AU2002305169A1 - Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer - Google Patents

Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer

Info

Publication number
AU2002305169A1
AU2002305169A1 AU2002305169A AU2002305169A AU2002305169A1 AU 2002305169 A1 AU2002305169 A1 AU 2002305169A1 AU 2002305169 A AU2002305169 A AU 2002305169A AU 2002305169 A AU2002305169 A AU 2002305169A AU 2002305169 A1 AU2002305169 A1 AU 2002305169A1
Authority
AU
Australia
Prior art keywords
protein
amino acid
cell
cancer
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2002305169A
Other versions
AU2002305169B2 (en
AU2002305169C1 (en
Inventor
Pia M. Challita-Eid
Mary Faris
Wangmao Ge
Rene S. Hubert
Aya Jakobovits
Karen Morrison
Robert Kendall Morrison
Arthur B. Raitano
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agensys Inc
Original Assignee
Agensys Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agensys Inc filed Critical Agensys Inc
Priority claimed from PCT/US2002/011544 external-priority patent/WO2002083916A2/en
Publication of AU2002305169A1 publication Critical patent/AU2002305169A1/en
Publication of AU2002305169B2 publication Critical patent/AU2002305169B2/en
Priority to AU2007237282A priority Critical patent/AU2007237282B2/en
Application granted granted Critical
Publication of AU2002305169C1 publication Critical patent/AU2002305169C1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

NUCLEIC ACID AND CORRESPONDING PROTEIN ENTITLED 162P1E6 USEFUL IN TREATMENT AND DETECTION OF CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority from United States Serial No. 60/283,112 filed April 10, 2001, and United States Serial No. 60/286,630, filed April 25, 2001. The contents of these applications are hereby incorporated by reference herein in their entirety.
STATEMENT OFRIGHTSTO INVENTIONSMADEUNDERFEDERALLYSPONSORED RESEARCH
Not applicable.
FIELD OF THE INVENTION
The invention described herein relates to a gene and its encoded protein, termed 162P1E6, expressed in certain cancers, and to diagnostic and therapeutic methods and compositions useful in the management of cancers that express 162P1E6.
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 Sep 2 (9): 1445-51), STEAP (Hubert, et al, Proc Natl Acad Sci U S A. 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 sequelae 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 162P1E6, that has now been found to be over- expressed in the cancer(s) listed in Table I. Northern blot expression analysis of 162P1E6 gene expression in normal tissues shows a restricted expression pattern in adult tissues. The nucleotide (Figure 2) and amino acid (Figure 2, and Figure 3) sequences of 162P1E6 are provided. The tissue-related profile of 162P1E6 in normal adult tissues, combined with the over-expression observed in the tissues listed in Table I, shows that 162P1E6 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 162P1E6 genes, mRNAs, and/or coding sequences, preferably in isolated form, including polynucleotides encoding 162PlE6-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 162PlE6-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 162P1E6 genes or mRNA sequences or parts thereof, and polynucleotides or oligonucleotides that hybridize to the 162P1E6 genes, mRNAs, or to 162PlE6-encoding polynucleotides. Also provided are means for isolating cDNAs and the genes encoding 162P1E6. Recombinant DNA molecules containing 162P1E6 polynucleotides, cells transformed or transduced with such molecules, and host-vector systems for the expression of 162P 1E6 gene products are also provided. The invention further provides antibodies that bind to 162P1E6 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 Figure 2 is not encoded and or the entire amino acid sequence of Figure 2 is not prepared. In certain embodiments, the entire nucleic acid sequence of Figure 2 is encoded and/or the entire amino acid sequence of Figure 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 162P1E6 polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express 162P1E6. A typical embodiment of this invention provides methods for monitoring 162P1E6 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 162P1E6 such as cancers of tissues listed in Table I, including therapies aimed at inhibiting the transcription, translation, processing or function of 162P1E6 as well as cancer vaccines. In one aspect, the invention provides compositions, and methods comprising them, for treating a cancer that expresses 162P1E6 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 162P1E6. Preferably, the carrier is a uniquely human carrier. In another aspect of the invention, the agent is a moiety that is immunoreactive with 162P1E6 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 162P1E6 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 162P1E6 as described above. The one or more than one nucleic acid molecule may also be, or encodes, a molecule that inhibits production of 162P1E6. Non-limiting examples of such molecules include, but are not limited to, those complementary to a nucleotide sequence essential for production of 162P1E6 (e.g. antisense sequences or molecules that form a triple helix with a nucleotide double helix essential for 162P1E6 production) or a ribozyme effective to lyse 162P1E6 mRNA.
Note that to determine the starting position of any peptide set forth in Tables V-XVTII and XXII to LI (collectively HLA Peptide Tables) respective to its parental protein, e.g., variant 1, variant 2, etc., reference is made to three factors: the particular variant, the length of the peptide in an HLA Peptide Table, and the Search Peptides in Table LII. Generally, a unique Search Peptide is used to obtain HLA peptides of a partiular for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table LII. Accordingly if a Search Peptide begins at position "X", one must add the value "X - 1 " to each position in Tables V-XVIII and XXII to LI to obtain the actual position of the HLA peptides in their parental molecule. For example if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150 - 1, i.e., 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
One embodiment of the mvention comprises an HLA peptide, that occurs at least twice in Tables V- XVIII and XXII to LI collectively, or an oligonucleotide that encodes the HLA peptide. Another embodiment of the invention comprises an HLA peptide that occurs at least once in Tables V-XViπ and at least once in tables XXII to LI, or an oligonucleotide that encodes the HLA peptide,.
Another embodiment of the invention is antibody epitopes which comprise a peptide regions, or an oligonucleotide encoding the peptide region, that has one two, three, four, or five of the following characteristics: i) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of Figure 5; ii) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of Figure 6; iii) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of Figure 7; iv) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of Figure 8; or v) a peptide region of at least 5 amino acids of a particular peptide of Figure 3, in any whole number increment up to the full length of that protein in Figure 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Beta-turn profile of Figure 9.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1. The 162P1E6 SSH sequence of 335 nucleotides.
Figure 2. The cDNA (SEQ ID. NO. :. _) and amino acid sequence (SEQ ID. NO. : ) of
162P1E6 variant 1 clone B (also called "162P1E6 v.l" or "162P1E6 variant 1") is shown in Figure 2A. The start methionine is underlined. The open reading frame extends from nucleic acid 2028-2468 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 2 (also called "162P1E6 v.2") is shown in Figure 2B. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 3 (also called
"162P1E6 v.3") is shown in Figure 2C. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 3-404 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 4 (also called "162P1E6 v.4") is shown in Figure 2D.
The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 388-696 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of
162P1E6 variant 5 (also called "162P1E6 v.5") is shown in Figure 2E. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 388-618 including the stop codon. The cDNA
(SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 6 (also called
"162P1E6 v.6") is shown in Figure 2F. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 388-600 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 7 (also called "162P1E6 v.7") is shown in
Figure 2G. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 480-788 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID.
NO. : ) of 162P1E6 variant 8 (also called "162P1E6 v.8") is shown in Figure 2H. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 480-692 including the stop codon. The cDNA (SEQ ID. NO. :, ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant
9 (also called "162P1E6 v.9") is shown in Figure 21. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 1535-1975 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 10 (also called "162P1E6 v.10") is shown in Figure 2J. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 1535-1975 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence
(SEQ ID. NO. : ) of 162P1E6 variant 11 (also called "162P1E6 v.l 1") is shown in Figure 2K. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ED. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of
162P1E6 variant 12 (also called "162P1E6 v.12") is shown in Figure 2L. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 13 (also called "162P1E6 v.13") is shown in Figure 2M. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO.
: ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 14 (also called "162P1E6 v.14") is shown in Figure 2N. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence
(SEQ ID. NO. : ) of 162P1E6 variant 15 (also called "162P1E6 v.15") is shown in Figure 20. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of
162P1E6 variant 16 (also called "162P1E6 v.16") is shown in Figure 2P. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 17 (also called "162P1E6 v.17") is shown in Figure 2Q. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO.
: ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 18 (also called "162P1E6 v.18") is shown in Figure 2R. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. :__ ) and amino acid sequence
(SEQ ID. NO. : ) of 162P1E6 variant 19 (also called "162P1E6 v.19") is shown in Figure 2S. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of
162P1E6 variant 20 (also called "162P1E6 v.20") is shown in Figure 2T. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. The cDNA (SEQ ID. NO. : ) and amino acid sequence (SEQ ID. NO. : ) of 162P1E6 variant 21 (also called "162P1E6 v.21") is shown in Figure 2U. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 2550-2990 including the stop codon. As used herein, a reference to 162P1E6 includes all variants thereof, including those shown in Figures 10 and 12.
Figure 3. The amino acid sequence of 162P1E6 v.l (SEQ ID. NO. : ) is shown in Figure 3A; it has 146 amino acids. The amino acid sequence of 162P1E6 v.3 (SEQ ID. NO. : ) is shown in Figure 3B; it has 133 amino acids. The amino acid sequence of 162P1E6 v.4 (SEQ ID. NO. : ) is shown in Figure
3C; it has 102 amino acids. The amino acid sequence of 162P1E6 v.5 (SEQ ID. NO. : ) is shown in
Figure 3D; it has 76 amino acids. The amino acid sequence of 162P1E6 v.6 (SEQ ID. NO. : ) is shown in
Figure 3E; it has 70 amino acids. The amino acid sequence of 162P1E6 v.18 (SEQ ID. NO. : ) is shown in Figure 3F; it has 146 amino acids. As used herein, a reference to 162P1E6 includes all variants thereof, including those shown in Figure 11.
Figure 4. The nucleic acid sequence alignment of 162P1E6 v.l with hypothetical gene XP_036612 (AK002208) is shown in Figure 4A. The amino acid sequence alignment of 162P1E6 v.l withwith hypothetical gene XP_036612 (AK002208) is shown in Figure 4B. The amino acid sequence alignment of 162P 1 E6 v.1 with putative Man7GlcNAc2-PP-dolichyl mannosyltransferase is shown in Figure 4C. The amino acid sequence alignment of 162P 1E6 v.1 with estrogen receptor beta2 splice variant is shown in Figure 4D. The amino acid sequence alignment of 162P1E6 v.3 with Alu subfamily is shown in Figure 4E. The amino acid sequence alignment of 162P1E6 v.3 with Zinc finger protein is shown in Figure 4F. The amino acid sequence alignemnt of 162P1E6 v.4 with Interleukin lbeta is shown in Figure 4G.
Figure 5. Hydrophilicity amino acid profile of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6, 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.
Figure 6. Hydropathicity amino acid profile of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6, determined by computer algorithm sequence analysis using the method of Kyte and Doolittle (Kyte J., Doolittle R.F., 1982. 1. Mol. Biol. 157: 105-132) accessed on the ProtScale website (www.expasy.ch/cgi-bin protscale.pl) through the ExPasy molecular biology server.
Figure 7. Percent accessible residues amino acid profile of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6, 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.
Figure 8. Average flexibility amino acid profile of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6, 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 (www.expasy.ch/cgi- bin/protscale.pl) through the ExPasy molecular biology server.
Figure 9. Beta-rum amino acid profile of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6, 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 (www.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
Figure 10. Schematic alignment of Single Nucleotide Polymorphism (SNP) variants of 162P1E6. Variants 162P1E6 v.12 through v.21 are variants with single nucleotide differences. Though these SNP variants are shown separately on the template of 162P1E6 v.2, they could also occur in any combinations and in any one of the transcript variants that contains the base pairs. Numbers correspond to those of 162P1E6 v.2. Black box shows the same sequence as 162P1E6 v.2. SNPs are indicated above the box. Figure 11. Schematic alignment of protein variants of 162P1E6. Nucleotide variants 162PlE6 v.l, v.2, v.9, v.10 and v.l 1 in Figure 12 code for the same protein 162P1E6 v.l. Variants 162P1E6 v.4 and v.7 code the same protein 162P1E6 v.4. Variant 162P1E6 v.6 and v.8 each code for the same protein 162P1E6 v.6. SNP variant 162P1E6 v.18 codes the same protein as variant 162P1E6 v.l except for one amino acid. All other SNP variants in Figure 10 code for the same protein as 162P1E6 v.l . Boxes with the same fill pattern represent the same sequence. Variant 162P1E6 v.4 and v.5 share the N-terminal 37 amino acids. Single amino acid differences are indicated above the box.
Figure 12. Schematic alignment of transcript variants of 162P1E6. Variant 162P1E6 v.2 is an alternative transcript. Variants 162P1E6 v.3 through v.l 1 are splice variants of transcript 162P1E6. Not all splice variants are shown here. Transcript 162P1E6 v.l may also have similar splicing pattern for the corresponding exons. Numbers in "( )" underneath the box correspond to those of 162P1E6 v.2. Boxes with the same fill pattern represent the same sequence.
Figure 13. Secondary structure prediction for 162P1E6. The secondary structure of A) 162P1E6 variant 1, B) 162P1E6 variant 3, C) 162P1E6 variant 4, D) 162P1E6 variant 5 and E) 162P1E6 variant 6 was predicted using the HNN - Hierarchical Neural Network method (Guermeur, 1997, http://pbil.ibcp.fr/cgi- bin/npsa_automat.pl?page=npsa_nn.html), accessed from the ExPasy molecular biology server (http://www.expasy.ch tools/). 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 listed.
Figure 14. Expression of 162P1E6 by RT-PCR. First strand cDNA was prepared from 1) vital pool 1 (liver, lung and kidney), 2) vital pool 2 (pancreas, colon and stomach), 3) LAPC xenograft pool (LAPC- 4AD, LAPC-4AI, LAPC-9 AD and LAPC-9AI), 4) prostate cancer pool, 5) bladder cancer pool, 6) lung cancer pool, 7) breast cancer pool, and 8) cancer metastasis pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi-quantitative PCR, using primers to 162P1E6, was performed at 26 and 30 cycles of amplification. Results show strong expression of 162P1E6 in bladder cancer pool, lung cancer pool, and breast cancer pool. Expression was also detected in prostate cancer pool and cancer metastasis pool, but not in the vital pools.
Figure 15. Expression of 162P1E6 in normal tissues. Two multiple tissue northern blots (Clontech) with 2 ug of mRNA/lane were probed with the 162P1E6 SSH fragment. Size standards in kilobases (kb) are indicated on the side. Results show expression of two approximately 4.4 kbl62PlE6 transcripts in placenta, prostate and thymus.
Figure 16. Expression of 162P1E6 in bladder cancer patient specimens. RNA was extracted from normal bladder (Nb), bladder cancer cell lines (CL: UM-UC-3, J82 and SCaBER), bladder cancer patient tumors (T) and normal tissue adjacent to bladder cancer (N). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in the bladder tumor tissues and in the SCaBER cancer cell line, but not in normal bladder, nor in the other cancer cell lines J82 and UM-UC-3.
Figure 17. Expression of 162P1E6 in prostate cancer patient specimens. RNA was extracted from LAPC-4AD, LAPC-4AI, LAPC-9 AD and LAPC-9 AI prostate cancer xenografts, normal prostate (N), prostate cancer patient tumors (T) and their normal adjacent tissues (NAT). Northern blot with 10 μg of total RNA/lane was probed with 162P1E6 SSH sequence. Size standards in kilobases (kb) are indicated on the side. The results show strong expression of 162P1E6 in normal prostate and in patient prostate cancer specimens. Weak expression was detected in the LAPC-4AD tissue, but not in the other prostate cancer xenografts.
Figure 18. Expression of 162P1E6 in kidney cancer patient tissues. RNA was extracted from kidney cancer cell lines (769-P, A498, SW839), normal kidney (N), kidney cancer patient tumors (T) and their normal adjacent tissues (NAT). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in 2 out of 2 papillary kidney tumor tissues but not in specimens of clear cell carcinoma, normal kidney nor in the kidney cancer cell lines.
Figure 19. Expression of 162P1E6 in lung cancer patient tissues. RNA was extracted from lung cancer cell lines (CALU-1, A427, NCI-H82, NCI-H146), normal lung (N), lung cancer patient tumors (T) and normal adjacent tissues (NAT) isolated from lung cancer patients. Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in the all lung tumor tissues tested, but not in normal lung nor in the lung cancer cell lines.
Figure 20. Expression of 162P 1 E6 in breast cancer patient tissues. RNA was extracted from breast cancer cell lines (DU4475, MCF7 and CAMA-1), normal breast (N), breast cancer patient tumors (T) and breast cancer metastasis to lymph node (Ml), and to ovary (M2). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show expression of 162P1E6 in normal breast, breast tumor tissues as well as in the cancer metastasis specimens, but not in the breast cancer cell lines tested.
Figure 21. 162P1E6 Expression in 293T Cells Following Transfection. 293T cells were transfected with either 162P1E6 .pcDNA3.1/mychis cones D7, D8, D9, D10 (A) or 162PlE6.pTag5 vector (B). 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 162P1E6 from the 4 different clone transfections of 162P1E6 .pcDNA3.1/mychis vector, and from the 2 different clone transfections of 162PlE6.pTag5 vector.
DETAILED DESCRIPTION OF THE INVENTION
Outline of Sections
I.) Definitions π.) 162P1E6 Polynucleotides
H.A.) Uses of 162P1E6 Polynucleotides
H.A.I.) Monitoring of Genetic Abnormalities π.A.2.) Antisense Embodiments
H.A.3.) Primers and Primer Pairs π.A.4.) Isolation of 162PlE6-Encoding Nucleic Acid Molecules π.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems HI.) 162PlE6-related Proteins IH.A.) Motif-bearing Protein Embodiments
III.B.) Expression of 162PlE6-related Proteins m.C.) Modifications of 162PlE6-related Proteins
III.D.) Uses of 162PlE6-related Proteins IV.) 162P1E6 Antibodies V.) 162P1E6 Cellular Immune Responses VI.)162P1E6 Transgenic Animals VH.) Methods for the Detection of 162P1E6
VIII.) Methods for Monitoring the Status of 162PlE6-related Genes and Their Products IX.) Identification of Molecules That Interact With 162P1E6 X.) Therapeutic Methods and Compositions
X.A.) Anti-Cancer Vaccines X.B.) 162P1E6 as a Target for Antibody-Based Therapy X.C.) 162P1E6 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 162P1E6. XH.) Inhibition of 162P1E6 Protein Function
Xπ.A.) Inhibition of 162P1E6 With Intracellular Antibodies
Xπ.B.) Inhibition of 162P1E6 with Recombinant Proteins
Xπ.C.) Inhibition of 162P1E6 Transcription or Translation XQ.D.) General Considerations for Therapeutic Strategies xm.) 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 Cl - 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 162P1E6 (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 162P1E6. 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 162PlE6-related protein). For example an analog of a 162P1E6 protein can be specifically bound by an antibody or T cell that specifically binds to 162P 1E6.
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-162P1E6 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-162PlE6 antibodies and clones thereof (including agonist, antagonist and neutralizing antibodies) and anti-162PlE6 antibody compositions withpolyepitopic 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, diphtlieria 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, 1131, 1125, 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, CA (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/6XSSC/0.1%> SDS/100 μg/ml ssDNA, in which temperatures for hybridization are above 37 degrees C and temperatures for washing in O.lXSSC/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 162P1E6 genes or that encode polypeptides other than 162P1E6 gene product or fragments thereof. A skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated 162P1E6 polynucleotide. A protein is said to be "isolated," for example, when physical, mechanical or chemical methods are employed to remove the 162P1E6 proteins from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated 162P1E6 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 TxNxM+ 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 humerus. 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 162P 1 E6-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 Figure 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 162P1E6, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit 162P1E6 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, 162P1E6 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 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.S), 0.1% sodium pyrophosphate, 5 xDenhardt's solution, sonicated salmon sperm DNA (50 μg/ml), 0.1% SDS, and 10% dextran sulfate at 42 °C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium, citrate) and 50% formamide at 55 °C, followed by a high- stringency wash consisting of 0.1 x 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 x SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/mL denatured sheared salmon sperm DNA, followed by washing the filters in 1 x 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- 150 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, or 150 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 ρosition(s) of a specifically described protein (e.g. the 162P1E6 protein shown in Figure 2 or Figure 3. An analog is an example of a variant protein. Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.
The "162PlE6-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 162P1E6 proteins or fragments thereof, as well as fusion proteins of a 162P1E6 protein and a heterologous polypeptide are also included. Such 162P1E6 proteins are collectively referred to as the 162PlE6-related proteins, the proteins of the invention, or 162P1E6. The term "162P1E6- related protein" refers to a polypeptide fragment or a 162P1E6 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, or 146 or more amino acids.
H 162P1E6 Polynucleotides
One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a 162P1E6 gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a 162PlE6-related protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a 162P1E6 gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a 162P1E6 gene, mRNA, or to a 162P1E6 encoding polynucleotide (collectively, "162P1E6 polynucleotides"). In all instances when referred to in this section, T can also be U in Figure 2.
