVNI
24.000
specified,


9
423
APTKEVSAAI
24.000
the


10
134
VVLRIWYTSL
20.000
length of


11
311
NPGKTMTIAM
20.000
each


12
16
CVSWSLYHDL
20.000
peptide


13
63
LVNKKWMLTL
20.000
is 10


14
86
APNAKLRLMV
18.000
amino


15
82
ASFQAPNAKL
18.000
acids,


16
187
TGMASRPNWL
12.000
the end


17
246
ASGLMLPSCL
12.000
position


18
269
TASAAGLLYL
12.000
for each


19
199
GAAFGSLVFL
12.000
peptide


20
614
YIMYRGLIRL
12.000
is the


21
496
STRYHTWGIM
10.000
start


22
188
GMASRPNWLL
6.000
position


23
363
NMLFGPKKNL
6.000
plus nine


24
28
MIYYFPLQTL
6.000


25
583
TSAPGSRDYL
6.000


26
54
FLTITPFWKL
6.000


27
288
CVFAIFTASM
5.000


28
560
LQAIAVSKLL
4.000


29
353
LGTMMLIIGL
4.000


30
4
LWREILLESL
4.000


31
1
MTSLWREILL
4.000


32
84
FQAPNAKLRL
4.000


33
253
SCLWFRGTGL
4.000


34
242
LLCLASGLML
4.000


35
570
KSSSNQVIFL
4.000


36
399
LLVGVGLLGL
4.000


37
293
FTASMWPQTL
4.000


38
396
FLWLLVGVGL
4.000


39
127
ILGQIVLVVL
4.000


40
266
VTGTASAAGL
4.000


41
12
SLLGCVSWSL
4.000


42
532
TLTVNISGKL
4.000


43
376
LQTKNSSKVL
4.000


44
150
QMSNKVILTL
4.000


45
124
WGFILGQIVL
4.000


46
46
SIAFLSPIFL
4.000


47
366
FGPKKNLDLL
4.000


48
474
MGNNDLTMWL
4.000


49
112
WSGSHLQRYL
4.000


50
315
TMTIAMIFYL
4.000
















TABLE XVII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - B35, 9-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
367
GPKKNLDLL
60.000
Portion



2
86
APNAKLRLM
40.000
of SEQ ID


3
471
KPYMGNNDL
40.000
NO: 3;


4
229
HPGPDPNPF
30.000
each


5
26
GPMIYYFPL
20.000
start


6
235
NPFGGAVLL
20.000
position


7
344
YARERSDVL
18.000
is


8
676
GSYKEGHNY
15.000
specified,


9
644
RIPDDPTNY
12.000
the


10
112
WSGSHLQRY
10.000
length of


11
445
SSLERSAHL
10.000
each


12
494
GPSTRYHTW
10.000
peptide


13
583
TSAPGSRDY
10.000
is 9


14
141
TSLNPIWSY
10.000
amino


15
570
KSSSNQVIF
10.000
acids,


16
88
NAKLRLMVL
9.000
the end


17
625
YARISHAEL
9.000
position


18
298
WPQTLGHLI
8.000
for each


19
181
KTGEVATGM
8.000
peptide


20
90
KLRLMVLAL
6.000
is the


21
348
RSDVLLGTM
6.000
start


22
269
TASAAGLLY
6.000
position


23
17
VSWSLYHDL
5.000
plus


24
2
TSLWREILL
5.000
eight


25
270
ASAAGLLYL
5.000


26
571
SSSNQVIFL
5.000


27
285
VSGCVFAIF
5.000


28
151
MSNKVILTL
5.000


29
512
IVKSEHHLL
4.500


30
192
RPNWLLAGA
4.000


31
632
ELSDSEIQM
4.000


32
233
DPNPFGGAV
4.000


33
482
WLGEKLGFY
4.000


34
197
LAGAAFGSL
3.000


35
96
LALGVSSSL
3.000


36
330
CAWCTAFKF
3.000


37
520
LPSPEGEIA
3.000


38
423
APTKEVSAA
3.000


39
120
YLRIWGFIL
3.000


40
466
ESDASKPYM
3.000


41
64
VNKKWMLTL
3.000


42
85
QAPNAKLRL
3.000


43
392
YMKLFLWLL
3.000


44
561
QAIAVSKLL
3.000


45
587
GSRDYLQLT
3.000


46
377
QTKNSSKVL
3.000


47
388
KSEKYMKLF
3.000


48
294
TASMWPQTL
3.000


49
135
VLRIWYTSL
3.000


50
199
GAAFGSLVF
3.000
















TABLE XVIII







HLA PEPTIDE SCORING RESULTS - 125P5C8 - B35, 10-MERS














SCORE (ESTIMATE OF HALF TIME OF




START
SUBSEQUENCE
DISASSOCIATION OF A MOLECULE


RANK
POSITION
RESIDUE LISTING
CONTAINING THIS SUBSEQUENCE)















1
367
GPKKNLDLLL
60.000
Portion



2
311
NPGKTMTIAM
40.000
of SEQ ID


3
585
APGSRDYLQL
30.000
NO: 3;


4
233
DPNPFGGAVL
20.000
each


5
510
YPIVKSEHHL
20.000
start


6
51
SPIFLTITPF
20.000
position


7
344
YARERSDVLL
18.000
is


8
19
WSLYHDLGPM
15.000
specified,


9
307
NSGTNPGKTM
10.000
the


10
445
SSLERSAHLL
10.000
length of


11
572
SSNQVIFLGY
10.000
each


12
570
KSSSNQVIFL
10.000
peptide


13
528
APAITLTVNI
8.000
is 10


14
423
APTKEVSAAI
8.000
amino


15
430
AAIWPFRFGY
6.000
acids,


16
496
STRYHTWGIM
6.000
the end


17
348
RSDVLLGTMM
6.000
position


18
85
QAPNAKLRLM
6.000
for each


19
428
VSAAIWPFRF
5.000
peptide


20
45
FSIAFLSPIF
5.000
is the


21
246
ASGLMLPSCL
5.000
start


22
583
TSAPGSRDYL
5.000
position


23
444
WSSLERSAHL
5.000
plus nine


24
112
WSGSHLQRYL
5.000


25
82
ASFQAPNAKL
5.000


26
176
KPEEKKTGEV
4.800


27
383
KVLFRKSEKY
4.000


28
471
KPYMGNNDLT
4.000


29
314
KTMTIAMIFY
4.000


30
540
KLVDFVVTHF
4.000


31
86
APNAKLRLMV
4.000


32
192
RPNWLLAGAA
4.000


33
281
WAAAVSGCVF
3.000


34
117
LQRYLRIWGF
3.000


35
377
QTKNSSKVLF
3.000


36
199
GAAFGSLVFL
3.000


37
388
KSEKYMKLFL
3.000


38
64
VNKKWMLTLL
3.000


39
269
TASAAGLLYL
3.000


40
675
FGSYKEGHNY
3.000


41
102
SSLIVQAVTW
2.500


42
522
SPEGEIAPAI
2.400


43
413
KAYERKLGKV
2.400


44
131
IVLVVLRIWY
2.000


45
235
NPFGGAVLLC
2.000


46
355
TMMLIIGLNM
2.000


47
114
GSHLQRYLRI
2.000


48
298
WPQTLGHLIN
2.000


49
406
LGLGLRHKAY
2.000


50
582
ITSAPGSRDY
2.000

































TABLE XIX







Pos
1
2
3
4
5
6
7
8
9






score











HLA Class I nonamer analysis of 125P5C8, variant 1 (aa 1-699) Listed



are scores which correlate with the ligation strength to a defined


HLA type for a sequence of amino acids. The algorithms used are


based on the book “MHC Ligands and Peptide Motifs” by H. G.


Rammensee, J. Bachmann and S. Stevanovic. The probability of being


processed and presented is given in order to predict T-cell


epitopes.










HLA-A*0201 nonamers


























126
F
I
L
G
Q

I

V
L
V






29
Portion of SEQ ID NO: 3, each



357
M
L
I
I
G

L

N
M
L






29
start position is specified -


398
W
L
L
V
G

V

G
L
L






28
the length of each peptide is


90
K
L
R
L
M

V

L
A
L






27
9 amino acids, the end


127
I
L
G
Q
I

V

L
V
V






27
position for each peptide is


319
A
M
I
F
Y

L

L
E
I






27
the start position plus eight.


615
I
M
Y
R
G

L

I
R
L






27


9
L
L
E
S
L

L

G
C
V






26


157
L
T
L
S
A

I

A
T
L






26


200
A
A
F
G
S

L

V
F
L






25


204
S
L
V
F
L

T

H
W
V






25


241
V
L
L
C
L

A

S
G
L






25


277
Y
L
H
T
W

A

A
A
V






25


13
L
L
G
C
V

S

W
S
L






24


49
F
L
S
P
I

F

L
T
I






24


92
R
L
M
V
L

A

L
G
V






24


392
Y
M
K
L
F

L

W
L
L






24


400
L
V
G
V
G

L

L
G
L






24


68
W
M
L
T
L

L

R
I
I






23


135
V
L
R
I
W

Y

T
S
L






23


351
V
L
L
G
T

M

M
L
I






23


364
M
L
F
G
P

K

K
N
L






23


519
L
L
P
S
P

E

G
E
I






23


555
D
L
D
R
K

L

Q
A
I






23


8
I
L
L
E
S

L

L
G
C






22


62
K
L
V
N
K

K

W
M
L






22


70
L
T
L
L
R

I

I
T
I






22


96
L
A
L
G
V

S

S
S
L






22


120
Y
L
R
I
W

G

F
I
L






22


188
G
M
A
S
R

P

N
W
L






22


261
G
L
I
W
W

V

T
G
T






22


354
G
T
M
M
L

I

I
G
L






22


395
L
F
L
W
L

L

V
G
V






22


446
S
L
E
R
S

A

H
L
L






22


568
L
L
K
S
S

S

N
Q
V






22


620
L
I
R
L
G

Y

A
R
I






22


20
S
L
Y
H
D

L

G
P
M






21


55
L
T
I
T
P

F

W
K
L






21


95
V
L
A
L
G

V

S
S
S






21


130
Q
I
V
L
V

V

L
R
I






21


211
W
V
F
G
E

V

S
L
V






21


254
C
L
W
F
R

G

T
G
L






21


270
A
S
A
A
G

L

L
Y
L






21


284
A
V
S
G
C

V

F
A
I






21


317
T
I
A
M
I

F

Y
L
L






21


352
L
L
G
T
M

M

L
I
I






21


504
I
M
A
L
S

R

Y
P
I






21


527
I
A
P
A
I

T

L
T
V






21


540
K
L
V
D
F

V

V
T
H






21


691
H
M
N
T
P

K

Y
F
L






21


12
S
L
L
G
C

V

S
W
S






20


97
A
L
G
V
S

S

S
L
I






20


154
K
V
I
L
T

L

S
A
I






20


160
S
A
I
A
T

L

D
R
I






20


275
L
L
Y
L
H

T

W
A
A






20


316
M
T
I
A
M

I

F
Y
L






20


323
Y
L
L
E
I

F

F
C
A






20


479
L
T
M
W
L

G

E
K
L






20


560
L
Q
A
I
A

V

S
K
L






20


625
Y
A
R
I
S

H

A
E
L






20


29
I
Y
Y
F
P

L

Q
T
L






19


56
T
I
T
P
F

W

K
L
V






19


262
L
I
W
W
V

T

G
T
A






19


350
D
V
L
L
G

T

M
M
L






19


439
Y
D
N
E
G

W

S
S
L






19


3
S
L
W
R
E

I

L
L
E






18


73
L
R
I
I
T

I

G
S
I






18


76
I
T
I
G
S

I

A
S
F






18


103
S
L
I
V
Q

A

V
T
W






18


122
R
I
W
G
F

I

L
G
Q






18


128
L
G
Q
I
V

L

V
V
L






18


151
M
S
N
K
V

I

L
T
L






18


155
V
I
L
T
L

S

A
I
A






18


197
L
A
G
A
A

F

G
S
L






18


208
L
T
H
W
V

F

G
E
V






18


244
C
L
A
S
G

L

M
L
P






18


301
T
L
G
H
L

I

N
S
G






18


344
Y
A
R
E
R

S

D
V
L






18


402
G
V
G
L
L

G

L
G
L






18


405
L
L
G
L
G

L

R
H
K






18


414
A
Y
E
R
K

L

G
K
V






18


445
S
S
L
E
R

S

A
H
L






18


457
T
G
A
D
F

I

T
I
L






18


499
Y
H
T
W
G

I

M
A
L






18


533
L
T
V
N
I

S

G
K
L






18


584
S
A
P
G
S

R

D
Y
L






18


31
Y
F
P
L
Q

T

L
E
L






17


42
L
E
G
F
S

I

A
F
L






17


47
I
A
F
L
S

P

I
F
L






17


83
S
F
Q
A
P

N

A
K
L






17


88
N
A
K
L
R

L

M
V
L






17


101
S
S
S
L
I

V

Q
A
V






17


150
Q
M
S
N
K

V

I
L
T






17


196
L
L
A
G
A

A

F
G
S






17


235
N
P
F
G
G

A

V
L
L






17


268
G
T
A
S
A

A

G
L
L






17


274
G
L
L
Y
L

H

T
W
A






17


296
S
M
W
P
Q

T

L
G
H






17


371
N
L
D
L
L

L

Q
T
K






17


374
L
L
L
Q
T

K

N
S
S






17


394
K
L
F
L
W

L

L
V
G






17


396
F
L
W
L
L

V

G
V
G






17


397
L
W
L
L
V

G

V
G
L






17


404
G
L
L
G
L

G

L
R
H






17


453
L
L
N
E
T

G

A
D
F






17


473
Y
M
G
N
N

D

L
T
M






17


482
W
L
G
E
K

L

G
F
Y






17


505
M
A
L
S
R

Y

P
I
V






17


526
E
I
A
P
A

I

T
L
T






17


536
N
I
S
G
K

L

V
D
F






17


537
I
S
G
K
L

V

D
F
V






17


577
I
F
L
G
Y

I

T
S
A






17


657
K
V
V
I
D

H

R
E
V






17


5
W
R
E
I
L

L

E
S
L






16


24
D
L
G
P
M

I

Y
Y
F






16


34
L
Q
T
L
E

L

T
G
L






16


39
L
T
G
L
E

G

F
S
I






16


45
F
S
I
A
F

L

S
P
I






16


71
T
L
L
R
I

I

T
I
G






16


109
V
T
W
W
S

G

S
H
L






16


138
I
W
Y
T
S

L

N
P
I






16


156
I
L
T
L
S

A

I
A
T






16


195
W
L
L
A
G

A

A
F
G






16


237
F
G
G
A
V

L

L
C
L






16


240
A
V
L
L
C

L

A
S
G






16


242
L
L
C
L
A

S

G
L
M






16


258
R
G
T
G
L

I

W
W
V






16


271
S
A
A
G
L

L

Y
L
H






16


281
W
A
A
A
V

S

G
C
V






16


294
T
A
S
M
W

P

Q
T
L






16


305
L
I
N
S
G

T

N
P
G






16


463
T
I
L
E
S

D

A
S
K






16


475
G
N
N
D
L

T

M
W
L






16


496
S
T
R
Y
H

T

W
G
I






16


511
P
I
V
K
S

E

H
H
L






16


512
I
V
K
S
E

H

H
L
L






16


525
G
E
I
A
P

A

I
T
L






16


529
P
A
I
T
L

T

V
N
I






16


571
S
S
S
N
Q

V

I
F
L






16


619
G
L
I
R
L

G

Y
A
R






16


67
K
W
M
L
T

L

L
R
I






15


72
L
L
R
I
I

T

I
G
S






15


80
S
I
A
S
F

Q

A
P
N






15


85
Q
A
P
N
A

K

L
R
L






15


116
H
L
Q
R
Y

L

R
I
W






15


125
G
F
I
L
G

Q

I
V
L






15


142
S
L
N
P
I

W

S
Y
Q






15


149
Y
Q
M
S
N

K

V
I
L






15


163
A
T
L
D
R

I

G
T
D






15


189
M
A
S
R
P

N

W
L
L






15


198
A
G
A
A
F

G

S
L
V






15


207
F
L
T
H
W

V

F
G
E






15


210
H
W
V
F
G

E

V
S
L






15


243
L
C
L
A
S

G

L
M
L






15


247
S
G
L
M
L

P

S
C
L






15


249
L
M
L
P
S

C

L
W
F






15


255
L
W
F
R
G

T

G
L
I






15


310
T
N
P
G
K

T

M
T
I






15


338
F
V
P
G
G

V

Y
A
R






15


356
M
M
L
I
I

G

L
N
M






15


358
L
I
I
G
L

N

M
L
F






15


375
L
L
Q
T
K

N

S
S
K






15


384
V
L
F
R
K

S

E
K
Y






15


387
R
K
S
E
K

Y

M
K
L






15


399
L
L
V
G
V

G

L
L
G






15


409
G
L
R
H
K

A

Y
E
R






15


417
R
K
L
G
K

V

A
P
T






15


420
G
K
V
A
P

T

K
E
V






15


431
A
I
W
P
F

R

F
G
Y






15


452
H
L
L
N
E

T

G
A
D






15


471
K
P
Y
M
G

N

N
D
L






15


518
H
L
L
P
S

P

E
G
E






15


559
K
L
Q
A
I

A

V
S
K






15


561
Q
A
I
A
V

S

K
L
L






15










HLA-A1 nonamers


























22
Y

H

D
L
G
P

M

I
Y






29
Portion of SEQ ID NO: 3, each



573
S

N

Q
V
I
F

L

G
Y






26
start position is specified -


583
T

S

A
P
G
S

R

D
Y






25
the length of each peptide is


269
T

A

S
A
A
G

L

L
Y






24
9 amino acids, the end


617
Y

R

G
L
I
R

L

G
Y






24
position for each peptide is


132
V

L

V
V
L
R

I

W
Y






22
the start position plus eight.


670
H

F

N
P
R
F

G

S
Y






22


23
H

D

L
G
P
M

I

Y
Y






21


431
A

I

W
P
F
R

F

G
Y






21


594
L

T

E
H
G
N

V

K
D






21


141
T

S

L
N
P
I

W

S
Y






20


490
Y

T

D
F
G
P

S

T
R






20


514
K

S

E
H
H
L

L

P
S






20


609
D

R

W
C
E
Y

I

M
Y






20


676
G

S

Y
K
E
G

H

N
Y






20


112
W

S

G
S
H
L

Q

R
Y






19


169
G

T

D
G
D
C

S

K
P






19


315
T

M

T
I
A
M

I

F
Y






19


336
F

K

F
V
P
G

G

V
Y






19


384
V

L

F
R
K
S

E

K
Y






19


605
S

T

D
H
D
R

W

C
E






19


36
T

L

E
L
T
G

L

E
G






18


588
S

R

D
Y
L
Q

L

T
E






18


635
D

S

E
I
Q
M

A

K
F






18


14
L

G

C
V
S
W

S

L
Y






17


190
A

S

R
P
N
W

L

L
A






17


388
K

S

E
K
Y
M

K

L
F






17


491
T

D

F
G
P
S

T

R
Y






17


689
H

F

H
M
N
T

P

K
Y






17


41
G

L

E
G
F
S

I

A
F






16


407
G

L

G
L
R
H

K

A
Y






16


458
G

A

D
F
I
T

I

L
E






16


482
W

L

G
E
K
L

G

F
Y






16


49
F

L

S
P
I
F

L

T
I






15


446
S

L

E
R
S
A

H

L
L






15


465
L

E

S
D
A
S

K

P
Y






15


502
W

G

I
M
A
L

S

R
Y






15


606
T

D

H
D
R
W

C

E
Y






15


633
L

S

D
S
E
I

Q

M
A






15


644
R

I

P
D
D
P

T

N
Y






15


646
P

D

D
P
T
N

Y

R
D






15


652
Y

R

D
N
Q
K

V

V
I






15


665
V

S

E
K
I
H

F

N
P






15










HLA-A26 nonamers


























76
I
T
I
G
S
I
A
S
F






31
Portion of SEQ ID NO: 3, each



24
D
L
G
P
M
I
Y
Y
F






30
start position is specified -


427
E
V
S
A
A
I
W
P
F






30
the length of each peptide is


536
N
I
S
G
K
L
V
D
F






28
9 amino acids, the end


320
M
I
F
Y
L
L
E
I
F






27
position for each peptide is


52
P
I
F
L
T
I
T
P
F






26
the start position plus eight.


350
D
V
L
L
G
T
M
M
L






26


358
L
I
I
G
L
N
M
L
F






26


541
L
V
D
F
V
V
T
H
F






26


55
L
T
I
T
P
F
W
K
L






25


482
W
L
G
E
K
L
G
F
Y






25


632
E
L
S
D
S
E
I
Q
M






25


157
L
T
L
S
A
I
A
T
L






24


316
M
T
I
A
M
I
F
Y
L






24


354
G
T
M
M
L
I
I
G
L






24


46
S
I
A
F
L
S
P
I
F






23


184
E
V
A
T
G
M
A
S
R






23


317
T
I
A
M
I
F
Y
L
L






23


357
M
L
I
I
G
L
N
M
L






23


400
L
V
G
V
G
L
L
G
L






23


431
A
I
W
P
F
R
F
G
Y






23


526
E
I
A
P
A
I
T
L
T






23


635
D
S
E
I
Q
M
A
K
F






23


644
R
I
P
D
D
P
T
N
Y






23


664
E
V
S
E
K
I
H
F
N






23


670
H
F
N
P
R
F
G
S
Y






23


205
L
V
F
L
T
H
W
V
F






22


384
V
L
F
R
K
S
E
K
Y






22


485
E
K
L
G
F
Y
T
D
F






22


555
D
L
D
R
K
L
Q
A
I






22


41
G
L
E
G
F
S
I
A
F






21


90
K
L
R
L
M
V
L
A
L






21


109
V
T
W
W
S
G
S
H
L






21


135
V
L
R
I
W
Y
T
S
L






21


181
K
T
G
E
V
A
T
G
M






21


314
K
T
M
T
I
A
M
I
F






21


364
M
L
F
G
P
K
K
N
L






21


453
L
L
N
E
T
G
A
D
F






21


511
P
I
V
K
S
E
H
H
L






21


609
D
R
W
C
E
Y
I
M
Y






21


20
S
L
Y
H
D
L
G
P
M






20


268
G
T
A
S
A
A
G
L
L






20


377
Q
T
K
N
S
S
K
V
L






20


398
W
L
L
V
G
V
G
L
L






20


402
G
V
G
L
L
G
L
G
L






20


456
E
T
G
A
D
F
I
T
I






20


479
L
T
M
W
L
G
E
K
L






20


512
I
V
K
S
E
H
H
L
L






20


667
E
K
I
H
F
N
P
R
F






20


132
V
L
V
V
L
R
I
W
Y






19


211
W
V
F
G
E
V
S
L
V






19


241
V
L
L
C
L
A
S
G
L






19


407
G
L
G
L
R
H
K
A
Y






19


460
D
F
I
T
I
L
E
S
D






19


478
D
L
T
M
W
L
G
E
K






19


533
L
T
V
N
I
S
G
K
L






19


1
M
T
S
L
W
R
E
I
L






18


62
K
L
V
N
K
K
W
M
L






18


126
F
I
L
G
Q
I
V
L
V






18


154
K
V
I
L
T
L
S
A
I






18


200
A
A
F
G
S
L
V
F
L






18


326
E
I
F
F
C
A
W
C
T






18


338
F
V
P
G
G
V
Y
A
R






18


502
W
G
I
M
A
L
S
R
Y






18


602
D
I
D
S
T
D
H
D
R






18


689
H
F
H
M
N
T
P
K
Y






18


13
L
L
G
C
V
S
W
S
L






17


31
Y
F
P
L
Q
T
L
E
L






17


38
E
L
T
G
L
E
G
F
S






17


112
W
S
G
S
H
L
Q
R
Y






17


122
R
I
W
G
F
I
L
G
Q






17


254
C
L
W
F
R
G
T
G
L






17


284
A
V
S
G
C
V
F
A
I






17


288
C
V
F
A
I
F
T
A
S






17


289
V
F
A
I
F
T
A
S
M






17


300
Q
T
L
G
H
L
I
N
S






17


321
I
F
Y
L
L
E
I
F
F






17


328
F
F
C
A
W
C
T
A
F






17


333
C
T
A
F
K
F
V
P
G






17


387
R
K
S
E
K
Y
M
K
L






17


446
S
L
E
R
S
A
H
L
L






17


466
E
S
D
A
S
K
P
Y
M






17


500
H
T
W
G
I
M
A
L
S






17


564
A
V
S
K
L
L
K
S
S






17


637
E
I
Q
M
A
K
F
R
I






17


7
E
I
L
L
E
S
L
L
G






16


8
I
L
L
E
S
L
L
G
C






16


16
C
V
S
W
S
L
Y
H
D






16


23
H
D
L
G
P
M
I
Y
Y






16


83
S
F
Q
A
P
N
A
K
L






16


99
G
V
S
S
S
L
I
V
Q






16


120
Y
L
R
I
W
G
F
I
L






16


125
G
F
I
L
G
Q
I
V
L






16


215
E
V
S
L
V
S
R
W
A






16


240
A
V
L
L
C
L
A
S
G






16


242
L
L
C
L
A
S
G
L
M






16


373
D
L
L
L
Q
T
K
N
S






16


385
L
F
R
K
S
E
K
Y
M






16


576
V
I
F
L
G
Y
I
T
S






16


620
L
I
R
L
G
Y
A
R
I






16


12
S
L
L
G
C
V
S
W
S






15


37
L
E
L
T
G
L
E
G
F






15


95
V
L
A
L
G
V
S
S
S






15


104
L
I
V
Q
A
V
T
W
W






15


118
Q
R
Y
L
R
I
W
G
F






15


130
Q
I
V
L
V
V
L
R
I






15


141
T
S
L
N
P
I
W
S
Y






15


163
A
T
L
D
R
I
G
T
D






15


169
G
T
D
G
D
C
S
K
P






15


208
L
T
H
W
V
F
G
E
V






15


244
C
L
A
S
G
L
M
L
P






15


261
G
L
I
W
W
V
T
G
T






15


285
V
S
G
C
V
F
A
I
F






15


297
M
W
P
Q
T
L
G
H
L






15


351
V
L
L
G
T
M
M
L
I






15


371
N
L
D
L
L
L
Q
T
K






15


388
K
S
E
K
Y
M
K
L
F






15


424
P
T
K
E
V
S
A
A
I






15


463
T
I
L
E
S
D
A
S
K






15


491
T
D
F
G
P
S
T
R
Y






15


492
D
F
G
P
S
T
R
Y
H






15


544
F
V
V
T
H
F
G
N
H






15


560
L
Q
A
I
A
V
S
K
L






15


573
S
N
Q
V
I
F
L
G
Y






15










HLA-A3 nonamers


























559
K
L

Q

A
I

A


V

S
K






34
Portion of SEQ ID NO: 3, each



418
K
L

G

K
V

A


P

T
K






31
start position is specified -


506
A
L

S

R
Y

P


I

V
K






31
the length of each peptide is


383
K
V

L

F
R

K


S

E
K






30
9 amino acids, the end


593
Q
L

T

E
H

G


N

V
K






30
position for each peptide is


375
L
L

Q

T
K

N


S

S
K






26
the start position plus eight.