Embodiments of a 162P1E6 polynucleotide include: a 162P1E6 polynucleotide having the sequence shown in Figure 2, the nucleotide sequence of 162P1E6 as shown in Figure 2 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in Figure 2; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in Figure 2 where T is U. For example, embodiments of 162P1E6 nucleotides comprise, without limitation:
(I) a polynucleotide comprising, consisting essentially of, or consisting of a sequence as shown in Figure 2 (SEQ ID NO: ), wherein T can also be U;
(II) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in
Figure 2A (SEQ ID NO: ), from nucleotide residue number 2028 through nucleotide residue number 2468, 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
Figure 2B (SEQ ID NO: ), from nucleotide residue number 2550 through nucleotide residue number 2990, 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
Figure 2C (SEQ ID NO: ), from nucleotide residue number 3 through nucleotide residue number 404, 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
Figure 2D (SEQ ID NO: ), from nucleotide residue number 388 through nucleotide residue number 696, 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
Figure 2E (SEQ ID NO: ), from nucleotide residue number 388 through nucleotide residue number 618, 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 Figure 2F (SEQ ID NO: ), from nucleotide residue number 388 through nucleotide residue number 600, including the stop codon, wherein T can also be U;
(VIII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figure 2G (SEQ ID NO: ), from nucleotide residue number 480 through nucleotide residue number 788, including the stop codon, wherein T can also be U; (IX) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in
Figure 2H (SEQ ID NO: ), from nucleotide residue number 480 through nucleotide residue number 692, including the stop codon, wherein T can also be U;
(X) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in
Figure 21 (SEQ ID NO: ), from nucleotide residue number 1535 through nucleotide residue number 1975, including the stop codon, wherein T can also be U;
(XI) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in
Figure 2J (SEQ ID NO: ), from nucleotide residue number 1535 through nucleotide residue number 1975, including the stop codon, wherein T can also be U;
(XII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in Figures 2K through 2U (SEQ ID NOs: ), from nucleotide residue number 2550 through nucleotide residue number 2990, including the stop codon, wherein T can also be U;
(XIII) a polynucleotide that encodes a 162PlE6-related protein that is at least 90% homologous to an entire amino acid sequence shown in Figures 2A-U (SEQ ID NO: );
(XIV) a polynucleotide that encodes a 162PlE6-related protein that is at least 90% identical to an entire amino acid sequence shown in Figures 2A-U (SEQ ID NO: );
(XV) a polynucleotide that encodes at least one peptide set forth in Tables V-XVTfl and XXII-LI;
(XVI) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of Figure 3A in any whole number increment up to 146 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5A; or of Figure 3B in any whole number increment up to 133 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5B; or of Figure 3C in any whole number increment up to 102 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5C; or of Figure 3D in any whole number increment up to 76 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5D; or of Figure 3E in any whole number increment up to 70 that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5E;
(XVII) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of Figure 3A in any whole number increment up to 146 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6A; or of Figure 3B in any whole number increment up to 133 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6B; or of Figure 3C in any whole number increment up to 102 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6C; or of Figure 3D in any whole number increment up to 76 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6D; or of Figure 3E in any whole number increment up to 70 that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6E;
(XVIII) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of Figure 3 A in any whole number increment up to 146 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7A; or of Figure 3B in any whole number increment up to 133 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7B; or of Figure 3C in any whole number increment up to 102 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7C; or of Figure 3D in any whole number increment up to 76 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7D; or of Figure 3E in any whole number increment up to 70 that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7E;
(XIX) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of Figure 3A in any whole number increment up to 146 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure 8 A; or of Figure 3B in any whole number increment up to 133 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure 8B; or of Figure 3C in any whole number increment up to 102 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure8C; or of Figure 3D in any whole number increment up to 76 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure 8D; or of Figure 3E in any whole number increment up to 70 that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure 8E;
(XX) a polynucleotide that encodes a peptide region of at least 5 amino acids of a peptide of Figure 3A in any whole number increment up to 146 that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figure 9A; or of Figure 3B in any whole number increment up to 133 that includes an amino acid position having a value greater than 0.5 in the Beta- turn profile of Figure 9B; or of Figure 3C in any whole number increment up to 102 that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figure 9C; or of Figure 3D in any whole number increment up to 76 that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figure 9D; or of Figure 3E in any whole number increment up to 70 that includes an amino acid position having a value greater than 0.5 in the Beta- turn profile of Figure 9E;
(XXI) a polynucleotide that encodes a 162P lE6-related protein whose sequence is encoded by the cDNAs contained in the plasmid deposited with American Type Culture Collection (ATCC) on March 28, 2002 as Accession No. ; (XXII) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XXI).
(XXIII) a peptide that is encoded by any of (I)-(XXII); and
(XXIV) a polynucleotide of any of (I)-(XXII) or peptide of (XXIII) 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 162P1E6 polynucleotides that encode specific portions of 162P1E6 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, or 146 or more contiguous amino acids of 162P1E6.
(b) 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, or 133 or more contiguous amino acids of 162P1E6 variant 3.
(c) 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, 5, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, or 102 contiguous amino acids of 162P1E6 variant 4;
(d) 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, or 76 contiguous amino acids of 162P1E6 variant 5; or
(e) 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, or 70 contiguous amino acids of 162P1E6 variant 6.
(f) 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, or 146 or more contiguous amino acids of 162P1E6 variant 18.
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 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 10 to about amino acid 20 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 20 to about amino acid 30 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 30 to about amino acid 40 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 40 to about amino acid 50 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 50 to about amino acid 60 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 60 to about amino acid 70 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 70 to about amino acid 80 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 80 to about amino acid 90 of the 162P1E6 protein shown in Figure 2 or Figure 3, polynucleotides encoding about amino acid 90 to about amino acid 100 of the 162P1E6 protein shown in Figure 2 or Figure 3, in increments of about 10 amino acids, ending at the carboxyl terminal amino acid set forth in Figure 2 or Figure 3. Accordingly polynucleotides encoding portions of the amino acid sequence (of about 10 amino acids), of amino acids 100 through tire carboxyl terminal amino acid of the 162P1E6 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 162P1E6 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 162P1E6 protein "or variant" shown in Figure 2 or Figure 3 can be generated by a variety of techniques well known in the art. These polynucleotide fragments can include any portion of the 162P1E6 sequence as shown in Figure 2.
Additional illustrative embodiments of the invention disclosed herein include 162P1E6 polynucleotide fragments encoding one or more of the biological motifs contained within a 162P1E6 protein "or variant" sequence, including one or more of the motif-bearing subsequences of a 162P1E6 protein "or variant" set forth in Tables V-XVIII and XXII-LI. In another embodiment, typical polynucleotide fragments of the invention encode one or more of the regions of 162P1E6 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 162P ΪE6 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.
H.A.) Uses of 162P1E6 Polynucleotides
II.A.l.) Monitoring of Genetic Abnormalities
The polynucleotides of the preceding paragraphs have a number of different specific uses. The human 162P1E6 gene maps to the chromosomal location set forth in the Example entitled "Chromosomal Mapping of 162P1E6." For example, because the 162P1E6 gene maps to this chromosome, polynucleotides that encode different regions of the 162P1E6 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 162P1E6 proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes 162P1E6 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 162P1E6 was shown to be highly expressed in bladder and other cancers, 162P1E6 polynucleotides are used in methods assessing the status of 162P1E6 gene products in normal versus cancerous tissues. Typically, polynucleotides that encode specific regions of the 162P1E6 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 162P1E6 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. π.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 162P1E6. 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 162P1E6 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., 162P1E6. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988). The 162P1E6 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-oligό) 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-l,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 162P1E6 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 162P1E6 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 162P1E6 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 162P1E6 mRNA and not to mRNA specifying other regulatory subunits of protein kinase. In one embodiment, 162P1E6 antisense oligonucleotides of the present invention are 15 to 30- mer fragments of the antisense DNA molecule that have a sequence that hybridizes to 162P1E6 mRNA. Optionally, 162P1E6 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 162P1E6. Alternatively, the antisense molecules are modified to employ ribozymes in the inhibition of 162P1E6 expression, see, e.g., L. A. Couture & D. T. Stinchcomb; Trends Genet 12: 510-515 (1996). π.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 162P1E6 polynucleotide in a sample and as a means for detecting a cell expressing a 162P1E6 protein.
Examples of such probes include polypeptides comprising all or part of the human 162P1E6 cDNA sequence shown in Figure 2. Examples of primer pairs capable of specifically amplifying 162P1E6 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 162PlE6 mRNA.
The 162P1E6 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 162P1E6 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 162P1E6 polypeptides; as tools for modulating or inhibiting the expression of the 162P1E6 gene(s) and/or translation of the 162P1E6 transcript(s); and as therapeutic agents.
The present invention includes the use of any probe as described herein to identify and isolate a 162P1E6 or 162P1E6 related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence er se, which would comprise all or most of the sequences found in the probe used. π.A.4.) Isolation of 162PlE6-Encoding Nucleic Acid Molecules
The 162P1E6 cDNA sequences described herein enable the isolation of other polynucleotides encoding 162P1E6 gene ρroduct(s), as well as the isolation of polynucleotides encoding 162P1E6 gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a 162P1E6 gene product as well as polynucleotides that encode analogs of 162PlE6-related proteins. Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a 162P1E6 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 162P1E6 gene cDNAs can be identified by probing with a labeled 162P1E6 cDNA or a fragment thereof. For example, in one embodiment, a 162P1E6 cDNA (e.g., Figure 2) or a portion thereof can be synthesized and used as a probe to retrieve overlapping and full-length cDNAs corresponding to a 162P1E6 gene. A 162P1E6 gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with 162P1E6 DNA probes or primers. π.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems
The invention also provides recombinant DNA or RNA molecules containing a 162P1E6 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 162P1E6 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 TsuPrl, 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 162P1E6 or a fragment, analog or homolog thereof can be used to generate 162P1E6 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 162P1E6 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 α/., 1991, MCB 11:1785). Using these expression vectors, 162P1E6 can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPrl. The host-vector systems of the invention are useful for the production of a 162P1E6 protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of 162P1E6 and 162P1E6 mutations or analogs.
Recombinant human 162P1E6 protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a 162PlE6-related nucleotide. For example, 293T cells can be transfected with an expression plasmid encoding 162P 1 E6 or fragment, analog or homolog thereof, a 162PlE6-related protein is expressed in the 293T cells, and the recombinant 162P1E6 protein is isolated using standard purification methods (e.g., affinity purification using anti-162PlE6 antibodies). In another embodiment, a 162P1E6 coding sequence is subcloned into the retroviral vector pSRαMSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPrl, 293 and rat-1 in order to establish 162P1E6 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 162P1E6 coding sequence can be used for the generation of a secreted form of recombinant 162P1E6 protein.
As discussed herein, redundancy in the genetic code permits variation in 162P1E6 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)).
m. 162PlE6-related Proteins
Another aspect of the present invention provides 162PlE6-related proteins. Specific embodiments of 162P1E6 proteins comprise a polypeptide having all or part of the amino acid sequence of human 162P1E6 as shown in Figure 2 or Figure 3. Alternatively, embodiments of 162P1E6 proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of 162P1E6 shown in Figure 2 or Figure 3.
In general, naturally occurring allelic variants of human 162P1E6 share a high degree of structural identity and homology (e.g., 90% or more homology). Typically, allelic variants of a 162P1E6 protein contain conservative amino acid substitutions within the 162P1E6 sequences described herein or contain a substitution of ah amino acid from a corresponding position in a homologue of 162P1E6. One class of 162P1E6 allelic variants are proteins that share a high degree of homology with at least a small region of a particular 162P1E6 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, orfhology 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 162P1E6 proteins such as polypeptides having amino acid insertions, deletions and substitutions. 162P1E6 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., 75:4331 (1986); Zoller et al, Nucl. Acids Res., 70:6487 (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 162P1E6 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, 162P1E6 variants, analogs or homologs, have the distinguishing attribute of having at least one epitope that is "cross reactive" with a 162P1E6 protein having an amino acid sequence of Figure 3. As used in this sentence, "cross reactive" means that an antibody or T cell that specifically binds to a 162P1E6 variant also specifically binds to a 162P1E6 protein having an amino acid sequence set forth in Figure 3. A polypeptide ceases to be a variant of a protein shown in Figure 3, when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting 162P1E6 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 162PlE6-related protein variants share 70%, 75%, 80%, 85%. or 90% or more similarity with an amino acid sequence of Figure 3, or a fragment thereof. Another specific class of 162P1E6 protein variants or analogs comprise one or more of the 162P1E6 biological motifs described herein or presently known in the art. Thus, encompassed by the present invention are analogs of 162P1E6 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 Figure 2 or Figure 3.
As discussed herein, embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a 162P1E6 protein shown in Figure 2 or Figure 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 162P1E6 protein shown in Figure 2 or Figure 3.
Moreover, representative embodiments of the invention disclosed herein include polypeptides consisting of about amino acid 1 to about amino acid 10 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 10 to about amino acid 20 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 20 to about amino acid 30 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 30 to about amino acid 40 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 40 to about amino acid 50 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 50 to about amino acid 60 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 60 to about amino acid 70 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 70 to about amino acid 80 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 80 to about amino acid 90 of a 162P1E6 protein shown in Figure 2 or Figure 3, polypeptides consisting of about amino acid 90 to about amino acid 100 of a 162P1E6 protein shown in Figure 2 or Figure 3, etc. throughout the entirety of a 162P1E6 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 162P1E6 protein shown in Figure 2 or Figure 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.
162P lE6-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 162PlE6-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a 162P1E6 protein (or variants, homologs or analogs thereof).
HLA.) Motif-bearing Protein Embodiments
Additional illustrative embodiments of the invention disclosed herein include 162P1E6 polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a 162P1E6 polypeptide sequence set forth in Figure 2 or Figure 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., URL addresses: pfam.wustl.edu/; http://searcrdauncher.bcnitmc.edu seq-search/struc-predict.html; psort.ims.u- tokyo.ac.jp/; www.cbs.dtu.dk/; www.ebi.ac.uk/interpro/scan.html; www.expasy.ch/tools/scnpsitl.html; Epimatrix™ and Epimer™, Brown University, www.brown.edu/Research/TB- HTV_Lab/φjmatrix/eρirnatrix.html; and BBVIAS, bimas.dcrt.nih.gov/.).
Motif bearing subsequences of all 162P1E6 variant proteins are set forth and identified in Tables V- XVIII and XXII-LI.
Table XIX sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu ). The columns of Table XIX 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 162P1E6 motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the 162P1E6 motifs discussed above are associated with growth dysregulation and because 162P1E6 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(l):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 and XXII-LI. CTL epitopes can be determined using specific algorithms to identify peptides within a 162P 1 E6 protein that are capable of optimally binding to specified HLA alleles (e.g., Table IV; Epimatrix™ and Epimer™, Brown University, URL www.brown.edu/Researcli/TB-HIV_Lab/epjjnatrbc/epiinatrk.httm and BIMAS, URL bimas.dcrt.nih.gov/.) 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 TV). 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 ppsition. 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 TV. 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 97/33602 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 XX, and or, one or more of the predicted CTL epitopes of Tables V-XVII and XXII- XLVII, and/or, one or more of the predicted HTL epitopes of Tables XLVπi-LI, 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. 162PlE6-related proteins are embodied in many forms, preferably in isolated form. A purified 162P1E6 protein molecule will be substantially free of other proteins or molecules that impair the binding of 162P 1E6 to antibody, T cell or other ligand. The nature and degree of isolation and purification will depend on the intended use. Embodiments of a 162PlE6-related proteins include purified 162PlE6-related proteins and functional, soluble 162PlE6-related proteins. In one embodiment, a functional, soluble 162P1E6 protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.
The invention also provides 162P1E6 proteins comprising biologically active fragments of a 162P1E6 amino acid sequence shown in Figure 2 or Figure 3. Such proteins exhibit properties of the starting 162P1E6 protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting 162P1E6 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.
162PlE6-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- olf analysis, or on the basis of immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-162PlE6 antibodies, or T cells or in identifying cellular factors that bind to 162P1E6. 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 162P1E6 protein that are capable of optimally binding to specified HLA alleles (e.g., by using the SYFPEriΗI site at World Wide Web URL syfpeithi.bmi-heidelberg.com/; the listings in Table IV(A)-(E); Epimatrix™ and Epimer™, Brown University, URL (www.brown.edu/Research/TB-HIV_Lab/ephnatrix/epirm1rix.^ and BIMAS, URL bimas.dcrt.nih.gov/). Illustrating this, peptide epitopes from 162P1E6 that are presented in the context of human MHC Class I molecules, e.g., HLA-A1, A2, A3, Al 1, A24, B7 and B35 were predicted (see, e.g., Tables V-XVIII, XXII-LI). Specifically, the complete amino acid sequence of the 162P1E6 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; in addition to the site SYFPEITHI, at URL s;yfpeithi.bmi- heidelberg.com/.
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 162P1E6 predicted binding peptides are shown in Tables V-XVIII and XXII-LI herein. In Tables V-XVIII and XXII-XLVTI, selected candidates, 9-mers and 10-mers, for each family member are shown along with then location, the amino acid sequence of each specific peptide, and an estimated binding score. In Tables XLVIII-LI, selected candidates, 15-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 syφeithi.bmi-heidelberg.com/, or BDVIAS, bimas.dcrt.nih.gov/) are to be "applied" to a 162P1E6 protein in accordance with the invention. As used in this context "applied" means that a 162P1E6 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 162P1E6 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.
IH.B.) Expression of 162PlE6-related Proteins
In an embodiment described in the examples that follow, 162P1E6 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 162P1E6 with a C-terminal 6XHis and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville TN). The Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted 162P1E6 protein in transfected cells. The secreted HIS- tagged 162P1E6 in the culture media can be purified, e.g., using a nickel column using standard techniques. m.C Modifications of 162PlE6-related Proteins
Modifications of 162PlE6-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 162P1E6 polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of a 162P1E6 protein. Another type of covalent modification of a 162P1E6 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 162P1E6 comprises linking a 162P1E6 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. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
The 162PlE6-related proteins of the present invention can also be modified to form a chimeric molecule comprising 162P1E6 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 rumor-associated antigen or fragment thereof. Alternatively, a protein in accordance with the invention can comprise a fusion of fragments of a 162P1E6 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 Figure 2 or Figure 3. Such a chimeric molecule can comprise multiples of the same subsequence of 162P1E6. A chimeric molecule can comprise a fusion of a 162PlE6-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 al62PlE6 protein. In an alternative embodiment, the chimeric molecule can comprise a fusion of a 162PlE6-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 162P 1E6 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, CHI, CH2 and CH3 regions of an IgGI molecule. For the production of immunoglobulin fusions see, e.g., U.S. Patent No. 5,428,130 issued June 27, 1995.
HI.D.) Uses of 162PlE6-related Proteins
The proteins of the invention have a number of different specific uses. As 162P1E6 is highly expressed in prostate and other cancers, 162PlE6-related proteins are used in methods that assess the status of 162P1E6 gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a 162P1E6 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 162PlE6-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a 162P1E6 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, 162PlE6-related proteins that contain the amino acid residues of one or more of the biological motifs in a 162P1E6 protein are used to screen for factors that interact with that region of 162P1E6.
162P 1E6 protein fragments/subsequences are particularly useful in generating and characterizing domain-specific antibodies (e.g., antibodies recognizing an extracellular or intracellular epitope of a 162P1E6 protein), for identifying agents or cellular factors that bind to 162P1E6 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 162P1E6 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 162P1E6 gene product. Antibodies raised against a 162P1E6 protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of 162P1E6 protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers. 162PlE6-related nucleic acids or proteins are also used in generating HTL or CTL responses.
Various immunological assays useful for the detection of 162P1E6 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 162PlE6-expressing cells (e.g., in radioscintigraphic imaging methods). 162P1E6 proteins are also particularly useful in generating cancer vaccines, as further described herein.
TV.) 162P1E6 Antibodies
Another aspect of the invention provides antibodies that bind to 162PlE6-related proteins. Preferred antibodies specifically bind to a 162PlE6-related protein and do not bind (or bind weakly) to peptides or proteins that are not 162PlE6-related proteins. For example, antibodies that bind 162P1E6 can bind 162PlE6-related proteins such as the homologs or analogs thereof.
162P1E6 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 162P1E6 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 162P1E6 is involved, such as advanced or metastatic prostate cancers.
The invention also provides various immunological assays useful for the detection and quantification of 162P1E6 and mutant 162PlE6-relate proteins. Such assays can comprise one or more 162P1E6 antibodies capable of recognizing and binding a 162PlE6-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 histocompatibiUty complex (MHC) binding assays.
In addition, immunological imaging methods capable of detecting prostate cancer and other cancers expressing 162P1E6 are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled 162P1E6 antibodies. Such assays are clinically useful in the detection, monitoring, and prognosis of 162P1E6 expressing cancers such as prostate cancer.
162P1E6 antibodies are also used in methods for purifying a 162PlE6-related protein and for isolating 162P1E6 homologues and related molecules. For example, a method of purifying a 162PlE6-related protein comprises incubating a 162P1E6 antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a 162PlE6-related protein under conditions that permit the 162P1E6 antibody to bind to the 162PlE6-related protein; washing the solid matrix to eliminate impurities; and eluting the 162PlE6-related protein from the coupled antibody. Other uses of 162P 1 E6 antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a 162P1E6 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 162PlE6-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 162P1E6 can also be used, such as a 162P1E6 GST-fusion protein. In a particular embodiment, a GST fusion protein comprising all or most of the amino acid sequence of Figure 2 or Figure 3 is produced, then used as an immunogen to generate appropriate antibodies. In another embodiment, a 162PlE6-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 162PlE6-related protein or 162P1E6 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 162P 1 E6 protein as shown in Figure 2 or Figure 3 can be analyzed to select specific regions of the 162P1E6 protein for generating antibodies. For example, hydrophobicity and hydrophihcity analyses of a 162P1E6 amino acid sequence are used to identify hydrophilic regions in the 162P1E6 structure. Regions of a 162P1E6 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 162P1E6 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, IL, are effective. Administration of a 162P1E6 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.
162P 1E6 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 162PlE6-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 162P1E6 protein can also be produced in the context of chimeric or complementarity detenmning region (CDR) grafted antibodies of multiple species origin. Humanized or human 162P1E6 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 etal, 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 α/., 1998, Nature Biotechnology 16: 535-539). Fully human 162P1E6 monoclonal antibodies can be generated using cloning technologies employing large human Ig gene combinatorial libraries (i.e., phage display) (Griffiths and Hoogeriboom, 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 162P1E6 monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application W098/24893, Kucherlapati and Jakobovits et al, published December 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 7(4): 607-614; U.S. patents 6,162,963 issued 19 December 2000; 6,150,584 issued 12 November 2000; and, 6,114598 issued 5 September 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.
Reactivity of 162P1E6 antibodies with a 162PlE6-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 162PlE6-related proteins, 162PlE6-expressing cells or extracts thereof. A 162P1E6 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 162P1E6 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.) 162P1E6 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. ~ ., 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 syφeithi.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 Nov; 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 ^Cr-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 aclministered 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 5 ^Cr-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 "Cτ release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.