463
T
I

L

E
S

D


A

S
K






26


540
K
L

V

D
F

V


V

T
H






26


103
S
L

I

V
Q

A


V

T
W






25


371
N
L

D

L
L

L


Q

T
K






25


562
A
I

A

V
S

K


L

L
K






25


404
G
L

L

G
L

G


L

R
H






24


619
G
L

I

R
L

G


Y

A
R






24


54
F
L

T

I
T

P


F

W
K






23


94
M
V

L

A
L

G


V

S
S






23


405
L
L

G

L
G

L


R

H
K






23


413
K
A

Y

E
R

K


L

G
K






23


532
T
L

T

V
N

I


S

G
K






23


184
E
V

A

T
G

M


A

S
R






22


195
W
L

L

A
G

A


A

F
G






22


218
L
V

S

R
W

A


V

S
G






22


74
R
I

I

T
I

G


S

I
A






21


90
K
L

R

L
M

V


L

A
L






21


154
K
V

I

L
T

L


S

A
I






21


240
A
V

L

L
C

L


A

S
G






21


361
G
L

N

M
L

F


G

P
K






21


394
K
L

F

L
W

L


L

V
G






21


409
G
L

R

H
K

A


Y

E
R






21


421
K
V

A

P
T

K


E

V
S






21


431
A
I

W

P
F

R


F

G
Y






21


453
L
L

N

E
T

G


A

D
F






21


478
D
L

T

M
W

L


G

E
K






21


622
R
L

G

Y
A

R


I

S
H






21


644
R
I

P

D
D

P


T

N
Y






21


49
F
L

S

P
I

F


L

T
I






20


92
R
L

M

V
L

A


L

G
V






20


108
A
V

T

W
W

S


G

S
H






20


217
S
L

V

S
R

W


A

V
S






20


342
G
V

Y

A
R

E


R

S
D






20


384
V
L

F

R
K

S


E

K
Y






20


407
G
L

G

L
R

H


K

A
Y






20


614
Y
I

M

Y
R

G


L

I
R






20


12
S
L

L

G
C

V


S

W
S






19


28
M
I

Y

Y
F

P


L

Q
T






19


95
V
L

A

L
G

V


S

S
S






19


127
I
L

G

Q
I

V


L

V
V






19


134
V
V

L

R
I

W


Y

T
S






19


205
L
V

F

L
T

H


W

V
F






19


265
W
V

T

G
T

A


S

A
A






19


338
F
V

P

G
G

V


Y

A
R






19


399
L
L

V

G
V

G


L

L
G






19


482
W
L

G

E
K

L


G

F
Y






19


536
N
I

S

G
K

L


V

D
F






19


599
N
V

K

D
I

D


S

T
D






19


658
V
V

I

D
H

R


E

V
S






19


671
F
N

P

R
F

G


S

Y
K






19


8
I
L

L

E
S

L


L

G
C






18


132
V
L

V

V
L

R


I

W
Y






18


241
V
L

L

C
L

A


S

G
L






18


304
H
L

I

N
S

G


T

N
P






18


358
L
I

I

G
L

N


M

L
F






18


464
I
L

E

S
D

A


S

K
P






18


567
K
L

L

K
S

S


S

N
Q






18


575
Q
V

I

F
L

G


Y

I
T






18


660
I
D

H

R
E

V


S

E
K






18


3
S
L

W

R
E

I


L

L
E






17


24
D
L

G

P
M

I


Y

Y
F






17


33
P
L

Q

T
L

E


L

T
G






17


36
T
L

E

L
T

G


L

E
G






17


41
G
L

E

G
F

S


I

A
F






17


120
Y
L

R

I
W

G


F

I
L






17


122
R
I

W

G
F

I


L

G
Q






17


135
V
L

R

I
W

Y


T

S
L






17


142
S
L

N

P
I

W


S

Y
Q






17


156
I
L

T

L
S

A


I

A
T






17


164
T
L

D

R
I

G


T

D
G






17


168
I
G

T

D
G

D


C

S
K






17


277
Y
L

H

T
W

A


A

A
V






17


350
D
V

L

L
G

T


M

M
L






17


357
M
L

I

I
G

L


N

M
L






17


402
G
V

G

L
L

G


L

G
L






17


446
S
L

E

R
S

A


H

L
L






17


620
L
I

R

L
G

Y


A

R
I






17


627
R
I

S

H
A

E


L

S
D






17










HLA-B*0702 nonamers


























235
N
P
F
G
G
A
V
L
L






24
Portion of SEQ ID NO: 3, each



26
G
P
M
I
Y
Y
F
P
L






22
start position is specified -


367
G
P
K
K
N
L
D
L
L






21
the length of each peptide is


423
A
P
T
K
E
V
S
A
A






21
9 amino acids, the end


471
K
P
Y
M
G
N
N
D
L






21
position for each peptide is


520
L
P
S
P
E
G
E
I
A






20
the start position plus eight.


522
S
P
E
G
E
I
A
P
A






20


86
A
P
N
A
K
L
R
L
M






19


229
H
P
G
P
D
P
N
P
F






19


233
D
P
N
P
F
G
G
A
V






19


311
N
P
G
K
T
M
T
I
A






19


192
R
P
N
W
L
L
A
G
A






18


90
K
L
R
L
M
V
L
A
L






17


200
A
A
F
G
S
L
V
F
L






17


298
W
P
Q
T
L
G
H
L
I






17


32
F
P
L
Q
T
L
E
L
T






16


270
A
S
A
A
G
L
L
Y
L






16


391
K
Y
M
K
L
F
L
W
L






16


645
I
P
D
D
P
T
N
Y
R






16


83
S
F
Q
A
P
N
A
K
L






15


189
M
A
S
R
P
N
W
L
L






15


364
M
L
F
G
P
K
K
N
L






15


528
A
P
A
I
T
L
T
V
N






15


29
I
Y
Y
F
P
L
Q
T
L






14


55
L
T
I
T
P
F
W
K
L






14


237
F
G
G
A
V
L
L
C
L






14


243
L
C
L
A
S
G
L
M
L






14


284
A
V
S
G
C
V
F
A
I






14


339
V
P
G
G
V
Y
A
R
E






14


345
A
R
E
R
S
D
V
L
L






14


402
G
V
G
L
L
G
L
G
L






14


525
G
E
I
A
P
A
I
T
L






14


571
S
S
S
N
Q
V
I
F
L






14


586
P
G
S
R
D
Y
L
Q
L






14


691
H
M
N
T
P
K
Y
F
L






14


1
M
T
S
L
W
R
E
I
L






13


31
Y
F
P
L
Q
T
L
E
L






13


42
L
E
G
F
S
I
A
F
L






13


64
V
N
K
K
W
M
L
T
L






13


120
Y
L
R
I
W
G
F
I
L






13


128
L
G
Q
I
V
L
V
V
L






13


135
V
L
R
I
W
Y
T
S
L






13


149
Y
Q
M
S
N
K
V
I
L






13


151
M
S
N
K
V
I
L
T
L






13


190
A
S
R
P
N
W
L
L
A






13


197
L
A
G
A
A
F
G
S
L






13


234
P
N
P
F
G
G
A
V
L






13


344
Y
A
R
E
R
S
D
V
L






13


366
F
G
P
K
K
N
L
D
L






13


387
R
K
S
E
K
Y
M
K
L






13


389
S
E
K
Y
M
K
L
F
L






13


397
L
W
L
L
V
G
V
G
L






13


400
L
V
G
V
G
L
L
G
L






13


584
S
A
P
G
S
R
D
Y
L






13


585
A
P
G
S
R
D
Y
L
Q






13


625
Y
A
R
I
S
H
A
E
L






13


642
K
F
R
I
P
D
D
P
T






13


47
I
A
F
L
S
P
I
F
L






12


49
F
L
S
P
I
F
L
T
I






12


58
T
P
F
W
K
L
V
N
K






12


85
Q
A
P
N
A
K
L
R
L






12


96
L
A
L
G
V
S
S
S
L






12


113
S
G
S
H
L
Q
R
Y
L






12


127
I
L
G
Q
I
V
L
V
V






12


176
K
P
E
E
K
K
T
G
E






12


210
H
W
V
F
G
E
V
S
L






12


227
H
P
H
P
G
P
D
P
N






12


231
G
P
D
P
N
P
F
G
G






12


251
L
P
S
C
L
W
F
R
G






12


254
C
L
W
F
R
G
T
G
L






12


267
T
G
T
A
S
A
A
G
L






12


294
T
A
S
M
W
P
Q
T
L






12


317
T
I
A
M
I
F
Y
L
L






12


368
P
K
K
N
L
D
L
L
L






12


411
R
H
K
A
Y
E
R
K
L






12


417
R
K
L
G
K
V
A
P
T






12


457
T
G
A
D
F
I
T
I
L






12


479
L
T
M
W
L
G
E
K
L






12


494
G
P
S
T
R
Y
H
T
W






12


536
N
I
S
G
K
L
V
D
F






12


560
L
Q
A
I
A
V
S
K
L






12


672
N
P
R
F
G
S
Y
K
E






12










HLA-B*08 nonamers


























88
N
A
K
L
R
L
M
V
L






33
Portion of SEQ ID NO: 3, each



344
Y
A
R
E
R
S
D
V
L






30
start position is specified -


367
G
P
K
K
N
L
D
L
L






27
the length of each peptide is


62
K
L
V
N
K
K
W
M
L






26
9 amino acids, the end


90
K
L
R
L
M
V
L
A
L






25
position for each peptide is


254
C
L
W
F
R
G
T
G
L






24
the start position plus eight.


409
G
L
R
H
K
A
Y
E
R






24


135
V
L
R
I
W
Y
T
S
L






23


389
S
E
K
Y
M
K
L
F
L






23


555
D
L
D
R
K
L
Q
A
I






23


640
M
A
K
F
R
I
P
D
D






23


120
Y
L
R
I
W
G
F
I
L






22


366
F
G
P
K
K
N
L
D
L






22


387
R
K
S
E
K
Y
M
K
L






22


445
S
S
L
E
R
S
A
H
L






22


64
V
N
K
K
W
M
L
T
L






21


377
Q
T
K
N
S
S
K
V
L






21


392
Y
M
K
L
F
L
W
L
L






21


512
I
V
K
S
E
H
H
L
L






21


536
N
I
S
G
K
L
V
D
F






21


625
Y
A
R
I
S
H
A
E
L






21


65
N
K
K
W
M
L
T
L
L






20


368
P
K
K
N
L
D
L
L
L






20


411
R
H
K
A
Y
E
R
K
L






20


188
G
M
A
S
R
P
N
W
L






19


424
P
T
K
E
V
S
A
A
I






19


2
T
S
L
W
R
E
I
L
L






18


176
K
P
E
E
K
K
T
G
E






18


398
W
L
L
V
G
V
G
L
L






18


407
G
L
G
L
R
H
K
A
Y






18


446
S
L
E
R
S
A
H
L
L






18


482
W
L
G
E
K
L
G
F
Y






18


26
G
P
M
I
Y
Y
F
P
L






17


58
T
P
F
W
K
L
V
N
K






17


235
N
P
F
G
G
A
V
L
L






17


241
V
L
L
C
L
A
S
G
L






17


364
M
L
F
G
P
K
K
N
L






17


375
L
L
Q
T
K
N
S
S
K






17


384
V
L
F
R
K
S
E
K
Y






17


471
K
P
Y
M
G
N
N
D
L






17


510
Y
P
I
V
K
S
E
H
H






17


620
L
I
R
L
G
Y
A
R
I






17


13
L
L
G
C
V
S
W
S
L






16


47
I
A
F
L
S
P
I
F
L






16


70
L
T
L
L
R
I
I
T
I






16


86
A
P
N
A
K
L
R
L
M






16


115
S
H
L
Q
R
Y
L
R
I






16


118
Q
R
Y
L
R
I
W
G
F






16


217
S
L
V
S
R
W
A
V
S






16


310
T
N
P
G
K
T
M
T
I






16


357
M
L
I
I
G
L
N
M
L






16


422
V
A
P
T
K
E
V
S
A






16


568
L
L
K
S
S
S
N
Q
V






16


584
S
A
P
G
S
R
D
Y
L






16










HLA-B*1510 nonamers


























499
Y
H
T
W
G
I
M
A
L






24
Portion of SEQ ID NO: 3, each



411
R
H
K
A
Y
E
R
K
L






22
start position is specified -


690
F
H
M
N
T
P
K
Y
F






17
the length of each peptide is


149
Y
Q
M
S
N
K
V
I
L






15
9 amino acids, the end


615
I
M
Y
R
G
L
I
R
L






15
position for each peptide is


29
I
Y
Y
F
P
L
Q
T
L






14
the start position plus eight.


125
G
F
I
L
G
Q
I
V
L






14


128
L
G
Q
I
V
L
V
V
L






14


209
T
H
W
V
F
G
E
V
S






14


235
N
P
F
G
G
A
V
L
L






14


294
T
A
S
M
W
P
Q
T
L






14


344
Y
A
R
E
R
S
D
V
L






14


457
T
G
A
D
F
I
T
I
L






14


551
N
H
E
D
D
L
D
R
K






14


669
I
H
F
N
P
R
F
G
S






14


22
Y
H
D
L
G
P
M
I
Y






13


47
I
A
F
L
S
P
I
F
L






13


64
V
N
K
K
W
M
L
T
L






13


85
Q
A
P
N
A
K
L
R
L






13


113
S
G
S
H
L
Q
R
Y
L






13


115
S
H
L
Q
R
Y
L
R
I






13


151
M
S
N
K
V
I
L
T
L






13


188
G
M
A
S
R
P
N
W
L






13


200
A
A
F
G
S
L
V
F
L






13


210
H
W
V
F
G
E
V
S
L






13


226
G
H
P
H
P
G
P
D
P






13


228
P
H
P
G
P
D
P
N
P






13


234
P
N
P
F
G
G
A
V
L






13


303
G
H
L
I
N
S
G
T
N






13


345
A
R
E
R
S
D
V
L
L






13


364
M
L
F
G
P
K
K
N
L






13


367
G
P
K
K
N
L
D
L
L






13


398
W
L
L
V
G
V
G
L
L






13


439
Y
D
N
E
G
W
S
S
L






13


517
H
H
L
L
P
S
P
E
G






13


525
G
E
I
A
P
A
I
T
L






13


547
T
H
F
G
N
H
E
D
D






13


552
H
E
D
D
L
D
R
K
L






13


1
M
T
S
L
W
R
E
I
L






12


31
Y
F
P
L
Q
T
L
E
L






12


88
N
A
K
L
R
L
M
V
L






12


90
K
L
R
L
M
V
L
A
L






12


157
L
T
L
S
A
I
A
T
L






12


189
M
A
S
R
P
N
W
L
L






12


268
G
T
A
S
A
A
G
L
L






12


270
A
S
A
A
G
L
L
Y
L






12


278
L
H
T
W
A
A
A
V
S






12


317
T
I
A
M
I
F
Y
L
L






12


354
G
T
M
M
L
I
I
G
L






12


377
Q
T
K
N
S
S
K
V
L






12


387
R
K
S
E
K
Y
M
K
L






12


397
L
W
L
L
V
G
V
G
L






12


400
L
V
G
V
G
L
L
G
L






12


475
G
N
N
D
L
T
M
W
L






12


512
I
V
K
S
E
H
H
L
L






12


516
E
H
H
L
L
P
S
P
E






12


560
L
Q
A
I
A
V
S
K
L






12


571
S
S
S
N
Q
V
I
F
L






12


584
S
A
P
G
S
R
D
Y
L






12


596
E
H
G
N
V
K
D
I
D






12


612
C
E
Y
I
M
Y
R
G
L






12


625
Y
A
R
I
S
H
A
E
L






12


629
S
H
A
E
L
S
D
S
E






12


687
N
H
H
F
H
M
N
T
P






12


691
H
M
N
T
P
K
Y
F
L






12










HLA-B*2705 nonamers


























118
Q
R
Y
L
R
I
W
G
F






27
Portion of SEQ ID NO: 3, each



345
A
R
E
R
S
D
V
L
L






24
start position is specified -


410
L
R
H
K
A
Y
E
R
K






24
the length of each peptide is


5
W
R
E
I
L
L
E
S
L






23
9 amino acids, the end


386
F
R
K
S
E
K
Y
M
K






23
position for each peptide is


73
L
R
I
I
T
I
G
S
I






22
the start position plus eight.


125
G
F
I
L
G
Q
I
V
L






22


76
I
T
I
G
S
I
A
S
F






21


617
Y
R
G
L
I
R
L
G
Y






21


497
T
R
Y
H
T
W
G
I
M






20


609
D
R
W
C
E
Y
I
M
Y






20


6
R
E
I
L
L
E
S
L
L






19


52
P
I
F
L
T
I
T
P
F






19


200
A
A
F
G
S
L
V
F
L






19


235
N
P
F
G
G
A
V
L
L






19


364
M
L
F
G
P
K
K
N
L






19


387
R
K
S
E
K
Y
M
K
L






19


404
G
L
L
G
L
G
L
R
H






19


491
T
D
F
G
P
S
T
R
Y






19


525
G
E
I
A
P
A
I
T
L






19


589
R
D
Y
L
Q
L
T
E
H






19


610
R
W
C
E
Y
I
M
Y
R






19


615
I
M
Y
R
G
L
I
R
L






19


652
Y
R
D
N
Q
K
V
V
I






19


662
H
R
E
V
S
E
K
I
H






19


58
T
P
F
W
K
L
V
N
K






18


96
L
A
L
G
V
S
S
S
L






18


129
G
Q
I
V
L
V
V
L
R






18


151
M
S
N
K
V
I
L
T
L






18


199
G
A
A
F
G
S
L
V
F






18


354
G
T
M
M
L
I
I
G
L






18


418
K
L
G
K
V
A
P
T
K






18


435
F
R
F
G
Y
D
N
E
G






18


471
K
P
Y
M
G
N
N
D
L






18


663
R
E
V
S
E
K
I
H
F






18


676
G
S
Y
K
E
G
H
N
Y






18


41
G
L
E
G
F
S
I
A
F






17


47
I
A
F
L
S
P
I
F
L






17


59
P
F
W
K
L
V
N
K
K






17


82
A
S
F
Q
A
P
N
A
K






17


157
L
T
L
S
A
I
A
T
L






17


358
L
I
I
G
L
N
M
L
F






17


383
K
V
L
F
R
K
S
E
K






17


409
G
L
R
H
K
A
Y
E
R






17


413
K
A
Y
E
R
K
L
G
K






17


463
T
I
L
E
S
D
A
S
K






17


481
M
W
L
G
E
K
L
G
F






17


619
G
L
I
R
L
G
Y
A
R






17


673
P
R
F
G
S
Y
K
E
G






17


29
I
Y
Y
F
P
L
Q
T
L






16


55
L
T
I
T
P
F
W
K
L






16


62
K
L
V
N
K
K
W
M
L






16


141
T
S
L
N
P
I
W
S
Y






16


205
L
V
F
L
T
H
W
V
F






16


321
I
F
Y
L
L
E
I
F
F






16


338
F
V
P
G
G
V
Y
A
R






16


350
D
V
L
L
G
T
M
M
L






16


357
M
L
I
I
G
L
N
M
L






16


367
G
P
K
K
N
L
D
L
L






16


380
N
S
S
K
V
L
F
R
K






16


391
K
Y
M
K
L
F
L
W
L






16


402
G
V
G
L
L
G
L
G
L






16


403
V
G
L
L
G
L
G
L
R






16


411
R
H
K
A
Y
E
R
K
L






16


485
E
K
L
G
F
Y
T
D
F






16


499
Y
H
T
W
G
I
M
A
L






16


508
S
R
Y
P
I
V
K
S
E






16


509
R
Y
P
I
V
K
S
E
H






16


550
G
N
H
E
D
D
L
D
R






16


559
K
L
Q
A
I
A
V
S
K






16


621
I
R
L
G
Y
A
R
I
S






16


643
F
R
I
P
D
D
P
T
N






16


644
R
I
P
D
D
P
T
N
Y






16


667
E
K
I
H
F
N
P
R
F






16


688
H
H
F
H
M
N
T
P
K






16


24
D
L
G
P
M
I
Y
Y
F






15


34
L
Q
T
L
E
L
T
G
L






15


37
L
E
L
T
G
L
E
G
F






15


42
L
E
G
F
S
I
A
F
L






15


83
S
F
Q
A
P
N
A
K
L






15


90
K
L
R
L
M
V
L
A
L






15


91
L
R
L
M
V
L
A
L
G






15


111
W
W
S
G
S
H
L
Q
R






15


114
G
S
H
L
Q
R
Y
L
R






15


121
L
R
I
W
G
F
I
L
G






15


166
D
R
I
G
T
D
G
D
C






15


168
I
G
T
D
G
D
C
S
K






15


172
G
D
C
S
K
P
E
E
K






15


173
D
C
S
K
P
E
E
K
K






15


181
K
T
G
E
V
A
T
G
M






15


210
H
W
V
F
G
E
V
S
L






15


241
V
L
L
C
L
A
S
G
L






15


249
L
M
L
P
S
C
L
W
F






15


270
A
S
A
A
G
L
L
Y
L






15


306
I
N
S
G
T
N
P
G
K






15


314
K
T
M
T
I
A
M
I
F






15


316
M
T
I
A
M
I
F
Y
L






15


320
M
I
F
Y
L
L
E
I
F






15


348
R
S
D
V
L
L
G
T
M






15


366
F
G
P
K
K
N
L
D
L






15


379
K
N
S
S
K
V
L
F
R






15


384
V
L
F
R
K
S
E
K
Y






15


397
L
W
L
L
V
G
V
G
L






15


398
W
L
L
V
G
V
G
L
L






15


416
E
R
K
L
G
K
V
A
P






15


439
Y
D
N
E
G
W
S
S
L






15


445
S
S
L
E
R
S
A
H
L






15


457
T
G
A
D
F
I
T
I
L






15


475
G
N
N
D
L
T
M
W
L






15


533
L
T
V
N
I
S
G
K
L






15


536
N
I
S
G
K
L
V
D
F






15


570
K
S
S
S
N
Q
V
I
F






15


588
S
R
D
Y
L
Q
L
T
E






15


634
S
D
S
E
I
Q
M
A
K






15


636
S
E
I
Q
M
A
K
F
R






15


660
I
D
H
R
E
V
S
E
K






15


13
L
L
G
C
V
S
W
S
L






14


15
G
C
V
S
W
S
L
Y
H






14


23
H
D
L
G
P
M
I
Y
Y






14


64
V
N
K
K
W
M
L
T
L






14


66
K
K
W
M
L
T
L
L
R






14


70
L
T
L
L
R
I
I
T
I






14


85
Q
A
P
N
A
K
L
R
L






14


88
N
A
K
L
R
L
M
V
L






14


128
L
G
Q
I
V
L
V
V
L






14


146
I
W
S
Y
Q
M
S
N
K






14


184
E
V
A
T
G
M
A
S
R






14


188
G
M
A
S
R
P
N
W
L






14


194
N
W
L
L
A
G
A
A
F






14


213
F
G
E
V
S
L
V
S
R






14


234
P
N
P
F
G
G
A
V
L






14


243
L
C
L
A
S
G
L
M
L






14


247
S
G
L
M
L
P
S
C
L






14


257
F
R
G
T
G
L
I
W
W






14


268
G
T
A
S
A
A
G
L
L






14


356
M
M
L
I
I
G
L
N
M






14


361
G
L
N
M
L
F
G
P
K






14


362
L
N
M
L
F
G
P
K
K






14


371
N
L
D
L
L
L
Q
T
K






14


375
L
L
Q
T
K
N
S
S
K






14


388
K
S
E
K
Y
M
K
L
F






14


400
L
V
G
V
G
L
L
G
L






14


405
L
L
G
L
G
L
R
H
K






14


427
E
V
S
A
A
I
W
P
F






14


441
N
E
G
W
S
S
L
E
R






14


448
E
R
S
A
H
L
L
N
E






14


490
Y
T
D
F
G
P
S
T
R






14


502
W
G
I
M
A
L
S
R
Y






14


511
P
I
V
K
S
E
H
H
L






14


532
T
L
T
V
N
I
S
G
K






14


540
K
L
V
D
F
V
V
T
H






14


541
L
V
D
F
V
V
T
H
F






14


552
H
E
D
D
L
D
R
K
L






14


560
L
Q
A
I
A
V
S
K
L






14


561
Q
A
I
A
V
S
K
L
L






14


562
A
I
A
V
S
K
L
L
K






14


622
R
L
G
Y
A
R
I
S
H






14


626
A
R
I
S
H
A
E
L
S






14


655
N
Q
K
V
V
I
D
H
R






14


681
G
H
N
Y
E
N
N
H
H






14


682
H
N
Y
E
N
N
H
H
F






14


2
T
S
L
W
R
E
I
L
L






13


26
G
P
M
I
Y
Y
F
P
L






13


31
Y
F
P
L
Q
T
L
E
L






13


46
S
I
A
F
L
S
P
I
F






13


65
N
K
K
W
M
L
T
L
L






13


67
K
W
M
L
T
L
L
R
I






13


113
S
G
S
H
L
Q
R
Y
L






13


115
S
H
L
Q
R
Y
L
R
I






13


119
R
Y
L
R
I
W
G
F
I






13


130
Q
I
V
L
V
V
L
R
I






13


135
V
L
R
I
W
Y
T
S
L






13


136
L
R
I
W
Y
T
S
L
N






13


154
K
V
I
L
T
L
S
A
I






13


189
M
A
S
R
P
N
W
L
L






13


221
R
W
A
V
S
G
H
P
H






13


237
F
G
G
A
V
L
L
C
L






13


267
T
G
T
A
S
A
A
G
L






13


271
S
A
A
G
L
L
Y
L
H






13


282
A
A
A
V
S
G
C
V
F






13


297
M
W
P
Q
T
L
G
H
L






13


313
G
K
T
M
T
I
A
M
I






13


330
C
A
W
C
T
A
F
K
F






13


336
F
K
F
V
P
G
G
V
Y






13


344
Y
A
R
E
R
S
D
V
L






13


347
E
R
S
D
V
L
L
G
T






13


349
S
D
V
L
L
G
T
M
M






13


368
P
K
K
N
L
D
L
L
L






13


377
Q
T
K
N
S
S
K
V
L






13


378
T
K
N
S
S
K
V
L
F






13


392
Y
M
K
L
F
L
W
L
L






13


479
L
T
M
W
L
G
E
K
L






13


501
T
W
G
I
M
A
L
S
R






13


506
A
L
S
R
Y
P
I
V
K






13


551
N
H
E
D
D
L
D
R
K






13


557
D
R
K
L
Q
A
I
A
V






13


558
R
K
L
Q
A
I
A
V
S






13


584
S
A
P
G
S
R
D
Y
L






13


593
Q
L
T
E
H
G
N
V
K






13


625
Y
A
R
I
S
H
A
E
L






13


635
D
S
E
I
Q
M
A
K
F






13


645
I
P
D
D
P
T
N
Y
R






13


649
P
T
N
Y
R
D
N
Q
K






13


654
D
N
Q
K
V
V
I
D
H






13


674
R
F
G
S
Y
K
E
G
H






13


691
H
M
N
T
P
K
Y
F
L






13










HLA-B*2709 nonamers


























345
A
R
E
R
S
D
V
L
L






22
Portion of SEQ ID NO: 3, each



5
W
R
E
I
L
L
E
S
L






20
start position is specified -


118
Q
R
Y
L
R
I
W
G
F






20
the length of each peptide is


497
T
R
Y
H
T
W
G
I
M






20
9 amino acids, the end


73
L
R
I
I
T
I
G
S
I






19
position for each peptide is


557
D
R
K
L
Q
A
I
A
V






19
the start position plus eight.