VI.) 162P1E6 Transgenic Animals
Nucleic acids that encode a 162PlE6-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 162P1E6 can be used to clone genomic DNA that encodes 162P1E6. The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode 162P1E6. 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. Patent Nos. 4,736,866 issued 12 April 1988, and 4,870,009 issued 26 September 1989. Typically, particular cells would be targeted for 162P1E6 transgene incorporation with tissue-specific enhancers.
Transgenic animals that include a copy of a transgene encoding 162P1E6 can be used to examine the effect of increased expression of DNA that encodes 162P1E6. 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 162P1E6 can be used to construct a 162P1E6 "knock out" animal that has a defective or altered gene encoding 162P1E6 as a result of homologous recombination between the endogenous gene encoding 162P1E6 and altered genomic DNA encoding 162P1E6 introduced into an embryonic cell of the animal. For example, cDNA that encodes 162P1E6 can be used to clone genomic DNA encoding 162P1E6 in accordance with established techniques. A portion of the genomic DNA encoding 162P1E6 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 then 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 162P1E6 polypeptide.
VH.) Methods for the Detection of 162P1E6
Another aspect of the present invention relates to methods for detecting 162P1E6 polynucleotides and 162PlE6-related proteins, as well as methods for identifying a cell that expresses 162P1E6. The expression profile of 162P1E6 makes it a diagnostic marker for metastasized disease. Accordingly, the status of 162P1E6 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 162P1E6 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 162P1E6 polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like. Detectable 162P1E6 polynucleotides include, for example, a 162P1E6 gene or fragment thereof, 162P1E6 mRNA, alternative splice variant 162P1E6 mRNAs, and recombinant DNA or RNA molecules mat contain a 162P1E6 polynucleotide. A number of methods for ainplifying and or detecting the presence of 162P1E6 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 162P1E6 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 162P1E6 polynucleotides as sense and antisense primers to amplify 162P1E6 cDNAs therein; and detecting the presence of the amplified 162P1E6 cDNA. Optionally, the sequence of the amplified 162P1E6 cDNA can be determined.
In another embodiment, a method of detecting a 162P1E6 gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using 162P1E6 polynucleotides as sense and antisense primers; and detecting the presence of the amplified 162P1E6 gene. Any number of appropriate sense and antisense probe combinations can be designed from a 162P1E6 nucleotide sequence (see, e.g., Figure 2) and used for this purpose.
The invention also provides assays for detecting the presence of a 162P1E6 protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like. Methods for detecting a 162PlE6-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 162PlE6-related protein in a biological sample comprises first contacting the sample with a 162P1E6 antibody, a 162PlE6-reactive fragment thereof, or a recombinant protein containing an antigen binding region of a 162P1E6 antibody; and then detecting the binding of 162PlE6-related protein in the sample.
Methods for identifying a cell that expresses 162P1E6 are also within the scope of the invention. In one embodiment, an assay for identifying a cell that expresses a 162P1E6 gene comprises detecting the presence of 162P1E6 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 162P1E6 riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for 162P1E6, 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 162P1E6 gene comprises detecting the presence of 162PlE6-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 162PlE6-related proteins and cells that express 162PlE6-related proteins.
162P1E6 expression analysis is also useful as a tool for identifying and evaluating agents that modulate 162P1E6 gene expression. For example, 162P1E6 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 162P1E6 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 162P1E6 expression by RT-PCR, nucleic acid hybridization or antibody binding.
VHI.) Methods for Monitoring the Status of 162PlE6-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 162P1E6 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 162P1E6 in a biological sample of interest can be compared, for example, to the status of 162P1E6 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 162P1E6 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. Patent No. 5,837,501) to compare 162P1E6 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 162P1E6 expressing cells) as well as the level, and biological activity of expressed gene products (such as 162P1E6 mRNA, polynucleotides and polypeptides). Typically, an alteration in the status of 162P1E6 comprises a change in the location of 162P1E6 and/or 162P1E6 expressing cells and/or an increase in 162P1E6 mRNA and/or protein expression.
162P1E6 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 162P1E6 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 162P1E6 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 162P1E6 gene), Northern analysis and/or PCR analysis of 162P1E6 mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of 162P1E6 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 162P1E6 proteins and/or associations of 162P1E6 proteins with polypeptide binding partners). Detectable 162P1E6 polynucleotides include, for example, a 162P1E6 gene or fragment thereof, 162P1E6 mRNA, alternative splice variants, 162P1E6 mRNAs, and recombinant DNA or RNA molecules containing a 162P1E6 polynucleotide.
The expression profile of 162P1E6 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 162P1E6 provides information useful for predicting susceptibility to particular disease stages, progression, and/or tumor aggressiveness. The invention provides methods and assays for determining 162P1E6 status and diagnosing cancers that express 162P1E6, such as cancers of the tissues listed in Table I. For example, because 162P1E6 mRNA is so highly expressed in prostate and other cancers relative to normal prostate tissue, assays that evaluate the levels of 162P1E6 mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with 162P1E6 deregulation, and can provide prognostic information useful in defining appropriate therapeutic options.
The expression status of 162P1E6 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 162P1E6 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 162P1E6 in a biological sample can be examined by a number of well-known procedures in the art. For example, the status of 162P1E6 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 162P1E6 expressing cells (e.g. those that express 162P1E6 mRNAs or proteins). This examination can provide evidence of dysregulated cellular growth, for example, when 162PlE6-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 162P1E6 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 Aug 154(2 Pt l):474-8).
In one aspect, the invention provides methods for monitoring 162P1E6 gene products by determining the status of 162P1E6 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 162P1E6 gene products in a corresponding normal sample. The presence of aberrant 162P1E6 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 162P1E6 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 162P1E6 mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I. The presence of significant 162P1E6 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 162P1E6 mRNA or express it at lower levels.
In a related embodiment, 162P 1E6 status is determined at the protein level rather than at the nucleic acid level. For example, such a method comprises determining the level of 162P1E6 protein expressed by cells in a test tissue sample and comparing the level so determined to the level of 162P1E6 expressed in a corresponding normal sample. In one embodiment, the presence of 162P1E6 protein is evaluated, for example, using immunohistochemical methods. 162P1E6 antibodies or binding partners capable of detecting 162P1E6 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 162P1E6 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 162P1E6 may be indicative of the presence or promotion of a tumor. Such assays therefore have diagnostic and predictive value where a mutation in 162P1E6 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 162P1E6 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. Patent Nos. 5,382,510 issued 7 September 1999, and 5,952,170 issued 17 January 1995).
Additionally, one can examine the methylation status of a 162P1E6 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 162P1E6. 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 162P1E6 expression. The presence of RT-PCR amplifiable 162P1E6 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; Ghossern etα/., 1995, J. Clin. Oncol. 13:1195-2000; Heston etα/., 1995, Clin. Chem 41:1687-1688).
A further aspect of the invention is an assessment of the susceptibihty that an individual has for developing cancer. In one embodiment, a method for predicting susceptibihty to cancer comprises detecting 162P1E6 mRNA or 162P1E6 protein in a tissue sample, its presence indicating susceptibihty to cancer, wherein the degree of 162P1E6 mRNA expression correlates to the degree of susceptibihty. In a specific embodiment, the presence of 162P1E6 in prostate or other tissue is examined, with the presence of 162P1E6 in the sample providing an indication of prostate cancer susceptibihty (or the emergence or existence of a prostate tumor). Similarly, one can evaluate the integrity 162P1E6 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 162P1E6 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 162P1E6 mRNA or 162P1E6 protein expressed by tumor cells, comparing the level so determined to the level of 162P1E6 mRNA or 162P1E6 protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of 162P1E6 mRNA or 162P1E6 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 162P1E6 is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors. Another embodiment is the evaluation of the integrity of 162P1E6 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 162P1E6 mRNA or 162P1E6 protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of 162P1E6 mRNA or 162P 1E6 protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of 162P1E6 mRNA or 162P1E6 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 162P1E6 expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity 162P1E6 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 162P1E6 gene and 162P1E6 gene products (or perturbations in 162P1E6 gene and 162P1E6 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., Booking 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 162P1E6 gene and 162P1E6 gene products (or perturbations in 162P1E6 gene and 162P1E6 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 162P1E6 gene and 162P1E6 gene products (or perturbations in 162P1E6 gene and 162P1E6 gene products) and another factor associated with malignancy entails detecting the overexpression of 162P1E6 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 162P1E6 mRNA or protein and PSA mRNA or protein overexpression (or PSCA or PSM expression). In a specific embodiment, the expression of 162P1E6 and PSA mRNA in prostate tissue is examined, where the coincidence of 162P1E6 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 162P1E6 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 162P1E6 mRNA include in situ hybridization using labeled 162P1E6 riboprobes, Northern blot and related techniques using 162P1E6 polynucleotide probes, RT- PCR analysis using primers specific for 162P1E6, 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 162P1E6 mRNA expression. Any number of primers capable of amplifying 162P1E6 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 162P1E6 protein can be used in an immunohistochemical assay of biopsied tissue.
IX.) Identification of Molecules That Interact With 162P1E6
The 162P1E6 protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with 162P1E6, as well as pathways activated by 162P1E6 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. Patent Nos. 5,955,280 issued 21 September 1999, 5,925,523 issued 20 July 1999, 5,846,722 issued 8 December 1998 and 6,004,746 issued 21 December 1999. Algorithms are also available in the art for genome-based predictions of protein function (see, e.g., Marcotte, et al, Nature 402: 4 November 1999, 83-86).
Alternatively one can screen peptide libraries to identify molecules that interact with 162P1E6 protein sequences. In such methods, peptides that bind to 162P1E6 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 162PlE6 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 162P1E6 protein sequences are disclosed for example in U.S. Patent Nos. 5,723,286 issued 3 March 1998 and 5,733,731 issued 31 March 1998.
Alternatively, cell lines that express 162P1E6 are used to identify protein-protein interactions mediated by 162P1E6. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B.J., et al. Biochem. Biophys. Res. Commun. 1999, 261:646-51). 162P1E6 protein can be immunoprecipitated from 162PlE6-expressing cell lines using anti-162PlE6 antibodies. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of 162P1E6 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, protem microsequencing, silver staining and two-dimensional gel electrophoresis.
Small molecules and ligands that interact with 162P1E6 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 162PlE6'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 162PlE6-related ion channel, protein pump, or cell communication functions are identified and used to treat patients that have a cancer that expresses 162P1E6 (see, e.g., Hille, B., Ionic Channels of Excitable Membranes 2nd Ed., Sinauer Assoc, Svmderland, MA, 1992). Moreover, ligands that regulate 162P1E6 function can be identified based on their ability to bind 162P1E6 and activate a reporter construct. Typical methods are discussed for example in U.S. Patent No. 5,928,868 issued 27 July 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 162P1E6 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 162P1E6.
An embodiment of this invention comprises a method of screening for a molecule that interacts with a 162P1E6 amino acid sequence shown in Figure 2 or Figure 3, comprising the steps of contacting a population of molecules with a 162P1E6 amino acid sequence, allowing the population of molecules and the 162P1E6 amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the 162P1E6 amino acid sequence, and then separating molecules that do not interact with the 162P1E6 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 162P1E6 amino acid sequence. The identified molecule can be used to modulate a function performed by 162P1E6. In a preferred embodiment, the 162P1E6 amino acid sequence is contacted with a library of peptides.
X.) Therapeutic Methods and Compositions
The identification of 162P1E6 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, 162P1E6 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 162P1E6 protein are useful for patients suffering from a cancer that expresses 162P1E6. These therapeutic approaches generally fall into two classes. One class comprises various methods for inhibiting the binding or association of a 162P1E6 protein with its binding partner or with other proteins. Another class comprises a variety of methods for inhibiting the transcription of a 162P1E6 gene or translation of 162P1E6 mRNA.
X.A.) Anti-Cancer Vaccines
The invention provides cancer vaccines comprising a 162PlE6-related protein or 162PlE6-related nucleic acid. In view of the expression of 162P1E6, cancer vaccines prevent and or treat 162PlE6-expressrng 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 etα/., 1997, J. Immunol. 159:3113-3117).
Such methods can be readily practiced by employing a 162PlE6-related protein, or a 162P1E6- encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the 162P1E6 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 Feb 31(l):66-78; Maruyama et al, Cancer Immunol Immunother 2000 Jun 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 162P1E6 protein shown in Figure 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 162P1E6 immunogen contains a biological motif, see e.g., Tables V-XVHI and XXII-LI, or a peptide of a size range from 162P1E6 indicated in Figure 5, Figure 6, Figure 7, Figure 8, and Figure 9.
The entire 162P1E6 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, Kaufinann, 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., Kofier, 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, Massachusetts) may also be used.
In patients with 162PlE6-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 162P 1E6 protein that bind corresponding HLA alleles (see e.g., Table IV; Epimer™ and Epimatrix™, Brown University (URL www.brown.edu/Research/TB-HIV_Lab/epimatrix/epnnatrix.html); and, BIMAS, (URL bimas.dcrt.nih.gov/; SYFPEITHI at URL syfbeithi.bmi-heidelberg.com/). In a preferred embodiment, a 162P1E6 immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables V-XVIII and XXII-LI 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 162P1E6 protein) so that an immune response is generated. A typical embodiment consists of a method for generating an immune response to 162P1E6 in a host, by contacting the host with a sufficient amount of at least one 162P1E6 B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the 162P1E6 B cell or cytotoxic T-cell epitope or analog thereof. A specific embodiment consists of a method of generating an immune response against a 162PlE6-related protein or a man-made multiepitopic peptide comprising: administering 162P1E6 immunogen (e.g. a 162P1E6 protein or a peptide fragment thereof, a 162P1E6 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. Patent No. 6, 146,635) or a universal helper epitope such as a PADRE™ peptide (Epimmune Inc., San Diego, CA; 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 162P1E6 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 162P1E6 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. Patent 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 162P1E6, 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 162P1E6. Constructs comprising DNA encoding a 162PlE6-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 162P1E6 protein/immunogen. Alternatively, a vaccine comprises a 162PlE6-related protein. Expression of the 162PlE6-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a 162P1E6 protein. Various prophylactic and therapeutic genetic immunization techniques known in the art can be used (for review, see information and references published at Internet address www.genweb.com). Nucleic acid-based delivery is described, for instance, in Wolff et. al, Science 247:1465 (1990) as well as U.S. Patent 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 (bupivicaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated ("gene gun") or pressure-mediated delivery (see, e.g., U.S. Patent No. 5,922,687).
For therapeutic or prophylactic immunization purposes, proteins of the invention can be expressed via vhal 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, lentivkus, and sindbis virus (see, e.g., Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Ins 87:982-990 (1995)). Non-viral delivery systems can also be employed by introducing naked DNA encoding a 162PlE6-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. Patent 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 162PlE6-related nucleic acid molecule. In one embodiment, the full-length human 162P1E6 cDNA is employed. In another embodiment, 162P1E6 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 162P 1E6 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 162P1E6 peptides to T cells in the context of MHC class I or II molecules. In one embodiment, autologous dendritic cells are pulsed with 162P1E6 peptides capable of binding to MHC class I and/or class II molecules. In another embodiment, dendritic cells are pulsed with the complete 162P1E6 protein. Yet another embodiment involves engineering the overexpression of a 162P1E6 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 162P1E6 can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.
X.B.) 162P1E6 as a Target for Antibodv-based Therapy
162P1E6 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 162P1E6 is expressed by cancer cells of various lineages relative to corresponding normal cells, systemic administration of 162P1E6- 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 162P1E6 are useful to treat 162PlE6-expressing cancers systemically, either as conjugates with a toxin or therapeutic agent, or as naked antibodies capable of inhibiting cell proliferation or function.
162P1E6 antibodies can be introduced into a patient such that the antibody binds to 162P1E6 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 162P1E6, 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 162P1E6 sequence shown in Figure 2 or Figure 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 (June 1, 1999)). When cytotoxic and/or therapeutic agents are delivered dhectly to cells, such as by conjugating them to antibodies specific for a molecule expressed by that cell (e.g. 162P 1E6), 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-162PlE6 antibody) that binds to a marker (e.g. 162P1E6) 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 162P1E6, comprising conjugating the cytotoxic agent to an antibody that immunospecifically binds to a 162P1E6 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-162PlE6 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 α/., 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, 162P1E6 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, NJ, 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, MA, also see e.g., US Patent 5,416,064).
Although 162P1E6 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 162P1E6 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 162P1E6 expression, preferably using immunohistochemical assessments of tumor tissue, quantitative 162P1E6 imaging, or other techniques that reliably indicate the presence and degree of 162P1E6 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-162P1E6 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-162PlE6 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-162PlE6 mAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express 162P1E6. 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-162PlE6 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 humanmouse 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 162P1E6 antigen with high affinity but exhibit low or no antigenicity in the patient.
Therapeutic methods of the invention contemplate the administration of single anti-162PlE6 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 dhectly cytotoxic mAbs with mAbs that rely on immune effector functionality. Such mAbs in combination can exhibit synergistic therapeutic effects. In addition, anti-162PlE6 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- 162P 1 E6 mAbs are administered in their "naked" or unconjugated form, or can have a therapeutic agent(s) conjugated to them.
Anti-162P1E6 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-162PlE6 antibody preparation, via an acceptable route of administration such as intravenous injection (TV), typically at a dose in the range of about 0.1, .2, .3, .4, .5, .6, .7, .8, .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-162PlE6 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 162P1E6 expression in the patient, the extent of circulating shed 162P1E6 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 162P1E6 in a given sample (e.g. the levels of circulating 162P1E6 antigen and/or 162P1E6 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 ItnmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).
Anti-idiotypic anti-162PlE6 antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a 162P lE6-related protein. In particular, the generation of anti-idiotypic antibodies is well known in file art; this methodology can readily be adapted to generate anti- idiotypic anti-162PlE6 antibodies that mimic an epitope on a 162PlE6-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.) 162P1E6 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 162P1E6 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 H 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, CA) molecule (described e.g., in U.S. Patent Number 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 then 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 162P1E6, the PADRE® universal helper T cell epitope or multiple HTL epitopes from 162P1E6, (see e.g., Tables V-XVHI and XXII to LI), 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 downstream 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. Patent 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, CA). Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows dhection 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) maybe 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, California). 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) aα^ministration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be deshable. 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 Feigner, et al, Proc. NaflAcad. 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. Patent 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 vhal delivery systems well known in the art, e.g., an expression construct encoding epitopes of the invention can be incorporated into a vhal 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 dhectly 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 dhectly 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: ), Plasmodiumfalciparum chcumsporozoite (CS) protein at positions 378-398
(DIEKXIAXMEKASSVFNVVNS; SEQ ID NO: ), and Streptococcus 18kD protein at positions 116-131
(GAVDSILGGVATYGAA; SEQ ID NO: ). 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, CA) 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: ), 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 theh 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 theh biological properties. For example, they can be modified to include D-amino acids to increase theh resistance to proteases and thus extend theh serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase theh 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 deshable 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 dhectly 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 theh surfaces.
The DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to 162P1E6. 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 162P1E6.
X.D. Adoptive Immunotherapy
Antigenic 162PlE6-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 theh 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 162P1E6. 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 administtation, 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 aheady bearing a tumor that expresses 162P1E6. 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 162P1E6- 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 162P1E6, a vaccine comprising 162P1E6- 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 administtation 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 administtation, 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, hfe-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 deshable 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, infrathecal, 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 requhed 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 administtation 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 a<hrιinistered 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, Pennsylvania, 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 BVI (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 a<hninistered 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 5x109 pfu.
For antibodies, a treatment generally involves repeated administration of the anti-162PlE6 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- 162P1E6 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 162P1E6 expression in the patient, the extent of chculating shed 162P1E6 antigen, the deshed 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, 500μg - lmg, lmg - 50mg, 50mg - lOOmg, lOOmg - 200mg, 200mg - 300mg, 400mg - 500mg, 500mg - 600mg, 600mg - 700mg, 700mg - 800mg, 800mg - 900mg, 900mg - lg, or lmg - 700mg. 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; 0.5mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, lOmg/kg body weight followed, e.g., in two, three or four weeks by weekly doses; 0.5 - lOmg/kg body weight, e.g., followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400mg m2 of body area weekly; l-600mg m2 of body area weekly; 225-400mg 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 a<iministration. 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 maybe 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 dhect 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 IO4 cells to about 10s cells, about IO6 cells to about 10s cells, about 10s to about 1011 cells, or about 10s to about 5 x 1010 cells. A dose may also about IO6 cells/m2 to about 1010 cells/m2, or about IO6 cells/m2 to about IO8 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 incoφorated 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 deshed peptide of the invention can be dhected 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 lability 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. Patent 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 deshed 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 deshed, as with, e.g., lecithin for intranasal delivery.
XI.) Diagnostic and Prognostic Embodiments of 162P1E6.
As disclosed herein, 162P1E6 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 the Example entitled "Expression analysis of 162P1E6 in normal tissues, and patient specimens").
162P1E6 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. Aug; 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 ρ53 and K-ras (see, e.g., Tulchinsky et al, Int J Mol Med 1999 Jul 4(1):99- 102 and Minimoto et al, Cancer Detect Prev 2000;24(1):1-12). Therefore, this disclosure of 162P1E6 polynucleotides and polypeptides (as well as 162P1E6 polynucleotide probes and anti-162PlE6 antibodies used to identify the presence of these molecules) and theh properties allows skilled artisans to utilize these molecules in methods that are analogous to those used, for example, in a variety of diagnostic assays dhected to examining conditions associated with cancer.
Typical embodiments of diagnostic methods which utilize the 162P1E6 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 162P1E6 polynucleotides described herein can be utilized in the same way to detect 162P1E6 overexpression or the metastasis of prostate and oilier 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 162P1E6 polypeptides described herein can be utilized to generate antibodies for use in detecting 162P1E6 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 162P1E6 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 162PlE6-expressing cells (lymph node) is found to contain 162P1E6- expressing cells such as the 162P1E6 expression seen in LAPC4 and LAPC9, xenografts isolated from lymph node and bone metastasis, respectively, this finding is indicative of metastasis.