652
Y
R
D
N
Q
K
V
V
I






19


6
R
E
I
L
L
E
S
L
L






15


119
R
Y
L
R
I
W
G
F
I






15


268
G
T
A
S
A
A
G
L
L






15


508
S
R
Y
P
I
V
K
S
E






15


525
G
E
I
A
P
A
I
T
L






15


90
K
L
R
L
M
V
L
A
L






14


92
R
L
M
V
L
A
L
G
V






14


125
G
F
I
L
G
Q
I
V
L






14


188
G
M
A
S
R
P
N
W
L






14


200
A
A
F
G
S
L
V
F
L






14


235
N
P
F
G
G
A
V
L
L






14


387
R
K
S
E
K
Y
M
K
L






14


402
G
V
G
L
L
G
L
G
L






14


411
R
H
K
A
Y
E
R
K
L






14


471
K
P
Y
M
G
N
N
D
L






14


615
I
M
Y
R
G
L
I
R
L






14


621
I
R
L
G
Y
A
R
I
S






14


626
A
R
I
S
H
A
E
L
S






14


643
F
R
I
P
D
D
P
T
N






14


663
R
E
V
S
E
K
I
H
F






14


26
G
P
M
I
Y
Y
F
P
L






13


29
I
Y
Y
F
P
L
Q
T
L






13


47
I
A
F
L
S
P
I
F
L






13


62
K
L
V
N
K
K
W
M
L






13


157
L
T
L
S
A
I
A
T
L






13


241
V
L
L
C
L
A
S
G
L






13


243
L
C
L
A
S
G
L
M
L






13


258
R
G
T
G
L
I
W
W
V






13


354
G
T
M
M
L
I
I
G
L






13


356
M
M
L
I
I
G
L
N
M






13


364
M
L
F
G
P
K
K
N
L






13


367
G
P
K
K
N
L
D
L
L






13


391
K
Y
M
K
L
F
L
W
L






13


397
L
W
L
L
V
G
V
G
L






13


398
W
L
L
V
G
V
G
L
L






13


475
G
N
N
D
L
T
M
W
L






13


586
P
G
S
R
D
Y
L
Q
L






13


2
T
S
L
W
R
E
I
L
L






12


67
K
W
M
L
T
L
L
R
I






12


85
Q
A
P
N
A
K
L
R
L






12


91
L
R
L
M
V
L
A
L
G






12


96
L
A
L
G
V
S
S
S
L






12


115
S
H
L
Q
R
Y
L
R
I






12


127
I
L
G
Q
I
V
L
V
V






12


130
Q
I
V
L
V
V
L
R
I






12


149
Y
Q
M
S
N
K
V
I
L






12


199
G
A
A
F
G
S
L
V
F






12


210
H
W
V
F
G
E
V
S
L






12


247
S
G
L
M
L
P
S
C
L






12


249
L
M
L
P
S
C
L
W
F






12


267
T
G
T
A
S
A
A
G
L






12


270
A
S
A
A
G
L
L
Y
L






12


313
G
K
T
M
T
I
A
M
I






12


314
K
T
M
T
I
A
M
I
F






12


317
T
I
A
M
I
F
Y
L
L






12


348
R
S
D
V
L
L
G
T
M






12


350
D
V
L
L
G
T
M
M
L






12


386
F
R
K
S
E
K
Y
M
K






12


420
G
K
V
A
P
T
K
E
V






12


435
F
R
F
G
Y
D
N
E
G






12


445
S
S
L
E
R
S
A
H
L






12


448
E
R
S
A
H
L
L
N
E






12


505
M
A
L
S
R
Y
P
I
V






12


512
I
V
K
S
E
H
H
L
L






12


533
L
T
V
N
I
S
G
K
L






12


570
K
S
S
S
N
Q
V
I
F






12


612
C
E
Y
I
M
Y
R
G
L






12


617
Y
R
G
L
I
R
L
G
Y






12


657
K
V
V
I
D
H
R
E
V






12


673
P
R
F
G
S
Y
K
E
G






12










HLA-B*4402 nonamers


























525
G

E

I
A
P
A
I
T

L







26
Portion of SEQ ID NO: 3, each



6
R

E

I
L
L
E
S
L

L







25
start position is specified -


465
L

E

S
D
A
S
K
P

Y







24
the length of each peptide is


552
H

E

D
D
L
D
R
K

L







24
9 amino acids, the end


37
L

E

L
T
G
L
E
G

F







23
position for each peptide is


42
L

E

G
F
S
I
A
F

L







23
the start position plus eight.


595
T

E

H
G
N
V
K
D

I







23


389
S

E

K
Y
M
K
L
F

L







22


523
P

E

G
E
I
A
P
A

I







22


214
G

E

V
S
L
V
S
R

W







21


612
C

E

Y
I
M
Y
R
G

L







21


663
R

E

V
S
E
K
I
H

F







21


200
A

A

F
G
S
L
V
F

L







18


273
A

G

L
L
Y
L
H
T

W







18


76
I

T

I
G
S
I
A
S

F







17


103
S

L

I
V
Q
A
V
T

W







17


235
N

P

F
G
G
A
V
L

L







17


431
A

I

W
P
F
R
F
G

Y







17


636
S

E

I
Q
M
A
K
F

R







17


24
D

L

G
P
M
I
Y
Y

F







16


49
F

L

S
P
I
F
L
T

I







16


52
P

I

F
L
T
I
T
P

F







16


55
L

T

I
T
P
F
W
K

L







16


70
L

T

L
L
R
I
I
T

I







16


90
K

L

R
L
M
V
L
A

L







16


125
G

F

I
L
G
Q
I
V

L







16


154
K

V

I
L
T
L
S
A

I







16


284
A

V

S
G
C
V
F
A

I







16


316
M

T

I
A
M
I
F
Y

L







16


319
A

M

I
F
Y
L
L
E

I







16


345
A

R

E
R
S
D
V
L

L







16


407
G

L

G
L
R
H
K
A

Y







16


427
E

V

S
A
A
I
W
P

F







16


561
Q

A

I
A
V
S
K
L

L







16


11
E

S

L
L
G
C
V
S

W







15


47
I

A

F
L
S
P
I
F

L







15


88
N

A

K
L
R
L
M
V

L







15


118
Q

R

Y
L
R
I
W
G

F







15


141
T

S

L
N
P
I
W
S

Y







15


151
M

S

N
K
V
I
L
T

L







15


157
L

T

L
S
A
I
A
T

L







15


194
N

W

L
L
A
G
A
A

F







15


229
H

P

G
P
D
P
N
P

F







15


249
L

M

L
P
S
C
L
W

F







15


270
A

S

A
A
G
L
L
Y

L







15


314
K

T

M
T
I
A
M
I

F







15


354
G

T

M
M
L
I
I
G

L







15


357
M

L

I
I
G
L
N
M

L







15


358
L

I

I
G
L
N
M
L

F







15


364
M

L

F
G
P
K
K
N

L







15


446
S

L

E
R
S
A
H
L

L







15


494
G

P

S
T
R
Y
H
T

W







15


499
Y

H

T
W
G
I
M
A

L







15


536
N

I

S
G
K
L
V
D

F







15


571
S

S

S
N
Q
V
I
F

L







15


613
E

Y

I
M
Y
R
G
L

I







15


667
E

K

I
H
F
N
P
R

F







15


670
H

F

N
P
R
F
G
S

Y







15


690
F

H

M
N
T
P
K
Y

F







15


2
T

S

L
W
R
E
I
L

L







14


31
Y

F

P
L
Q
T
L
E

L







14


41
G

L

E
G
F
S
I
A

F







14


53
I

F

L
T
I
T
P
F

W







14


60
F

W

K
L
V
N
K
K

W







14


73
L

R

I
I
T
I
G
S

I







14


205
L

V

F
L
T
H
W
V

F







14


282
A

A

A
V
S
G
C
V

F







14


290
F

A

I
F
T
A
S
M

W







14


377
Q

T

K
N
S
S
K
V

L







14


384
V

L

F
R
K
S
E
K

Y







14


388
K

S

E
K
Y
M
K
L

F







14


390
E

K

Y
M
K
L
F
L

W







14


391
K

Y

M
K
L
F
L
W

L







14


457
T

G

A
D
F
I
T
I

L







14


485
E

K

L
G
F
Y
T
D

F







14


502
W

G

I
M
A
L
S
R

Y







14


529
P

A

I
T
L
T
V
N

I







14


570
K

S

S
S
N
Q
V
I

F







14


573
S

N

Q
V
I
F
L
G

Y







14


583
T

S

A
P
G
S
R
D

Y







14


584
S

A

P
G
S
R
D
Y

L







14


586
P

G

S
R
D
Y
L
Q

L







14


617
Y

R

G
L
I
R
L
G

Y







14


631
A

E

L
S
D
S
E
I

Q







14


1
M

T

S
L
W
R
E
I

L







13


10
L

E

S
L
L
G
C
V

S







13


22
Y

H

D
L
G
P
M
I

Y







13


23
H

D

L
G
P
M
I
Y

Y







13


65
N

K

K
W
M
L
T
L

L







13


67
K

W

M
L
T
L
L
R

I







13


85
Q

A

P
N
A
K
L
R

L







13


113
S

G

S
H
L
Q
R
Y

L







13


116
H

L

Q
R
Y
L
R
I

W







13


128
L

G

Q
I
V
L
V
V

L







13


131
I

V

L
V
V
L
R
I

W







13


149
Y

Q

M
S
N
K
V
I

L







13


160
S

A

I
A
T
L
D
R

I







13


178
E

E

K
K
T
G
E
V

A







13


188
G

M

A
S
R
P
N
W

L







13


189
M

A

S
R
P
N
W
L

L







13


247
S

G

L
M
L
P
S
C

L







13


248
G

L

M
L
P
S
C
L

W







13


255
L

W

F
R
G
T
G
L

I







13


257
F

R

G
T
G
L
I
W

W







13


269
T

A

S
A
A
G
L
L

Y







13


294
T

A

S
M
W
P
Q
T

L







13


317
T

I

A
M
I
F
Y
L

L







13


320
M

I

F
Y
L
L
E
I

F







13


324
L

L

E
I
F
F
C
A

W







13


325
L

E

I
F
F
C
A
W

C







13


336
F

K

F
V
P
G
G
V

Y







13


346
R

E

R
S
D
V
L
L

G







13


350
D

V

L
L
G
T
M
M

L







13


367
G

P

K
K
N
L
D
L

L







13


368
P

K

K
N
L
D
L
L

L







13


392
Y

M

K
L
F
L
W
L

L







13


397
L

W

L
L
V
G
V
G

L







13


398
W

L

L
V
G
V
G
L

L







13


415
Y

E

R
K
L
G
K
V

A







13


426
K

E

V
S
A
A
I
W

P







13


445
S

S

L
E
R
S
A
H

L







13


456
E

T

G
A
D
F
I
T

I







13


481
M

W

L
G
E
K
L
G

F







13


484
G

E

K
L
G
F
Y
T

D







13


491
T

D

F
G
P
S
T
R

Y







13


533
L

T

V
N
I
S
G
K

L







13


555
D

L

D
R
K
L
Q
A

I







13


615
I

M

Y
R
G
L
I
R

L







13


644
R

I

P
D
D
P
T
N

Y







13


666
S

E

K
I
H
F
N
P

R







13


689
H

F

H
M
N
T
P
K

Y







13










HLA-B*5101 nonamers


























527
I
A
P
A
I
T
L
T
V






25
Portion of SEQ ID NO: 3, each



160
S
A
I
A
T
L
D
R
I






24
start position is specified -


96
L
A
L
G
V
S
S
S
L






23
the length of each peptide is


344
Y
A
R
E
R
S
D
V
L






23
9 amino acids, the end


47
I
A
F
L
S
P
I
F
L






22
position for each peptide is


233
D
P
N
P
F
G
G
A
V






22
the start position plus eight.


471
K
P
Y
M
G
N
N
D
L






22


529
P
A
I
T
L
T
V
N
I






22


88
N
A
K
L
R
L
M
V
L






21


128
L
G
Q
I
V
L
V
V
L






21


200
A
A
F
G
S
L
V
F
L






21


235
N
P
F
G
G
A
V
L
L






21


281
W
A
A
A
V
S
G
C
V






21


298
W
P
Q
T
L
G
H
L
I






21


505
M
A
L
S
R
Y
P
I
V






21


630
H
A
E
L
S
D
S
E
I






21


138
I
W
Y
T
S
L
N
P
I






20


197
L
A
G
A
A
F
G
S
L






20


294
T
A
S
M
W
P
Q
T
L






20


561
Q
A
I
A
V
S
K
L
L






20


661
D
H
R
E
V
S
E
K
I






20


85
Q
A
P
N
A
K
L
R
L






19


538
S
G
K
L
V
D
F
V
V






19


584
S
A
P
G
S
R
D
Y
L






19


625
Y
A
R
I
S
H
A
E
L






19


70
L
T
L
L
R
I
I
T
I






18


98
L
G
V
S
S
S
L
I
V






18


189
M
A
S
R
P
N
W
L
L






18


237
F
G
G
A
V
L
L
C
L






18


366
F
G
P
K
K
N
L
D
L






18


367
G
P
K
K
N
L
D
L
L






18


26
G
P
M
I
Y
Y
F
P
L






17


413
K
A
Y
E
R
K
L
G
K






17


563
I
A
V
S
K
L
L
K
S






17


569
L
K
S
S
S
N
Q
V
I






17


127
I
L
G
Q
I
V
L
V
V






16


130
Q
I
V
L
V
V
L
R
I






16


147
W
S
Y
Q
M
S
N
K
V






16


148
S
Y
Q
M
S
N
K
V
I






16


258
R
G
T
G
L
I
W
W
V






16


267
T
G
T
A
S
A
A
G
L






16


310
T
N
P
G
K
T
M
T
I






16


457
T
G
A
D
F
I
T
I
L






16


528
A
P
A
I
T
L
T
V
N






16


555
D
L
D
R
K
L
Q
A
I






16


650
T
N
Y
R
D
N
Q
K
V






16


652
Y
R
D
N
Q
K
V
V
I






16


29
I
Y
Y
F
P
L
Q
T
L






15


32
F
P
L
Q
T
L
E
L
T






15


49
F
L
S
P
I
F
L
T
I






15


58
T
P
F
W
K
L
V
N
K






15


68
W
M
L
T
L
L
R
I
I






15


124
W
G
F
I
L
G
Q
I
V






15


185
V
A
T
G
M
A
S
R
P






15


198
A
G
A
A
F
G
S
L
V






15


247
S
G
L
M
L
P
S
C
L






15


330
C
A
W
C
T
A
F
K
F






15


334
T
A
F
K
F
V
P
G
G






15


339
V
P
G
G
V
Y
A
R
E






15


351
V
L
L
G
T
M
M
L
I






15


352
L
L
G
T
M
M
L
I
I






15


422
V
A
P
T
K
E
V
S
A






15


456
E
T
G
A
D
F
I
T
I






15


468
D
A
S
K
P
Y
M
G
N






15


595
T
E
H
G
N
V
K
D
I






15


615
I
M
Y
R
G
L
I
R
L






15


623
L
G
Y
A
R
I
S
H
A






15


648
D
P
T
N
Y
R
D
N
Q






15


67
K
W
M
L
T
L
L
R
I






14


73
L
R
I
I
T
I
G
S
I






14


86
A
P
N
A
K
L
R
L
M






14


115
S
H
L
Q
R
Y
L
R
I






14


126
F
I
L
G
Q
I
V
L
V






14


151
M
S
N
K
V
I
L
T
L






14


176
K
P
E
E
K
K
T
G
E






14


199
G
A
A
F
G
S
L
V
F






14


245
L
A
S
G
L
M
L
P
S






14


282
A
A
A
V
S
G
C
V
F






14


311
N
P
G
K
T
M
T
I
A






14


395
L
F
L
W
L
L
V
G
V






14


424
P
T
K
E
V
S
A
A
I






14


454
L
N
E
T
G
A
D
F
I






14


510
Y
P
I
V
K
S
E
H
H






14


557
D
R
K
L
Q
A
I
A
V






14


607
D
H
D
R
W
C
E
Y
I






14


645
I
P
D
D
P
T
N
Y
R






14


651
N
Y
R
D
N
Q
K
V
V






14


21
L
Y
H
D
L
G
P
M
I






13


45
F
S
I
A
F
L
S
P
I






13


51
S
P
I
F
L
T
I
T
P






13


81
I
A
S
F
Q
A
P
N
A






13


113
S
G
S
H
L
Q
R
Y
L






13


123
I
W
G
F
I
L
G
Q
I






13


157
L
T
L
S
A
I
A
T
L






13


162
I
A
T
L
D
R
I
G
T






13


202
F
G
S
L
V
F
L
T
H






13


208
L
T
H
W
V
F
G
E
V






13


243
L
C
L
A
S
G
L
M
L






13


251
L
P
S
C
L
W
F
R
G






13


255
L
W
F
R
G
T
G
L
I






13


269
T
A
S
A
A
G
L
L
Y






13


284
A
V
S
G
C
V
F
A
I






13


290
F
A
I
F
T
A
S
M
W






13


318
I
A
M
I
F
Y
L
L
E






13


319
A
M
I
F
Y
L
L
E
I






13


376
L
Q
T
K
N
S
S
K
V






13


397
L
W
L
L
V
G
V
G
L






13


406
L
G
L
G
L
R
H
K
A






13


423
A
P
T
K
E
V
S
A
A






13


450
S
A
H
L
L
N
E
T
G






13


494
G
P
S
T
R
Y
H
T
W






13


504
I
M
A
L
S
R
Y
P
I






13


519
L
L
P
S
P
E
G
E
I






13


523
P
E
G
E
I
A
P
A
I






13


537
I
S
G
K
L
V
D
F
V






13


579
L
G
Y
I
T
S
A
P
G






13


586
P
G
S
R
D
Y
L
Q
L






13


592
L
Q
L
T
E
H
G
N
V






13


620
L
I
R
L
G
Y
A
R
I






13


672
N
P
R
F
G
S
Y
K
E






13


39
L
T
G
L
E
G
F
S
I






12


40
T
G
L
E
G
F
S
I
A






12


55
L
T
I
T
P
F
W
K
L






12


119
R
Y
L
R
I
W
G
F
I






12


144
N
P
I
W
S
Y
Q
M
S






12


154
K
V
I
L
T
L
S
A
I






12


211
W
V
F
G
E
V
S
L
V






12


216
V
S
L
V
S
R
W
A
V






12


229
H
P
G
P
D
P
N
P
F






12


260
T
G
L
I
W
W
V
T
G






12


271
S
A
A
G
L
L
Y
L
H






12


272
A
A
G
L
L
Y
L
H
T






12


273
A
G
L
L
Y
L
H
T
W






12


277
Y
L
H
T
W
A
A
A
V






12


313
G
K
T
M
T
I
A
M
I






12


350
D
V
L
L
G
T
M
M
L






12


360
I
G
L
N
M
L
F
G
P






12


393
M
K
L
F
L
W
L
L
V






12


414
A
Y
E
R
K
L
G
K
V






12


419
L
G
K
V
A
P
T
K
E






12


429
S
A
A
I
W
P
F
R
F






12


430
A
A
I
W
P
F
R
F
G






12


437
F
G
Y
D
N
E
G
W
S






12


520
L
P
S
P
E
G
E
I
A






12


522
S
P
E
G
E
I
A
P
A






12


560
L
Q
A
I
A
V
S
K
L






12


568
L
L
K
S
S
S
N
Q
V






12


574
N
Q
V
I
F
L
G
Y
I






12


612
C
E
Y
I
M
Y
R
G
L






12


640
M
A
K
F
R
I
P
D
D






12








HLA Class I nonamer analysis of 125P5C8, variants 2-5.



Listed are scores which correlate with the ligation


strength to a defined HLA type for a sequence of


amino acids. The algorithms used are based on the book


“MHC Ligands and Peptide Motifs” by H. G.


Rammensee, J. Bachmann and S. Stevanovic.


The probability of being processed and presented


is given in order to predict T-cell epitopes.


variant 2 (aa 1-10)










HLA-A26 nonamers


























1
M
P
S
L
W
R
E
I
L






8
Portion of SEQ ID NO: 5, each



2
P
S
L
W
R
E
I
L
L






8
start position is specified - the



















length of each peptide is 9 amino



















acids, the end position for each



















peptide is the start position



















plus eight.










HLA-B*0702 nonamers


























1
M

P

S
L
W
R
E
I

L







23
Portion of SEQ ID NO: 5, each




















start position is specified - the



















length of each peptide is 9 amino



















acids, the end position for each



















peptide is the start position



















plus eight.










HLA-B*08 nonamers




















2
P
S

L

W

R

E
I
L

L

18
Portion of SEQ ID NO: 5, each



1
M
P

S

L

W

R
E
I

L

17
start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*1510 nonamers




















1
M

P

S
L
W
R
E
I

L

12
Portion of SEQ ID NO: 5, each














start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*2705 nonamers




















2
P

S

L
W
R
E
I
L

L

13
Portion of SEQ ID NO: 5, each



1
M

P

S
L
W
R
E
I

L

11
start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*2709 nonamers




















2
P
S
L
W
R
E
I
L
L
12
Portion of SEQ ID NO: 5, each



1
M
P
S
L
W
R
E
I
L
10
start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*4402 nonamers




















2
P

S

L
W
R
E
I
L

L

14
Portion of SEQ ID NO: 5, each



1
M

P

S
L
W
R
E
I

L

13
start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*5101 nonamers




















1
M

P

S
L
W
R
E
I

L

17
Portion of SEQ ID NO: 5, each














start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.







variant 3 (aa 674-690)










HLA-A1 nonamers




















676
G

S

Y
K
E
G

P

N
Y
20
Portion of SEQ ID NO: 7, each



678
Y

K

E
G
P
N

Y

E
N
15
start position is specified -













the length of each peptide is 9













amino acids, the end position













for each peptide is the start













position plus eight.