Alternatively 162P1E6 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 162P1E6 or express 162P1E6 at a different level are found to express 162P1E6 or have an increased expression of 162P1E6 (see, e.g., the 162P1E6 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 162P1E6) 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, 162P1E6 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-AnoUes, G. Biotechmques 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 the Example entitled "Expression analysis of 162P1E6 in normal tissues, and patient specimens," where a 162P1E6 polynucleotide fragment is used as a probe to show the expression of 162P1E6 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 Nov-Dec 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 162P1E6 polynucleotide shown in Figure 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. 162P1E6 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 epitoρe(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. Patent No. 5,840,501 and U.S. Patent No. 5,939,533). For example it may be preferable to utilize a polypeptide comprising one of the 162P1E6 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 162P1E6 polypeptide shown in Figure 3).
As shown herein, the 162P1E6 polynucleotides and polypeptides (as well as the 162P1E6 polynucleotide probes and anti-162PlE6 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 162P1E6 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 162P1E6 polynucleotides and polypeptides (as well as the 162P1E6 polynucleotide probes and anti-162PlE6 antibodies used to identify the presence of these molecules) need to be employed to confirm a metastases of prostatic origin.
Finally, in addition to theh use in diagnostic assays, the 162P1E6 polynucleotides disclosed herein have a number of other utilities such as theh use in the identification of oncogenetic associated chromosomal abnormalities in the chromosomal region to which the 162P1E6 gene maps (see the Example entitled "Chromosomal Mapping of 162P1E6" below). Moreover, in addition to theh use in diagnostic assays, the 162PlE6-related proteins and polynucleotides disclosed herein have other utilities such as theh use in the forensic analysis of tissues of unknown origin (see, e.g., Takahama K Forensic Sci Int 1996 Jun 28;80(l-2): 63-9).
Additionally, 162PlE6-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of 162P1E6. For example, the amino acid or nucleic acid sequence of Figure 2 or Figure 3, or fragments of either, can be used to generate an immune response to a 162P1E6 antigen. Antibodies or other molecules that react with 162P1E6 can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.
XII.) Inhibition of 162P1E6 Protein Function
The invention includes various methods and compositions for inhibiting the binding of 162P1E6 to its binding partner or its association with other protein(s) as well as methods for inhibiting 162P1E6 function.
XII.A.) Inhibition of 162P1E6 With Intracellular Antibodies
In one approach, a recombinant vector that encodes single chain antibodies that specifically bind to 162P1E6 are introduced into 162P1E6 expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti-162PlE6 antibody is expressed intracellularly, binds to 162P1E6 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, 1. 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 deshed 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 theh natural cellular destination.
In one embodiment, intrabodies are used to capture 162P1E6 in the nucleus, thereby preventing its activity within the nucleus. Nuclear targeting signals are engineered into such 162P1E6 intrabodies in order to achieve the deshed targeting. Such 162P1E6 intrabodies are designed to bind specifically to a particular 162P1E6 domain. In another embodiment, cytosolic intrabodies that specifically bind to a 162P1E6 protein are used to prevent 162P1E6 from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing 162P1E6 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. Patent No. 5,919,652 issued 6 July 1999). XH.B.) Inhibition of 162P1E6 with Recombinant Proteins
In another approach, recombinant molecules bind to 162P1E6 and thereby inhibit 162P1E6 function. For example, these recombinant molecules prevent or inhibit 162P1E6 from accessing/binding to its binding partner(s) or associating with other ρrotein(s). Such recombinant molecules can, for example, contain the reactive part(s) of a 162P1E6 specific antibody molecule. In a particular embodiment, the 162P1E6 binding domain of a 162P1E6 binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two 162P1E6 ligand binding domains linked to the Fc portion of a human IgG, such as human IgGl. 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 162P1E6, whereby the dimeric fusion protein specifically binds to 162P1E6 and blocks 162P1E6 interaction with a binding partner. Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.
Xπ.C.) Inhibition of 162P1E6 Transcription or Translation
The present invention also comprises various methods and compositions for inhibiting the transcription of the 162P1E6 gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of 162P1E6 mRNA into protein.
In one approach, a method of inhibiting the transcription of the 162P1E6 gene comprises contacting the 162P1E6 gene with a 162P1E6 antisense polynucleotide. In another approach, a method of inhibiting 162P1E6 mRNA translation comprises contacting a 162P1E6 mRNA with an antisense polynucleotide. In another approach, a 162P1E6 specific ribozyme is used to cleave a 162P1E6 message, thereby inhibiting translation. Such antisense and ribozyme based methods can also be dhected to the regulatory regions of the 162P1E6 gene, such as 162P1E6 promoter and/or enhancer elements. Similarly, proteins capable of inhibiting a 162P1E6 gene transcription factor are used to inhibit 162P1E6 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 162P1E6 by interfering with 162P1E6 transcriptional activation are also useful to treat cancers expressing 162P1E6. Similarly, factors that interfere with 162P1E6 processing are useful to treat cancers that express 162P1E6. Cancer treatment methods utilizing such factors are also within the scope of the invention.
Xπ.D.) General Considerations for Therapeutic Strategies
Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing 162P1E6 (i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 162P1E6 inhibitory molecules). A number of gene therapy approaches are known in the art. Recombinant vectors encoding 162P1E6 antisense polynucleotides, ribozymes, factors capable of interfering with 162P1E6 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, ribθ2yme, 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 deteπrrinhig the extent to which a therapeutic composition will inhibit the binding of 162P1E6 to a binding partner, etc.
In vivo, the effect of a 162P1E6 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 W098/16628 and U.S. Patent 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 deshed 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, inttatumor, 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 162PlE6-related protein or a 162P1E6 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 Figure 2 or Figure 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 deshable 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 162P1E6 Gene
To isolate genes that are over-expressed in bladder cancer we used the Suppression Subtractive Hybridization (SSH) procedure using cDNA derived from bladder cancer patient tissues.
The 162P1E6 SSH cDNA sequence was derived from a subtraction consisting of a baldder cancer minus normal bladder and a mixture of 9 normal tissues: stomach, skeletal muscle, lung, brain, liver, kidney, pancreas, small intestine and heart. The 162P1E6 SSH cDNA sequence of 335 bp, listed in Figure 1, did not show homology to any known gene.
The full-length 162P1E6 v.l clone B was cloned from bladder cancer cDNA, revealing an ORF of 146 amino acids (Figure 2 and Figure 3). Other variants of 162P1E6 were also identified and these are listed in Figures 2 and 3.
Materials and Methods
Human Tissues:
The patient cancer and normal tissues were purchased from different sources such as the NDRI (Philadelphia, PA). mRNA for some normal tissues were purchased from Clontech, Palo Alto, CA.
RNA Isolation:
Tissues were homogenized in Trizol reagent (Life Technologies, Gibco BRL) using 10 ml/ g tissue 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): 5'TTTTGATCAAGCTT303' (SEQ ID NO: )
Adaptor 1 :
5'CTAATACGACTCACTATAGGGCTCGAGCGGCCGCCCGGGCAG3' (SEQ ID NO: )
3OGCCCGTCCTAG5' (SEQ ID NO: )
Adaptor 2:
5'GTAATACGACTCACTATAGGGCAGCGTGGTCGCGGCCGAG3' (SEQ IDNO: )
3'CGGCTCCTAG5' (SEQ IDNO: )
PCRprimer 1:
5'CTAATACGACTCACTATAGGGC3' (SEQ ID NO: Nested primer (NP) 1 : 5'TCGAGCGGCCGCCCGGGCAGGA3' (SEQ ID NO: )
Nested primer (NP)2: 5ΑGCGTGGTCGCGGCCGAGGA3' (SEQ ID NO: )
Suppression Subtractive Hybridization:
Suppression Subtractive Hybridization (SSH) was used to identify cDNAs corresponding to genes that may be differentially expressed in bladder cancer. The SSH reaction utilized cDNA from bladder cancer and normal tissues.
The gene 162P1E6 was derived from bladder cancer minus normal tissue cDNA subtraction. The 162P1E6 SSH DNA sequence (Figure 1) was identified.
The cDNA derived from of pool of normal tissues was used as the source of the "driver" cDNA, while the cDNA from a pool of bladder cancer tissues was used as the source of the "tester" cDNA. Double stranded cDNAs corresponding to tester and driver cDNAs were synthesized from 2 μg of ρoly(A)+ RNA isolated from the relevant xenograft tissue, as described above, using CLONTECH's PCR-Select cDNA Subtraction Kit and 1 ng of oligonucleotide DPNCDN as primer. Fhst- and second-strand synthesis were carried out as described in the Kit's user manual protocol (CLONTECH Protocol No. PT1117-1, Catalog No. KI 804-1). 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.
Driver cDNA was generated by combining in a 1 : 1 ratio Dpn II digested cDNA from the relevant tissue source (see above) with a mix of digested cDNAs derived from the nine normal tissues: stomach, skeletal muscle, lung, brain, liver, kidney, pancreas, small intestine, and heart.
Tester cDNA was generated by diluting 1 μl 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 200 μl 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 x reaction buffer (CLONTECH) and 0.5 μl 50 x 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 ρCR2.1 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 elecfrophoresis.
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:
Fhst 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. Fhst strand cDNAs from 16 different normal human tissues can be obtained from Clontech.
Normalization of the fhst sttand cDNAs from multiple tissues was performed by using the primers
5'atatcgccgcgctcgtcgtcgacaa3' (SEQ TD NO: ) and 5'agccacacgcagctcattgtagaagg 3' (SEQ T NO: ) to amplify β-actin. Fhst strand cDNA (5 μl) were amplified in a total volume of 50 μl containing 0.4 μM primers, 0.2 μM each dNTPs, lXPCR buffer (Clontech, 10 mM Tris-HCL, 1.5 mM MgCl2, 50 mM KCl, pH8.3) and IX 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 b.p. 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 requhed to achieve equal band intensities in all tissues after 22 cycles of PCR.
To determine expression levels of the 162P1E6 gene, 5 μl of normalized fhst 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. The primers used for RT-PCR were designed using the 162P1E6 SSH sequence and are listed below:
162P1E6.1
5 '- CTCAGGATTACGTCCCAAGTGTCT - 3 ' (SEQ ID NO: )
162P1E6.2
5 '- ATAAGGTGGGTGCTGACCAGTTT - 3 ' (SEQ ID NO: ) A typical RT-PCR expression analysis is shown in Figure 14. Fhst 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-9 AD and LAPC-9 AI), prostate cancer pool, bladder cancer pool, lung cancer pool, breast cancer pool, and cancer metastasis pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi- quantitative PCR, using primers to 162P1E6, was performed at 26 and 30 cycles of amplification. Results show strong expression of 162P1E6 in bladder cancer pool, lung cancer pool, and breast cancer pool. Expression was also detected in prostate cancer pool and cancer metastasis pool, but not in the vital pools.
Example 2; Full Length Cloning of 162P1E6
To isolate genes that are over-expressed in bladder cancer we used the Suppression Subtractive Hybridization (SSH) procedure using cDNA derived from bladder cancer patient tissues.
The 162P1E6 SSH cDNA sequence was derived from a subtraction consisting of a bladder cancer minus normal bladder and a mixture of 9 normal tissues: stomach, skeletal muscle, lung, brain, liver, kidney, pancreas, small intestine and heart. The 162P1E6 SSH cDNA sequence of 335 bp, listed in Figure 1, did not show homology to any known gene.
The full-length 162P1E6 v.l clone B was cloned from bladder cancer cDNA, revealing an ORF of 146 amino acids (Figure 2A and Figure 3A). 162P1E6 v.l showed 99%> identity over 1860 nucleotides (from 1345 to 3204 of 162P1E6 v.l) with the hypothetical gene XP_036612 (AK002208) (Figure 4A). 162P1E6 v.l protein showed 100% identity over 146 amino acids with the hypothetical protein XP_036612 (AK002208) of unknown function (Figure 4B). Also, 162P1E6 has 35% identity over a 71 amino acid region to the Man7GlcNAc2-PP-dolichyl mannosyltransferase, and 38% identity over a 39 amino acid region homology to the estrogen receptor beta2 splice variant (Figures 4C and 4D, respectively).
Other variants of 162P1E6 were also identified and these are listed in Figures 2 and 3. 162P1E6 v.3, v.4, v.5, and v.6 code for proteins that are different from 162P1E6 v.l. The 162P1E6 v.3, v.4, v.5, and v.6 are novel and have not been previously described in public databases. 162P1E6 v.18 codes for the same protein as 162P1E6 v.l except for one amino acid at position 130.
Example 3; Chromosomal Mapping of 162P1E6
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 Al), human-rodent somatic cell hybrid panels such as is available from the Coriell Institute (Camden, New Jersey), and genomic viewers utilizing BLAST homologies to sequenced and mapped genomic clones (NCBI, Bethesda, Maryland).
162P1E6 maps to chromosome lq32.2 using 162P1E6 sequence and the NCBI BLAST tool: (htφ://www.ncbi.nlm.nih.gov/genome/seq/page.cgi?F=HsBlast.html&&ORG=Hs).
Example 4: Expression Analysis of 162P1E6 in Normal Tissues and Patient Specimens
Expression analysis by RT-PCR demonstrated that 162P1E6 is strongly expressed in cancer patient specimens (Figure 14). Fhst 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, lung cancer pool, breast cancer pool, and cancer metastasis pool. Normalization was performed by PCR using primers to actin and GAPDH. Semi-quantitative PCR, using primers to 162P1E6, was performed at 26 and 30 cycles of amplification. Results show strong expression of 162P1E6 in bladder cancer pool, lung cancer pool, and breast cancer pool. Expression was also detected in prostate cancer pool and cancer metastasis pool, but not in the vital pools.
Extensive northern blot analysis of 162P1E6 in multiple human normal tissues is shown in Figure 15. Two multiple tissue northern blots (Clontech) both with 2 ug of mRNA/lane were probed with the 162P1E6 SSH sequence. Size standards in kilobases (kb) are indicated on the side. Results show expression of two approximately 4.4 kbl62PlE6 transcripts in placenta, prostate and thymus.
Expression of 162P1E6 in patient bladder cancer specimens is shown in Figure 16 RNA was extracted from normal bladder (Nb), bladder cancer cell lines (CL: UM-UC-3, J82 and SCaBER), bladder cancer patient tumors (T) and normal tissue adjacent to bladder cancer (N). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in the bladder tumor tissues and in the SCaBER cancer cell line, but not in normal bladder, nor in the other cancer cell lines J82 and UM-UC-3.
Figure 17 shows that 162P1E6 was expressed in prostate cancer patient specimens. RNA was extracted from LAPC-4AD, LAPC-4AI, LAPC-9 AD and LAPC-9AI prostate cancer xenografts, normal prostate (N), prostate cancer patient tumors (T) and theh normal adjacent tissues (NAT). Northern blot with 10 ug of total RNA/lane was probed with 162P1E6 SSH sequence. Size standards in kilobases (kb) are indicated on the side. The results show strong expression of 162P1E6 in normal prostate and in patient prostate cancer specimens. Weak expression was detected in the LAPC-4AD tissue, but not in the other prostate cancer xenografts.
Expression of 162P1E6 was also detected in kidney cancer patient specimens (Figure 18). RNA was extracted from kidney cancer cell lines (769-P, A498, SW839), normal kidney (N), kidney cancer patient tumors (T) and theh normal adjacent tissues (NAT). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in 2 out of 2 papillary kidney tumor tissues but not in specimens of renal clear cell carcinoma, normal kidney, nor in the kidney cancer cell lines.
Figure 19 shows that 162P1E6 was expressed in lung cancer patient specimens. RNA was extracted from lung cancer cell lines (CALU-1, A427, NCI-H82, NCI-H146), normal lung (N), lung cancer patient tumors (T) and normal adjacent tissues (NAT) isolated from lung cancer patients. Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Size standards in kilobases are indicated on the side. Results show strong expression of 162P1E6 in the all lung tumor tissues tested, but not in normal lung nor in the lung cancer cell lines.
In Figure 20, expression of 162P1E6 was tested in breast cancer patient specimens. RNA was extracted from breast cancer cell lines (DU4475, MCF7 and CAMA-1), normal breast (N), breast cancer patient tumors (T), breast cancer metastasis to lymph node (Ml), and to ovary (M2). Northern blots with 10 ug of total RNA were probed with the 162P1E6 SSH fragment. Results show expression of 162P1E6 in normal breast, breast tumor tissues as well as in the cancer metastasis specimens, but not in the breast cancer cell lines tested.
The restricted expression of 162P1E6 in normal tissues and the expression detected in bladder cancer, breast cancer, lung, prostate, kidney and cancer metastases suggest that 162P1E6 is a potential therapeutic target and a diagnostic marker for human cancers.
Example 5: Transcript Variants of 162P1E6
Transcript variants are variants of matured mRNA from the same gene by alternative transcription or alternative splicing. Alternative transcripts are transcripts from the same gene but start ttanscription at different points. Splice variants are mRNA variants spliced differently from the same transcript. In eukaryotes, when a multi-exon gene is transcribed from genomic DNA, the initial RNA is 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 alternative transcripts and each transcript can have zero to many splice variants. Each transcript variant has a unique exon makeup, and can have different coding and/or non-coding (5 ' or 3' end) portions, from the original ttanscript. Transcript variants can code for similar or different 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, or at different times, proteins encoded by transcript variants can have similar or different cellular or extracellular localizations, i.e., be secreted.
Transcript variants are identified by a variety of art-accepted methods. For example, alternative transcripts and splice variants are identified in a full-length cloning experiment, or by use of full-length transcript and EST sequences. Fhst, all human ESTs were grouped into clusters which show dhect 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 original gene sequence is compared to the consensus sequence(s) or other full-length sequences. Each consensus sequence is a potential splice variant for that gene (see, e.g., http://www.doubletwist.com/products/cl l_agentsOverview.jhtml). 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 transcript variants based on genomic sequences. Genomic-based transcript 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 (http://compbio.ornl.gov/Grail-bin/EmptyGrailForm) and GenScan (http://genes.mit.edu GENSCAN.html). 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 U S A. 2000 Nov 7; 97(23): 12690-3.
To further confirm the parameters of a transcript 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., et al, Albumin banks peninsula: a new termination variant characterized by electtospray mass spectrometry, Biochem Biophys Acta. 1999 Aug 17;1433(l-2):321-6; Ferranti P, et al, Differential splicing of pre-messenger RNA produces multiple forms of mature caprine alpha(sl)-casein, Eur J Biochem. 1997 Oct l;249(l):l-7. For 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 Apr;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(l-2):211-8. For 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, Biochem 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 a gene maps is modulated in a particular cancer, the alternative ttanscripts or splice variants of the gene are modulated as well. Disclosed herein is that 162P1E6 has a particular expression profile. Alternative transcripts and splice variants of 162P1E6 that are structurally and/or functionally similar to 162P1E6 share this expression pattern, thus serving as tumor associated markers/antigens.
The exon composition of the original transcript is designated as 162P1E6 v.l. Using the full-length gene and EST sequences, an alternative transcript, designated as 162P1E6 v.2, and nine splice variants of this alternative transcript were identified, designated as 162P1E6 v.3 through 162P1E6 v.l 1. In comparison with 162P1E6 v.l the alternative transcript 162P1E6 v.2 had an additional 522 bp at the 5' end. Both 162P1E6 v.l and v.2 were single exon ttanscripts. Based on the splicing pattern for transcript 162P1E6 v.l and v.2 can, they may be divided into splicing segments as indicated in Table LIII(A), LIII(B) and Figure 12. Since 162P1E6 v.l and v.2 share the same 3240 bp sequence, 162P1E6 v.l may also be spliced in a similar pattern to generate similar splice variants. Each different combination of exons in spatial order, e.g. exons 1, 2, 3, 4 and 7, is a potential splice variant. Figure 12 provides the schematic alignment of the exons of 162P1E6 v.l through v.l 1.
Tables LIII through LVII are set forth on a variant-by-variant basis. Table LIV shows the nucleotide sequence of transcript variants 2-11 (Tables LIV(A)-LIV(J), respectively). Table LV provides alignments of the transcript variant, 162P1E6 v.2, with the following nucleic acid sequences: of 162P1E6 v.l (LV(A)), 162P1E6 v.3 (Table LV(B)), 162P1E6 v.4 (Table LV(C)), 162P1E6 v.5 (Table LV(D)), 162P1E6 v.6 (Table LV(E)), 162P1E6 v.7 (Table LV(F)), 162P1E6 v.8 (Table LV(G)), 162P1E6 v.9 (Table LV(H)), 162P1E6 v.10 (Table LV(I)), and 162P1E6 v.11 (Table LV(J)). Table LVI (A-J) provides the amino acid translations of 162P1E6 variant 2 through variant 11 for theh identified reading frame orientations. Table LVII provides alignments of the amino acid sequence encoded by the transcript variant, 162P1E6 v.2, with that of 162P1E6 v.l (Table LVπ(A)), 162P1E6 v.3 (Table LVTI(B)), 162P1E6 v.4 (Table LVII(C)), 162P1E6 v.5 (Table LVH(D)), 162P1E6 v.6 (Table LVII(E)), 162P1E6 v.7 (Table LVTI(F)), 162P1E6 v.8 (Table LVII(G)), 162P1E6 v.9 (Table LVII(H)), 162P1E6 v.10 (Table LVTI(I)), and 162P1E6 v.ll (Table LVH(J)).
Example 6: Single Nucleotide Polymorphisms of 162P1E6
A Single Nucleotide Polymorphism (SNP) is a single base pah variation in nucleotide sequences. At a specific point of the genome, there are four possible nucleotide base pahs: A/T, C/G, G/C and T/A. Genotype refers to the base pah make-up of one or more spots in the genome of an individual, while haplotype refers to base pah 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 disearses 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 Oct; 11(5):637-641; M. Pirmohamed and B. K. Park, "Genetic susceptibility to adverse drug reactions," Trends Pharmacol. Sci. 2001 Jun; 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 Feb; l(l):39-47; R. Judson, J. C. Stephens and A. Windemuth, "The predictive power of haplotypes in clinical response," Pharmacogenomics. 2000 feb; 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 Oct-Nov; 20(9):18-20; K. M. Weiss, "In search of human variation," Genome Res. 1998 Jul; 8(7):691-697; M. M. She, "Enabling large-scale pharmacogenetic studies by high- throughput mutation detection and genotyping technologies," Clin. Chem. 2001 Feb; 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 dhect 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 dhect 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 Dec; 5(4):329-340).