HLA-B*2705 nonamers




















676
G
S
Y
K
E
G
P
N
Y
19
Portion of SEQ ID NO: 7, each



681
G
P
N
Y
E
N
N
H
H
14
start position is specified - the


682
P
N
Y
E
N
N
H
H
F
14
length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*4402 nonamers




















679
K
E
G
P
N
Y
E
N
N
15
Portion of SEQ ID NO: 7, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*5101 nonamers




















681
G
P
N
Y
E
N
N
H
H
13
Portion of SEQ ID NO: 7, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.







variant 4 (aa 679-695)










HLA-B*1510 nonamers




















687
T
H
H
F
H
M
N
T
P
13
Portion of SEQ ID NO: 9, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*2705 nonamers




















681
G
H
N
Y
E
N
T
H
H
15
Portion of SEQ ID NO: 9, each



682
H
N
Y
E
N
T
H
H
F
13
start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*4402 nonamers




















682
H
N
Y
E
N
T
H
H
F
13
Portion of SEQ ID NO: 9, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.







variant 5 (aa 681-697)










HLA-A1 nonamers




















689
N
F
H
M
N
T
P
K
Y
17
Portion of SEQ ID NO: 11, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-A26 nonamers




















689
N
F
H
M
N
T
P
K
Y
18
Portion of SEQ ID NO: 11, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*1510 nonamers




















690
F
H
M
N
T
P
K
Y
F
17
Portion of SEQ ID NO: 11, each



687
N
H
N
F
H
M
N
T
P
12
start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*2705 nonamers




















682
H
N
Y
E
N
N
H
N
F
16
Portion of SEQ ID NO: 11, each



688
H
N
F
H
M
N
T
P
K
16
start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*2709 nonamers




















682
H
N
Y
E
N
N
H
N
F
11
Portion of SEQ ID NO: 11, each














start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.










HLA-B*4402 nonamers




















690
F
H
M
N
T
P
K
Y
F
15
Portion of SEQ ID NO: 11, each



689
N
F
H
M
N
T
P
K
Y
14
start position is specified - the













length of each peptide is 9 amino













acids, the end position for each













peptide is the start position













plus eight.






















Pos
1
2
3
4
5
6
7
8
9
0
score











HLA Class I decamer analysis of 125P5C8, variant 1 (aa 1-699)



Listed are scores which correlate with the ligation strength


to a defined HLA type for a sequence of amino acids. The


algorithms used are based on the book “MHC Ligands and Peptide


Motifs” by H. G. Rammensee, J. Bachmann and S. Stevanovic.


The probability of being processed and presented is given in


order to predict T-cell epitopes.










HLA-A*0201 decamers





















8
I
L
L
E
S

L

L
G
C
V
30
Portion of SEQ ID NO: 3,



399
L
L
V
G
V

G

L
L
G
L
30
each start position is


156
I
L
T
L
S

A

I
A
T
L
29
specified - the length of


394
K
L
F
L
W

L

L
V
G
V
29
each peptide is 10 amino


12
S
L
L
G
C

V

S
W
S
L
28
acids, the end position for


41
G
L
E
G
F

S

I
A
F
L
28
each peptide is the start


126
F
I
L
G
Q

I

V
L
V
V
28
position plus nine.


127
I
L
G
Q
I

V

L
V
V
L
28


196
L
L
A
G
A

A

F
G
S
L
28


296
S
M
W
P
Q

T

L
G
H
L
27


559
K
L
Q
A
I

A

V
S
K
L
27


72
L
L
R
I
I

T

I
G
S
I
26


95
V
L
A
L
G

V

S
S
S
L
25


150
Q
M
S
N
K

V

I
L
T
L
25


356
M
M
L
I
I

G

L
N
M
L
25


396
F
L
W
L
L

V

G
V
G
L
25


28
M
I
Y
Y
F

P

L
Q
T
L
24


351
V
L
L
G
T

M

M
L
I
I
24


375
L
L
Q
T
K

N

S
S
K
V
24


413
K
A
Y
E
R

K

L
G
K
V
24


518
H
L
L
P
S

P

E
G
E
I
24


536
N
I
S
G
K

L

V
D
F
V
24


567
K
L
L
K
S

S

S
N
Q
V
24


614
Y
I
M
Y
R

G

L
I
R
L
24


69
M
L
T
L
L

R

I
I
T
I
23


97
A
L
G
V
S

S

S
L
I
V
23


122
R
I
W
G
F

I

L
G
Q
I
23


242
L
L
C
L
A

S

G
L
M
L
23


453
L
L
N
E
T

G

A
D
F
I
23


504
I
M
A
L
S

R

Y
P
I
V
23


619
G
L
I
R
L

G

Y
A
R
I
23


20
S
L
Y
H
D

L

G
P
M
I
22


33
P
L
Q
T
L

E

L
T
G
L
22


54
F
L
T
I
T

P

F
W
K
L
22


199
G
A
A
F
G

S

L
V
F
L
22


207
F
L
T
H
W

V

F
G
E
V
22


318
I
A
M
I
F

Y

L
L
E
I
22


526
E
I
A
P
A

I

T
L
T
V
22


591
Y
L
Q
L
T

E

H
G
N
V
22


46
S
I
A
F
L

S

P
I
F
L
21


49
F
L
S
P
I

F

L
T
I
T
21


137
R
I
W
Y
T

S

L
N
P
I
21


188
G
M
A
S
R

P

N
W
L
L
21


315
T
M
T
I
A

M

I
F
Y
L
21


532
T
L
T
V
N

I

S
G
K
L
21


3
S
L
W
R
E

I

L
L
E
S
20


4
L
W
R
E
I

L

L
E
S
L
20


63
L
V
N
K
K

W

M
L
T
L
20


89
A
K
L
R
L

M

V
L
A
L
20


142
S
L
N
P
I

W

S
Y
Q
M
20


275
L
L
Y
L
H

T

W
A
A
A
20


344
Y
A
R
E
R

S

D
V
L
L
20


392
Y
M
K
L
F

L

W
L
L
V
20


478
D
L
T
M
W

L

G
E
K
L
20


506
A
L
S
R
Y

P

I
V
K
S
20


576
V
I
F
L
G

Y

I
T
S
A
20


125
G
F
I
L
G

Q

I
V
L
V
19


132
V
L
V
V
L

R

I
W
Y
T
19


134
V
V
L
R
I

W

Y
T
S
L
19


240
A
V
L
L
C

L

A
S
G
L
19


261
G
L
I
W
W

V

T
G
T
A
19


269
T
A
S
A
A

G

L
L
Y
L
19


283
A
A
V
S
G

C

V
F
A
I
19


293
F
T
A
S
M

W

P
Q
T
L
19


404
G
L
L
G
L

G

L
R
H
K
19


405
L
L
G
L
G

L

R
H
K
A
19


503
G
I
M
A
L

S

R
Y
P
I
19


562
A
I
A
V
S

K

L
L
K
S
19


568
L
L
K
S
S

S

N
Q
V
I
19


30
Y
Y
F
P
L

Q

T
L
E
L
18


38
E
L
T
G
L

E

G
F
S
I
18


96
L
A
L
G
V

S

S
S
L
I
18


103
S
L
I
V
Q

A

V
T
W
W
18


249
L
M
L
P
S

C

L
W
F
R
18


254
C
L
W
F
R

G

T
G
L
I
18


266
V
T
G
T
A

S

A
A
G
L
18


309
G
T
N
P
G

K

T
M
T
I
18


363
N
M
L
F
G

P

K
K
N
L
18


397
L
W
L
L
V

G

V
G
L
L
18


55
L
T
I
T
P

F

W
K
L
V
17


75
I
I
T
I
G

S

I
A
S
F
17


80
S
I
A
S
F

Q

A
P
N
A
17


82
A
S
F
Q
A

P

N
A
K
L
17


90
K
L
R
L
M

V

L
A
L
G
17


161
A
I
A
T
L

D

R
I
G
T
17


200
A
A
F
G
S

L

V
F
L
T
17


241
V
L
L
C
L

A

S
G
L
M
17


274
G
L
L
Y
L

H

T
W
A
A
17


276
L
Y
L
H
T

W

A
A
A
V
17


316
M
T
I
A
M

I

F
Y
L
L
17


323
Y
L
L
E
I

F

F
C
A
W
17


421
K
V
A
P
T

K

E
V
S
A
17


445
S
S
L
E
R

S

A
H
L
L
17


622
R
L
G
Y
A

R

I
S
H
A
17


624
G
Y
A
R
I

S

H
A
E
L
17


84
F
Q
A
P
N

A

K
L
R
L
16


91
L
R
L
M
V

L

A
L
G
V
16


100
V
S
S
S
L

I

V
Q
A
V
16


129
G
Q
I
V
L

V

V
L
R
I
16


155
V
I
L
T
L

S

A
I
A
T
16


159
L
S
A
I
A

T

L
D
R
I
16


197
L
A
G
A
A

F

G
S
L
V
16


217
S
L
V
S
R

W

A
V
S
G
16


334
T
A
F
K
F

V

P
G
G
V
16


342
G
V
Y
A
R

E

R
S
D
V
16


352
L
L
G
T
M

M

L
I
I
G
16


353
L
G
T
M
M

L

I
I
G
L
16


374
L
L
L
Q
T

K

N
S
S
K
16


391
K
Y
M
K
L

F

L
W
L
L
16


482
W
L
G
E
K

L

G
F
Y
T
16


511
P
I
V
K
S

E

H
H
L
L
16


519
L
L
P
S
P

E

G
E
I
A
16


540
K
L
V
D
F

V

V
T
H
F
16


594
L
T
E
H
G

N

V
K
D
I
16


629
S
H
A
E
L

S

D
S
E
I
16


632
E
L
S
D
S

E

I
Q
M
A
16


16
C
V
S
W
S

L

Y
H
D
L
15


47
I
A
F
L
S

P

I
F
L
T
15


48
A
F
L
S
P

I

F
L
T
I
15


62
K
L
V
N
K

K

W
M
L
T
15


66
K
K
W
M
L

T

L
L
R
I
15


92
R
L
M
V
L

A

L
G
V
S
15


99
G
V
S
S
S

L

I
V
Q
A
15


108
A
V
T
W
W

S

G
S
H
L
15


209
T
H
W
V
F

G

E
V
S
L
15


245
L
A
S
G
L

M

L
P
S
C
15


253
S
C
L
W
F

R

G
T
G
L
15


257
F
R
G
T
G

L

I
W
W
V
15


271
S
A
A
G
L

L

Y
L
H
T
15


300
Q
T
L
G
H

L

I
N
S
G
15


301
T
L
G
H
L

I

N
S
G
T
15


319
A
M
I
F
Y

L

L
E
I
F
15


358
L
I
I
G
L

N

M
L
F
G
15


359
I
I
G
L
N

M

L
F
G
P
15


361
G
L
N
M
L

F

G
P
K
K
15


365
L
F
G
P
K

K

N
L
D
L
15


366
F
G
P
K
K

N

L
D
L
L
15


384
V
L
F
R
K

S

E
K
Y
M
15


401
V
G
V
G
L

L

G
L
G
L
15


456
E
T
G
A
D

F

I
T
I
L
15


498
R
Y
H
T
W

G

I
M
A
L
15


510
Y
P
I
V
K

S

E
H
H
L
15


533
L
T
V
N
I

S

G
K
L
V
15


554
D
D
L
D
R

K

L
Q
A
I
15


555
D
L
D
R
K

L

Q
A
I
A
15


570
K
S
S
S
N

Q

V
I
F
L
15


593
Q
L
T
E
H

G

N
V
K
D
15


615
I
M
Y
R
G

L

I
R
L
G
15


639
Q
M
A
K
F

R

I
P
D
D
15


659
V
I
D
H
R

E

V
S
E
K
15










HLA-A*0203 decamers





















275
L

L

Y
L
H
T

W

A
A
A
27
Portion of SEQ ID NO: 3,



192
R

P

N
W
L
L

A

G
A
A
19
each start position is


264
W

W

V
T
G
T

A

S
A
A
19
specified - the length of


274
G

L

L
Y
L
H

T

W
A
A
19
each peptide is 10 amino


422
V

A

P
T
K
E

V

S
A
A
19
acids, the end position for


154
K

V

I
L
T
L

S

A
I
A
18
each peptide is the start


191
S

R

P
N
W
L

L

A
G
A
18
position plus nine.


263
I

W

W
V
T
G

T

A
S
A
18


521
P

S

P
E
G
E

I

A
P
A
18


555
D

L

D
R
K
L

Q

A
I
A
18


193
P

N

W
L
L
A

G

A
A
F
17


265
W

V

T
G
T
A

S

A
A
G
17


276
L

Y

L
H
T
W

A

A
A
V
17


423
A

P

T
K
E
V

S

A
A
I
17










HLA-A1 decamers





















490
Y

T

D
F
G
P

S

T
R
Y
33
Portion of SEQ ID NO: 3,















each start position is














specified - the length of














each peptide is 10 amino














acids, the end position for














each peptide is the start














position plus nine.


490
Y

T

D
F
G
P

S

T
R
Y
33
Portion of SEQ ID NO: 3,


605
S

T

D
H
D
R

W

C
E
Y
33
each start position is


22
Y

H

D
L
G
P

M

I
Y
Y
31
specified - the length of


268
G

T

A
S
A
A

G

L
L
Y
30
each peptide is 10 amino


572
S

S

N
Q
V
I

F

L
G
Y
30
acids, the end position for


464
I

L

E
S
D
A

S

K
P
Y
26
each peptide is the start


314
K

T

M
T
I
A

M

I
F
Y
25
position plus nine.


140
Y

T

S
L
N
P

I

W
S
Y
23


616
M

Y

R
G
L
I

R

L
G
Y
23


446
S

L

E
R
S
A

H

L
L
N
21


582
I

T

S
A
P
G

S

R
D
Y
21


131
I

V

L
V
V
L

R

I
W
Y
20


345
A

R

E
R
S
D

V

L
L
G
20


388
K

S

E
K
Y
M

K

L
F
L
20


608
H

D

R
W
C
E

Y

I
M
Y
20


643
F

R

I
P
D
D

P

T
N
Y
20


13
L

L

G
C
V
S

W

S
L
Y
18


335
A

F

K
F
V
P

G

G
V
Y
18


665
V

S

E
K
I
H

F

N
P
R
18


669
I

H

F
N
P
R

F

G
S
Y
18


169
G

T

D
G
D
C

S

K
P
E
17


231
G

P

D
P
N
P

F

G
G
A
17


383
K

V

L
F
R
K

S

E
K
Y
17


430
A

A

I
W
P
F

R

F
G
Y
17


440
D

N

E
G
W
S

S

L
E
R
17


594
L

T

E
H
G
N

V

K
D
I
17


675
F

G

S
Y
K
E

G

H
N
Y
17


688
H

H

F
H
M
N

T

P
K
Y
17


111
W

W

S
G
S
H

L

Q
R
Y
16


348
R

S

D
V
L
L

G

T
M
M
16


406
L

G

L
G
L
R

H

K
A
Y
16


458
G

A

D
F
I
T

I

L
E
S
16


476
N

N

D
L
T
M

W

L
G
E
16


481
M

W

L
G
E
K

L

G
F
Y
16


553
E

D

D
L
D
R

K

L
Q
A
16










HLA-A26 decamers





















456
E
T
G
A
D
F
I
T
I
L
28
Portion of SEQ ID NO: 3,



284
A
V
S
G
C
V
F
A
I
F
27
each start position is


75
I
I
T
I
G
S
I
A
S
F
26
specified - the length of


140
Y
T
S
L
N
P
I
W
S
Y
25
each peptide is 10 amino


196
L
L
A
G
A
A
F
G
S
L
24
acids, the end position for


314
K
T
M
T
I
A
M
I
F
Y
24
each peptide is the start


316
M
T
I
A
M
I
F
Y
L
L
24
position plus nine.


540
K
L
V
D
F
V
V
T
H
F
24


28
M
I
Y
Y
F
P
L
Q
T
L
23


36
T
L
E
L
T
G
L
E
G
F
23


41
G
L
E
G
F
S
I
A
F
L
23


63
L
V
N
K
K
W
M
L
T
L
23


134
V
V
L
R
I
W
Y
T
S
L
23


399
L
L
V
G
V
G
L
L
G
L
23


478
D
L
T
M
W
L
G
E
K
L
23


614
Y
I
M
Y
R
G
L
I
R
L
23


16
C
V
S
W
S
L
Y
H
D
L
22


127
I
L
G
Q
I
V
L
V
V
L
22


268
G
T
A
S
A
A
G
L
L
Y
22


288
C
V
F
A
I
F
T
A
S
M
22


320
M
I
F
Y
L
L
E
I
F
F
22


377
Q
T
K
N
S
S
K
V
L
F
22


490
Y
T
D
F
G
P
S
T
R
Y
22


559
K
L
Q
A
I
A
V
S
K
L
22


605
S
T
D
H
D
R
W
C
E
Y
22


7
E
I
L
L
E
S
L
L
G
C
21


33
P
L
Q
T
L
E
L
T
G
L
21


54
F
L
T
I
T
P
F
W
K
L
21


156
I
L
T
L
S
A
I
A
T
L
21


293
F
T
A
S
M
W
P
Q
T
L
21


350
D
V
L
L
G
T
M
M
L
I
21


357
M
L
I
I
G
L
N
M
L
F
21


383
K
V
L
F
R
K
S
E
K
Y
21


632
E
L
S
D
S
E
I
Q
M
A
21


24
D
L
G
P
M
I
Y
Y
F
P
20


108
A
V
T
W
W
S
G
S
H
L
20


131
I
V
L
V
V
L
R
I
W
Y
20


236
P
F
G
G
A
V
L
L
C
L
20


240
A
V
L
L
C
L
A
S
G
L
20


266
V
T
G
T
A
S
A
A
G
L
20


582
I
T
S
A
P
G
S
R
D
Y
20


637
E
I
Q
M
A
K
F
R
I
P
20


12
S
L
L
G
C
V
S
W
S
L
19


13
L
L
G
C
V
S
W
S
L
Y
19


46
S
I
A
F
L
S
P
I
F
L
19


95
V
L
A
L
G
V
S
S
S
L
19


142
S
L
N
P
I
W
S
Y
Q
M
19


184
E
V
A
T
G
M
A
S
R
P
19


327
I
F
F
C
A
W
C
T
A
F
19


338
F
V
P
G
G
V
Y
A
R
E
19


347
E
R
S
D
V
L
L
G
T
M
19


452
H
L
L
N
E
T
G
A
D
F
19


464
I
L
E
S
D
A
S
K
P
Y
19


511
P
I
V
K
S
E
H
H
L
L
19


526
E
I
A
P
A
I
T
L
T
V
19


602
D
I
D
S
T
D
H
D
R
W
19


664
E
V
S
E
K
I
H
F
N
P
19


1
M
T
S
L
W
R
E
I
L
L
18


111
W
W
S
G
S
H
L
Q
R
Y
18


204
S
L
V
F
L
T
H
W
V
F
18


248
G
L
M
L
P
S
C
L
W
F
18


326
E
I
F
F
C
A
W
C
T
A
18


358
L
I
I
G
L
N
M
L
F
G
18


384
V
L
F
R
K
S
E
K
Y
M
18


390
E
K
Y
M
K
L
F
L
W
L
18


396
F
L
W
L
L
V
G
V
G
L
18


496
S
T
R
Y
H
T
W
G
I
M
18


535
V
N
I
S
G
K
L
V
D
F
18


634
S
D
S
E
I
Q
M
A
K
F
18


38
E
L
T
G
L
E
G
F
S
I
17


57
I
T
P
F
W
K
L
V
N
K
17


99
G
V
S
S
S
L
I
V
Q
A
17


122
R
I
W
G
F
I
L
G
Q
I
17


145
P
I
W
S
Y
Q
M
S
N
K
17


215
E
V
S
L
V
S
R
W
A
V
17


233
D
P
N
P
F
G
G
A
V
L
17


242
L
L
C
L
A
S
G
L
M
L
17


300
Q
T
L
G
H
L
I
N
S
G
17


333
C
T
A
F
K
F
V
P
G
G
17


335
A
F
K
F
V
P
G
G
V
Y
17


427
E
V
S
A
A
I
W
P
F
R
17


532
T
L
T
V
N
I
S
G
K
L
17


543
D
F
V
V
T
H
F
G
N
H
17


576
V
I
F
L
G
Y
I
T
S
A
17


659
V
I
D
H
R
E
V
S
E
K
17


669
I
H
F
N
P
R
F
G
S
Y
17


23
H
D
L
G
P
M
I
Y
Y
F
16


51
S
P
I
F
L
T
I
T
P
F
16


94
M
V
L
A
L
G
V
S
S
S
16


126
F
I
L
G
Q
I
V
L
V
V
16


218
L
V
S
R
W
A
V
S
G
H
16


241
V
L
L
C
L
A
S
G
L
M
16


279
H
T
W
A
A
A
V
S
G
C
16


291
A
I
F
T
A
S
M
W
P
Q
16


319
A
M
I
F
Y
L
L
E
I
F
16


365
L
F
G
P
K
K
N
L
D
L
16


386
F
R
K
S
E
K
Y
M
K
L
16


387
R
K
S
E
K
Y
M
K
L
F
16


394
K
L
F
L
W
L
L
V
G
V
16


431
A
I
W
P
F
R
F
G
Y
D
16


481
M
W
L
G
E
K
L
G
F
Y
16


501
T
W
G
I
M
A
L
S
R
Y
16


531
I
T
L
T
V
N
I
S
G
K
16


536
N
I
S
G
K
L
V
D
F
V
16


555
D
L
D
R
K
L
Q
A
I
A
16


562
A
I
A
V
S
K
L
L
K
S
16


572
S
S
N
Q
V
I
F
L
G
Y
16


575
Q
V
I
F
L
G
Y
I
T
S
16


619
G
L
I
R
L
G
Y
A
R
I
16


689
H
F
H
M
N
T
P
K
Y
F
16


4
L
W
R
E
I
L
L
E
S
L
15


22
Y
H
D
L
G
P
M
I
Y
Y
15


39
L
T
G
L
E
G
F
S
I
A
15


76
I
T
I
G
S
I
A
S
F
Q
15


87
P
N
A
K
L
R
L
M
V
L
15


117
L
Q
R
Y
L
R
I
W
G
F
15


296
S
M
W
P
Q
T
L
G
H
L
15


317
T
I
A
M
I
F
Y
L
L
E
15


323
Y
L
L
E
I
F
F
C
A
W
15


359
I
I
G
L
N
M
L
F
G
P
15


373
D
L
L
L
Q
T
K
N
S
S
15


404
G
L
L
G
L
G
L
R
H
K
15


421
K
V
A
P
T
K
E
V
S
A
15


426
K
E
V
S
A
A
I
W
P
F
15


460
D
F
I
T
I
L
E
S
D
A
15


484
G
E
K
L
G
F
Y
T
D
F
15


492
D
F
G
P
S
T
R
Y
H
T
15


607
D
H
D
R
W
C
E
Y
I
M
15


608
H
D
R
W
C
E
Y
I
M
Y
15


627
R
I
S
H
A
E
L
S
D
S
15


643
F
R
I
P
D
D
P
T
N
Y
15


658
V
V
I
D
H
R
E
V
S
E
15


8
I
L
L
E
S
L
L
G
C
V
14


43
E
G
F
S
I
A
F
L
S
P
14


49
F
L
S
P
I
F
L
T
I
T
14


64
V
N
K
K
W
M
L
T
L
L
14


70
L
T
L
L
R
I
I
T
I
G
14


80
S
I
A
S
F
Q
A
P
N
A
14


103
S
L
I
V
Q
A
V
T
W
W
14


130
Q
I
V
L
V
V
L
R
I
W
14


137
R
I
W
Y
T
S
L
N
P
I
14


199
G
A
A
F
G
S
L
V
F
L
14


211
W
V
F
G
E
V
S
L
V
S
14


212
V
F
G
E
V
S
L
V
S
R
14


223
A
V
S
G
H
P
H
P
G
P
14


244
C
L
A
S
G
L
M
L
P
S
14


265
W
V
T
G
T
A
S
A
A
G
14


351
V
L
L
G
T
M
M
L
I
I
14


354
G
T
M
M
L
I
I
G
L
N
14


366
F
G
P
K
K
N
L
D
L
L
14


400
L
V
G
V
G
L
L
G
L
G
14


402
G
V
G
L
L
G
L
G
L
R
14


424
P
T
K
E
V
S
A
A
I
W
14


462
I
T
I
L
E
S
D
A
S
K
14


463
T
I
L
E
S
D
A
S
K
P
14


498
R
Y
H
T
W
G
I
M
A
L
14


506
A
L
S
R
Y
P
I
V
K
S
14


524
E
G
E
I
A
P
A
I
T
L
14


585
A
P
G
S
R
D
Y
L
Q
L
14


594
L
T
E
H
G
N
V
K
D
I
14


622
R
L
G
Y
A
R
I
S
H
A
14


688
H
H
F
H
M
N
T
P
K
Y
14










HLA-A3 decamers


























374
L
L

L

Q
T

K


N

S
S
K





27
Portion of SEQ ID NO: 3,



417
R
K

L

G
K

V


A

P
T
K





25
each start position is


558
R
K

L

Q
A

I


A

V
S
K





25
specified - the length of


167
R
I

G

T
D

G


D

C
S
K





24
each peptide is 10 amino


404
G
L

L

G
L

G


L

R
H
K





24
acids, the end position for


361
G
L

N

M
L

F


G

P
K
K





23
each peptide is the start


409
G
L

R

H
K

A


Y

E
R
K





23
position plus nine.