Using the methods described above, ten SNPs were identified in the transcripts. Using 162P1E6 v.2 as template, the SNPs were located at positions 218 (G/A), 1197 (C/G), 1832 (G/A), 2314 (C/A), 2570 (T/A), 2630 (G/A), 2938 (A/G), 3597 (G/A), 3629 (A/C) and 3692 (A/G) (see Figure 12). The transcripts or proteins with alternative alleles were designated as variants 162P1E6 v.12, v.13, v.14, v.15, v.16, v.17, v.18, v.19, v.20 and v.21. Figure 10 shows the schematic alignment of the SNP variants. Figure 11 shows the schematic alignment of protein variants, corresponding to ttanscript variants and SNP variants. Nucleotide variants that code for the same amino acid sequence as variant 1 are not shown in Figure 11. These alleles of the SNPs, though shown separately here, can occur in different combinations (haplotypes) and in any one of the transcript variants that contains the sequence context of the SNPs, e.g., 162P1E6 v.l, 162P1E6 v.2 or 162PlE6 v.ll. Example 7: Production of Recombinant 162P1E6 in Prokaryotic Systems
To express recombinant 162P1E6 and 162P1E6 variants in prokaryotic cells, the full or partial length 162P1E6 and 162P1E6 variant cDNA sequences are cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 162P1E6 variants are expressed: the full length sequence presented in Figures 2 and 3, 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 162P1E6, variants, or analogs thereof.
A. In vitro transcription and translation constructs: pCRII: To generate 162P1E6 sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad CA) are generated encoding either all or fragments of the 162P1E6 cDNA. The pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of 162P1E6 RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of 162P1E6 at the RNA level. Transcribed 162P1E6 RNA representing the cDNA amino acid coding region of the 162P1E6 gene is used in in vitro translation systems such as the TnT™ Coupled Reticulolysate System (Promega, Corp., Madison, WI) to synthesize 162P1E6 protein.
B. Bacterial Constructs: pGEX Constructs: To generate recombinant 162P1E6 proteins in bacteria that are fused to the Glutathione S-transferase (GST) protein, all or parts of the 162P1E6 cDNA protein coding sequence are cloned into the pGEX family of GST-fusion vectors (Amersham Pharmacia Biotech, Piscataway, NJ). These constructs allow controlled expression of recombinant 162P1E6 protein sequences with GST fused at the ammo-terminus and a six histidine epitope (6X His) at the carboxyl-terminus. The GST and 6X 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 6X 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-l, may be employed such that it permits cleavage of the GST tag from 162PlE6-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 162P1E6 proteins that are fused to maltose-binding protein (MBP), all or parts of the 162P1E6 cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, MA). These constructs allow controlled expression of recombinant 162P1E6 protein sequences with MBP fused at the amino-terminus and a 6X His epitope tag at the carboxyl-terminus. The MBP and 6X 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 6X 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 162P1E6. The ρMAL-c2X and pMAL-ρ2X 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 162P1E6 in bacterial cells, all or parts of the 162P1E6 cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, WI). These vectors allow tightly controlled expression of recombinant 162P1E6 protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6X 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 162P1E6 protein are expressed as arnmo-teiminal fusions to NusA.
C. Yeast Constructs: pESC Constructs: To express 162P1E6 in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the 162P1E6 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, CA). 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 162P1E6. 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 162P1E6 in the yeast species Saccharomyces pombe, all or parts of the 162P1E6 cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a 162P1E6 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 162P1E6 in Eukaryotic Systems A. Mammalian Constructs:
To express recombinant 162P1E6 in eukaryotic cells, the full or partial length 162P1E6 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 162P1E6 are expressed in these constructs, amino acids 1 to 146 of 162P1E6 v.l and v.18, amino acids 1 to 133 of 162P1E6 v.3, amino acids 1 to 102 of 162P1E6 v.4, amino acids 1 to 76 of 162P1E6 v.5, amino acids 1 to 70 of 162P1E6 v.6, 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, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more contiguous amino acids from 162P1E6, variants, or analogs thereof. In certain embodiments a region of a specific variant of 162P1E6 is expressed that encodes an amino acid at a specific position which differs from the amino acid of any other variant found at that position. In other embodiments, a region of a variant of 162P1E6 is expressed that lies partly or entirely within a sequence that is unique to that variant.
The constructs can be transfected into any one of a wide variety of mammahan cells such as 293T cells. Transfected 293T cell lysates can be probed with the anti-162PlE6 polyclonal serum, described herein. pcDNA4/HisMax Constructs: To express 162P1E6 in mammalian cells, a 162P1E6 ORF, or portions thereof, of 162P1E6 are cloned into pcDNA4 HisMax Version A (Invitrogen, Carlsbad, CA). Protein expression is driven from the cytomegalovirus (CMV) promoter and the SP16 translational enhancer. The recombinant protein has Xpress™ and six histidine (6X His) epitopes fused to the ammo-terminus. The pcDNA4/HisMax vector also contains the bovine growth hormone (BGH) polyadenylation signal and ttanscription 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 ColEl origin permits selection and maintenance of the plasmid in E. coli. pcDNA3.1/MvcHis Constructs: To express 162P1E6 in mammalian cells, a 162P1E6 ORF, or portions thereof, of 162P1E6 with a consensus Kozak translation initiation site was cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, CA). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant protein has the myc epitope and 6X 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 was used, as it allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColEl origin permits selection and maintenance of the plasmid in E. coli. Results of expression from 162PlE6.pcDNA3.1/MycHis construct are shown in Figure 21A. pcDNA3.1/CT-GFP-TOPO Construct:' To express 162P1E6 in mammalian cells and to allow detection of the recombinant proteins using fluorescence, a 162P1E6 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 ttanscription 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 ColE 1 origin permits selection and maintenance of the plasmid in E. coli. Additional constructs with an amino-terminal GFP fusion are made inpcDNA3.1/NT-GFP-TOPO spanning the enthe length of a 162P1E6 protein.
PAPtag: A 162P1E6 ORF, or portions thereof, is cloned into pAPtag-5 (GenHunter Corp. Nashville, TN). This construct generates an alkaline phosphatase fusion at the carboxyl-terminus of a 162P1E6 protein while fusing the IgGκ signal sequence to the amino-terminus. Constructs are also generated in which alkaline phosphatase with an amino-terminal IgGκ signal sequence is fused to the amino-terminus of a 162P1E6 protein. The resulting recombinant 162P1E6 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 162P1E6 proteins. Protein expression is driven from the CMV promoter and the recombinant proteins also contain myc and 6X 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 162P1E6 ORF was cloned into pTag-5. This vector is similar to pAPtag but without the alkaline phosphatase fusion. This construct generated 162P1E6 protein with an amino-terminal IgGκ signal sequence and myc and 6X His epitope tags at the carboxyl-terminus that facilitate detection and affinity purification. The resulting recombinant 162P1E6 protein was 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 162P1E6 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. Results of expression from 162PlE6.ρTag5 construct are shown in Figure 21B.
PsecFc: A 162P1E6 ORF, or portions thereof, is also cloned into psecFc The psecFc vector was assembled by cloning the human immunoglobulin GI (IgG) Fc (hinge, CH2, CH3 regions) into pSecTag2 (Invitrogen, California). This construct generates an IgGl Fc fusion at the carboxyl-terminus of the 162P1E6 proteins, while fusing the IgGK signal sequence to N-terminus. 162P1E6 fusions utilizing the murine IgGl Fc region are also used. The resulting recombinant 162P1E6 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 162P1E6 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 162P1E6 constitutively, 162P1E6 ORF, or portions thereof, of 162P1E6 are cloned into pSRα constructs. Amphotropic and ecotropic retroviruses are generated by transfection of pSRα constructs into the 293T-10A1 packaging line or co- ttansfection of pSRα and a helper plasmid (containing deleted packaging sequences) into the 293 cells, respectively. The retrovirus is used to infect a variety of mammalian cell lines, resulting in the integration of the cloned gene, 162P1E6, into the host cell-lines. Protein expression is 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 ColEl origin permit selection and maintenance of die plasmid in E. coli. The retroviral vectors can thereafter be used for infection and generation of various cell lines using, for example, PC3, NIH 3T3, TsuPrl, 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 162P1E6 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: ) is added to cloning primer at the 3' end of the ORF. Additional pSRα constructs are made to produce both ammo-terminal and carboxyl-terminal GFP and myc/6X His fusion proteins of the full-length 162P1E6 proteins.
Additional Viral Vectors: Additional constructs are made for vhal-mediated delivery and expression of 162P1E6. High virus titer leading to high level expression of 162P1E6 is achieved in vhal delivery systems such as adenovhal vectors and herpes amplicon vectors. A 162P1E6 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 adenovhal vectors. Alternatively, 162P1E6 coding sequences or fragments thereof are cloned into the HSV-1 vector (Imgenex) to generate herpes vhal vectors. The vhal 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 162P1E6 in mammalian cells, coding sequences of 162P1E6, 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 162P1E6. These vectors are thereafter used to control expression of 162P1E6 in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
B. Baculovirus Expression Systems
To generate recombinant 162P1E6 proteins in a baculovirus expression system, 162P1E6 ORF, or portions thereof, are cloned into the baculovirus transfer vector pBlueBac 4.5 (Invittogen), which provides a His-tag at the N-terminus. Specifically, ρBlueBac-162PlE6 is co-transfected with helper plasmid pBac-N- Blue (Invitrogen) into SF9 (Spodoptera fi-ugiperda) insect cells to generate recombinant baculovirus (see Invittogen instruction manual for details). Baculovirus is then collected from cell supernatant and purified by plaque assay.
Recombinant 162P1E6 protehi is then generated by infection of HighFive insect cells (Invitrogen) with purified baculovirus. Recombinant 162P1E6 protein can be detected using anti-162PlE6 or anti-His-tag antibody. 162P1E6 protein can be purified and used in various cell-based assays or as immunogen to generate polyclonal and monoclonal antibodies specific for 162P1E6.
Example 9: Antigenicitv Profiles and Secondary Structure
Figure 5 A-E, Figure 6A-E, Figure 7 -E, Figure 8 A-E, and Figure 9 A-E depict graphically five amino acid profiles of the 162P1E6 variants 1, 3, 4, 5, and 6, respectively, each assessment available by accessing the ProtScale website (URL www.expasy.ch/cgi-bin protscale.pl) on the ExPasy molecular biology server.
These profiles: Figure 5, Hydrophilicity, (Hopp T.P., Woods K.R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828); Figure 6, Hydropathicity, (Kyte J., Doolittle R.F., 1982. J. Mol. Biol. 157:105-132); Figure 7, Percentage Accessible Residues (Janin J., 1979 Nature 277:491-492); Figure 8, Average Flexibility, (Bhaskaran R., and Ponnuswamy P.K., 1988. Int. J. Pept. Protein Res. 32:242-255); Figure 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 162P1E6 protein. Each of the above amino acid profiles of 162P1E6 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 he between 0 and 1.
Hydrophilicity (Figure 5), Hydropathicity (Figure 6) and Percentage Accessible Residues (Figure 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 (Figure 8) and Beta-turn (Figure 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 162P1E6 variant proteins indicated, e.g., by the profiles set forth in Figure 5A-E, Figure 6A-E, Figure 7A-E, Figure 8A-E, and/or Figure 9A-E are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-162PlE6 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 162P1E6 protein variants listed in Figures 2 and 3 (Variants 1, 3, 4, 5, and 6). In particular, peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profiles of Figure 5; a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figures 6 ; a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profiles of Figure 7; a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profiles on Figure 8 ; and, a peptide region of at least 5 amino acids of Figures 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figures 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 162P1E6 variant proteins 1, 3, 4, 5, and 6, 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, http://pbil.ibcp.fr/cgi- bin/npsa_automat.pl?page=npsa_nn.html), accessed from the ExPasy molecular biology server (http://www.expasy.ch/tools/). The analysis indicates that 162P1E6 variant 1 is composed of 21.92% alpha helix, 28.08% extended strand, and 50.00% random coil (Figure 13A). Variant 3 is composed of 29.32%, alpha helix, 19.55% extended strand, and 51.13% random coil (Figure 13B). Variant 4 is composed of 37.25% alpha helix, 13.73% extended strand, and 49.02% random coil (Figure 13C). Variant 5 is composed of 11.84% alpha helix, 19.74% extended strand, and 68.42% random coil (Figure 13D). Variant 6 is composed of 14.29% alpha helix, 21.43% extended strand, and 64.29% random coil (Figure 13E).
Analysis for the potential presence of transmembrane domains in the 162P1E6 variant proteins was carried out using a variety of transmembrane prediction algorithms accessed from the ExPasy molecular biology server (http://www.expasy.ch/tools/). The programs do not predict the presence of transmembrane domains in the 162P1E6 protein variants, suggesting that they are soluble proteins.
Example 10: Generation of 162P1E6 Polyclonal Antibodies
Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if deshed, 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 a full length 162P1E6 protein variant, 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 hnmunized host (see the Example entitled "Antigenicity Profiles"). 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., Figure 5A-E, Figure 6A-E, Figure 7A-E, Figure 8A-E, or Figure 9A-E for amino acid profiles that indicate such regions of 162P1E6 protein variants).
For example, recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta- turn regions of 162P1E6 protein variants are used as antigens to generate polyclonal antibodies in New Zealand White rabbits. For example, in 162P1E6 variant 1, such regions include, but are not limited to, amino acids 1-15, amino acids 25-38, amino acids 44-54, and amino acids 122-132. 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 1-15 of 162P1E6 variant 1 is conjugated to KLH and used to immunize the rabbit. Alternatively the immunizing agent may include all or portions of the 162P1E6 variant proteins, analogs or fusion proteins thereof. For example, the 162P1E6 variant 1 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 GST-fusion protein encoding the full length 162P1E6 variant 1 gene, amino acids 1-146, is produced and purified and used as immunogen. Other recombinant bacterial fusion proteins that may be employed include maltose binding protein, LacZ, thioredoxin, NusA, or an immunoglobulin constant region (see the section entitled "Production of 162P1E6 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 162P1E6 in Eukaryotic Systems"), and retain post- translational modifications such as glycosylations found in native protein. In one embodiment, the full length sequence of variant 1, amino acids 1-146, is cloned into the Tag5 mammalian secretion vector. The recombinant protein is purified by metal chelate chromatography from tissue culture supematants of 293T cells stably expressing the recombinant vector. The purified Tag5 162P1E6 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 ttehalose 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 (IF A). 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 a KLH-conjugated peptide encoding amino acids 1-15 of variant 1, the full-length 162P1E6 variant 1 cDNA is cloned into pCDNA 3.1 myc-his expression vector (Invittogen, see the Example entitled "Production of Recombinant 162P1E6 in Eukaryotic Systems"). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-162PlE6 serum and with anti-His antibody (Santa Cruz Biotechnologies, Santa Cruz, CA) to determine specific reactivity to denatured 162P1E6 protein using the Western blot technique. Figure 21 shows expression of Myc His epitope tagged 162P1E6 variant 1 protein in 293T cells as detected by an anti-His antibody. In addition, the immune serum is tested by fluorescence microscopy, flow cytometry and immunoprecipitation against 293T and other recombinant 162P1E6- expressing cells to determine specific recognition of native protein. Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express 162P1E6 are also carried out to test reactivity and specificity.
Anti-serum from rabbits immunized with 162P1E6 variant 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. For example, antiserum derived from a GST-162P1E6 variant 1 fusion protein encoding amino acids 1-146 is first purified by passage over a column of GST protein covalently coupled to AffiGel matrix (BioRad, Hercules, Calif.). The antiserum is then affinity purified by passage over a column composed of a MBP- fusion protein also encoding amino acids 1-146 covalently coupled to Affigel matrix. The serum is then further purified by protein G affinity chromatography to isolate the IgG fraction. 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 162P1E6 Monoclonal Antibodies mAbs)
In one embodiment, therapeutic mAbs to 162P1E6 variants comprise those that react with epitopes specific for each variant protein or specific to sequences in common between the variants that would disrupt or modulate the biological function of the 162P1E6 variants, for example those that would disrupt the interaction with ligands and binding partners. Immunogens for generation of such mAbs include those designed to encode or contain the entire 162P1E6 protein variant sequence, regions of the 162P1E6 protein variants predicted to be antigenic from computer analysis of the amino acid sequence (see, e.g., Figure 5A-E, Figure 6A-E, Figure 7A-E, Figure 8A-E, or Figure 9A-E, and the Example entitled "Antigenicity Profiles"). Immunogens include peptides, recombinant bacterial proteins, and mammalian expressed Tag 5 proteins and human and murine IgG FC fusion proteins. In addition, cells engineered to express high levels of a respective 162P1E6 variant, such as 293T-162P1E6 variant 1 or 300.19-162P1E6 variant lmurine Pre-B cells, are used to immunize mice.
To generate mAbs to a 162P1E6 variant, mice are first immunized intraperitoneally (IP) with, typically, 10-50 μg of protein immunogen or IO7 162PlE6-expressrng cells mixed in complete Freund's adjuvant. Mice are then subsequently immunized EP every 2-4 weeks with, typically, 10-50 μg of protein immunogen or IO7 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 a 162P1E6 variant sequence is used to immunize mice by dhect injection of the plasmid DNA. For example, the full length variant 1 sequence, encoding amino acids 1-146, 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 162P1E6 variant 1 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 the respective 162P1E6 variant.
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 162P1E6 monoclonal antibodies, a Tag5-162P1E6 variant 1 antigen encoding amino acids 1-146, is expressed and purified from stably transfected 293T cells. Balb C mice are initially immunized intraperitoneally with 25 μg of the Tag5-162P1E6 variant 1 protein mixed in complete Freund's adjuvant. Mice are subsequently immunized every two weeks with 25 μg of the antigen mixed in incomplete Freund's adjuvant for a total of three immunizations. ELISA using the Tag5 antigen determines the titer of serum from immunized mice. Reactivity and specificity of serum to full length 162P 1E6 variant 1 protehi is monitored by Western blotting, immunoprecipitation and flow cytometry using 293T cells transfected with an expression vector encoding the 162P1E6 variant 1 cDNA (see e.g., the Example entitled "Production of Recombinant 162P1E6 in Eukaryotic Systems" and Figure 21). Other recombinant 162P1E6 variant 1-expressing cells or cells endogenously expressing 162P1E6 variant 1 are also used. Mice showing the strongest reactivity are rested and given a final injection of Tag5 antigen in PBS and then sacrificed four days later. The spleens of the sacrificed mice are harvested and fused to SPO/2 myeloma cells using standard procedures (Harlow and Lane, 1988). Supernatants from HAT selected growth wells are screened by ELISA, Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometry to identify 162P1E6 specific antibody-producing clones.
The binding affinity of a 162P1E6 monoclonal antibody is determined using standard technologies. Affinity measurements quantify the strength of antibody to epitope binding and are used to help define which 162P1E6 monoclonal antibodies preferred for diagnostic or therapeutic use, as appreciated by one of skill in the art. The BIAcore system (Uppsala, Sweden) is a 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 inliibition and dhect 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 (see Table TV).
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 the Example entitled "Antigenicity Profiles" and Tables V-XVIII and XXII-LI employ the protein sequence data from the gene product of 162P1E6 set forth in Figures 2 and 3, the specific peptides used to generate the tables are listed in table LII.
Computer searches for epitopes bearing HLA Class I or Class II supermotifs or motifs are performed as follows. All translated 162P1E6 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 theh 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" = a x a2i x a3; x a,„- where a,-,- is a coefficient which represents the effect of the presence of a given amino acid (j) at a given position (0 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 v occurs at position i in the peptide, it is assumed to contribute a constant amount/,- 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 αl, J. Mol Biol 267:1258-126, 1997; (see also Sidney et αl, 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, is calculated relative to the remainder of the group, and used as the estimate of ,-. 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 deshed.
Selection of HLA-A2 supertype cross-reactive peptides
Protein sequences from 162P1E6 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 theh 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 162P1E6 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 162P1E6 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-suρertype alleles tested are thereby identified.
Selection of Al and A24 motif-bearing epitopes
To further increase population coverage, HLA-A1 and -A24 epitopes can also be incoφorated into vaccine compositions. An analysis of the 162P1E6 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-bearrng 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 .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 ttansfectants 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 x IO6 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 200-250xl06 PBMC are processed to obtain 24xl06 CD8+ T-cells (enough for a 48-well plate culture). Briefly, the PBMCs are thawed in RPMI with 30μg/ml DNAse, washed once with PBS containing 1% human AB serum and resuspended in PBS/1% AB serum at a concentration of 20xl06cells/ml. The magnetic beads are washed 3 times with PBS/AB serum, added to the cells (140μl beads/20xl06 cells) and incubated for 1 hour at 4°C with continuous mixing. The beads and cells are washed 4x with PBS/AB serum to remove the nonadherent cells and resuspended at lOOxlO6 cells/ml (based on the original cell number) in PBS/AB serum containing lOOμl/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 φm for 5-7 minutes, washed once with PBS with 1% BSA, counted and pulsed with 40μg/ml of peptide at a cell concenttation of l-2xl06/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 1x10s cells/ml) are co-cultured with 0.25ml of CD8+ T-cells (at 2x106 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 5xl06 cells/ml and irradiated at ~4200 rads. The PBMCs are plated at 2xl06 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 lOμg/ml of peptide in the presence of 3 μg/ml J32 microglobulin in 0.25ml RPMI/5%AB per well for 2 hours at 37°C. Peptide solution from each well is asphated and the wells are washed once with RPMI. Most of the media is asphated 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 50IU/ml (Tsai et al, Critical Reviews in Immunology 18(l-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 lvtic activity by 51Cr release.
Seven days after the second restimulation, cytotoxicity is deteπnined 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 lOμg/ml peptide overnight at 37°C.