421
K
V

A

P
T

K


E

V
S
A





23


452
H
L

L

N
E

T


G

A
D
F





23


131
I
V

L

V
V

L


R

I
W
Y





22


240
A
V

L

L
C

L


A

S
G
L





22


284
A
V

S

G
C

V


F

A
I
F





22


145
P
I

W

S
Y

Q


M

S
N
K





21


383
K
V

L

F
R

K


S

E
K
Y





21


462
I
T

I

L
E

S


D

A
S
K





21


659
V
I

D

H
R

E


V

S
E
K





21


75
I
I

T

I
G

S


I

A
S
F





20


90
K
L

R

L
M

V


L

A
L
G





20


127
I
L

G

Q
I

V


L

V
V
L





20


154
K
V

I

L
T

L


S

A
I
A





20


156
I
L

T

L
S

A


I

A
T
L





20


217
S
L

V

S
R

W


A

V
S
G





20


218
L
V

S

R
W

A


V

S
G
H





20


277
Y
L

H

T
W

A


A

A
V
S





20


342
G
V

Y

A
R

E


R

S
D
V





20


370
K
N

L

D
L

L


L

Q
T
K





20


506
A
L

S

R
Y

P


I

V
K
S





20


561
Q
A

I

A
V

S


K

L
L
K





20


658
V
V

I

D
H

R


E

V
S
E





20


13
L
L

G

C
V

S


W

S
L
Y





19


20
S
L

Y

H
D

L


G

P
M
I





19


92
R
L

M

V
L

A


L

G
V
S





19


94
M
V

L

A
L

G


V

S
S
S





19


108
A
V

T

W
W

S


G

S
H
L





19


126
F
I

L

G
Q

I


V

L
V
V





19


158
T
L

S

A
I

A


T

L
D
R





19


204
S
L

V

F
L

T


H

W
V
F





19


211
W
V

F

G
E

V


S

L
V
S





19


248
G
L

M

L
P

S


C

L
W
F





19


275
L
L

Y

L
H

T


W

A
A
A





19


357
M
L

I

I
G

L


N

M
L
F





19


526
E
I

A

P
A

I


T

L
T
V





19


540
K
L

V

D
F

V


V

T
H
F





19


575
Q
V

I

F
L

G


Y

I
T
S





19


592
L
Q

L

T
E

H


G

N
V
K





19


619
G
L

I

R
L

G


Y

A
R
I





19


657
K
V

V

I
D

H


R

E
V
S





19


670
H
F

N

P
R

F


G

S
Y
K





19


56
T
I

T

P
F

W


K

L
V
N





18


122
R
I

W

G
F

I


L

G
Q
I





18


134
V
V

L

R
I

W


Y

T
S
L





18


242
L
L

C

L
A

S


G

L
M
L





18


261
G
L

I

W
W

V


T

G
T
A





18


305
L
I

N

S
G

T


N

P
G
K





18


335
A
F

K

F
V

P


G

G
V
Y





18


394
K
L

F

L
W

L


L

V
G
V





18


396
F
L

W

L
L

V


G

V
G
L





18


398
W
L

L

V
G

V


G

L
L
G





18


464
I
L

E

S
D

A


S

K
P
Y





18


534
T
V

N

I
S

G


K

L
V
D





18


559
K
L

Q

A
I

A


V

S
K
L





18


564
A
V

S

K
L

L


K

S
S
S





18


567
K
L

L

K
S

S


S

N
Q
V





18


3
S
L

W

R
E

I


L

L
E
S





17


9
L
L

E

S
L

L


G

C
V
S





17


12
S
L

L

G
C

V


S

W
S
L





17


38
E
L

T

G
L

E


G

F
S
I





17


63
L
V

N

K
K

W


M

L
T
L





17


72
L
L

R

I
I

T


I

G
S
I





17


95
V
L

A

L
G

V


S

S
S
L





17


99
G
V

S

S
S

L


I

V
Q
A





17


135
V
L

R

I
W

Y


T

S
L
N





17


142
S
L

N

P
I

W


S

Y
Q
M





17


184
E
V

A

T
G

M


A

S
R
P





17


195
W
L

L

A
G

A


A

F
G
S





17


198
A
G

A

A
F

G


S

L
V
F





17


288
C
V

F

A
I

F


T

A
S
M





17


351
V
L

L

G
T

M


M

L
I
I





17


446
S
L

E

R
S

A


H

L
L
N





17


463
T
I

L

E
S

D


A

S
K
P





17


505
M
A

L

S
R

Y


P

I
V
K





17


531
I
T

L

T
V

N


I

S
G
K





17


593
Q
L

T

E
H

G


N

V
K
D





17


599
N
V

K

D
I

D


S

T
D
H





17


622
R
L

G

Y
A

R


I

S
H
A





17


644
R
I

P

D
D

P


T

N
Y
R





17


8
I
L

L

E
S

L


L

G
C
V





16


28
M
I

Y

Y
F

P


L

Q
T
L





16


53
I
F

L

T
I

T


P

F
W
K





16


57
I
T

P

F
W

K


L

V
N
K





16


74
R
I

I

T
I

G


S

I
A
S





16


97
A
L

G

V
S

S


S

L
I
V





16


103
S
L

I

V
Q

A


V

T
W
W





16


105
I
V

Q

A
V

T


W

W
S
G





16


196
L
L

A

G
A

A


F

G
S
L





16


265
W
V

T

G
T

A


S

A
A
G





16


358
L
I

I

G
L

N


M

L
F
G





16


360
I
G

L

N
M

L


F

G
P
K





16


402
G
V

G

L
L

G


L

G
L
R





16


427
E
V

S

A
A

I


W

P
F
R





16


512
I
V

K

S
E

H


H

L
L
P





16


518
H
L

L

P
S

P


E

G
E
I





16


555
D
L

D

R
K

L


Q

A
I
A





16


36
T
L

E

L
T

G


L

E
G
F





15


41
G
L

E

G
F

S


I

A
F
L





15


62
K
L

V

N
K

K


W

M
L
T





15


69
M
L

T

L
L

R


I

I
T
I





15


71
T
L

L

R
I

I


T

I
G
S





15


102
S
S

L

I
V

Q


A

V
T
W





15


116
H
L

Q

R
Y

L


R

I
W
G





15


133
L
V

V

L
R

I


W

Y
T
S





15


205
L
V

F

L
T

H


W

V
F
G





15


223
A
V

S

G
H

P


H

P
G
P





15


324
L
L

E

I
F

F


C

A
W
C





15


328
F
F

C

A
W

C


T

A
F
K





15


338
F
V

P

G
G

V


Y

A
R
E





15


373
D
L

L

L
Q

T


K

N
S
S





15


382
S
K

V

L
F

R


K

S
E
K





15


399
L
L

V

G
V

G


L

L
G
L





15


418
K
L

G

K
V

A


P

T
K
E





15


431
A
I

W

P
F

R


F

G
Y
D





15


508
S
R

Y

P
I

V


K

S
E
H





15


562
A
I

A

V
S

K


L

L
K
S





15


568
L
L

K

S
S

S


N

Q
V
I





15


616
M
Y

R

G
L

I


R

L
G
Y





15


618
R
G

L

I
R

L


G

Y
A
R





15


620
L
I

R

L
G

Y


A

R
I
S





15


627
R
I

S

H
A

E


L

S
D
S





15


7
E
I

L

L
E

S


L

L
G
C





14


49
F
L

S

P
I

F


L

T
I
T





14


81
I
A

S

F
Q

A


P

N
A
K





14


120
Y
L

R

I
W

G


F

I
L
G





14


155
V
I

L

T
L

S


A

I
A
T





14


201
A
F

G

S
L

V


F

L
T
H





14


244
C
L

A

S
G

L


M

L
P
S





14


281
W
A

A

A
V

S


G

C
V
F





14


304
H
L

I

N
S

G


T

N
P
G





14


323
Y
L

L

E
I

F


F

C
A
W





14


350
D
V

L

L
G

T


M

M
L
I





14


385
L
F

R

K
S

E


K

Y
M
K





14


407
G
L

G

L
R

H


K

A
Y
E





14


412
H
K

A

Y
E

R


K

L
G
K





14


453
L
L

N

E
T

G


A

D
F
I





14


482
W
L

G

E
K

L


G

F
Y
T





14


500
H
T

W

G
I

M


A

L
S
R





14


530
A
I

T

L
T

V


N

I
S
G





14


539
G
K

L

V
D

F


V

V
T
H





14


581
Y
I

T

S
A

P


G

S
R
D





14


643
F
R

I

P
D

D


P

T
N
Y





14


648
D
P

T

N
Y

R


D

N
Q
K





14










HLA-B*0702 decamers


























585
A
P
G
S
R
D
Y
L
Q
L





25
Portion of SEQ ID NO: 3,



233
D
P
N
P
F
G
G
A
V
L





23
each start position is


367
G
P
K
K
N
L
D
L
L
L





23
specified - the length of


86
A
P
N
A
K
L
R
L
M
V





22
each peptide is 10 amino


528
A
P
A
I
T
L
T
V
N
I





22
acids, the end position for


423
A
P
T
K
E
V
S
A
A
I





21
each peptide is the start


311
N
P
G
K
T
M
T
I
A
M





20
position plus nine.


510
Y
P
I
V
K
S
E
H
H
L





20


522
S
P
E
G
E
I
A
P
A
I





20


231
G
P
D
P
N
P
F
G
G
A





19


251
L
P
S
C
L
W
F
R
G
T





19


51
S
P
I
F
L
T
I
T
P
F





18


176
K
P
E
E
K
K
T
G
E
V





18


192
R
P
N
W
L
L
A
G
A
A





18


471
K
P
Y
M
G
N
N
D
L
T





17


89
A
K
L
R
L
M
V
L
A
L





16


127
I
L
G
Q
I
V
L
V
V
L





15


235
N
P
F
G
G
A
V
L
L
C





15


236
P
F
G
G
A
V
L
L
C
L





15


269
T
A
S
A
A
G
L
L
Y
L





15


388
K
S
E
K
Y
M
K
L
F
L





15


570
K
S
S
S
N
Q
V
I
F
L





15


30
Y
Y
F
P
L
Q
T
L
E
L





14


41
G
L
E
G
F
S
I
A
F
L





14


84
F
Q
A
P
N
A
K
L
R
L





14


150
Q
M
S
N
K
V
I
L
T
L





14


196
L
L
A
G
A
A
F
G
S
L





14


199
G
A
A
F
G
S
L
V
F
L





14


229
H
P
G
P
D
P
N
P
F
G





14


284
A
V
S
G
C
V
F
A
I
F





14


344
Y
A
R
E
R
S
D
V
L
L





14


365
L
F
G
P
K
K
N
L
D
L





14


390
E
K
Y
M
K
L
F
L
W
L





14


399
L
L
V
G
V
G
L
L
G
L





14


401
V
G
V
G
L
L
G
L
G
L





14


456
E
T
G
A
D
F
I
T
I
L





14


520
L
P
S
P
E
G
E
I
A
P





14


26
G
P
M
I
Y
Y
F
P
L
Q





13


63
L
V
N
K
K
W
M
L
T
L





13


82
A
S
F
Q
A
P
N
A
K
L





13


87
P
N
A
K
L
R
L
M
V
L





13


198
A
G
A
A
F
G
S
L
V
F





13


246
A
S
G
L
M
L
P
S
C
L





13


339
V
P
G
G
V
Y
A
R
E
R





13


391
K
Y
M
K
L
F
L
W
L
L





13


444
W
S
S
L
E
R
S
A
H
L





13


494
G
P
S
T
R
Y
H
T
W
G





13


559
K
L
Q
A
I
A
V
S
K
L





13


583
T
S
A
P
G
S
R
D
Y
L





13


1
M
T
S
L
W
R
E
I
L
L





12


4
L
W
R
E
I
L
L
E
S
L





12


12
S
L
L
G
C
V
S
W
S
L





12


16
C
V
S
W
S
L
Y
H
D
L





12


32
F
P
L
Q
T
L
E
L
T
G





12


46
S
I
A
F
L
S
P
I
F
L





12


64
V
N
K
K
W
M
L
T
L
L





12


108
A
V
T
W
W
S
G
S
H
L





12


126
F
I
L
G
Q
I
V
L
V
V





12


148
S
Y
Q
M
S
N
K
V
I
L





12


156
I
L
T
L
S
A
I
A
T
L





12


187
T
G
M
A
S
R
P
N
W
L





12


209
T
H
W
V
F
G
E
V
S
L





12


227
H
P
H
P
G
P
D
P
N
P





12


234
P
N
P
F
G
G
A
V
L
L





12


240
A
V
L
L
C
L
A
S
G
L





12


242
L
L
C
L
A
S
G
L
M
L





12


266
V
T
G
T
A
S
A
A
G
L





12


282
A
A
A
V
S
G
C
V
F
A





12


298
W
P
Q
T
L
G
H
L
I
N





12


316
M
T
I
A
M
I
F
Y
L
L





12


343
V
Y
A
R
E
R
S
D
V
L





12


376
L
Q
T
K
N
S
S
K
V
L





12


396
F
L
W
L
L
V
G
V
G
L





12


498
R
Y
H
T
W
G
I
M
A
L





12


526
E
I
A
P
A
I
T
L
T
V





12


536
N
I
S
G
K
L
V
D
F
V





12


547
T
H
F
G
N
H
E
D
D
L





12


624
G
Y
A
R
I
S
H
A
E
L





12


645
I
P
D
D
P
T
N
Y
R
D





12


672
N
P
R
F
G
S
Y
K
E
G





12


690
F
H
M
N
T
P
K
Y
F
L





12










HLA-B*4402 decamers


























426
K
E
V
S
A
A
I
W
P
F





24
Portion of SEQ ID NO: 3,



10
L
E
S
L
L
G
C
V
S
W





23
each start position is


389
S
E
K
Y
M
K
L
F
L
W





23
specified - the length of


455
N
E
T
G
A
D
F
I
T
I





23
each peptide is 10 amino


636
S
E
I
Q
M
A
K
F
R
I





22
acids, the end position for


484
G
E
K
L
G
F
Y
T
D
F





21
each peptide is the start


666
S
E
K
I
H
F
N
P
R
F





21
position plus nine.


612
C
E
Y
I
M
Y
R
G
L
I





20


89
A
K
L
R
L
M
V
L
A
L





19


30
Y
Y
F
P
L
Q
T
L
E
L





17


51
S
P
I
F
L
T
I
T
P
F





17


150
Q
M
S
N
K
V
I
L
T
L





17


272
A
A
G
L
L
Y
L
H
T
W





17


319
A
M
I
F
Y
L
L
E
I
F





17


430
A
A
I
W
P
F
R
F
G
Y





17


525
G
E
I
A
P
A
I
T
L
T





17


643
F
R
I
P
D
D
P
T
N
Y





17


6
R
E
I
L
L
E
S
L
L
G





16


82
A
S
F
Q
A
P
N
A
K
L





16


240
A
V
L
L
C
L
A
S
G
L





16


284
A
V
S
G
C
V
F
A
I
F





16


406
L
G
L
G
L
R
H
K
A
Y





16


498
R
Y
H
T
W
G
I
M
A
L





16


552
H
E
D
D
L
D
R
K
L
Q





16


631
A
E
L
S
D
S
E
I
Q
M





16


23
H
D
L
G
P
M
I
Y
Y
F





15


48
A
F
L
S
P
I
F
L
T
I





15


102
S
S
L
I
V
Q
A
V
T
W





15


103
S
L
I
V
Q
A
V
T
W
W





15


140
Y
T
S
L
N
P
I
W
S
Y





15


156
I
L
T
L
S
A
I
A
T
L





15


178
E
E
K
K
T
G
E
V
A
T





15


198
A
G
A
A
F
G
S
L
V
F





15


246
A
S
G
L
M
L
P
S
C
L





15


296
S
M
W
P
Q
T
L
G
H
L





15


316
M
T
I
A
M
I
F
Y
L
L





15


445
S
S
L
E
R
S
A
H
L
L





15


456
E
T
G
A
D
F
I
T
I
L





15


524
E
G
E
I
A
P
A
I
T
L





15


535
V
N
I
S
G
K
L
V
D
F





15


585
A
P
G
S
R
D
Y
L
Q
L





15


1
M
T
S
L
W
R
E
I
L
L





14


40
T
G
L
E
G
F
S
I
A
F





14


41
G
L
E
G
F
S
I
A
F
L





14


52
P
I
F
L
T
I
T
P
F
W





14


69
M
L
T
L
L
R
I
I
T
I





14


84
F
Q
A
P
N
A
K
L
R
L





14


115
S
H
L
Q
R
Y
L
R
I
W





14


124
W
G
F
I
L
G
Q
I
V
L





14


186
A
T
G
M
A
S
R
P
N
W





14


228
P
H
P
G
P
D
P
N
P
F





14


233
D
P
N
P
F
G
G
A
V
L





14


234
P
N
P
F
G
G
A
V
L
L





14


255
L
W
F
R
G
T
G
L
I
W





14


283
A
A
V
S
G
C
V
F
A
I





14


314
K
T
M
T
I
A
M
I
F
Y





14


323
Y
L
L
E
I
F
F
C
A
W





14


325
L
E
I
F
F
C
A
W
C
T





14


327
I
F
F
C
A
W
C
T
A
F





14


335
A
F
K
F
V
P
G
G
V
Y





14


353
L
G
T
M
M
L
I
I
G
L





14


357
M
L
I
I
G
L
N
M
L
F





14


363
N
M
L
F
G
P
K
K
N
L





14


365
L
F
G
P
K
K
N
L
D
L





14


387
R
K
S
E
K
Y
M
K
L
F





14


390
E
K
Y
M
K
L
F
L
W
L





14


391
K
Y
M
K
L
F
L
W
L
L





14


397
L
W
L
L
V
G
V
G
L
L





14


510
Y
P
I
V
K
S
E
H
H
L





14


551
N
H
E
D
D
L
D
R
K
L





14


570
K
S
S
S
N
Q
V
I
F
L





14


583
T
S
A
P
G
S
R
D
Y
L





14


663
R
E
V
S
E
K
I
H
F
N





14


669
I
H
F
N
P
R
F
G
S
Y





14


688
H
H
F
H
M
N
T
P
K
Y





14


12
S
L
L
G
C
V
S
W
S
L





13


22
Y
H
D
L
G
P
M
I
Y
Y





13


45
F
S
I
A
F
L
S
P
I
F





13


46
S
I
A
F
L
S
P
I
F
L





13


59
P
F
W
K
L
V
N
K
K
W





13


67
K
W
M
L
T
L
L
R
I
I





13


75
I
I
T
I
G
S
I
A
S
F





13


108
A
V
T
W
W
S
G
S
H
L





13


111
W
W
S
G
S
H
L
Q
R
Y





13


117
L
Q
R
Y
L
R
I
W
G
F





13


127
I
L
G
Q
I
V
L
V
V
L





13


129
G
Q
I
V
L
V
V
L
R
I





13


130
Q
I
V
L
V
V
L
R
I
W





13


131
I
V
L
V
V
L
R
I
W
Y





13


187
T
G
M
A
S
R
P
N
W
L





13


193
P
N
W
L
L
A
G
A
A
F





13


202
F
G
S
L
V
F
L
T
H
W





13


236
P
F
G
G
A
V
L
L
C
L





13


247
S
G
L
M
L
P
S
C
L
W





13


248
G
L
M
L
P
S
C
L
W
F





13


256
W
F
R
G
T
G
L
I
W
W





13


269
T
A
S
A
A
G
L
L
Y
L





13


309
G
T
N
P
G
K
T
M
T
I





13


315
T
M
T
I
A
M
I
F
Y
L





13


320
M
I
F
Y
L
L
E
I
F
F





13


349
S
D
V
L
L
G
T
M
M
L





13


356
M
M
L
I
I
G
L
N
M
L





13


366
F
G
P
K
K
N
L
D
L
L





13


376
L
Q
T
K
N
S
S
K
V
L





13


377
Q
T
K
N
S
S
K
V
L
F





13


383
K
V
L
F
R
K
S
E
K
Y





13


399
L
L
V
G
V
G
L
L
G
L





13


415
Y
E
R
K
L
G
K
V
A
P





13


423
A
P
T
K
E
V
S
A
A
I





13


435
F
R
F
G
Y
D
N
E
G
W





13


470
S
K
P
Y
M
G
N
N
D
L





13


481
M
W
L
G
E
K
L
G
F
Y





13


493
F
G
P
S
T
R
Y
H
T
W





13


522
S
P
E
G
E
I
A
P
A
I





13


528
A
P
A
I
T
L
T
V
N
I





13


540
K
L
V
D
F
V
V
T
H
F





13


547
T
H
F
G
N
H
E
D
D
L





13


554
D
D
L
D
R
K
L
Q
A
I





13


559
K
L
Q
A
I
A
V
S
K
L





13


560
L
Q
A
I
A
V
S
K
L
L





13


572
S
S
N
Q
V
I
F
L
G
Y





13


614
Y
I
M
Y
R
G
L
I
R
L





13


616
M
Y
R
G
L
I
R
L
G
Y





13


634
S
D
S
E
I
Q
M
A
K
F





13


684
Y
E
N
N
H
H
F
H
M
N





13


689
H
F
H
M
N
T
P
K
Y
F





13


16
C
V
S
W
S
L
Y
H
D
L





12


21
L
Y
H
D
L
G
P
M
I
Y





12


28
M
I
Y
Y
F
P
L
Q
T
L





12


33
P
L
Q
T
L
E
L
T
G
L





12


36
T
L
E
L
T
G
L
E
G
F





12


37
L
E
L
T
G
L
E
G
F
S





12


42
L
E
G
F
S
I
A
F
L
S





12


54
F
L
T
I
T
P
F
W
K
L





12


63
L
V
N
K
K
W
M
L
T
L





12


64
V
N
K
K
W
M
L
T
L
L





12


87
P
N
A
K
L
R
L
M
V
L





12


119
R
Y
L
R
I
W
G
F
I
L





12


134
V
V
L
R
I
W
Y
T
S
L





12


148
S
Y
Q
M
S
N
K
V
I
L





12


153
N
K
V
I
L
T
L
S
A
I





12


196
L
L
A
G
A
A
F
G
S
L





12


199
G
A
A
F
G
S
L
V
F
L





12


204
S
L
V
F
L
T
H
W
V
F





12


242
L
L
C
L
A
S
G
L
M
L





12


253
S
C
L
W
F
R
G
T
G
L





12


266
V
T
G
T
A
S
A
A
G
L





12


267
T
G
T
A
S
A
A
G
L
L





12


268
G
T
A
S
A
A
G
L
L
Y





12


293
F
T
A
S
M
W
P
Q
T
L





12


312
P
G
K
T
M
T
I
A
M
I





12


329
F
C
A
W
C
T
A
F
K
F





12


343
V
Y
A
R
E
R
S
D
V
L





12


344
Y
A
R
E
R
S
D
V
L
L





12


346
R
E
R
S
D
V
L
L
G
T





12


367
G
P
K
K
N
L
D
L
L
L





12


388
K
S
E
K
Y
M
K
L
F
L





12


396
F
L
W
L
L
V
G
V
G
L





12


401
V
G
V
G
L
L
G
L
G
L





12


410
L
R
H
K
A
Y
E
R
K
L





12


438
G
Y
D
N
E
G
W
S
S
L





12


441
N
E
G
W
S
S
L
E
R
S





12







HLA Class I decamer analysis of 125P5C8, variants 2-5. Listed are


scores which correlate with the ligation strength to a defined


HLA type for a sequence of amino acids. The algorithms used are


based on the book “MHC Ligands and peptide Motifs” by


H. G. Rammensee, J. Bachmann and S. Stevanovic. The probability


of being processed and presented is given in order to predict


T-cell epitopes.


variant 2 (aa 1-11)










HLA-A1 decamers


























2
P

S

L
W
R
E

I

L
L
E





12
Portion of SEQ ID NO: 5,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.










HLA-B*0702 decamers


























1
M
P
S
L
W
R
E
I
L
L





22
Portion of SEQ ID NO: 5,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.










HLA-B*4402 decamers


























1
M
P
S
L
W
R
E
I
L
L





14
Portion of SEQ ID NO: 5,



2
P
S
L
W
R
E
I
L
L
E





6
each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.







variant 3 (aa 673-691)










HLA-A1 decamers


























675
F

G

S
Y
K
E

G

P
N
Y





17
Portion of SEQ ID NO: 7,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.










HLA-B*0702 decamers


























681
G
P
N
Y
E
N
N
H
H
F





16
Portion of SEQ ID NO: 7,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.










HLA-B*4402 decamers


























679
K
E
G
P
N
Y
E
N
N
H





15
Portion of SEQ ID NO: 7,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.







variant 4 (aa 678-696)










HLA-A26 decamers


























687
N
T
H
H
F
H
M
N
T
P





14
Portion of SEQ ID NO: 9,




















each start position is



















specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.










HLA-B*4402 decamers


























67
K
E
G
H
N
Y
E
N
T
H





13
Portion of SEQ ID NO: 9,



684
Y
E
N
T
H
H
F
H
M
N





13
each start position is


681
G
H
N
Y
E
N
T
H
H
F





12
specified - the length of



















each peptide is 10 amino



















acids, the end position for



















each peptide is the start



















position plus nine.







variant 5 (aa 680-698)


No significant matches found



























Pos
1
2
3
4
5
6
7
8
9
0
1
2
3
4
5
score











HLA Class II analysis of 125P5C8 variant 1. Listed are



scores that fall within the top 50% (rounded up) of all


scores for the selected allele.










HLA-DRB1*0101 15 - mers


























394
K
L
F
L
W
L
L
V
G
V
G
L
L
G
L
35
Portion of SEQ



459
A
D
F
I
T
I
L
E
S
D
A
S
K
P
Y
35
ID NO: 3, each


36
T
L
E
L
T
G
L
E
G
F
S
I
A
F
L
34
start position


121
L
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
33
is specified -


137
R
I
W
Y
T
S
L
N
P
I
W
S
Y
Q
M
33
the length of


7
E
I
L
L
E
S
L
L
G
C
V
S
W
S
L
32
each peptide is


28
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
L
32
15 amino acids,


117
L
Q
R
Y
L
R
I
W
G
F
I
L
G
Q
I
32
the end


44
G
F
S
I
A
F
L
S
P
I
F
L
T
I
T
31
position for


144
N
P
I
W
S
Y
Q
M
S
N
K
V
I
L
T
31
each peptide is


260
T
G
L
I
W
W
V
T
G
T
A
S
A
A
G
31
the start


353
L
G
T
M
M
L
I
I
G
L
N
M
L
F
G
31
position plus


69
M
L
T
L
L
R
I
I
T
I
G
S
I
A
S
30
fourteen.


239
G
A
V
L
L
C
L
A
S
G
L
M
L
P
S
30


272
A
A
G
L
L
Y
L
H
T
W
A
A
A
V
S
30


72
L
L
R
I
I
T
I
G
S
I
A
S
F
Q
A
29


140
Y
T
S
L
N
P
I
W
S
Y
Q
M
S
N
K
29


31
Y
F
P
L
Q
T
L
E
L
T
G
L
E
G
F
28


75
I
I
T
I
G
S
I
A
S
F
Q
A
P
N
A
28


286
S
G
C
V
F
A
I
F
T
A
S
M
W
P
Q
28


389
S
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
28


554
D
D
L
D
R
K
L
Q
A
I
A
V
S
K
L
28


618
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
E
28


58
T
P
F
W
K
L
V
N
K
K
W
M
L
T
L
27


70
L
T
L
L
R
I
I
T
I
G
S
I
A
S
F
27


238
G
G
A
V
L
L
C
L
A
S
G
L
M
L
P
27


262
L
I
W
W
V
T
G
T
A
S
A
A
G
L
L
27


359
I
I
G
L
N
M
L
F
G
P
K
K
N
L
D
27


399
L
L
V
G
V
G
L
L
G
L
G
L
R
H
K
27


412
H
K
A
Y
E
R
K
L
G
K
V
A
P
T
K
27


476
N
N
D
L
T
M
W
L
G
E
K
L
G
F
Y
27


2
T
S
L
W
R
E
I
L
L
E
S
L
L
G
C
26


67
K
W
M
L
T
L
L
R
I
I
T
I
G
S
I
26


91
L
R
L
M
V
L
A
L
G
V
S
S
S
L
I
26


193
P
N
W
L
L
A
G
A
A
F
G
S
L
V
F
26


202
F
G
S
L
V
F
L
T
H
W
V
F
G
E
V
26


274
G
L
L
Y
L
H
T
W
A
A
A
V
S
G
C
26


295
A
S
M
W
P
Q
T
L
G
H
L
I
N
S
G
26


333
C
T
A
F
K
F
V
P
G
G
V
Y
A
R
E
26


354
G
T
M
M
L
I
I
G
L
N
M
L
F
G
P
26


395
L
F
L
W
L
L
V
G
V
G
L
L
G
L
G
26


397
L
W
L
L
V
G
V
G
L
L
G
L
G
L
R
26


451
A
H
L
L
N
E
T
G
A
D
F
I
T
I
L
26


562
A
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
26


573
S
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
26


19
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
P
25


89
A
K
L
R
L
M
V
L
A
L
G
V
S
S
S
25


92
R
L
M
V
L
A
L
G
V
S
S
S
L
I
V
25


106
V
Q
A
V
T
W
W
S
G
S
H
L
Q
R
Y
25


190
A
S
R
P
N
W
L
L
A
G
A
A
F
G
S
25


191
S
R
P
N
W
L
L
A
G
A
A
F
G
S
L
25


240
A
V
L
L
C
L
A
S
G
L
M
L
P
S
C
25


261
G
L
I
W
W
V
T
G
T
A
S
A
A
G
L
25


346
R
E
R
S
D
V
L
L
G
T
M
M
L
I
I
25


347
E
R
S
D
V
L
L
G
T
M
M
L
I
I
G
25


461
F
I
T
I
L
E
S
D
A
S
K
P
Y
M
G
25


507
L
S
R
Y
P
I
V
K
S
E
H
H
L
L
P
25


517
H
H
L
L
P
S
P
E
G
E
I
A
P
A
I
25


617
Y
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
25


18
S
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
24


78
I
G
S
I
A
S
F
Q
A
P
N
A
K
L
R
24


90
K
L
R
L
M
V
L
A
L
G
V
S
S
S
L
24


153
N
K
V
I
L
T
L
S
A
I
A
T
L
D
R
24


182
T
G
E
V
A
T
G
M
A
S
R
P
N
W
L
24


212
V
F
G
E
V
S
L
V
S
R
W
A
V
S
G
24


252
P
S
C
L
W
F
R
G
T
G
L
I
W
W
V
24


275
L
L
Y
L
H
T
W
A
A
A
V
S
G
C
V
24


278
L
H
T
W
A
A
A
V
S
G
C
V
F
A
I
24


318
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
24


341
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
T
24


370
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
24


416
E
R
K
L
G
K
V
A
P
T
K
E
V
S
A
24


450
S
A
H
L
L
N
E
T
G
A
D
F
I
T
I
24


458
G
A
D
F
I
T
I
L
E
S
D
A
S
K
P
24


501
T
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
24


530
A
I
T
L
T
V
N
I
S
G
K
L
V
D
F
24


531
I
T
L
T
V
N
I
S
G
K
L
V
D
F
V
24


565
V
S
K
L
L
K
S
S
S
N
Q
V
I
F
L
24


575
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
24


622
R
L
G
Y
A
R
I
S
H
A
E
L
S
D
S
24


93
L
M
V
L
A
L
G
V
S
S
S
L
I
V
Q
23


102
S
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
23


132
V
L
V
V
L
R
I
W
Y
T
S
L
N
P
I
23


152
S
N
K
V
I
L
T
L
S
A
I
A
T
L
D
23


162
I
A
T
L
D
R
I
G
T
D
G
D
C
S
K
23


213
F
G
E
V
S
L
V
S
R
W
A
V
S
G
H
23


371
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
23


373
D
L
L
L
Q
T
K
N
S
S
K
V
L
F
R
23


557
D
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
23


10
L
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
22


22
Y
H
D
L
G
P
M
I
Y
Y
F
P
L
Q
T
22


51
S
P
I
F
L
T
I
T
P
F
W
K
L
V
N
22


57
I
T
P
F
W
K
L
V
N
K
K
W
M
L
T
22


95
V
L
A
L
G
V
S
S
S
L
I
V
Q
A
V
22


124
W
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
22


125
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
W
22


206
V
F
L
T
H
W
V
F
G
E
V
S
L
V
S
22


209
T
H
W
V
F
G
E
V
S
L
V
S
R
W
A
22


259
G
T
G
L
I
W
W
V
T
G
T
A
S
A
A
22


279
H
T
W
A
A
A
V
S
G
C
V
F
A
I
F
22


294
T
A
S
M
W
P
Q
T
L
G
H
L
I
N
S
22


524
E
G
E
I
A
P
A
I
T
L
T
V
N
I
S
22


597
H
G
N
V
K
D
I
D
S
T
D
H
D
R
W
22


614
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
R
I
22


16
C
V
S
W
S
L
Y
H
D
L
G
P
M
I
Y
21


42
L
E
G
F
S
I
A
F
L
S
P
I
F
L
T
21


52
P
I
F
L
T
I
T
P
F
W
K
L
V
N
K
21


150
Q
M
S
N
K
V
I
L
T
L
S
A
I
A
T
21


208
L
T
H
W
V
F
G
E
V
S
L
V
S
R
W
21


244
C
L
A
S
G
L
M
L
P
S
C
L
W
F
R
21


287
G
C
V
F
A
I
F
T
A
S
M
W
P
Q
T
21


413
K
A
Y
E
R
K
L
G
K
V
A
P
T
K
E
21


539
G
K
L
V
D
F
V
V
T
H
F
G
N
H
E
21


577
I
F
L
G
Y
I
T
S
A
P
G
S
R
D
Y
21


623
L
G
Y
A
R
I
S
H
A
E
L
S
D
S
E
21


34
L
Q
T
L
E
L
T
G
L
E
G
F
S
I
A
20


65
N
K
K
W
M
L
T
L
L
R
I
I
T
I
G
20


73
L
R
I
I
T
I
G
S
I
A
S
F
Q
A
P
20


94
M
V
L
A
L
G
V
S
S
S
L
I
V
Q
A
20


115
S
H
L
Q
R
Y
L
R
I
W
G
F
I
L
G
20


122
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
L
20


151
M
S
N
K
V
I
L
T
L
S
A
I
A
T
L
20


156
I
L
T
L
S
A
I
A
T
L
D
R
I
G
T
20


178
E
E
K
K
T
G
E
V
A
T
G
M
A
S
R
20


179
E
K
K
T
G
E
V
A
T
G
M
A
S
R
P
20


192
R
P
N
W
L
L
A
G
A
A
F
G
S
L
V
20


291
A
I
F
T
A
S
M
W
P
Q
T
L
G
H
L
20


301
T
L
G
H
L
I
N
S
G
T
N
P
G
K
T
20


313
G
K
T
M
T
I
A
M
I
F
Y
L
L
E
I
20


319
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
20


426
K
E
V
S
A
A
I
W
P
F
R
F
G
Y
D
20


448
E
R
S
A
H
L
L
N
E
T
G
A
D
F
I
20


478
D
L
T
M
W
L
G
E
K
L
G
F
Y
T
D
20


509
R
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
20


576
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
D
20


588
S
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
20


649
P
T
N
Y
R
D
N
Q
K
V
V
I
D
H
R
20


655
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
I
20


3
S
L
W
R
E
I
L
L
E
S
L
L
G
C
V
19


61
W
K
L
V
N
K
K
W
M
L
T
L
L
R
I
19


71
T
L
L
R
I
I
T
I
G
S
I
A
S
F
Q
19


81
I
A
S
F
Q
A
P
N
A
K
L
R
L
M
V
19


123
I
W
G
F
I
L
G
Q
I
V
L
V
V
L
R
19


133
L
V
V
L
R
I
W
Y
T
S
L
N
P
I
W
19


146
I
W
S
Y
Q
M
S
N
K
V
I
L
T
L
S
19


219
V
S
R
W
A
V
S
G
H
P
H
P
G
P
D
19


251
L
P
S
C
L
W
F
R
G
T
G
L
I
W
W
19


320
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
T
19


325
L
E
I
F
F
C
A
W
C
T
A
F
K
F
V
19


355
T
M
M
L
I
I
G
L
N
M
L
F
G
P
K
19


357
M
L
I
I
G
L
N
M
L
F
G
P
K
K
N
19


369
K
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
19


392
Y
M
K
L
F
L
W
L
L
V
G
V
G
L
L
19


430
A
A
I
W
P
F
R
F
G
Y
D
N
E
G
W
19


436
R
F
G
Y
D
N
E
G
W
S
S
L
E
R
S
19


441
N
E
G
W
S
S
L
E
R
S
A
H
L
L
N
19


484
G
E
K
L
G
F
Y
T
D
F
G
P
S
T
R
19


496
S
T
R
Y
H
T
W
G
I
M
A
L
S
R
Y
19


499
Y
H
T
W
G
I
M
A
L
S
R
Y
P
I
V
19


522
S
P
E
G
E
I
A
P
A
I
T
L
T
V
N
19


611
W
C
E
Y
I
M
Y
R
G
L
I
R
L
G
Y
19


666
S
E
K
I
H
F
N
P
R
F
G
S
Y
K
E
19


668
K
I
H
F
N
P
R
F
G
S
Y
K
E
G
H
19


46
S
I
A
F
L
S
P
I
F
L
T
I
T
P
F
18


49
F
L
S
P
I
F
L
T
I
T
P
F
W
K
L
18


85
Q
A
P
N
A
K
L
R
L
M
V
L
A
L
G
18


131
I
V
L
V
V
L
R
I
W
Y
T
S
L
N
P
18


149
Y
Q
M
S
N
K
V
I
L
T
L
S
A
I
A
18


154
K
V
I
L
T
L
S
A
I
A
T
L
D
R
I
18


159
L
S
A
I
A
T
L
D
R
I
G
T
D
G
D
18


175
S
K
P
E
E
K
K
T
G
E
V
A
T
G
M
18


199
G
A
A
F
G
S
L
V
F
L
T
H
W
V
F
18


221
R
W
A
V
S
G
H
P
H
P
G
P
D
P
N
18


231
G
P
D
P
N
P
F
G
G
A
V
L
L
C
L
18


232
P
D
P
N
P
F
G
G
A
V
L
L
C
L
A
18


234
P
N
P
F
G
G
A
V
L
L
C
L
A
S
G
18


253
S
C
L
W
F
R
G
T
G
L
I
W
W
V
T
18


264
W
W
V
T
G
T
A
S
A
A
G
L
L
Y
L
18


299
P
Q
T
L
G
H
L
I
N
S
G
T
N
P
G
18


311
N
P
G
K
T
M
T
I
A
M
I
F
Y
L
L
18


329
F
C
A
W
C
T
A
F
K
F
V
P
G
G
V
18


332
W
C
T
A
F
K
F
V
P
G
G
V
Y
A
R
18


351
V
L
L
G
T
M
M
L
I
I
G
L
N
M
L
18


361
G
L
N
M
L
F
G
P
K
K
N
L
D
L
L
18


363
N
M
L
F
G
P
K
K
N
L
D
L
L
L
Q
18


374
L
L
L
Q
T
K
N
S
S
K
V
L
F
R
K
18


378
T
K
N
S
S
K
V
L
F
R
K
S
E
K
Y
18


390
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
G
18


393
M
K
L
F
L
W
L
L
V
G
V
G
L
L
G
18


400
L
V
G
V
G
L
L
G
L
G
L
R
H
K
A
18


402
G
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
18


403
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
R
18


434
P
F
R
F
G
Y
D
N
E
G
W
S
S
L
E
18


470
S
K
P
Y
M
G
N
N
D
L
T
M
W
L
G
18


486
K
L
G
F
Y
T
D
F
G
P
S
T
R
Y
H
18


487
L
G
F
Y
T
D
F
G
P
S
T
R
Y
H
T
18


535
V
N
I
S
G
K
L
V
D
F
V
V
T
H
F
18


538
S
G
K
L
V
D
F
V
V
T
H
F
G
N
H
18


558
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
S
18


574
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
18


589
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
I
18


633
L
S
D
S
E
I
Q
M
A
K
F
R
I
P
D
18


635
D
S
E
I
Q
M
A
K
F
R
I
P
D
D
P
18


681
G
H
N
Y
E
N
N
H
H
F
H
M
N
T
P
18


29
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
L
E
17


40
T
G
L
E
G
F
S
I
A
F
L
S
P
I
F
17


48
A
F
L
S
P
I
F
L
T
I
T
P
F
W
K
17


64
V
N
K
K
W
M
L
T
L
L
R
I
I
T
I
17


88
N
A
K
L
R
L
M
V
L
A
L
G
V
S
S
17


100
V
S
S
S
L
I
V
Q
A
V
T
W
W
S
G
17


128
L
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
17


130
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
N
17


205
L
V
F
L
T
H
W
V
F
G
E
V
S
L
V
17


215
E
V
S
L
V
S
R
W
A
V
S
G
H
P
H
17


246
A
S
G
L
M
L
P
S
C
L
W
F
R
G
T
17


265
W
V
T
G
T
A
S
A
A
G
L
L
Y
L
H
17


273
A
G
L
L
Y
L
H
T
W
A
A
A
V
S
G
17


305
L
I
N
S
G
T
N
P
G
K
T
M
T
I
A
17


317
T
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
17


324
L
L
E
I
F
F
C
A
W
C
T
A
F
K
F
17


327
I
F
F
C
A
W
C
T
A
F
K
F
V
P
G
17


334
T
A
F
K
F
V
P
G
G
V
Y
A
R
E
R
17


340
P
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
17


348
R
S
D
V
L
L
G
T
M
M
L
I
I
G
L
17


382
S
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
17


421
K
V
A
P
T
K
E
V
S
A
A
I
W
P
F
17


435
F
R
F
G
Y
D
N
E
G
W
S
S
L
E
R
17


493
F
G
P
S
T
R
Y
H
T
W
G
I
M
A
L
17


502
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
E
17


504
I
M
A
L
S
R
Y
P
I
V
K
S
E
H
H
17


510
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
E
17


526
E
I
A
P
A
I
T
L
T
V
N
I
S
G
K
17


542
V
D
F
V
V
T
H
F
G
N
H
E
D
D
L
17


560
L
Q
A
I
A
V
S
K
L
L
K
S
S
S
N
17


566
S
K
L
L
K
S
S
S
N
Q
V
I
F
L
G
17


579
L
G
Y
I
T
S
A
P
G
S
R
D
Y
L
Q
17


609
D
R
W
C
E
Y
I
M
Y
R
G
L
I
R
L
17


610
R
W
C
E
Y
I
M
Y
R
G
L
I
R
L
G
17


612
C
E
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
17


625
Y
A
R
I
S
H
A
E
L
S
D
S
E
I
Q
17


630
H
A
E
L
S
D
S
E
I
Q
M
A
K
F
R
17


1
M
T
S
L
W
R
E
I
L
L
E
S
L
L
G
16


4
L
W
R
E
I
L
L
E
S
L
L
G
C
V
S
16


6
R
E
I
L
L
E
S
L
L
G
C
V
S
W
S
16


14
L
G
C
V
S
W
S
L
Y
H
D
L
G
P
M
16


26
G
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
16


33
P
L
Q
T
L
E
L
T
G
L
E
G
F
S
I
16


39
L
T
G
L
E
G
F
S
I
A
F
L
S
P
I
16


66
K
K
W
M
L
T
L
L
R
I
I
T
I
G
S
16


87
P
N
A
K
L
R
L
M
V
L
A
L
G
V
S
16


97
A
L
G
V
S
S
S
L
I
V
Q
A
V
T
W
16


98
L
G
V
S
S
S
L
I
V
Q
A
V
T
W
W
16


120
Y
L
R
I
W
G
F
I
L
G
Q
I
V
L
V
16


127
I
L
G
Q
I
V
L
V
V
L
R
I
W
Y
T
16


135
V
L
R
I
W
Y
T
S
L
N
P
I
W
S
Y
16


148
S
Y
Q
M
S
N
K
V
I
L
T
L
S
A
I
16


164
T
L
D
R
I
G
T
D
G
D
C
S
K
P
E
16


180
K
K
T
G
E
V
A
T
G
M
A
S
R
P
N
16


186
A
T
G
M
A
S
R
P
N
W
L
L
A
G
A
16


194
N
W
L
L
A
G
A
A
F
G
S
L
V
F
L
16


195
W
L
L
A
G
A
A
F
G
S
L
V
F
L
T
16


210
H
W
V
F
G
E
V
S
L
V
S
R
W
A
V
16


217
S
L
V
S
R
W
A
V
S
G
H
P
H
P
G
16


223
A
V
S
G
H
P
H
P
G
P
D
P
N
P
F
16


230
P
G
P
D
P
N
P
F
G
G
A
V
L
L
C
16


236
P
F
G
G
A
V
L
L
C
L
A
S
G
L
M
16


242
L
L
C
L
A
S
G
L
M
L
P
S
C
L
W
16


282
A
A
A
V
S
G
C
V
F
A
I
F
T
A
S
16


309
G
T
N
P
G
K
T
M
T
I
A
M
I
F
Y
16


326
E
I
F
F
C
A
W
C
T
A
F
K
F
V
P
16


349
S
D
V
L
L
G
T
M
M
L
I
I
G
L
N
16


356
M
M
L
I
I
G
L
N
M
L
F
G
P
K
K
16


381
S
S
K
V
L
F
R
K
S
E
K
Y
M
K
L
16


396
F
L
W
L
L
V
G
V
G
L
L
G
L
G
L
16


417
R
K
L
G
K
V
A
P
T
K
E
V
S
A
A
16


419
L
G
K
V
A
P
T
K
E
V
S
A
A
I
W
16


423
A
P
T
K
E
V
S
A
A
I
W
P
F
R
F
16


443
G
W
S
S
L
E
R
S
A
H
L
L
N
E
T
16


480
T
M
W
L
G
E
K
L
G
F
Y
T
D
F
G
16


495
P
S
T
R
Y
H
T
W
G
I
M
A
L
S
R
16


500
H
T
W
G
I
M
A
L
S
R
Y
P
I
V
K
16


516
E
H
H
L
L
P
S
P
E
G
E
I
A
P
A
16


520
L
P
S
P
E
G
E
I
A
P
A
I
T
L
T
16


527
I
A
P
A
I
T
L
T
V
N
I
S
G
K
L
16


534
T
V
N
I
S
G
K
L
V
D
F
V
V
T
H
16


553
E
D
D
L
D
R
K
L
Q
A
I
A
V
S
K
16


555
D
L
D
R
K
L
Q
A
I
A
V
S
K
L
L
16


563
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
I
16


578
F
L
G
Y
I
T
S
A
P
G
S
R
D
Y
L
16


620
L
I
R
L
G
Y
A
R
I
S
H
A
E
L
S
16


627
R
I
S
H
A
E
L
S
D
S
E
I
Q
M
A
16


638
I
Q
M
A
K
F
R
I
P
D
D
P
T
N
Y
16


654
D
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
16


662
H
R
E
V
S
E
K
I
H
F
N
P
R
F
G
16


23
H
D
L
G
P
M
I
Y
Y
F
P
L
Q
T
L
15


47
I
A
F
L
S
P
I
F
L
T
I
T
P
F
W
15


50
L
S
P
I
F
L
T
I
T
P
F
W
K
L
V
15


101
S
S
S
L
I
V
Q
A
V
T
W
W
S
G
S
15


112
W
S
G
S
H
L
Q
R
Y
L
R
I
W
G
F
15


145
P
I
W
S
Y
Q
M
S
N
K
V
I
L
T
L
15


196
L
L
A
G
A
A
F
G
S
L
V
F
L
T
H
15


245
L
A
S
G
L
M
L
P
S
C
L
W
F
R
G
15


263
I
W
W
V
T
G
T
A
S
A
A
G
L
L
Y
15


288
C
V
F
A
I
F
T
A
S
M
W
P
Q
T
L
15


302
L
G
H
L
I
N
S
G
T
N
P
G
K
T
M
15


307
N
S
G
T
N
P
G
K
T
M
T
I
A
M
I
15


310
T
N
P
G
K
T
M
T
I
A
M
I
F
Y
L
15


315
T
M
T
I
A
M
I
F
Y
L
L
E
I
F
F
15


337
K
F
V
P
G
G
V
Y
A
R
E
R
S
D
V
15


383
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
L
15


422
V
A
P
T
K
E
V
S
A
A
I
W
P
F
R
15


442
E
G
W
S
S
L
E
R
S
A
H
L
L
N
E
15


456
E
T
G
A
D
F
I
T
I
L
E
S
D
A
S
15


462
I
T
I
L
E
S
D
A
S
K
P
Y
M
G
N
15


468
D
A
S
K
P
Y
M
G
N
N
D
L
T
M
W
15


472
P
Y
M
G
N
N
D
L
T
M
W
L
G
E
K
15


514
K
S
E
H
H
L
L
P
S
P
E
G
E
I
A
15


615
I
M
Y
R
G
L
I
R
L
G
Y
A
R
I
S
15


632
E
L
S
D
S
E
I
Q
M
A
K
F
R
I
P
15


640
M
A
K
F
R
I
P
D
D
P
T
N
Y
R
D
15


658
V
V
I
D
H
R
E
V
S
E
K
I
H
F
N
15


11
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
L
14


25
L
G
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
14


38
E
L
T
G
L
E
G
F
S
I
A
F
L
S
P
14


41
G
L
E
G
F
S
I
A
F
L
S
P
I
F
L
14


43
E
G
F
S
I
A
F
L
S
P
I
F
L
T
I
14


103
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
L
14


118
Q
R
Y
L
R
I
W
G
F
I
L
G
Q
I
V
14


129
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
14


134
V
V
L
R
I
W
Y
T
S
L
N
P
I
W
S
14


174
C
S
K
P
E
E
K
K
T
G
E
V
A
T
G
14


183
G
E
V
A
T
G
M
A
S
R
P
N
W
L
L
14


185
V
A
T
G
M
A
S
R
P
N
W
L
L
A
G
14


201
A
F
G
S
L
V
F
L
T
H
W
V
F
G
E
14


218
L
V
S
R
W
A
V
S
G
H
P
H
P
G
P
14


226
G
H
P
H
P
G
P
D
P
N
P
F
G
G
A
14


235
N
P
F
G
G
A
V
L
L
C
L
A
S
G
L
14


283
A
A
V
S
G
C
V
F
A
I
F
T
A
S
M
14


314
K
T
M
T
I
A
M
I
F
Y
L
L
E
I
F
14


321
I
F
Y
L
L
E
I
F
F
C
A
W
C
T
A
14


345
A
R
E
R
S
D
V
L
L
G
T
M
M
L
I
14


350
D
V
L
L
G
T
M
M
L
I
I
G
L
N
M
14


498
R
Y
H
T
W
G
I
M
A
L
S
R
Y
P
I
14


513
V
K
S
E
H
H
L
L
P
S
P
E
G
E
I
14


571
S
S
S
N
Q
V
I
F
L
G
Y
I
T
S
A
14


594
L
T
E
H
G
N
V
K
D
I
D
S
T
D
H
14


634
S
D
S
E
I
Q
M
A
K
F
R
I
P
D
D
14


642
K
F
R
I
P
D
D
P
T
N
Y
R
D
N
Q
14










HLA-DRB1*0301 (DR17)