Adherent target cells are removed from culture flasks with trypsin-EDTA. Target cells are labeled with 200μCi of 51Cr sodium chromate (Dupont, Wilmington, DE) for 1 hour at 37°C. Labeled target cells are resuspended at IO6 per ml and diluted 1:10 withK562 cells at a concentration of 3.3xl06/ml (an NK-sensitive erythroblastoma cell line used to reduce non-specific lysis). Target cells (100 μl) and effectors (lOOμl) 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)] x 100. Maximum and spontaneous τelease 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 NaHC03, 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 lxl 06 cells/ml. The plates are incubated for 48 hours at 37°C with 5%. C02.
Recombinant human IFN-gamma is added to the standard wells starting at 400 pg or 1200pg/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 6x 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 H3P0 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, 5xl04 CD8+ cells are added to a T25 flask containing the following: lxlO6 irradiated (4,200 rad) PBMC (autologous or allogeneic) per ml, 2xl05 irradiated (8,000 rad) EBV- transformed cells per ml, and OKT3 (anti-CD3) at 30ng per ml 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 200IU/ml and every three days thereafter with fresh media at 501U/ml. The cells are split if the cell concentration exceeds lxl06/ml and the cultures are assayed between days 13 and 15 at E:T ratios of 30, 10, 3 and 1:1 in the 5lCr release assay or at lxl06/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 5xl04 CD8+ cells are added to a T25 flask containing the following: lxlO6 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); 2x105 irradiated (8,000 rad) EBV-ttansformed cells per ml RPMI-1640 containing 10%(v/v) human AB serum, non-essential AA, sodium pyruvate, 25mM 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 162P1E6. 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/A 11 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 demonsttated 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 5000nM or less, to three of more A2 supertype alleles. The rationale for this requhement 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 3/5 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*l 1 (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-suρertype 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 162PlE6-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 Vhal 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 162PlE6-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 162PlE6-derived, HLA class II HTL epitopes, a 162P1E6 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 bhiding 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 162PlE6-derived peptides identified above are tested for theh 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 βl, DR2w2 β2, DR6wl9, 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 DR4wl5, DR5wl 1, 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. 162PlE6-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 theh DR3 binding capacity. However, in view of the binding specificity of Hie DR3 motif, peptides bhiding only to DR3 can also be considered as candidates for inclusion in a vaccine formulation.
To efficiently identify peptides that bind DR3, target 162P1E6 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 lμM 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 162PlE6-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 162PlE6-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=l-(SQRT(l-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=l-(l-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*(l-A)). Confirmed members of the A3-like supertype are A3, Al 1, 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 supeitype-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 Al and A24 motifs. On average, Al 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 Al 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, MJ. 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 5ICr labeled Jurkat-A2.1/Kb target cells in the absence or presence of peptide, and also tested on 5ICr labeled target cells bearing the endogenously synthesized antigen, i.e. cells that are stably transfected with 162P1E6 expression vectors.
The results demonstrate that CTL lines obtained from animals primed with peptide epitope recognize endogenously synthesized 162P1E6 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 Al 1, 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 162P1E6- derived CTL and HTL peptide vaccine compositions. The vaccine composition used herein comprise peptides to be administered to a patient with a 162PlE6-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 deshed.
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 (30x106 cells/flask) are co-cultured at 37°C with syngeneic, irradiated (3000 rads), peptide coated lymphoblasts (lOxlO6 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 1.5xl06) 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 requhed at a concentration of 1 μg/ml. For the assay, IO4 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 x (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 requhed 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., 5xl05 effector cells for 10,000 targets) in the absence of peptide and 5:1 (i.e., 5x104 effector cells for 10,000 targets) in the presence of peptide, the specific lytic units would be: [(1/50,000)-(1/500,000)] x IO6 = 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 the Example entitled "Confirmation of Immunogenicity." 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 administtation of such compositions.
Example 21: Selection of CTL and HTL epitopes for inclusion in a 162PlE6-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 162P1E6 clearance. The number of epitopes used depends on observations of patients who spontaneously clear 162P1E6. For example, if it has been observed that patients who spontaneously clear 162P1E6- expressing cells generate an immune response to at least three (3) epitopes from 162P1E6 antigen, then at least three 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 deshable 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 162P1E6, 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 162P1E6.
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 162P1E6, are selected such that multiple supermotifs/motifs are represented to ensure broad population coverage. Similarly, HLA class II epitopes are selected from 162P1E6 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 incoφorated into a minigene for expression in an expression vector.
Such a construct may additionally include sequences that dhect 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 dhected 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 Perkm/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 fhst PCR reaction, 5 μg of each of two oligonucleotides are annealed and extended: In an example using eight oligonucleotides, i.e., four pahs of primers, oligonucleotides 1+2, 3+4, 5+6, and 7+8 are combined in 100 μl reactions containing Pfu polymerase buffer (lx= 10 mM KCL, 10 mM (NH4)2S04, 20 mM Tris-chloride, pH 8.75, 2 mM MgS04, 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 dhectly 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 dhected against the A2-resrricted 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 dhected 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- restticted 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 incoφoration 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 EVI with 100 μg of a DNA mmigene 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 IO7 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 mmigene, 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 the Example entitled "Induction of CTL Responses Using a Prime Boost Protocol." Example 24: Peptide Compositions for Prophylactic Uses
Vaccine compositions of the present invention can be used to prevent 162P1E6 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 162PlE6-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 requhed. The composition is found to be both safe and efficacious as a prophylaxis against 162PlE6-associated disease.
Alternatively, a composition typically comprising teansfecting 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 162P1E6 Sequences
A native 162P1E6 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 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 concenttation 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 162P1E6 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(s) that is presently unknown. Furthermore, this embodiment (excluding an analoged embodiment) dhects the immune response to multiple peptide sequences that are actually present in native 162P1E6, 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 162P1E6 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 tteatment of cancer that expresses 162P1E6 and such other antigens. For example, a vaccine composition can be provided as a single polypeptide that incoφorates multiple epitopes from 162P1E6 as well as tumor- associated antigens that are often expressed with a target cancer associated with 162P1E6 expression, or can be administered as a composition comprising a cocktail of one or more discrete epitopes. Alternatively, the vaccine can be adrninistered 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 dhected to 162P1E6. 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 proposes, 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, 162P1E6 HLA-A*0201-specific CTL frequencies from HLA A*0201 -positive individuals at different stages of disease or following immunization comprising a 162P1E6 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 ttansmembrane-cytosolic tail and COOH-terminal addition of a sequence containing a BhA 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 BhA in the presence of biotin (Sigma, St. Louis, Missouri), adenosine 5' triphosphate and magnesium. Steeptavidin- 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 tettamer-phycoerythrin.
For the analysis of patient blood samples, approximately one million PBMCs are centrifuged at 300g for 5 minutes and resuspended in 50 μl of cold phosphate-buffered saline. Tri-color analysis is performed with the teteamer-phycoeiythrin, along with anti-CD8-Tricolor, and anti-CD38. The PBMCs are incubated with tettamer 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 tettamer 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 162P1E6 epitope, and thus the status of exposure to 162P1E6, 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 162PlE6-associated disease or who have been vaccinated with a 162P1E6 vaccine.
For example, the class I restricted CTL response of persons who have been vaccinated may be analyzed. The vaccine may be any 162P1E6 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 (2mM), penicillin (50U/ml), streptomycin (50 μg/ml), and Hepes (lOmM) containing 10% heat-inactivated human AB serum (complete RPMI) and plated using microculrure 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 x IO5 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 10s irradiated (3,000 rad) autologous feeder cells. The cultures are tested for cytotoxic activity on day 14. A positive CTL response requhes 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 at. J. Clin. Invest. 98:1432-1440, 1996).
Target cell lines are autologous and allogeneic EBV-ttansformed B-LCL that are either purchased from the American Society for Histocompatibility and Immunogenetics (ASHI, Boston, MA) 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-niatched or autologous EBV-ttansformed 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 Coφ., Arlington Heights, IL) 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 x [(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 162P1E6 or a 162P1E6 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 1.5x105 cells/well and are stimulated with 10 μg/ml synthetic peptide of the invention, whole 162P1E6 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 lOU/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 incoφoration. Antigen-specific T cell proliferation is calculated as the ratio of 3H-thymidine incoφoration in the presence of antigen divided by the 3H-thymidine incoφoration 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 162P1E6
Phase II trials are performed to study the effect of administering the CTL-HTL peptide compositions to patients having cancer that expresses 162P1E6. The main objectives of the trial are to determine an effective dose and regimen for inducing CTLs in cancer patients that express 162P1E6, 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 fhst 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 162P1E6.
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 162PlE6-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 the Example entitled "The Plasmid Construct and the Degree to Which It Induces Immunogenicity," can also be used for the administration of the vaccine to humans. Such a vaccine regimen can include an initial administtation 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 the Example entitled "Construction of "Minigene" Multi-Epitope DNA Plasmids" in the form of naked nucleic acid administered EVI (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 5xl09 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 achninistered. For evaluation of vaccine efficacy, patient blood samples are obtained before hnmvmization 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 162P1E6 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 162P1E6 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 x IO6 DC per patient are typically administered, larger number of DC, such as IO7 or 10s 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 10s 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 x IO6 DC, then the patient will be injected with a total of 2.5 x 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 162P1E6 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 theh 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. 162P1E6. 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 theh 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 ttansfected with an expression construct encoding a single HLA allele. These cells can then be used as described, i.e., they can then be ttansfected with nucleic acids that encode 162P1E6 to isolate peptides corresponding to 162P1E6 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 162PlE6-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring 162P1E6. Although use of oligonucleotides comprising from about 15 to 30 base pahs 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 162P1E6. 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 162PlE6-encoding transcript.
Example 35: Purification of Naturally-occurring or Recombinant 162P1E6 Using 162P1E6- Specific Antibodies
Naturally occurring or recombinant 162P1E6 is substantially purified by immunoaffinity chromatography using antibodies specific for 162P1E6. An immunoaffinity column is constructed by covalently coupling anti-162PlE6 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 162P1E6 are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of 162P1E6 (e.g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/162P1E6 binding (e.g., a buffer of pH 2 to pH 3, or a high concenttation of a chaofrope, such as urea or thiocyanate ion), and GCR.P is collected.
Example 36: Identification of Molecules Which Interact with 162P1E6
162P1E6, 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 162P1E6, washed, and any wells with labeled 162P1E6 complex are assayed. Data obtained using different concentrations of 162P1E6 are used to calculate values for the number, affinity, and association of 162P1E6 with the candidate molecules.
Example 37: In Vivo Assay for 162P1E6 Tumor Growth Promotion
The effect of the 162P1E6 protein on tumor cell growth is evaluated in vivo by evaluating tumor development and growth of cells expressing or lacking 162P1E6. For example, SCTD mice are injected subcutaneously on each flank with 1 x 106 of either 3T3 , prostate, bladder, kidney, lung or breast cancer cell lines (e.g. UM-UC3, J82, 769-P, CaKil, CaLu, NCI-H82 or MCF7 cells) containing tkNeo empty vector or 162P1E6. At least two strategies may be used: (1) Constitutive 162P1E6 expression under regulation of a promoter such as a constitutive promoter obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 July 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, tettacycline, 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 162PlE6-expressing cells grow at a faster rate and whether tumors produced by 162PlE6-expressing cells demonstrate characteristics of altered aggressiveness (e.g., enhanced metastasis, vascularization, reduced responsiveness to chemotherapeutic drugs).
Additionally, mice can be implanted with 1 x 105 of the same cells orthotopically to determine if 162P1E6 has an effect on local growth in prostate, bladder, kidney, lung or breast, and whether 162P1E6 affects the ability of the cells to metastasize, specifically to lymph nodes, adrenal, liver and bone (Miki T et al, Oncol Res. 2001;12:209; Fu X et al, Int J Cancer. 1991, 49:938; Kiguchi Ket al, Clin Exp Metastasis. 1998, 16:751).
The assay is also useful to determine the 162P1E6 inhibitory effect of candidate therapeutic compositions, such as for example, 162P1E6 antibodies, 162P1E6 intrabodies, 162P1E6 antisense molecules and ribozymes.
Example 38: 162P1E6 Monoclonal Antibody-mediated Inhibition of Tumor Growth and Metastasis In Vivo
The significant expression of 162P1E6 in cancer tissues, together with its restrictive expression in normal tissues makes 162P1E6 a good target for antibody therapy. Similarly, 162P1E6 is a target for T cell- based immunotherapy. Thus, the therapeutic efficacy of anti-162PlE6 mAbs in human bladder cancer xenograft mouse models is evaluated by using recombinant cell lines UM-UC3-162P1E6, J82-162P1E6, 769- P-162P1E6, CaKil-162PlE6, CaLu-162PlE6, NCI-H82-162P1E6 or MCF7-162P1E6 cells, and 3T3- 162P1E6 (see, e.g., Kaighn, M.E., et al, Invest Urol, 1979. 17(1): p. 16-23). Similarly, anti-162PlE6 mAbs are evaluated in human kidney, bladder, lung, breast and prostate cancer xenograft models using recombinant cell lines such as UM-UC3-162P1E6, J82-162P1E6, 769-P-162P1E6, CaKil-162PlE6, CaLu-162PlE6, NCI- H82-162P1E6 and MCF7-162P1E6 cells.
Antibody efficacy on tumor growth and metastasis formation is studied, e.g., in a mouse orthotopic bladder cancer xenograft model, a orthotopic kidney cancer, orthotopic mammary cancer model and orthotopic lung cancer xenograft model in addition to the prostate cancer xenograft model. The antibodies can be unconjugated, as discussed in this Example, or can be conjugated to a therapeutic modality, as appreciated in the art. Anti-162P1E6 mAbs inhibit formation of kidney, bladder, lung and breast xenografts. Anti-162P1E6 mAbs also retard the growth of established orthotopic tumors and prolonged survival of tumor- bearing mice. Anti-162P1E6 mAbs can also regulate the growth and metastasis of prostate cancer xenograft tumors. These results indicate the utility of anti-162PlE6 mAbs in the treatment of local and advanced stages of kidney, bladder, lung and breast cancer. (See, e.g., Saffran, D., et al., PNAS 10:1073-1078 or www.pnas.org/cgi/doi/10.1073/pnas.051624698).
Administration of the anti-162PlE6 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, specially in mice bearing kidney, bladder, lung and breast tumors. These studies indicate that 162P1E6 as an attractive target for immunotherapy and demonstrate the therapeutic potential of anti-162PlE6 mAbs for the treatment of local and metastatic cancer. This example demonstrates that unconjugated 162P1E6 monoclonal antibodies are effective to inhibit the growth of human bladder, kidney, lung and breast tumor xenografts grown in SCID mice; accordingly a combination of such efficacious monoclonal antibodies is also effective.
Tumor inhibition using multiple unconjugated 162P1E6 mAbs Materials and Methods
1 2P1E6 Monoclonal Antibodies:
Monoclonal antibodies are raised against 162P1E6 as described in the Example entitled "Generation of 162P1E6 Monoclonal Antibodies (mAbs)." The antibodies are characterized by ELISA, Western blot, FACS, and immunoprecipitation for theh capacity to bind 162P1E6. Epitope mapping data for the anti- 162P1E6 mAbs, as determined by ELISA and Western analysis, recognize epitopes on the 162P1E6 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, CA). 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 UM-UC3, J82, 769-P, CaKil, CaLu, NCI-H82 or MCF7 cells tumor xenografts. Cancer xenograft and Cell Lines
The LAPC-4AD 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 bladder, kidney, lung and breast carcinoma cell lines, as well as the fibroblast line NIH 3T3 (American Type Culture Collection) are maintained in DMEM supplemented with L-glutamine and 10% FBS. Prostate cancer cell lines (American Type Culture Collection) are maintained in RPMI supplemented with L-glutamine and 10% FBS.
UM-UC3-162P1E6, J82-162P1E6, 769-P-162P1E6, CaKil-162PlE6, CaLu-162PlE6, NCI-H82- 162P1E6 or MCF7-162P1E6 cells 3T3-162P1E6 cell populations are generated by retroviral gene transfer as described in Hubert, R.S., et al., Proc Natl Acad Sci U S A, 1999. 96(25): 14523.
Xenograft Mouse Models.
Subcutaneous (s.c.) tumors are generated by injection of 1 x 10 6 cancer 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. Tumor sizes are determined by caliper measurements, and the tumor volume is calculated as length x width x height. Mice with s.c. tumors greater than 1.5 cm in diameter are sacrificed.
Orthotopic injections are performed under anesthesia by using ketamine/xylazine. For bladder and breast orthotopic studies, an incision is made through the abdomen to expose the bladder or the breast, and tumor cells (5 x 10s) mixed with Matrigel are injected into the bladder/breast wall in a 10-μl volume. For kidney orthopotic models, an incision is made through the abdominal muscles to expose the kidney. Tumor cells mixed with Matrigel are injected under the kidney capsule in a 10-μl volume (Yoshida Y et al, Anticancer Res. 1998, 18:327; Ahn et al, Tumour Biol. 2001, 22:146). For prostate orthotopic studies, an incision is made through the abdominal muscles to expose the dorsal prostate. Tumor cells (5 x 105 ) mixed with Matrigel are injected into each dorsal lobe in a 10-μl volume. To monitor tumor growth, mice are palpated and blood is collected on a weekly basis measuring G250, BTA, PSA and TPA (Tissue Polypeptide Antigen) levels (Stephan C et al, Urology. 2002, 59:2; Buccheri G, Ferrigno D. Lung Cancer. 2001;34 Suppl 2:S65; Ross JS, Cohen MB. Adv Anat Pathol. 2001, 8:37). The mice are segregated into groups for the appropriate treatments, with anti-162PlE6 or control mAbs being injected i.p.
Anti-162P1E6 mAbs Inhibit Growth of 162P1E6-Exρressing Xeno graft-Cancer Tumors
The effect of anti- 162P 1 E6 mAbs on tumor formation is tested on the growth and progression of bladder, kidney, lung, prostate and breast cancer xenografts using cell line orthotopic models, as stated above. As compared with the s.c. tumor model, the orthotopic model, which requhes injection of tumor cells dhectly in the mouse bladder, kidney and ovary, 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.
Ill Accordingly, tumor cells are injected into the mouse bladder, kidney, lung, prostate or breast, and 2 days later, the mice are segregated into two groups and treated with either: a) 200-500μg, of anti-162PlE6 Ab, or b) PBS three times per week for two to five weeks.
A major advantage of the orthotopic cancer models is the ability to study the development of metastases. Formation of metastasis in mice bearing established orthotopic tumors is studies by EEC analysis on liver, lung and bone sections using an antibody against a tumor-specific cell-surface protein such as anti- CK20 for bladder cancer, anti-G250 for kidney cancer, anti-STEAP-1 for prostate cancer and anti-TPA antibody for lung cancer models (Lin S et al, Cancer Detect Prev. 2001;25:202; McCluggage W et al, Histopathol 2001, 38:542).
Mice bearing established orthotopic tumors are administered lOOOμg injections of either anti- 162P1E6 mAb or PBS over a 4-week period. Mice in both groups are allowed to establish a high tumor burden, to ensure a high frequency of metastasis formation in mouse lungs, livers and bones. Mice then are killed and theh bladders, livers, bone and lungs are analyzed for the presence of tumor cells by IHC analysis.
These studies demonstrate a broad anti-tumor efficacy of anti-162PlE6 antibodies on initiation and progression of prostate and kidney cancer in xenograft mouse models. Anti-162P1E6 antibodies inhibit tumor formation of tumors as well as retarding the growth of aheady established tumors and prolong the survival of treated mice. Moreover, anti-162PlE6 mAbs demonstrate a dramatic inhibitory effect on the spread of local bladder, kidney, lung and breast tumor to distal sites, even in the presence of a large tumor burden. Thus, anti-162PlE6 mAbs are efficacious on major clinically relevant end points (tumor growth), prolongation of survival, and health.
Example 39: Therapeutic and Diagnostic use of Anti-162P1E6 Antibodies in Humans.
Anti-162P1E6 monoclonal antibodies are safely and effectively used for diagnostic, prophylactic, prognostic and/or therapeutic puφoses in humans. Western blot and immunohistochemical analysis of cancer tissues and cancer xenografts with anti-162PlE6 mAb show strong extensive staining in carcinoma but significantly lower or undetectable levels in normal tissues. Detection of 162P1E6 in carcinoma and in metastatic disease demonstrates the usefulness of the mAb as a diagnostic and/or prognostic indicator. Anti- 162P1E6 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-162PlE6 mAb specifically binds to carcinoma cells. Thus, anti-162PlE6 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 162P1E6. Shedding or release of an extracellular domain of 162P1E6 into the extracellular milieu, such as that seen for alkaline phosphodiesterase B10 (Meerson, N. R., Hepatology 27:563-568 (1998)), allows diagnostic detection of 162P1E6 by anti-162PlE6 antibodies in serum and/or urine samples from suspect patients.
Anti-162P1E6 antibodies that specifically bind 162P1E6 are used in therapeutic applications for the treatment of cancers that express 162P1E6. Anti-162P1E6 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-162PlE6 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., the Example entitled "162P1E6 Monoclonal Antibody-mediated Inhibition of Bladder and Lung Tumors In Vivo "). Conjugated and unconjugated anti-162PlE6 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-162P1E6 Antibodies In vivo
Antibodies are used in accordance with the present invention which recognize an epitope on 162P1E6, and are used in the treatment of certain tumors such as those listed in Table I. Based upon a number of factors, including 162P1E6 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-162PlE6 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-162PlE6 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-162P1E6 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-162PlE6 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- 162P1E6 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 162P1E6. In connection with the use of the anti-162PlE6 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 postoperative follow-up to determine what tumor remains and or returns. In one embodiment, a (U1 In)-162P1E6 antibody is used as an imaging agent in a Phase I human clinical trial in patients having a carcinoma that expresses 162P1E6 (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 Administtation
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-162PlE6 antibodies can be administered with doses in the range of 5 to 400 mg/m 2 , with the lower doses used, e.g., in connection with safety studies. The affinity of anti-162PlE6 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-162PlE6 antibodies that are fully human antibodies, as compared to the chimeric antibody, have slower clearance; accordingly, dosing in patients with such fully human anti-162PlE6 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-162PlE6 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 aάininistration 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 vascularure 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-162PlE6 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-162PlE6 antibodies. As will be appreciated, one criteria that can be utilized in connection with enrollment of patients is 162P1E6 expression levels in theh 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 162P1E6. Standard tests and follow-up are utilized to monitor each of these safety concerns. Anti-162P1E6 antibodies are found to be safe upon human administration.