15 - mers

























462
I
T
I
L
E
S
D
A
S
K
P
Y
M
G
N
29
Portion of SEQ



361
G
L
N
M
L
F
G
P
K
K
N
L
D
L
L
28
ID NO: 3, each


129
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
27
start position


186
A
T
G
M
A
S
R
P
N
W
L
L
A
G
A
27
is specified -


355
T
M
M
L
I
I
G
L
N
M
L
F
G
P
K
27
the length of


478
D
L
T
M
W
L
G
E
K
L
G
F
Y
T
D
27
each peptide is


655
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
I
27
15 amino acids,


318
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
25
the end


363
N
M
L
F
G
P
K
K
N
L
D
L
L
L
Q
25
position for


434
P
F
R
F
G
Y
D
N
E
G
W
S
S
L
E
24
each peptide is


238
G
G
A
V
L
L
C
L
A
S
G
L
M
L
P
22
the start


354
G
T
M
M
L
I
I
G
L
N
M
L
F
G
P
22
position plus


381
S
S
K
V
L
F
R
K
S
E
K
Y
M
K
L
22
fourteen.


476
N
N
D
L
T
M
W
L
G
E
K
L
G
F
Y
22


642
K
F
R
I
P
D
D
P
T
N
Y
R
D
N
Q
22


18
S
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
21


52
P
I
F
L
T
I
T
P
F
W
K
L
V
N
K
21


73
L
R
I
I
T
I
G
S
I
A
S
F
Q
A
P
21


154
K
V
I
L
T
L
S
A
I
A
T
L
D
R
I
21


165
L
D
R
I
G
T
D
G
D
C
S
K
P
E
E
21


240
A
V
L
L
C
L
A
S
G
L
M
L
P
S
C
21


246
A
S
G
L
M
L
P
S
C
L
W
F
R
G
T
21


397
L
W
L
L
V
G
V
G
L
L
G
L
G
L
R
21


450
S
A
H
L
L
N
E
T
G
A
D
F
I
T
I
21


509
R
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
21


591
Y
L
Q
L
T
E
H
G
N
V
K
D
I
D
S
21


641
A
K
F
R
I
P
D
D
P
T
N
Y
R
D
N
21


10
L
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
20


11
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
L
20


26
G
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
20


34
L
Q
T
L
E
L
T
G
L
E
G
F
S
I
A
20


39
L
T
G
L
E
G
F
S
I
A
F
L
S
P
I
20


106
V
Q
A
V
T
W
W
S
G
S
H
L
Q
R
Y
20


132
V
L
V
V
L
R
I
W
Y
T
S
L
N
P
I
20


317
T
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
20


371
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
20


394
K
L
F
L
W
L
L
V
G
V
G
L
L
G
L
20


516
E
H
H
L
L
P
S
P
E
G
E
I
A
P
A
20


530
A
I
T
L
T
V
N
I
S
G
K
L
V
D
F
20


549
F
G
N
H
E
D
D
L
D
R
K
L
Q
A
I
20


566
S
K
L
L
K
S
S
S
N
Q
V
I
F
L
G
20


629
S
H
A
E
L
S
D
S
E
I
Q
M
A
K
F
20


14
L
G
C
V
S
W
S
L
Y
H
D
L
G
P
M
19


31
Y
F
P
L
Q
T
L
E
L
T
G
L
E
G
F
19


44
G
F
S
I
A
F
L
S
P
I
F
L
T
I
T
19


54
F
L
T
I
T
P
F
W
K
L
V
N
K
K
W
19


61
W
K
L
V
N
K
K
W
M
L
T
L
L
R
I
19


66
K
K
W
M
L
T
L
L
R
I
I
T
I
G
S
19


93
L
M
V
L
A
L
G
V
S
S
S
L
I
V
Q
19


125
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
W
19


148
S
Y
Q
M
S
N
K
V
I
L
T
L
S
A
I
19


202
F
G
S
L
V
F
L
T
H
W
V
F
G
E
V
19


213
F
G
E
V
S
L
V
S
R
W
A
V
S
G
H
19


282
A
A
A
V
S
G
C
V
F
A
I
F
T
A
S
19


294
T
A
S
M
W
P
Q
T
L
G
H
L
I
N
S
19


313
G
K
T
M
T
I
A
M
I
F
Y
L
L
E
I
19


362
L
N
M
L
F
G
P
K
K
N
L
D
L
L
L
19


369
K
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
19


372
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
F
19


403
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
R
19


419
L
G
K
V
A
P
T
K
E
V
S
A
A
I
W
19


472
P
Y
M
G
N
N
D
L
T
M
W
L
G
E
K
19


538
S
G
K
L
V
D
F
V
V
T
H
F
G
N
H
19


557
D
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
19


612
C
E
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
19


194
N
W
L
L
A
G
A
A
F
G
S
L
V
F
L
18


227
H
P
H
P
G
P
D
P
N
P
F
G
G
A
V
18


333
C
T
A
F
K
F
V
P
G
G
V
Y
A
R
E
18


375
L
L
Q
T
K
N
S
S
K
V
L
F
R
K
S
18


405
L
L
G
L
G
L
R
H
K
A
Y
E
R
K
L
18


454
L
N
E
T
G
A
D
F
I
T
I
L
E
S
D
18


486
K
L
G
F
Y
T
D
F
G
P
S
T
R
Y
H
18


528
A
P
A
I
T
L
T
V
N
I
S
G
K
L
V
18


532
T
L
T
V
N
I
S
G
K
L
V
D
F
V
V
18


543
D
F
V
V
T
H
F
G
N
H
E
D
D
L
D
18


551
N
H
E
D
D
L
D
R
K
L
Q
A
I
A
V
18


553
E
D
D
L
D
R
K
L
Q
A
I
A
V
S
K
18


603
I
D
S
T
D
H
D
R
W
C
E
Y
I
M
Y
18


656
Q
K
V
V
I
D
H
R
E
V
S
E
K
I
H
18


59
P
F
W
K
L
V
N
K
K
W
M
L
T
L
L
17


80
S
I
A
S
F
Q
A
P
N
A
K
L
R
L
M
17


114
G
S
H
L
Q
R
Y
L
R
I
W
G
F
I
L
17


234
P
N
P
F
G
G
A
V
L
L
C
L
A
S
G
17


303
G
H
L
I
N
S
G
T
N
P
G
K
T
M
T
17


325
L
E
I
F
F
C
A
W
C
T
A
F
K
F
V
17


340
P
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
17


382
S
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
17


501
T
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
17


562
A
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
17


666
S
E
K
I
H
F
N
P
R
F
G
S
Y
K
E
17


3
S
L
W
R
E
I
L
L
E
S
L
L
G
C
V
16


82
A
S
F
Q
A
P
N
A
K
L
R
L
M
V
L
16


110
T
W
W
S
G
S
H
L
Q
R
Y
L
R
I
W
16


146
I
W
S
Y
Q
M
S
N
K
V
I
L
T
L
S
16


159
L
S
A
I
A
T
L
D
R
I
G
T
D
G
D
16


207
F
L
T
H
W
V
F
G
E
V
S
L
V
S
R
16


299
P
Q
T
L
G
H
L
I
N
S
G
T
N
P
G
16


383
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
L
16


458
G
A
D
F
I
T
I
L
E
S
D
A
S
K
P
16


468
D
A
S
K
P
Y
M
G
N
N
D
L
T
M
W
16


508
S
R
Y
P
I
V
K
S
E
H
H
L
L
P
S
16


567
K
L
L
K
S
S
S
N
Q
V
I
F
L
G
Y
16


581
Y
I
T
S
A
P
G
S
R
D
Y
L
Q
L
T
16


614
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
R
I
16


648
D
P
T
N
Y
R
D
N
Q
K
V
V
I
D
H
16


672
N
P
R
F
G
S
Y
K
E
G
H
N
Y
E
N
16


5
W
R
E
I
L
L
E
S
L
L
G
C
V
S
W
15


57
I
T
P
F
W
K
L
V
N
K
K
W
M
L
T
15


122
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
L
15


386
F
R
K
S
E
K
Y
M
K
L
F
L
W
L
L
15


390
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
G
15


408
L
G
L
R
H
K
A
Y
E
R
K
L
G
K
V
15


428
V
S
A
A
I
W
P
F
R
F
G
Y
D
N
E
15


490
Y
T
D
F
G
P
S
T
R
Y
H
T
W
G
I
15


660
I
D
H
R
E
V
S
E
K
I
H
F
N
P
R
15


664
E
V
S
E
K
I
H
F
N
P
R
F
G
S
Y
15










HLA-DRB1*0401




(DR4Dw4) 15-mers

























28
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
L
28
Portion of SEQ ID NO: 3,



278
L
H
T
W
A
A
A
V
S
G
C
V
F
A
I
28
each start position is


341
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
T
28
specified - the length of


622
R
L
G
Y
A
R
I
S
H
A
E
L
S
D
S
28
each peptide is 15 amino


66
K
K
W
M
L
T
L
L
R
I
I
T
I
G
S
26
acids, the end position


69
M
L
T
L
L
R
I
I
T
I
G
S
I
A
S
26
for each peptide is the


72
L
L
R
I
I
T
I
G
S
I
A
S
F
Q
A
26
start position plus


75
I
I
T
I
G
S
I
A
S
F
Q
A
P
N
A
26
fourteen.


102
S
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
26


140
Y
T
S
L
N
P
I
W
S
Y
Q
M
S
N
K
26


159
L
S
A
I
A
T
L
D
R
I
G
T
D
G
D
26


209
T
H
W
V
F
G
E
V
S
L
V
S
R
W
A
26


213
F
G
E
V
S
L
V
S
R
W
A
V
S
G
H
26


239
G
A
V
L
L
C
L
A
S
G
L
M
L
P
S
26


272
A
A
G
L
L
Y
L
H
T
W
A
A
A
V
S
26


286
S
G
C
V
F
A
I
F
T
A
S
M
W
P
Q
26


299
P
Q
T
L
G
H
L
I
N
S
G
T
N
P
G
26


369
K
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
26


459
A
D
F
I
T
I
L
E
S
D
A
S
K
P
Y
26


501
T
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
26


524
E
G
E
I
A
P
A
I
T
L
T
V
N
I
S
26


655
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
I
26


2
T
S
L
W
R
E
I
L
L
E
S
L
L
G
C
22


42
L
E
G
F
S
I
A
F
L
S
P
I
F
L
T
22


51
S
P
I
F
L
T
I
T
P
F
W
K
L
V
N
22


57
I
T
P
F
W
K
L
V
N
K
K
W
M
L
T
22


58
T
P
F
W
K
L
V
N
K
K
W
M
L
T
L
22


117
L
Q
R
Y
L
R
I
W
G
F
I
L
G
Q
I
22


121
L
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
22


137
R
I
W
Y
T
S
L
N
P
I
W
S
Y
Q
M
22


144
N
P
I
W
S
Y
Q
M
S
N
K
V
I
L
T
22


234
P
N
P
F
G
G
A
V
L
L
C
L
A
S
G
22


261
G
L
I
W
W
V
T
G
T
A
S
A
A
G
L
22


320
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
T
22


326
E
I
F
F
C
A
W
C
T
A
F
K
F
V
P
22


389
S
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
22


395
L
F
L
W
L
L
V
G
V
G
L
L
G
L
G
22


434
P
F
R
F
G
Y
D
N
E
G
W
S
S
L
E
22


436
R
F
G
Y
D
N
E
G
W
S
S
L
E
R
S
22


441
N
E
G
W
S
S
L
E
R
S
A
H
L
L
N
22


458
G
A
D
F
I
T
I
L
E
S
D
A
S
K
P
22


486
K
L
G
F
Y
T
D
F
G
P
S
T
R
Y
H
22


490
Y
T
D
F
G
P
S
T
R
Y
H
T
W
G
I
22


507
L
S
R
Y
P
I
V
K
S
E
H
H
L
L
P
22


588
S
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
22


614
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
R
I
22


675
F
G
S
Y
K
E
G
H
N
Y
E
N
N
H
H
22


681
G
H
N
Y
E
N
N
H
H
F
H
M
N
T
P
22


1
M
T
S
L
W
R
E
I
L
L
E
S
L
L
G
20


7
E
I
L
L
E
S
L
L
G
C
V
S
W
S
L
20


10
L
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
20


18
S
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
20


26
G
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
20


31
Y
F
P
L
Q
T
L
E
L
T
G
L
E
G
F
20


36
T
L
E
L
T
G
L
E
G
F
S
I
A
F
L
20


39
L
T
G
L
E
G
F
S
I
A
F
L
S
P
I
20


44
G
F
S
I
A
F
L
S
P
I
F
L
T
I
T
20


47
I
A
F
L
S
P
I
F
L
T
I
T
P
F
W
20


54
F
L
T
I
T
P
F
W
K
L
V
N
K
K
W
20


67
K
W
M
L
T
L
L
R
I
I
T
I
G
S
I
20


70
L
T
L
L
R
I
I
T
I
G
S
I
A
S
F
20


78
I
G
S
I
A
S
F
Q
A
P
N
A
K
L
R
20


90
K
L
R
L
M
V
L
A
L
G
V
S
S
S
L
20


91
L
R
L
M
V
L
A
L
G
V
S
S
S
L
I
20


92
R
L
M
V
L
A
L
G
V
S
S
S
L
I
V
20


93
L
M
V
L
A
L
G
V
S
S
S
L
I
V
Q
20


95
V
L
A
L
G
V
S
S
S
L
I
V
Q
A
V
20


120
Y
L
R
I
W
G
F
I
L
G
Q
I
V
L
V
20


125
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
W
20


129
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
20


130
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
N
20


132
V
L
V
V
L
R
I
W
Y
T
S
L
N
P
I
20


133
L
V
V
L
R
I
W
Y
T
S
L
N
P
I
W
20


152
S
N
K
V
I
L
T
L
S
A
I
A
T
L
D
20


153
N
K
V
I
L
T
L
S
A
I
A
T
L
D
R
20


156
I
L
T
L
S
A
I
A
T
L
D
R
I
G
T
20


162
I
A
T
L
D
R
I
G
T
D
G
D
C
S
K
20


165
L
D
R
I
G
T
D
G
D
C
S
K
P
E
E
20


194
N
W
L
L
A
G
A
A
F
G
S
L
V
F
L
20


202
F
G
S
L
V
F
L
T
H
W
V
F
G
E
V
20


240
A
V
L
L
C
L
A
S
G
L
M
L
P
S
C
20


259
G
T
G
L
I
W
W
V
T
G
T
A
S
A
A
20


260
T
G
L
I
W
W
V
T
G
T
A
S
A
A
G
20


289
V
F
A
I
F
T
A
S
M
W
P
Q
T
L
G
20


303
G
H
L
I
N
S
G
T
N
P
G
K
T
M
T
20


313
G
K
T
M
T
I
A
M
I
F
Y
L
L
E
I
20


353
L
G
T
M
M
L
I
I
G
L
N
M
L
F
G
20


354
G
T
M
M
L
I
I
G
L
N
M
L
F
G
P
20


359
I
I
G
L
N
M
L
F
G
P
K
K
N
L
D
20


372
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
F
20


390
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
G
20


394
K
L
F
L
W
L
L
V
G
V
G
L
L
G
L
20


397
L
W
L
L
V
G
V
G
L
L
G
L
G
L
R
20


402
G
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
20


403
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
R
20


444
W
S
S
L
E
R
S
A
H
L
L
N
E
T
G
20


462
I
T
I
L
E
S
D
A
S
K
P
Y
M
G
N
20


509
R
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
20


510
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
E
20


530
A
I
T
L
T
V
N
I
S
G
K
L
V
D
F
20


539
G
K
L
V
D
F
V
V
T
H
F
G
N
H
E
20


543
D
F
V
V
T
H
F
G
N
H
E
D
D
L
D
20


557
D
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
20


562
A
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
20


565
V
S
K
L
L
K
S
S
S
N
Q
V
I
F
L
20


566
S
K
L
L
K
S
S
S
N
Q
V
I
F
L
G
20


573
S
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
20


576
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
D
20


589
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
I
20


597
H
G
N
V
K
D
I
D
S
T
D
H
D
R
W
20


617
Y
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
20


620
L
I
R
L
G
Y
A
R
I
S
H
A
E
L
S
20


625
Y
A
R
I
S
H
A
E
L
S
D
S
E
I
Q
20


642
K
F
R
I
P
D
D
P
T
N
Y
R
D
N
Q
20


666
S
E
K
I
H
F
N
P
R
F
G
S
Y
K
E
20


80
S
I
A
S
F
Q
A
P
N
A
K
L
R
L
M
18


94
M
V
L
A
L
G
V
S
S
S
L
I
V
Q
A
18


99
G
V
S
S
S
L
I
V
Q
A
V
T
W
W
S
18


122
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
L
18


150
Q
M
S
N
K
V
I
L
T
L
S
A
I
A
T
18


179
E
K
K
T
G
E
V
A
T
G
M
A
S
R
P
18


201
A
F
G
S
L
V
F
L
T
H
W
V
F
G
E
18


212
V
F
G
E
V
S
L
V
S
R
W
A
V
S
G
18


296
S
M
W
P
Q
T
L
G
H
L
I
N
S
G
T
18


300
Q
T
L
G
H
L
I
N
S
G
T
N
P
G
K
18


347
E
R
S
D
V
L
L
G
T
M
M
L
I
I
G
18


370
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
18


417
R
K
L
G
K
V
A
P
T
K
E
V
S
A
A
18


421
K
V
A
P
T
K
E
V
S
A
A
I
W
P
F
18


442
E
G
W
S
S
L
E
R
S
A
H
L
L
N
E
18


472
P
Y
M
G
N
N
D
L
T
M
W
L
G
E
K
18


540
K
L
V
D
F
V
V
T
H
F
G
N
H
E
D
18


550
G
N
H
E
D
D
L
D
R
K
L
Q
A
I
A
18


558
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
S
18


563
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
I
18


631
A
E
L
S
D
S
E
I
Q
M
A
K
F
R
I
18


648
D
P
T
N
Y
R
D
N
Q
K
V
V
I
D
H
18


654
D
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
18


658
V
V
I
D
H
R
E
V
S
E
K
I
H
F
N
18


363
N
M
L
F
G
P
K
K
N
L
D
L
L
L
Q
17


19
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
P
16


27
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
16


29
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
L
E
16


46
S
I
A
F
L
S
P
I
F
L
T
I
T
P
F
16


65
N
K
K
W
M
L
T
L
L
R
I
I
T
I
G
16


108
A
V
T
W
W
S
G
S
H
L
Q
R
Y
L
R
16


123
I
W
G
F
I
L
G
Q
I
V
L
V
V
L
R
16


136
L
R
I
W
Y
T
S
L
N
P
I
W
S
Y
Q
16


146
I
W
S
Y
Q
M
S
N
K
V
I
L
T
L
S
16


192
R
P
N
W
L
L
A
G
A
A
F
G
S
L
V
16


199
G
A
A
F
G
S
L
V
F
L
T
H
W
V
F
16


208
L
T
H
W
V
F
G
E
V
S
L
V
S
R
W
16


210
H
W
V
F
G
E
V
S
L
V
S
R
W
A
V
16


219
V
S
R
W
A
V
S
G
H
P
H
P
G
P
D
16


262
L
I
W
W
V
T
G
T
A
S
A
A
G
L
L
16


274
G
L
L
Y
L
H
T
W
A
A
A
V
S
G
C
16


287
G
C
V
F
A
I
F
T
A
S
M
W
P
Q
T
16


290
F
A
I
F
T
A
S
M
W
P
Q
T
L
G
H
16


295
A
S
M
W
P
Q
T
L
G
H
L
I
N
S
G
16


319
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
16


325
L
E
I
F
F
C
A
W
C
T
A
F
K
F
V
16


329
F
C
A
W
C
T
A
F
K
F
V
P
G
G
V
16


333
C
T
A
F
K
F
V
P
G
G
V
Y
A
R
E
16


383
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
L
16


393
M
K
L
F
L
W
L
L
V
G
V
G
L
L
G
16


470
S
K
P
Y
M
G
N
N
D
L
T
M
W
L
G
16


479
L
T
M
W
L
G
E
K
L
G
F
Y
T
D
F
16


487
L
G
F
Y
T
D
F
G
P
S
T
R
Y
H
T
16


496
S
T
R
Y
H
T
W
G
I
M
A
L
S
R
Y
16


575
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
16


640
M
A
K
F
R
I
P
D
D
P
T
N
Y
R
D
16


649
P
T
N
Y
R
D
N
Q
K
V
V
I
D
H
R
16


672
N
P
R
F
G
S
Y
K
E
G
H
N
Y
E
N
16


186
A
T
G
M
A
S
R
P
N
W
L
L
A
G
A
15


252
P
S
C
L
W
F
R
G
T
G
L
I
W
W
V
15


340
P
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
15


373
D
L
L
L
Q
T
K
N
S
S
K
V
L
F
R
15


553
E
D
D
L
D
R
K
L
Q
A
I
A
V
S
K
15


5
W
R
E
I
L
L
E
S
L
L
G
C
V
S
W
14


6
R
E
I
L
L
E
S
L
L
G
C
V
S
W
S
14


11
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
L
14


14
L
G
C
V
S
W
S
L
Y
H
D
L
G
P
M
14


25
L
G
P
M
I
Y
Y
F
P
L
Q
T
L
E
L
14


50
L
S
P
I
F
L
T
I
T
P
F
W
K
L
V
14


52
P
I
F
L
T
I
T
P
F
W
K
L
V
N
K
14


97
A
L
G
V
S
S
S
L
I
V
Q
A
V
T
W
14


103
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
L
14


106
V
Q
A
V
T
W
W
S
G
S
H
L
Q
R
Y
14


114
G
S
H
L
Q
R
Y
L
R
I
W
G
F
I
L
14


124
W
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
14


128
L
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
14


135
V
L
R
I
W
Y
T
S
L
N
P
I
W
S
Y
14


143
L
N
P
I
W
S
Y
Q
M
S
N
K
V
I
L
14


154
K
V
I
L
T
L
S
A
I
A
T
L
D
R
I
14


193
P
N
W
L
L
A
G
A
A
F
G
S
L
V
F
14


205
L
V
F
L
T
H
W
V
F
G
E
V
S
L
V
14


221
R
W
A
V
S
G
H
P
H
P
G
P
D
P
N
14


238
G
G
A
V
L
L
C
L
A
S
G
L
M
L
P
14


242
L
L
C
L
A
S
G
L
M
L
P
S
C
L
W
14


263
I
W
W
V
T
G
T
A
S
A
A
G
L
L
Y
14


273
A
G
L
L
Y
L
H
T
W
A
A
A
V
S
G
14


275
L
L
Y
L
H
T
W
A
A
A
V
S
G
C
V
14


294
T
A
S
M
W
P
Q
T
L
G
H
L
I
N
S
14


302
L
G
H
L
I
N
S
G
T
N
P
G
K
T
M
14


315
T
M
T
I
A
M
I
F
Y
L
L
E
I
F
F
14


318
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
14


321
I
F
Y
L
L
E
I
F
F
C
A
W
C
T
A
14


322
F
Y
L
L
E
I
F
F
C
A
W
C
T
A
F
14


324
L
L
E
I
F
F
C
A
W
C
T
A
F
K
F
14


348
R
S
D
V
L
L
G
T
M
M
L
I
I
G
L
14


349
S
D
V
L
L
G
T
M
M
L
I
I
G
L
N
14


355
T
M
M
L
I
I
G
L
N
M
L
F
G
P
K
14


356
M
M
L
I
I
G
L
N
M
L
F
G
P
K
K
14


361
G
L
N
M
L
F
G
P
K
K
N
L
D
L
L
14


371
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
14


392
Y
M
K
L
F
L
W
L
L
V
G
V
G
L
L
14


400
L
V
G
V
G
L
L
G
L
G
L
R
H
K
A
14


416
E
R
K
L
G
K
V
A
P
T
K
E
V
S
A
14


419
L
G
K
V
A
P
T
K
E
V
S
A
A
I
W
14


425
T
K
E
V
S
A
A
I
W
P
F
R
F
G
Y
14


429
S
A
A
I
W
P
F
R
F
G
Y
D
N
E
G
14


450
S
A
H
L
L
N
E
T
G
A
D
F
I
T
I
14


451
A
H
L
L
N
E
T
G
A
D
F
I
T
I
L
14


476
N
N
D
L
T
M
W
L
G
E
K
L
G
F
Y
14


502
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
E
14


504
I
M
A
L
S
R
Y
P
I
V
K
S
E
H
H
14


516
E
H
H
L
L
P
S
P
E
G
E
I
A
P
A
14


517
H
H
L
L
P
S
P
E
G
E
I
A
P
A
I
14


528
A
P
A
I
T
L
T
V
N
I
S
G
K
L
V
14


538
S
G
K
L
V
D
F
V
V
T
H
F
G
N
H
14


560
L
Q
A
I
A
V
S
K
L
L
K
S
S
S
N
14


574
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
14


579
L
G
Y
I
T
S
A
P
G
S
R
D
Y
L
Q
14


591
Y
L
Q
L
T
E
H
G
N
V
K
D
I
D
S
14


600
V
K
D
I
D
S
T
D
H
D
R
W
C
E
Y
14


618
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
E
14


630
H
A
E
L
S
D
S
E
I
Q
M
A
K
F
R
14


635
D
S
E
I
Q
M
A
K
F
R
I
P
D
D
P
14


637
E
I
Q
M
A
K
F
R
I
P
D
D
P
T
N
14










HLA-DRB1*1101 15-mers


























562
A
I
A
V
S
K
L
L
K
S
S
S
N
Q
V
26
Portion of SEQ ID NO: 3,



219
V
S
R
W
A
V
S
G
H
P
H
P
G
P
D
25
each start position is


329
F
C
A
W
C
T
A
F
K
F
V
P
G
G
V
25
specified - the length of


58
T
P
F
W
K
L
V
N
K
K
W
M
L
T
L
24
each peptide is 15 amino


137
R
I
W
Y
T
S
L
N
P
I
W
S
Y
Q
M
24
acids, the end position


441
N
E
G
W
S
S
L
E
R
S
A
H
L
L
N
24
for each peptide is the


614
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
R
I
24
start position plus


622
R
L
G
Y
A
R
I
S
H
A
E
L
S
D
S
24
fourteen.


393
M
K
L
F
L
W
L
L
V
G
V
G
L
L
G
23


458
G
A
D
F
I
T
I
L
E
S
D
A
S
K
P
22


15
G
C
V
S
W
S
L
Y
H
D
L
G
P
M
I
21


66
K
K
W
M
L
T
L
L
R
I
I
T
I
G
S
21


88
N
A
K
L
R
L
M
V
L
A
L
G
V
S
S
21


129
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
21


202
F
G
S
L
V
F
L
T
H
W
V
F
G
E
V
21


403
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
R
21


510
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
E
21


655
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
I
21


90
K
L
R
L
M
V
L
A
L
G
V
S
S
S
L
20


114
G
S
H
L
Q
R
Y
L
R
I
W
G
F
I
L
20


159
L
S
A
I
A
T
L
D
R
I
G
T
D
G
D
20


213
F
G
E
V
S
L
V
S
R
W
A
V
S
G
H
20


221
R
W
A
V
S
G
H
P
H
P
G
P
D
P
N
20


371
N
L
D
L
L
L
Q
T
K
N
S
S
K
V
L
20


413
K
A
Y
E
R
K
L
G
K
V
A
P
T
K
E
20


459
A
D
F
I
T
I
L
E
S
D
A
S
K
P
Y
20


501
T
W
G
I
M
A
L
S
R
Y
P
I
V
K
S
20


539
G
K
L
V
D
F
V
V
T
H
F
G
N
H
E
20


662
H
R
E
V
S
E
K
I
H
F
N
P
R
F
G
20


666
S
E
K
I
H
F
N
P
R
F
G
S
Y
K
E
20


7
E
I
L
L
E
S
L
L
G
C
V
S
W
S
L
19


57
I
T
P
F
W
K
L
V
N
K
K
W
M
L
T
19


146
I
W
S
Y
Q
M
S
N
K
V
I
L
T
L
S
19


350
D
V
L
L
G
T
M
M
L
I
I
G
L
N
M
19


649
P
T
N
Y
R
D
N
Q
K
V
V
I
D
H
R
19


22
Y
H
D
L
G
P
M
I
Y
Y
F
P
L
Q
T
18


69
M
L
T
L
L
R
I
I
T
I
G
S
I
A
S
18


72
L
L
R
I
I
T
I
G
S
I
A
S
F
Q
A
18


75
I
I
T
I
G
S
I
A
S
F
Q
A
P
N
A
18


92
R
L
M
V
L
A
L
G
V
S
S
S
L
I
V
18


103
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
L
18


108
A
V
T
W
W
S
G
S
H
L
Q
R
Y
L
R
18


199
G
A
A
F
G
S
L
V
F
L
T
H
W
V
F
18


260
T
G
L
I
W
W
V
T
G
T
A
S
A
A
G
18


272
A
A
G
L
L
Y
L
H
T
W
A
A
A
V
S
18


356
M
M
L
I
I
G
L
N
M
L
F
G
P
K
K
18


383
K
V
L
F
R
K
S
E
K
Y
M
K
L
F
L
18


490
Y
T
D
F
G
P
S
T
R
Y
H
T
W
G
I
18


499
Y
H
T
W
G
I
M
A
L
S
R
Y
P
I
V
18


573
S
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
18


576
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
D
18


608
H
D
R
W
C
E
Y
I
M
Y
R
G
L
I
R
18


681
G
H
N
Y
E
N
N
H
H
F
H
M
N
T
P
18


19
W
S
L
Y
H
D
L
G
P
M
I
Y
Y
F
P
17


28
M
I
Y
Y
F
P
L
Q
T
L
E
L
T
G
L
17


54
F
L
T
I
T
P
F
W
K
L
V
N
K
K
W
17


117
L
Q
R
Y
L
R
I
W
G
F
I
L
G
Q
I
17


121
L
R
I
W
G
F
I
L
G
Q
I
V
L
V
V
17


261
G
L
I
W
W
V
T
G
T
A
S
A
A
G
L
17


333
C
T
A
F
K
F
V
P
G
G
V
Y
A
R
E
17


338
F
V
P
G
G
V
Y
A
R
E
R
S
D
V
L
17


389
S
E
K
Y
M
K
L
F
L
W
L
L
V
G
V
17


395
L
F
L
W
L
L
V
G
V
G
L
L
G
L
G
17


412
H
K
A
Y
E
R
K
L
G
K
V
A
P
T
K
17


2
T
S
L
W
R
E
I
L
L
E
S
L
L
G
C
16


16
C
V
S
W
S
L
Y
H
D
L
G
P
M
I
Y
16


51
S
P
I
F
L
T
I
T
P
F
W
K
L
V
N
16


128
L
G
Q
I
V
L
V
V
L
R
I
W
Y
T
S
16


192
R
P
N
W
L
L
A
G
A
A
F
G
S
L
V
16


209
T
H
W
V
F
G
E
V
S
L
V
S
R
W
A
16


210
H
W
V
F
G
E
V
S
L
V
S
R
W
A
V
16


262
L
I
W
W
V
T
G
T
A
S
A
A
G
L
L
16


319
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
16


320
M
I
F
Y
L
L
E
I
F
F
C
A
W
C
T
16


362
L
N
M
L
F
G
P
K
K
N
L
D
L
L
L
16


396
F
L
W
L
L
V
G
V
G
L
L
G
L
G
L
16


432
I
W
P
F
R
F
G
Y
D
N
E
G
W
S
S
16


434
P
F
R
F
G
Y
D
N
E
G
W
S
S
L
E
16


506
A
L
S
R
Y
P
I
V
K
S
E
H
H
L
L
16


507
L
S
R
Y
P
I
V
K
S
E
H
H
L
L
P
16


528
A
P
A
I
T
L
T
V
N
I
S
G
K
L
V
16


540
K
L
V
D
F
V
V
T
H
F
G
N
H
E
D
16


575
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
R
16


588
S
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
16


600
V
K
D
I
D
S
T
D
H
D
R
W
C
E
Y
16


634
S
D
S
E
I
Q
M
A
K
F
R
I
P
D
D
16


654
D
N
Q
K
V
V
I
D
H
R
E
V
S
E
K
16


111
W
W
S
G
S
H
L
Q
R
Y
L
R
I
W
G
15


125
G
F
I
L
G
Q
I
V
L
V
V
L
R
I
W
15


212
V
F
G
E
V
S
L
V
S
R
W
A
V
S
G
15


296
S
M
W
P
Q
T
L
G
H
L
I
N
S
G
T
15


379
K
N
S
S
K
V
L
F
R
K
S
E
K
Y
M
15


409
G
L
R
H
K
A
Y
E
R
K
L
G
K
V
A
15


418
K
L
G
K
V
A
P
T
K
E
V
S
A
A
I
15


444
W
S
S
L
E
R
S
A
H
L
L
N
E
T
G
15


509
R
Y
P
I
V
K
S
E
H
H
L
L
P
S
P
15


532
T
L
T
V
N
I
S
G
K
L
V
D
F
V
V
15


550
G
N
H
E
D
D
L
D
R
K
L
Q
A
I
A
15


559
K
L
Q
A
I
A
V
S
K
L
L
K
S
S
S
15


610
R
W
C
E
Y
I
M
Y
R
G
L
I
R
L
G
15


36
T
L
E
L
T
G
L
E
G
F
S
I
A
F
L
14


67
K
W
M
L
T
L
L
R
I
I
T
I
G
S
I
14


102
S
S
L
I
V
Q
A
V
T
W
W
S
G
S
H
14


172
G
D
C
S
K
P
E
E
K
K
T
G
E
V
A
14


182
T
G
E
V
A
T
G
M
A
S
R
P
N
W
L
14


248
G
L
M
L
P
S
C
L
W
F
R
G
T
G
L
14


250
M
L
P
S
C
L
W
F
R
G
T
G
L
I
W
14


259
G
T
G
L
I
W
W
V
T
G
T
A
S
A
A
14


263
I
W
W
V
T
G
T
A
S
A
A
G
L
L
Y
14


271
S
A
A
G
L
L
Y
L
H
T
W
A
A
A
V
14


340
P
G
G
V
Y
A
R
E
R
S
D
V
L
L
G
14


361
G
L
N
M
L
F
G
P
K
K
N
L
D
L
L
14


378
T
K
N
S
S
K
V
L
F
R
K
S
E
K
Y
14


394
K
L
F
L
W
L
L
V
G
V
G
L
L
G
L
14


398
W
L
L
V
G
V
G
L
L
G
L
G
L
R
H
14


404
G
L
L
G
L
G
L
R
H
K
A
Y
E
R
K
14


405
L
L
G
L
G
L
R
H
K
A
Y
E
R
K
L
14


478
D
L
T
M
W
L
G
E
K
L
G
F
Y
T
D
14


492
D
F
G
P
S
T
R
Y
H
T
W
G
I
M
A
14


551
N
H
E
D
D
L
D
R
K
L
Q
A
I
A
V
14


560
L
Q
A
I
A
V
S
K
L
L
K
S
S
S
N
14


589
R
D
Y
L
Q
L
T
E
H
G
N
V
K
D
I
14


613
E
Y
I
M
Y
R
G
L
I
R
L
G
Y
A
R
14


617
Y
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
14


618
R
G
L
I
R
L
G
Y
A
R
I
S
H
A
E
14


635
D
S
E
I
Q
M
A
K
F
R
I
P
D
D
P
14


4
L
W
R
E
I
L
L
E
S
L
L
G
C
V
S
13


11
E
S
L
L
G
C
V
S
W
S
L
Y
H
D
L
13


41
G
L
E
G
F
S
I
A
F
L
S
P
I
F
L
13


47
I
A
F
L
S
P
I
F
L
T
I
T
P
F
W
13


118
Q
R
Y
L
R
I
W
G
F
I
L
G
Q
I
V
13


130
Q
I
V
L
V
V
L
R
I
W
Y
T
S
L
N
13


149
Y
Q
M
S
N
K
V
I
L
T
L
S
A
I
A
13


150
Q
M
S
N
K
V
I
L
T
L
S
A
I
A
T
13


152
S
N
K
V
I
L
T
L
S
A
I
A
T
L
D
13


203
G
S
L
V
F
L
T
H
W
V
F
G
E
V
S
13


206
V
F
L
T
H
W
V
F
G
E
V
S
L
V
S
13


235
N
P
F
G
G
A
V
L
L
C
L
A
S
G
L
13


236
P
F
G
G
A
V
L
L
C
L
A
S
G
L
M
13


239
G
A
V
L
L
C
L
A
S
G
L
M
L
P
S
13


275
L
L
Y
L
H
T
W
A
A
A
V
S
G
C
V
13


286
S
G
C
V
F
A
I
F
T
A
S
M
W
P
Q
13


315
T
M
T
I
A
M
I
F
Y
L
L
E
I
F
F
13


318
I
A
M
I
F
Y
L
L
E
I
F
F
C
A
W
13


324
L
L
E
I
F
F
C
A
W
C
T
A
F
K
F
13


359
I
I
G
L
N
M
L
F
G
P
K
K
N
L
D
13


369
K
K
N
L
D
L
L
L
Q
T
K
N
S
S
K
13


391
K
Y
M
K
L
F
L
W
L
L
V
G
V
G
L
13


400
L
V
G
V
G
L
L
G
L
G
L
R
H
K
A
13


402
G
V
G
L
L
G
L
G
L
R
H
K
A
Y
E
13


416
E
R
K
L
G
K
V
A
P
T
K
E
V
S
A
13


461
F
I
T
I
L
E
S
D
A
S
K
P
Y
M
G
13


473
Y
M
G
N
N
D
L
T
M
W
L
G
E
K
L
13


477
N
D
L
T
M
W
L
G
E
K
L
G
F
Y
T
13


504
I
M
A
L
S
R
Y
P
I
V
K
S
E
H
H
13


517
H
H
L
L
P
S
P
E
G
E
I
A
P
A
I
13


527
I
A
P
A
I
T
L
T
V
N
I
S
G
K
L
13


553
E
D
D
L
D
R
K
L
Q
A
I
A
V
S
K
13


557
D
R
K
L
Q
A
I
A
V
S
K
L
L
K
S
13


574
N
Q
V
I
F
L
G
Y
I
T
S
A
P
G
S
13







HLA Class II analysis of 125P5C8 variants 2-5. Listed are


scores that fall within the top 50% (rounded up) of all scores


for the selected allele.


variant 2 (aa 1-16)










HLA-DRB1*0101 15-mers


























2
P
S
L
W
R
E
I
L
L
E
S
L
L
G
C
26
Portion of SEQ ID NO: 5, each



1
M
P
S
L
W
R
E
I
L
L
E
S
L
L
G
16
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*0401




(DR4Dw4) 15-mers

























2
P
S
L
W
R
E
I
L
L
E
S
L
L
G
C
22
Portion of SEQ ID NO: 5, each



1
M
P
S
L
W
R
E
I
L
L
E
S
L
L
G
20
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*1101 15-mers


























2
P
S
L
W
R
E
I
L
L
E
S
L
L
G
C
16
Portion of SEQ ID NO: 5, each



1
M
P
S
L
W
R
E
I
L
L
E
S
L
L
G
12
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.







variant 3 (aa 668-696)










HLA-DRB1*0101 15-mers


























668

K

I
H

F

N

P

R
F

G

S
Y
K
E
G
P
19
Portion of SEQ ID NO: 7, each



681

G

P
N

Y

E

N

N
H

H

F
H
M
N
T
P
18
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*0301




(DR17) 15-mers

























672

N

P
R

F

G

S

Y
K

E

G
P
N
Y
E
N
16
Portion of SEQ ID NO: 7, each




















start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*0401 (DR4Dw4)




15-mers

























675

F

G
S

Y

K

E


G

P

N

Y
E
N
N
H
H
22
Portion of SEQ ID NO: 7, each



681

G

P
N

Y

E

N


N

H

H

F
H
M
N
T
P
22
start position is specified -


672

N

P
R

F

G

S


Y

K

E

G
P
N
Y
E
N
16
the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*1101 15-mers


























681

G

P
N

Y

E

N

N
H

H

F
H
M
N
T
P
18
Portion of SEQ ID NO: 7, each



671

F

N
P

R

F

G

S
Y

K

E
G
P
N
Y
E
15
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.







variant 4 (aa 673-699)










HLA-DRB1*0101 15-mers


























681
G
H
N
Y
E
N
T
H
H
F
H
M
N
T
P
18
Portion of SEQ ID NO: 9, each



679
K
E
G
H
N
Y
E
N
T
H
H
F
H
M
N
15
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*0401




(DR4Dw4) 15-mers

























675
F
G
S
Y
K
E
G
H
N
Y
E
N
T
H
H
22
Portion of SEQ ID NO: 9, each



681
G
H
N
Y
E
N
T
H
H
F
H
M
N
T
P
22
start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.










HLA-DRB1*1101 15-mers


























681
G
H
N
Y
E
N
T
H
H
F
H
M
N
T
P
18
Portion of SEQ ID NO: 9, each




















start position is specified -



















the length of each peptide is



















15 amino acids, the end



















position for each peptide is



















the start position plus



















fourteen.







variant 5 (aa 675-699)










HLA-DRB1*0101 15-mers


























681
G
H
N
Y
E
N
N
H
N
F
H
M
N
T
P
18
Portion of SEQ ID NO: 11,




















each start position is



















specified - the length of



















each peptide is 15 amino



















acids, the end position for



















each peptide is the start



















position plus fourteen.










HLA-DRB1*0301




(DR17) 15-mers

























679
K
E
G
H
N
Y
E
N
N
H
N
F
H
M
N
15
Portion of SEQ ID NO: 11,




















each start position is



















specified - the length of



















each peptide is 15 amino



















acids, the end position for



















each peptide is the start



















position plus fourteen.










HLA-DRB1*0401




(DR4Dw4) 15-mers

























675
F
G
S
Y
K
E
G
H
N
Y
E
N
N
H
N
22
Portion of SEQ ID NO: 11,



681
G
H
N
Y
E
N
N
H
N
F
H
M
N
T
P
22
each start position is



















specified - the length of



















each peptide is 15 amino



















acids, the end position for



















each peptide is the start



















position plus fourteen.
















TABLE XX







Frequently Occurring Motifs












avrg. %





Name
identity
Description
Potential Function





zf-C2H2
34%
Zinc finger, C2H2
Nucleic acid-binding protein





type
functions as transcription





factor, nuclear location probable





cytochrome b N
68%
Cytochrome b(N-
membrane bound oxidase, generate




terminal)/b6/petB
superoxide





ig
19%
Immunoglobulin
domains are one hundred amino




domain
acids long and include a





conserved intradomain disulfide





bond.





WD40
18%
WD domain, G-beta
tandem repeats of about 40




repeat
residues, each containing a Trp-





Asp motif. Function in signal





transduction and protein





interaction





PDZ
23%
PDZ domain
may function in targeting





signaling molecules to sub-





membranous sites





LRR
28%
Leucine Rich
short sequence motifs involved in




Repeat
protein-protein interactions





pkinase
23%
Protein kinase
conserved catalytic core common




domain
to both serine/threonine and





tyrosine protein kinases





containing an ATP binding site





and a catalytic site





PH
16%
PH domain
pleckstrin homology involved in





intracellular signaling or as





constituents of the cytoskeleton





EGF
34%
EGF-like domain
30-40 amino-acid long found in





the extracellular domain of





membrane-bound proteins or in





secreted proteins





rvt
49%
Reverse




transcriptase




(RNA-dependent DNA




polymerase)





ank
25%
Ank repeat
Cytoplasmic protein, associates





integral membrane proteins to the





cytoskeleton





oxidored q1
32%
NADH-
membrane associated. Involved in




Ubiquinone/plastoquinone
proton translocation across the




(complex
membrane




I), various chains





efhand
24%
EF hand
calcium-binding domain, consists of





a12 residue loop flanked on both





sides by a 12 residue alpha-helical





domain





rvp
79%
Retroviral aspartyl
Aspartyl or acid proteases,




protease
centered on a catalytic aspartyl





residue





Collagen
42%
Collagen triple
extracellular structural proteins




helix repeat (20
involved in formation of connective




copies)
tissue. The sequence consists of





the G-X-Y and the polypeptide





chains forms a triple helix.





fn3
20%
Fibronectin type
Located in the extracellular




III domain
ligand-binding region of receptors





and is about 200 amino acid





residues long with two pairs of





cysteines involved in disulfide





bonds





7tm 1
19%
7 transmembrane
seven hydrophobic transmembrane




receptor (rhodopsin
regions, with the N-terminus




family)
located extracellularly while the





C-terminus is cytoplasmic. Signal





through G proteins
















TABLE XXI





Motifs and Post-translational Modifications of


125P5C8







N-glycosylation site.








380-383
NSSK (SEQ ID NO: 34)





455-458
NETG (SEQ ID NO: 35)





536-539
NISG (SEQ ID NO: 36)










Protein kinase C phosphorylation site.








152-154
SnK





381-383
SsK





389-391
SeK





496-498
StR





538-540
SgK





666-668
SeK





677-679
SyK





694-696
TpK










Casein kinase II phosphorylation site.








40-43
TGLE (SEQ ID NO: 37)





170-173
TDGD (SEQ ID NO: 38)





175-178
SKPE (SEQ ID NO: 39)





445-448
SSLE (SEQ ID NO: 40)





457-460
TGAD (SEQ ID NO: 41)





463-466
TILE (SEQ ID NO: 42)





606-609
TDED (SEQ ID NO: 43)





629-632
SHAE (SEQ ID NO: 44)





634-637
SDSE (SEQ ID NO: 45)





677-680
SYKE (SEQ ID NO: 46)










Tyrosine kinase phosphorylation site.








610-617
RWC.EYIMY (SEQ ID NO: 47)





644-652
RIPDDPTNY (SEQ ID NO: 48)










N-myristoylation site.








79-84
GSIASF (SEQ ID NO: 49)





 99-104
GVSSSL (SEQ ID NO: 50)





199-204
GAAFGS (SEQ ID NO: 51)





268-273
GTASAA (SEQ ID NO: 52)





287-292
GCVFAI (SEQ ID NO: 53)





309-314
GTNPGK (SEQ ID NO: 54)





341-346
GGVYAR (SEQ ID NO: 55)
















TABLE XXII







Properties of 125P5C8










Bioinformatic




Program
Outcome















ORF
ORF finder
2099



Protein length

699 aa



Transmembrane
TM Pred
11TM, N terminus



region

intracellular. TM





helices at aa 7-29,





39-61, 68-87, 91-





112, 120-139, 195-





213, 236-257, 280-





298, 321-342, 351-





357, 392-410




HMMTop
10TM, N terminus





extracellular. TM





helices at aa 44-61,





93-110 125-142, 195-





212, 235-252, 259-





276, 281-298, 315-





368, 392-410




Sosui
10TM, TM helices at





aa 1-23, 42-64, 94-





116, 120-142, 189-





211, 238-260, 269-





291, 318-340, 350-





368, 390-411




TMHMM
10TM, N terminus





intracellular. TM





helices at aa 13-32,





42-64, 91-113, 123-





140, 195-214, 237-





259, 272-294, 317-





339, 351-368, 395-





412



Signal Peptide
Signal P
yes, cleavage site





between aa 335-336



pI
pI/MW tool
8.75



Molecular
pI/MW tool
78.56 kDa



weight



Localization
PSORT
60% plasma membrane,





51% mitochondrial





inner membrane,




PSORT II
65.2% plasma





membrane, 17.4%





endoplasmic





reticulum



Motifs
Pfam
phosphoribulokinase/





uridine-kinase





family




Prints

C. elegans Srg family






integral membrane





protein signature




Blocks
No significant motif









Claims
  • 1. A method for detecting the presence of a 125P5C8 protein or polynucleotide in a test sample, comprising: (a) contacting the test sample with an antibody that specifically binds to a protein of SEQ ID NO: 3, 5, 7, 9, or 11, or a nucleic acid that specifically hybridizes to a polynucleotide of SEQ ID NO:2, 4, 6, 8, or 10; and(b) detecting binding of protein or polynucleotide in the sample to the antibody or nucleic acid, respectively.
  • 2. The method of claim 1, wherein the protein or polynucleotide in the sample is an mRNA.
  • 3. The method of claim 1, further comprising contacting a normal sample with the antibody or nucleic acid and detecting an amount or absence of binding in the normal sample thereto.
  • 4. The method of claim 1, further comprising comparing the binding detected in step (b) to an amount or absence of binding of the antibody or nucleic acid to protein or polynucleotide in a corresponding normal sample.
  • 5. The method of claim 4, wherein an elevated amount of binding to the test sample compared to the normal sample indicates the presence of cancer.
  • 6. The method of claim 4, wherein the presence of elevated 125P5C8 polynucleotide or protein in the test sample relative to the normal sample indicates the presence of cancer.
  • 7. The method of claim 6, wherein the cancer is selected from the group consisting of cancers of the prostate, bladder, kidney, ovary, breast, and colon.
  • 8. The method of claim 1, wherein the test sample is from a tissue selected from the group consisting of prostate, bladder, kidney, colon, ovary, and breast.
  • 9. The method of claim 1, wherein the test sample is from a patient with cancer.
  • 10. A method of inhibiting growth of a cell, comprising contacting the cell with an antibody or antigen binding fragment thereof that specifically binds to a protein comprising the amino acid sequence of SEQ ID NO: 3, 5, 7, 9, or 11, thereby inhibiting growth of the cell.
  • 11. The method of claim 10, wherein the antibody or fragment thereof is monoclonal.
  • 12. The method of claim 10, wherein the antibody or fragment thereof is human or humanized.
  • 13. The method of claim 10, wherein the antibody or fragment thereof is an Fab, F(ab′)2, Fv or Sfv fragment.
  • 14. The method of claim 10, wherein the antibody or fragment thereof is a recombinant protein.
  • 15. The method of claim 10, wherein the cell is a cancer cell.
  • 16. The method of claim 10, wherein the cell is from a tissue source selected from the group consisting of prostate, bladder, kidney, colon, ovary, and breast.
  • 17. A method of delivering a cytotoxic agent to a cell, comprising providing to the cell an effective amount of an antibody or fragment thereof that immunospecifically binds to an epitope on a protein of SEQ ID NO: 3, 5, 7, 9, or 11, wherein the antibody or fragment is coupled to a cytotoxic agent.
  • 18. The method of claim 17, wherein the cytotoxic agent is a toxin, a chemotherapeutic agent or a radioisotope.
  • 19. The method of claim 18, wherein the radioisotope is selected from the group consisting of 212Bi, 131I, 131In, 90Y, 186Re, 211At, 125I, 188Re, 153Sm, 213Bi, 32P, and Lu.
  • 20. The method of claim 17, wherein the cytotoxic agent is selected from the group consisting of auristatins, auromycins, maytansinoids, yttrium, bismuth, ricin, ricin A-chain, combrestatin, duocarmycins, dolostatins, doxorubicin, daunorubicin, taxol, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, Sapaonaria officinalis inhibitor, and glucocorticoid.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. Ser. No. 12/820,083, filed Jun. 21, 2010, now allowed, which is a divisional of U.S. Ser. No. 12/235,444, filed Sep. 22, 2008, now U.S. Pat. No. 7,759,474, issued Jul. 20, 2010, which is a divisional of U.S. Ser. No. 10/099,460, filed Mar. 13, 2002, now U.S. Pat. No. 7,488,479, issued Feb. 10, 2009, which is a continuation-in-part of U.S. Ser. No. 09/809,638, filed Mar. 14, 2001, now U.S. Pat. No. 7,271,240, issued Sep. 18, 2007. The content of these applications is hereby incorporated by reference herein in their entirety.

Provisional Applications (2)
Number Date Country
60283112 Apr 2001 US
60286630 Apr 2001 US
Divisions (1)
Number Date Country
Parent 10121016 Apr 2002 US
Child 11841126 US
Continuations (1)
Number Date Country
Parent 11841126 Aug 2007 US
Child 13052002 US