Example 41:Human Clinical Trial Adjunctive Therapy with Human Anti-162P1E6 Antibody and Chemotherapeutic Agent
A phase I human clinical trial is initiated to assess the safety of six intravenous doses of a human anti-162PlE6 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-162P 1E6 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-162PlE6 antibody with dosage of antibody escalating from approximately about 25 mg/m 2 to about 275 mg/m 2 over the course of the tteatment in accordance with the following schedule: Day O Day 7 Day 14 Day 21 Day 28 Day 35
mAb Dose 25 75 125 175 225 275 mg/m 2 mg/m 2 mg/m 2 mg/m 2 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 162P1E6. 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 MRl or other imaging.
The anti-162PlE6 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-162P1E6 Antibody
Anti-162P1E6 antibodies are safe in connection with the above-discussed adjunctive trial, a Phase II human clinical trial coih ms 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-162PlE6 antibodies.
Example 43: Human Clinical Trial: Diagnostic Imaging with Anti-162P1E6 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-162PlE6 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 162P1E6 to Known Sequences
Five variants of 162P1E6 have been identified. The 162P1E6 v.l gene exhibits homology to a previously cloned human gene of no known function named hypothetical protein XP-036612 (gi 14720533), showing 100% identity over the enthe length of the protein (Figure B). 162P1E6 v.l shows some homology to human Man7GlcNAc2-PP-dolichyl mannosylttansferase (gi 15864569), displaying 35% identity and 49% homology to the last segment of that protein (Figure 4C). 162P1E6 v.l is a 146 aa soluble protein, primarily localized to the cytoplasm, with potential localization to the nucleus and microbodies (Table XXI). While PFam and PRINTS analysis fail to identify known protein motifs within 162P1E6 v.l, BLOCKs analysis demonstrates that 162P1E6 v.l and v.4 carry a Synapsin 9 motif at amino acid 38-55 (Table XXI). Synapsins are phosphoproteins that associate with cytoskeletal proteins and function in the regulation of neurotransmitter release (Rosahl TW et al, Nature. 1995, 375:488).
The 162P1E6 v.3 protein exhibits 41% identity and 43% homology to the human Alu subfamily SQ (gi 728837), a protein of no known function (Figure 4E). The 162P1E6 v.3 protehi shows 43% identity and 54% homology the human zinc finger protein 195 (gi6005974) (Figure 4F). 162P1E6 v.3 is a transmembrane protein with a helix located at amino acid 40-70 (Table XXI). The 162P1E6 v.4 protein exhibits 36% identity and 54% homology to the Caφ interleukin lβ protein (gi2821975) (Figure 4G). IL-1 is an inflammatory cytokine, that plays a role in the progression, drag resistance and survival of cancer cells (Arlt A, et al, Cancer Res. 2002, 62:910; Suganuma M, et al, Int J Oncol. 2002, 20:131). In addition, IL-lβ induces the activation of several MAPK cascades in gastric tumors, resulting in the regulation of gene expression (Fan X et al, J Gasfroenterol Hepatol. 2001, 16:1098). While 162P1E6 v.5 shows some homology to an unknown protein (gi 16331181), it also shares a common sequence with 162P1E6 v.4 (See Figure 11), and may function in a similar manner.
The presence of a synapsin motif and its homology interleukin-1 beta indicate that 162P1E6 participates in the process of tumor formation and progression. By way of its synapsin domain, 162P1E6 functions in regulating protein interactions and cell adhesion. Based on its homology to IL-1 β, 162P1E6 regulates signal transduction in mammalian cells, thereby regulating gene expression and cellular outcomes, including cell proliferation, survival, drug resistance, etc, all of which have a dhect effect on tumor growth and progression.
Accordingly, when 162P1E6 functions as a regulator of protein interactions, cell growth, tumor formation, or cell signaling, 162P1E6 is used for therapeutic, diagnostic, prognostic and/or preventative puφoses.
Example 45: Regulation of Transcription
The localization of 162P1E6 coupled to the presence of protein interaction domains within its sequence and homology to IL-1 indicate that 162P1E6 modulates the transcriptional regulation of eukaryotic genes. Regulation of gene expression is confirmed, e.g., by studying gene expression in cells expressing or lacking 162P1E6. For this piupose, two types of experiments are performed.
In the first set of experiments, RNA from parental and 162PlE6-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, ELKl-luc, ARE-luc, ρ53-luc, and CRE-luc 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, 162P1E6 plays a role in gene regulation, and it is used as a target for diagnostic, prognostic, preventative and/or therapeutic puφoses.
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, protein interaction motifs have been instrumental in inducing kinase activation, recruitment of proteins and complex formation (Samelson L. Annu Rev Immunol. 2002;20:371). In addition, IL-1 has been shown to regulate multiple signaling cascades that control gene expression and cell survival (Oncogene. 1999, 18:6087). In addition, the 162P1E6 protein contains several phosphorylation sites (see Table XX) indicating an association with specific signaling cascades. Based on the presence of a protein interaction motif and similarity to IL-1, 162P1E6 regulates signaling pathways important for cell growth and survival. Using immunoprecipitation and Western blotting techniques, proteins are identified that associate with 162P1E6 and mediate signaling events. Several pathways known to play a role in cancer biology can be regulated by 162P1E6, including phospholipid pathways such as PI3K, AKT, etc, adhesion and migration pathways, including FAK, Rho, Rac-1, β-catenin, 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 162P1E6 dhectly 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.
NFkB-luc, NFkB/Rel; Ik-kinase/SAPK; growth/apoptosis/stress SRE-luc, SRF/TCF/ELKl; MAPK/SAPK; growth/differentiation AP-l-luc, FOS/JUN; MAPK/SAPK/PKC; growth/apoptosis/stress ARE-luc, androgen receptor; steroids/MAPK; growth/differentiation/apoptosis ρ53-luc, ρ53; SAPK; growth/differentiation/apoptosis CRE-luc, CREB/ATF2; PKA/p38; growth/apoptosis/stress TCF-luc, TCF/Lef; β-catenin, Adhesion/invasion
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 162P1E6 are mapped and used for the identification and validation of therapeutic targets. When 162P1E6 is involved in cell signaling, it is used as target for diagnostic, prognostic, preventative and/or therapeutic proposes. Example 47: Involvement in Tumor Progression
Based on the role of protopoφhyrinogen oxidase in tumor formation (Germanaud J, above), the 162P1E6 gene can contribute to tumor initiation and progression. The role of 162P1E6 in tumor growth is confirmed in a variety of primary and ttansfected cell lines including bladder, kidney and ovary cell lines, as well as NIH 3T3 cells engineered to stably express 162P1E6. Parental cells lacking 162P1E6 and cells expressing 162P1E6 are evaluated for cell growth using a well-documented proliferation assay (Fraser SP, Grimes JA, Djamgoz MB. Prostate. 2000;44:61, Johnson DE, Ochieng J, Evans SL. Anticancer Drags. 1996, 7:288).
To confirm the role of 162P1E6 in the transformation process, its effect in colony forming assays is investigated. Parental NTH-3T3 cells lacking 162P1E6 are compared to NIH-3T3 cells expressing 162P1E6, 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 162P1E6 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 bladder, ovary and kidney cell lines lacking 162P1E6 are compared to cells expressing 162P1E6. 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 enthe cell population.
162P1E6 can also play a role in cell cycle and apoptosis. Parental cells and cells expressing 162P1E6 are compared for differences in cell cycle regulation using a well-established BrdU assay (Abdel- Malek ZA. 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 GI, S, and G2M phases of the cell cycle. Alternatively, the effect of stress on apoptosis is evaluated in control parental cells and cells expressing 162P1E6, including normal and tumor bladder, kidney and ovary cells. Engineered and parental cells are treated with various chemotherapeutic agents, such as etoposide, taxol, 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 162P1E6 can play a critical role in regulating tumor progression and tumor load.
When 162P1E6 plays a role in cell growth, transformation, invasion or apoptosis, it is used as a target for diagnostic, prognostic, preventative and/or therapeutic puφoses.
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, 162P1E6 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 162P1E6 in angiogenesis, enhancement or inhibition, is confirmed. For example, endothelial cells engineered to express 162P1E6 are evaluated using tube formation and proliferation assays. The effect of 162P1E6 is also confirmed in animal models in vivo. For example, cells either expressing or lacking 162P1E6 are implanted subcutaneously in immunocompromised mice. Endothelial cell migration and angiogenesis are evaluated 5-15 days later using immunohistochemistry teclmiques. 162P1E6 affects angiogenesis, and it is used as a target for diagnostic, prognostic, preventative and/or therapeutic pinposes
Example 49: Involvement in Protein-Protein Interactions
Synapsin motifs have been shown to mediate interaction with other proteins, specially cytoskeletal protein and SH3 containing proteins (Onofri F et al, J Biol Chem. 2000, 275:29857). Using immunoprecipitation techniques as well as two yeast hybrid systems, proteins are identified that associate with 162P1E6. Immunoprecipitates from cells expressing 162P1E6 and cells lacking 162P1E6 are compared for specific protein-protein associations.
Studies are performed to confirm the extent of association of 162P1E6 with effector molecules, such as nuclear proteins, transcription factors, kinases, phsophates etc. Studies comparing 162P1E6 positive and 162P1E6 negative cells as well as studies comparing unstimulated/resting cells and cells treated with epithelial cell activators, such as cytokines, growth factors and anti-integrin Ab reveal unique interactions.
In addition, protein-protein interactions are confirmed using two yeast hybrid methodology (Curr Opin ChemBiol. 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 162PlE6-DNA-binding domain fusion protein and a reporter construct. Protein-protein interaction is detected by colorhnetric reporter activity. Specific association with effector molecules and transcription factors directs one of skill to the mode of action of 162P1E6, 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 162P1E6.
Thus it is found that 162P1E6 associates with proteins and small molecules. Accordingly, 162PlE6and these proteins and small molecules are used for diagnostic, prognostic, preventative and/or therapeutic proposes.
Throughout this application, various website data content, publications, patent applications and patents are referenced. (Websites are referenced by theh Uniform Resource Locator, or URL, addresses on the World Wide Web.) The disclosures of each of these references are hereby incoφorated by reference herein in theh 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 162P1E6 When Malignant
- Bladder
- Prostate
- Kidney
- Lung
- Breast
TABLE H: Amino Acid Abbreviations
TABLE HI: Amino Acid Substitution Matrix
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. (See URL www.ikp.unibe.ch/manualblosum62.html )
A C D E F G H I K L M N P Q R S T V 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
TABLE IV HLA Class I/II Motifs/Supermotifs
TABLE IV (A): HLA Class I Supermotifs/Motifs
Bolded residues are preferred, italicized residues are less prefenred: 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 H Supermotif
Docket No. 511582007740
TABLE TV (C): HLA Class H Motifs
MOTIFS 1° anchor 1 2 3 4 5 1° anchor 6 7 8 9
DR4 preferred ΕMYLIVW M T I YSTCPALIM 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 TVMSACTPL M IV deleterious C G GRD N G
DR3 MOTIFS 1° anchor 1 1° anchor 4 1° anchor 6 motif a LIVMFY D preferred motif b LIVMFAY DNQEST KRH preferred
DR MFLIVWY VMSTACPLI
Supermotif
Italicized residues indicate less preferred or "tolerated" residues
DocketNo. 511582007740
TABLE TV (D): HLA Class I Supermotifs
POSITION: C-terminus
SUPERMOTIFS
Al 1° Anchor 1° Anchor TILVMS FWY
A2 1° Anchor 1° Anchor UVMATQ LΓVMAT
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 YF WIVLMT FTYWLM
B7 preferred FWY (5/5) 1° Anchor FWY FWY 1° Anchor
LIVM (3/5) P (4/5) (3/5) VT FMWYA deleterious DE (3/5); DE G QN DE
P(5/5); (3/5) (4/5) (4/5) (4/5)
G(4/5);
A(3/5);
QN(3/5)
B27 1° Anchor l°Anchor RHK FY WMIVA
B44 F Anchor 1° Anchor ED FWYLIMVA
B58 1° Anchor 1° Anchor ATS ΕWYLIVMA
B62 1° Anchor 1° Anchor Q IVMP FWYMIVLA
Itahcized residues indicate less preferred or "tolerated" residues
Docket No. 511582007740
TABLE TV (E): HLA Class I Motifs
POSITION: C-terminus or C-terminus
Al preferred GFY 1° Anchor DEA YFW P DEQN YFW 1 "Anchor
9-mer W STM Y deleterious DE RHKLIVMP A G A
Al preferred GRHK ASTCLIVM l°Anchor GSTC ASTC LIVM DE l°Anchor
9-mer DE4S Y deleterious A RHKDEPY DE PQN RHK PG GP
FW
Al preferred YFW l°Anchor DEAQN A YFWQN PASTC GDE P 1 "Anchor
10-mer STM Y deleterious GP RHKGLIVM DE RHK QNA RHKYFW RHK A
Al preferred YFW STCLΓVM 1 "Anchor A YFW PG G YFW 1° Anchor
10-mer ΌΈAS Y deleterious RHK RHKDEPY P G PRHK QN
FW
A2.1 preferred YFW 1° Anchor YFW STC YFW A P l°Anchor
9-mer IMIVQAT VLIMAT deleterious DEP DERKH RKH DERKH
Itahcized residues indicate less preferred or "tolerated" residues
Docket No: 511582007740
TABLE TV (E): HLA Class I Motifs, continued
POSITION: 1 2 3 4 5 6 7 8 9 C-Teπninus
A2.1 preferred AYFW rAnchor LVΓM G G FYWL 1 "Anchor 10-mer IMIVQA VIM VLIMAT T deleterious DEP DE RKHA P RKH DERKH RKH
A3 preferred RHK 1 "Anchor YFW PRHKYFW A YFW P 1 "Anchor
LMVTSA KYRHFA
TFCGD deleterious DEP DE
All preferred A 1 "Anchor YFW YFW YFW YFW 1 "Anchor VTLMIS KRYH AGNCDF deleterious DEP G
A24 preferred YFWRHK 1 "Anchor STC YFW YFW 1 "Anchor
9-mer YFWM FLIW deleterious DEG DE G QNP DERH G AQN K
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
MVALF7. RK
ST deleterious GP DE
Itahcized residues indicate less preferred or "tolerated" residues
DocketNo. 511582007740
TABLE TV (E): HLA Class I Motifs, continued
POSITION C-Terminus
A6801 preferred YFWSTC 1 "Anchor YFWLIV YFW 1 "Anchor
AVTMSLI M RK deleterious GP DEG RHK
B0702 preferred RHKFW 1 "Anchor RHK RHK RHK RHK PA 1 "Anchor
Y P IMFWYAIV deleterious DEQNP DEP DE DE GDE QN DE
B3501 preferred FWYLIV l°Anchor FWY FWY 1 "Anchor
M P IMFWYIV
A deleterious AGP G G
B51 preferred LIVMFW 1 "Anchor FWY STC FWY G FWY 1 "Anchor
Y P UVFWYAM deleterious AGPDER DE G DEQN GDE HKSTC
B5301 preferred LIVMFW 1 "Anchor FWY STC FWY LIVMFWY FWY 1 "Anchor
Y P IMFWY Z, V deleterious AGPQN RHKQN DE
B5401 preferred FWY 1 "Anchor FWYL LIVM ALIVM FWYAP l"Anchor P ΓVM ATΓVLM
WY 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 XX: Motifs and Post-translational Modifications of 162P1E6 v.l
Protein kinase C phosphorylation site 2-4 TnK 123 - 125 SsR
124 - 126 SrK
Casein kinase II phosphorylation site
2-5 TnkE 25-28 SflD
52-55 SsqE
124 - 127 SrkD
137 - 140 TqwD Amidation site
71-74 iGKR
TABLE XXI: Protein Properties of 162P1E6
162P1E6 variant 1 Bioinforma URL Outcome tic Program
ORF ORF finder bp2028-2468 (includes stop)
Protein length 146 aa
Transmembrane region TM Pred http://www.ch.embnet.org/ no TM
HMMTop http://www.enzim.hu/hmmtop/ no TM
Sosui http://www.genome.ad.jp/SOSui/ soluble protein
TMHMM http://www.cbs.dtu.dk/services/TMHMM no TM, extracellular
Signal Peptide Signal P http://www.cbs.dtu.dk/services/SignalP/ none pi pI/MW tool http://www.expasy.ch/tools/ 10.2 pi
Molecular weight pI/MW tool http://www.expasy.ch/tools/ 16.6 kDa
Localization PSORT http://psort.nibb.ac.jp/ 64% microbody, 45% cytoplasmic
PSORT II http://psort.nibb.ac.jp/ 65% cytoplasmic, 21% nuclear,
Motifs Pfam http://www.sanger.ac.uk/Pfam/ no significant motif
Prints http://www.biochem.ucl.ac.uk/ no significant motif
Blocks http.7/www.blocks.fhcrc.org no significant motif
162P1E6 variant 3 Bioinforma URL Outcome tic Program
ORF ORF finder bp3-404 (includes stop)
Protein length 133 aa Transmembrane region TM Pred http://www.ch.embnet.org/ 1 TM, TM helix at 40-70aa, N terminus extracellular HMMTop http://www.enzim.hu/hmmtop/ 1 TM, TM helix at 41-64aa, N terminus extracellular
Sosui http://www.genome.ad.jp/SOSui/ soluble protein
TMHMM http://www.cbs.dtu.dk/services/TMHMM no TM, extracellular
Signal Peptide Signal P http://www.cbs.dtu.dk/services/SignalP/ none pi pI/MW tool http://www. expasy. ch/tools/ 8.8pl
Molecular weight pI/MW tool http://www.expasy.ch/tools/ 14.5 kDa
Localization PSORT http://psort.nibb.ac.jp/ 64% peroxisome, 45% cytoplasmic
PSORT II http://psort.nibb.ac.jp/ 43.5% nuclear, 30% cytoplasmic
Motifs Pfam http://www.sanger.ac.uk/Pfam/ no significant motif
Prints http://www.biochem.ucl.ac.uk/ no significant motif
Blocks http://www.blocks.fhcrc.org/ no significant motif
TABLE XXI, continued: Protein Properties of 162P1E6
162P1E6 variant 4 Bioinforma URL Outcome tic Program
ORF ORF finder bp388-696 (includes stop)
Protein length 102 aa
Transmembrane region TM Pred http://www.ch.embnet.org/ 1 TM,aa 79-97, N-terminus inside
HMMTop 1 TM,aa 71-95, N-terminus inside
Sosui http://www.genome.ad.jp/SOSui/ membrane protein
TMHMM http://www.cbs.dtu.dk/services/TMHMM no TM, extracellular
Signal Peptide Signal P http://www.cbs.dtu.dk/services/SignalP/ none pi pI/MW tool http ://www. expasy. ch/tools/ 10.8pl
Molecular weight pI/MW tool http://www.expasy.ch/tools/ 10.9 kDa
Localization PSORT http://psort.nibb.ac.jp/ 81% lysosome, 60% peroxisome
PSORT II http://psort.nibb.ac.jp/ 56% cytoplasmic, 21% mitochondrial
Motifs Pfam http ://www. sanger.ac.uk/Pfam/ no significant motif
Prints http://www.biochem.ucl.ac.uk/ no significant motif
Blocks http://www.blocks.fhcrc.org/ Synapsin 9 galactose-phosphate uridyl transferase family 1
Table LH: Search Peptides
162P1E6 v.l: For all 162P1E6 v.l nonamers, decamers and 15-mers (aa 1-146)
MTNKEIVESF SRHILGRMWG HWRLSFLDKS LGVRTRSLTL LCPPTPMNGP GSSQELWFFL SSSPISSGFH IGKRGCKVLF VLFGQCLVER NAHAPAFQGL GKQAQSSWIF LKQLQNTCFF FVSSRKDQPH RAQLWHTQWD LDKGRG
162P1E6 v.3: For all 162P1E6 v.3 nonamers, decamers, and 15-mers (aa 1-133)
LKWAESLLLT LDLEKPVSLL LSVTNLYSKN SAQFSTILQT LSFPATFTPS PSIPLSSAYF FFFSDRVSLC RPGRSAVAQS WAHCSLNLPE AGFHHVAQTG LELLSLSNPP ASASQSVGIT GVSHRIRPHV LFH 162P1E6 v.4: For all 162P 1 E6 v.4 nonamers, decamers and 15-mers (aa 1 -102)
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRTLS SLKYPSWRVR TPHEDFSGVK FRRHGADNHE ASAATATTAA ATTVAAAAAA AAAAAAARVT LT 162P1E6 v.5:
162P1E6 v.5 Nonamers (aa 30-76)
A ELGALYRKGP TTPSSVMAHT VGPRQRERVTDIPTRFQWSE VQEAWS 162P1E6 v.5 Decamers (aa 29-76)
PA ELGALYRKGP TTPSSVMAHT VGPRQRERVTDIPTRFQWSE VQEAWS
162P1E6 v.5 15-mers (aa 24-76)
SVPHRPA ELGALYRKGP TTPSSVMAHT VGPRQRERVTDIPTRFQWSE VQEAWS
162P1E6 v.6:
162P1E6 v.6 Nonamers (aa 47-70)
WRVR TPHEERTNHTELSYGTHSGT
162P1E6 v.6 Decamers (aa 46-70)
SWRVR TPHEERTNHTELSYGTHSGT
162P1E6 v.6 15-mers (aa 41-70) SLKYPSWRVR TPHEERTNHTELSYGTHSGT
Table LIV(A). Nucleotide sequence of transcript variant 162P1E6 v.2 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccGgg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgact aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg gtaagtgttt 600 ctcatttttt gttccctgtc ctcaagcctt aggggcaaaa gaaacatcca agatttgaaa 660 tttcttttct tcttctcatc tgcatggctg tagccatctc tctgttctgc attatcttat 720 gacaaaaaaa aaaaattctt attttgaagc aaactcaaag ctaggtcctg atgtctcaag 780 gcacaggtac tcgtacttaa aggtgagtct gaaatctgtg gatttgggga actttggaaa 840 aacaaagatg agtggctaga tcagggggct cattgggcag gaagaggaga ctggaaaatg 900 ccatattcac tgcaagtcaa ttatcaactt cctccaaggc taaaatagct gaacctgctg 960 cattttaaac caatcctcag ccactttggt gttttctcaa ggatttccag ggatcccagg 1020 cagtaaattc tgctgataat aggaattggt gtgataaggt gggtgctgag cagtttaagc 1080 accaagattg tagctctgtc tggttttgtg gagatttact caactagaag aacagagatt 1140 tggctggttt ttcagtcctg gggtgcaggg tgcacctgta ctggaaaatt taggacCtgg 1200 tttcattctt tgagtctcat gttcaagttg gttttaatgt tatgaagaca cttgggacgt 1260 aatcctgagg gcagctgggg ggaagaaagt ggtcactgga tggacttacc ctgtagcgag 1320 cccatgcatg gtttgttctc tgatcgtgca tgtgcttggc tctagaccca tgtaaccatg 1380 gtgaaggcca ctgggggatt cagttggcaa aggcatagtg ggcagaagaa tcttgaacaa 1440 ggagtccaga gcaggtcaag tctcctgata caggttgtga ctcatggttt ttgtctctgc 1500 ctgtagcagc tacaggtctg taaagcaagg ggagagtgat aaggaaagaa ctcacctttc 1560 tggggctctc tgacattaat gccacctccc atttgctttt tgcagacact gtcatctctc 1620 aagtacccat cttggagggt acggacccca catgagggtg aggctctctg cacactccag 1680 agtgaggact ttaataatct agtggactgt acatgttggg aggggaagag cggggtgccg 1740 agggtctgga gggagaagaa ttgactgccc cttttgctct tggagttaag cagaaatcta 1800 aagagaaggc aaagaatctt gccttcctgg cGtcatttcc tcctaccatc ccaggccatc 1860 atttatttat tacagccaac agactggcct ctttcttccc tttgactggg aatgggtcaa 1920 aggcggtgca ggaggaggat ctggtccaga taattcacaa gcagggtgca ttttcctctc 1980 attattgaga actgtgagtg tttatcaaga aggcagagca ggagaagatg aaccagtctt 2040 cttcccctca ctacccagat ctctgcctgc caacaagccc cgtgttcacc ctggcaaaga 2100 gtctttacat tcagaccaag gagagtgtga ctccttctca gcactagcta gaaacctcaa 2160 gcccttgctt aagggccttt ttcagagaga cccaatgccc agaaggctag atgcgtgggg 2220 aggagccaca tacgagaaac tgcctccctg cttcgggtca gaacaagccc caggaagaaa 2280 gtatttcaaa caacaaggtg catctgcccc aacCcatcca gcctgcatgt tggtgctgag 2340 aacagccttt tatggggctt gcactgagcc atgggcatgt ctgaacacaa caaggaagag 2400 gccagagcag caacagcacg caaagggttg atgggcattt cttttaagac agagcagaaa 2460 actcttagat actttgcgtc cttcctattt gactcagtct atgaaagcca ggttagcttg 2520 ctttcttcct ccctaaatcc tccatcctca tgaccaacaa agaaatagtT gaatcatttt 2580 ccaggcacat cttggggagg atgtggggcc attggaggct gtccttcctG gataagtctt 2640 taggagtgag aacaaggagt cttaccctcc tctgtccacc cacccccatg aatgggcctg 2700 gctccagcca ggagttgtgg tttttcctga gctcctcacc tatctcttct ggatttcaca 2760 ttggcaaacg gggttgcaaa gtgctcttcg tgctctttgg acagtgcctt gtggagagga 2820 atgcccatgc ccctgcattc caaggccttg gtaagcaagc tcagagtagc tggatttttc 2880 taaagcaatt gcagaacacc tgctttttct ttgtttcctc tagaaaggac caaccacAcc 2940 gagctcagtt atggcacaca cagtgggacc tagaσaaagg gagagggtga ccgacatccc 3000 aactaggtaa acacagagga ggttccacat ggacttatct gggtggctgt tttgaaaacg 3060 agaaacagtc aagagtccct ggccccacag acccacctcc ccaactcagc actgtctgtc 3120 tgtgcagcag gtgcaaggac gtgttgaact agctctctgc agcctccttg gaggatgtga 3180 tcctatggga ggggtaggag tattcaggtc cttgacatct cccaaatgtg tgattccggg 3240 atgccaaagg cctttggcca ggtaatgcag tgtctacagg ctgaggttga catgcatccc 3300 caccctctga gaaaaagatc ctcagacaat ccatgtgctt ctcttgtcct tcattccacc 3360 ggagtctgtc tcatacccaa ccagatttca gtggagtgaa gttcaggagg catggagctg 3420 acaaccatga ggcctcggca gccaccgcca ccaccgccgc cgccaccacc gtagcagcag 3480 cagcagcagc agcagcagca gcagcagcag caagagtaac tctgacttag gaatagagac 3540 agccagagag aaatgtgatc aatgaaggag acatctggag tgtgcgtgct tcttcaGagg 3600 gacgggtgat gggcagattg gaaaaagcAc cgcagatggg aaccttaatc tttcttttct 3660 aaaattgatg ctatgaaaat ttgcgttttc tGtaacttgt aaaaactaaa agttgcttgt 3720 ctactgaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aa 3762 Table LIV(B) . Nucleotide sequence of transcript variant 162P1E6 v.3 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag aggcggggtt tcaccatgtt gcccagactg 300 gtcttgaact cctgagctta agcaatccac ctgcctcggc ctcccaaagt gttgggatca 360 caggcgtgag ccaccgcatc cggcctcatg ttctttttca ttaaagagag aaatcaacta 420 ttcaggaccg gcccccacct ttcctcagga gtcatttctg ttccgcacag gcctgctgaa 480 ctgggtgctt tatataggat ttcagtggag tgaagttcag gaggcatgga gctgacaacc 540 atgaggcctc ggcagccacc gccaccaccg ccgccgccac caccgtagca gcagcagcag 600 cagcagcagc agcagcagca gcagcaagag taactctgac ttaggaatag agacagccag 660 agagaaatgt gatcaatgaa ggagacatct ggagtgtgcg tgcttcttca gagggacggg 720 tgatgggcag attggaaaaa gcaccgcaga tgggaacctt aatctttctt ttctaaaatt 780 gatgctatga aaatttgcgt tttctgtaac ttgtaaaaac taaaagttgc ttgtctactg 840 aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaa 876
Table LIV(C). Nucleotide sequence of transcript variant 162P1E6 v.4 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag aggcggggtt tcaccatgtt gcccagactg 300 gtcttgaact cctgagctta agcaatccac ctgcctcggc ctcccaaagt gttgggatca 360 caggcgtgag ccaccgcatc cggcctcatg ttctttttca ttaaagagag aaatcaacta 420 ttcaggaccg gcccccacct ttcctcagga gtcatttctg ttccgcacag gcctgctgaa 480 ctgggtgctt tatataggac actgtcatct ctcaagtacc catcttggag ggtacggacc 540 ccacatgagg atttcagtgg agtgaagttc aggaggcatg gagctgacaa ccatgaggcc 600 tcggcagcca ccgccaccac cgccgccgcc accaccgtag cagcagcagc agcagcagca 660 gcagcagcag cagcagcaag agtaactctg acttaggaat agagacagcc agagagaaat 720 gtgatcaatg aaggagacat ctggagtgtg cgtgcttctt cagagggacg ggtgatgggc 780 agattggaaa aagcaccgca gatgggaacc ttaatctttc ttttctaaaa ttgatgctat 840 gaaaatttgc gttttctgta acttgtaaaa actaaaagtt gcttgtctac tgaaaaaaaa 900 aaaaaaaaaa aaaaaaaaaa aaaaaaaa 928
Table LIV(D). Nucleotide sequence of transcript variant 162P1E6 v.5 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag aggcggggtt tcaccatgtt gcccagactg 300 gtcttgaact cctgagctta agcaatccac ctgcctcggc ctcccaaagt gttgggatca 360 caggcgtgag ccaccgcatc cggcctcatg ttctttttca ttaaagagag aaatcaacta 420 ttcaggaccg gcccccacct ttcctcagga gtcatttctg ttccgcacag gcctgctgaa 480 ctgggtgctt tatataggaa aggaccaacc acaccgagct cagttatggc acacacagtg 540 ggacctagac aaagggagag ggtgaccgac atcccaacta gatttcagtg gagtgaagtt 600 caggaggcat ggagctgaca accatgaggc ctcggcagcc accgccacca ccgccgccgc 660 caccaccgta gcagcagcag cagcagcagc agcagcagca gcagcagcaa gagtaactct 720 gacttaggaa tagagacagc cagagagaaa tgtgatcaat gaaggagaca tctggagtgt 780 gcgtgcttct tcagagggac gggtgatggg cagattggaa aaagcaccgc agatgggaac 840 cttaatcttt cttttctaaa attgatgcta tgaaaatttg cgttttctgt aacttgtaaa 900 aactaaaagt tgcttgtcta ctgaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaa 959
Table LTV(E). Nucleotide sequence of transcript variant 162P1E6 v.6 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag aggcggggtt tcaccatgtt gcccagactg 300 gtcttgaact cctgagctta agcaatccac ctgcctcggc ctcccaaagt gttgggatca 360 caggcgtgag ccaccgcatc cggcctcatg ttctttttca ttaaagagag aaatcaacta 420 ttcaggaccg gcccccacct ttcctcagga gtcatttctg ttccgcacag gcctgctgaa 480 ctgggtgctt tatataggac actgtcatct ctcaagtacc catcttggag ggtacggacc 540 ccacatgagg aaaggaccaa ccacaccgag ctcagttatg gcacacacag tgggacctag 600 acaaagggag agggtgaccg acatcccaac tagatttcag tggagtgaag ttcaggaggc 660 atggagctga caaccatgag gcctcggcag ccaccgccac caccgccgcc gccaccaccg 720 tagcagcagc agcagcagca gcagcagcag cagcagcagc aagagtaact ctgacttagg 780 aatagagaca gccagagaga aatgtgatca atgaaggaga catctggagt gtgcgtgctt 840 cttcagaggg acgggtgatg ggcagattgg aaaaagcacc gcagatggga accttaatct 900 ttcttttcta aaattgatgc tatgaaaatt tgcgttttct gtaacttgta aaaactaaaa 960 gttgcttgtc tactgaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa a 1011
Table LIV(F). Nucleotide sequence oftranscript variant 162P1E6 v.7 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgacfc aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg acactgtcat 600 ctctcaagta cccatcttgg agggtacgga ccccacatga ggatttcagt ggagtgaagt 660 tcaggaggca tggagctgac aaccatgagg cctcggcagc caccgccacc accgccgccg 720 ccaccaccgt agcagcagca gcagcagcag cagcagcagc agcagcagca agagtaactc 780 tgacttagga atagagacag ccagagagaa atgtgatcaa tgaaggagac atctggagtg 840 tgcgtgcttc ttcagaggga cgggtgatgg gcagattgga aaaagcaccg cagatgggaa 900 ccttaatctt tcttttctaa aattgatgct atgaaaattt gcgttttctg taacttgtaa 960 aaactaaaag ttgcttgtct actgaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 1020
Table LIV(G). Nucleotide sequence oftranscript variant 162P1E6 v.8 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgact aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg acactgtcat 600 ctctcaagta cccatcttgg agggtacgga ccccacatga ggaaaggacc aaccacaccg 660 agctcagtta tggcacacac agtgggacct agacaaaggg agagggtgac cgacatccca 720 actagatttc agtggagtga agttcaggag gcatggagct gacaaccatg aggcctcggc 780 agccaccgcc accaccgccg ccgccaccac cgtagcagca gcagcagcag cagcagcagc 840 agcagcagca gcaagagtaa ctctgactta ggaatagaga cagccagaga gaaatgtgat 900 caatgaagga gacatctgga gtgtgcgtgc ttcttcagag ggacgggtga tgggcagatt 960 ggaaaaagca ccgcagatgg gaaccttaat ctttcttttc taaaattgat gctatgaaaa 1020 tttgcgtttt ctgtaacttg taaaaactaa aagttgcttg tctactgaaa aaaaaaaaaa 1080 aaaaaaaaaa aaaaaaaaaa aaa 1103
Table LIV(H). Nucleotide sequence oftranscript variant 162P1E6 v.9 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgact aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg acactgtcat 600 ctctcaagta cccatcttgg agggtacgga ccccacatga gggtgaggct ctctgcacac 660 tccagagtga ggactttaat aatctagtgg actgtacatg ttgggagggg aagagcgggg 720 tgccgagggt ctggagggag aagaattgac tgcccctttt gctcttggag ttaagcagaa 780 atctaaagag aaggcaaaga atcttgcctt cctggcgtca tttcctccta ccatcccagg 840 ccatcattta tttattacag ccaacagact ggcctctttc ttccctttga ctgggaatgg 900 gtcaaaggcg gtgcaggagg aggatctggt ccagataatt cacaagcagg gtgcattttc 960 ctctcattat tgagaactgt gagtgtttat caagaaggca gagcaggaga agatgaacca 1020 gtcttcttcc cctcactacc cagatctctg cctgccaaca agccccgtgt tcaccctggc 1080 aaagagtctt tacattcaga ccaaggagag tgtgactcct tctcagcact agctagaaac 1140 ctcaagccct tgcttaaggg cctttttcag agagacccaa tgcccagaag gctagatgcg 1200 tggggaggag ccacatacga gaaactgcct ccctgcttcg ggtcagaaca agccccagga 1260 agaaagtatt tcaaacaaca aggtgcatct gccccaaccc atccagcctg catgttggtg 1320 ctgagaacag ccttttatgg ggcttgcact gagccatggg catgtctgaa cacaacaagg 1380 aagaggccag agcagcaaca gcacgcaaag ggttgatggg catttctttt aagacagagc 1440 agaaaactct tagatacttt gcgtccttcc tatttgactc agtctatgaa agccaggtta 1500 gcttgctttc ttcctcccta aatcctccat cctcatgacc aacaaagaaa tagttgaatc 1560 attttccagg cacatcttgg ggaggatgtg gggccattgg aggctgtcct tcctggataa 1620 gtctttagga gtgagaacaa ggagtcttac cctcctctgt ccacccaccc ccatgaatgg 1680 gcctggctcc agccaggagt tgtggttttt cctgagctcc tcacctatct cttctggatt 1740 tcacattggc aaacggggtt gcaaagtgct cttcgtgctc tttggacagt gccttgtgga 1800 gaggaatgcc catgcccctg cattccaagg ccttggtaag caagctcaga gtagctggat 1860 ttttctaaag caattgcaga acacctgctt tttctttgtt tcctctagaa aggaccaacc 1920 acaccgagct cagttatggc acacacagtg ggacctagac aaagggagag ggtgaccgac 1980 atcccaacta gatttcagtg gagtgaagtt caggaggcat ggagctgaca accatgaggc 2040 ctcggcagcc accgccacca ccgccgccgc caccaccgta gcagcagcag cagcagcagc 2100 agcagcagca gcagcagcaa gagtaactct gacttaggaa tagagacagc cagagagaaa 2160 tgtgatcaat gaaggagaca tctggagtgt gcgtgcttct tcagagggac gggtgatggg 2220 cagattggaa aaagcaccgc agatgggaac cttaatcttt cttttctaaa attgatgcta 2280 tgaaaatttg cgttttctgt aacttgtaaa aactaaaagt tgcttgtcta ctgaaaaaaa 2340 aaaaaaaaaa aaaaaaaaaa aaaaaaaaa 2369
Table LIV(I). Nucleotide sequence oftranscript variant 162P1E6 v.10 ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgact aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg acactgtcat 600 ctctcaagta cccatcttgg agggtacgga ccccacatga gggtgaggct ctctgcacac 660 tccagagtga ggactttaat aatctagtgg actgtacatg ttgggagggg aagagcgggg 720 tgccgagggt ctggagggag aagaattgac tgcccctttt gctcttggag ttaagcagaa 780 atctaaagag aaggcaaaga atcttgcctt cctggcgtca tttcctccta ccatcccagg 840 ccatcattta tttattacag ccaacagact ggcctctttc ttccctttga ctgggaatgg 900 gtcaaaggcg gtgcaggagg aggatctggt ccagataatt cacaagcagg gtgcattttc 960 ctctcattat tgagaactgt gagtgtttat caagaaggca gagcaggaga agatgaacca 1020 gtcttcttcc cctcactacc cagatctctg cctgccaaca agccccgtgt tcaccctggc 1080 aaagagtctt tacattcaga ccaaggagag tgtgactcct tctcagcact agctagaaac 1140 ctcaagccct tgcttaaggg cctttttcag agagacccaa tgcccagaag gctagatgcg 1200 tggggaggag ccacatacga gaaactgcct ccctgcttcg ggtcagaaca agccccagga 1260 agaaagtatt tcaaacaaca aggtgcatct gccccaaccc atccagcctg catgttggtg 1320 ctgagaacag ccttttatgg ggcttgcact gagccatggg catgtctgaa cacaacaagg 1380 aagaggccag agcagcaaca gcacgcaaag ggttgatggg catttctttt aagacagagc 1440 agaaaactct tagatacttt gcgtccttcc tatttgactc agtctatgaa agccaggtta 1500 gcttgctttc ttcctcccta aatcctccat cctcatgacc aacaaagaaa tagttgaatc 1560 attttccagg cacatcttgg ggaggatgtg gggccattgg aggctgtcct tcctggataa 1620 gtctttagga gtgagaacaa ggagtcttac cctcctctgt ccacccaccc ccatgaatgg 1680 gcctggctcc agccaggagt tgtggttttt cctgagctcc tcacctatct cttctggatt 1740 tcacattggc aaacggggtt gcaaagtgct cttcgtgctc tttggacagt gccttgtgga 1800 gaggaatgcc catgcccctg cattccaagg ccttggtaag caagctcaga gtagctggat 1860 ttttctaaag caattgcaga acacctgctt tttctttgtt tcctctagaa aggaccaacc 1920 acaccgagct cagttatggc acacacagtg ggacctagac aaagggagag ggtgaccgac 1980 atcccaacta ggtaaacaca gaggaggttc cacatggact tatctgggtg gctgttttga 2040 aaacgagaaa cagtcaagag tccctggccc cacagaccca cctccccaac tcagcactgt 2100 ctgtctgtgc agcaggtgca aggacgtgtt gaactagctc tctgcagcct ccttggagga 2160 tgtgatccta tgggaggggt aggagtattc aggtccttga catctcccaa atgtgtgatt 2220 ccgggatgcc aaaggccttt ggccaggtaa tgcagtgtct acaggctgag gttgacatgc 2280 atccccaccc tctgagaaaa agatcctcag acaatccatg tgcttctctt gtccttcatt 2340 ccaccggagt ctgtctcata cccaaccaga tttcagtgga gtgaagttca ggaggcatgg 2400 agctgacaac catgaggcct cggcagccac cgccaccacc gccgccgcca ccaccgtagc 2460 agcagcagca gcagcagcag cagcagcagc agcagcaaga gtaactctga cttaggaata 2520 gagacagcca gagagaaatg tgatcaatga aggagacatc tggagtgtgc gtgcttcttc 2580 agagggacgg gtgatgggca gattggaaaa agcaccgcag atgggaacct taatctttct 2640 tttctaaaat tgatgctatg aaaatttgcg ttttctgtaa cttgtaaaaa ctaaaagttg 2700 cttgtctact gaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaa 2747
Table LIV(J). Nucleotide sequence of transcript variant 162P1E6 v.ll ccttgaaatg ggctgagtcc ctcttgctca cccttgactt ggaaaaacca gtttctcttt 60 tattgtctgt tactaatctc tattctaaaa attcagctca attctcaacc atactccaaa 120 ctctctcttt tccagctacc tttactccct ctccttcaat tccactttcc tctgcttact 180 tttttttttt ttctgacagg gtctcacttt gtcgcccggg caggagtgca gtggctcaat 240 cttgggctca ctgcagcctc aacctcccag gttcaagcga ttctcctgcc tcagcccctc 300 aagtagctgg gactacaagc gcacaccacc acgcctgact aattttttgt atttttttgt 360 agaggcgggg tttcaccatg ttgcccagac tggtcttgaa ctcctgagct taagcaatcc 420 acctgcctcg gcctcccaaa gtgttgggat cacaggcgtg agccaccgca tccggcctca 480 tgttcttttt cattaaagag agaaatcaac tattcaggac cggcccccac ctttcctcag 540 gagtcatttc tgttccgcac aggcctgctg aactgggtgc tttatatagg gtaagtgttt 600 ctcatttttt gttccctgtc ctcaagcctt aggggcaaaa gaaacatcca agatttgaaa 660 tttcttttct tcttctcatc tgcatggctg tagccatctc tctgttctgc attatcttat 720 gacaaaaaaa aaaaattctt attttgaagc aaactcaaag ctaggtcctg atgtctcaag 780 gcacaggtac tcgtacttaa aggtgagtct gaaatctgtg gatttgggga actttggaaa 840 aacaaagatg agtggctaga tcagggggct cattgggcag gaagaggaga ctggaaaatg 900 ccatattcac tgcaagtcaa ttatcaactt cctccaaggc taaaatagct gaacctgctg 960 cattttaaac caatcctcag ccactttggt gttttctcaa ggatttccag ggatcccagg 1020 cagtaaattc tgctgataat aggaattggt gtgataaggt gggtgctgag cagtttaagc 1080 accaagattg tagctctgtc tggttttgtg gagatttact caactagaag aacagagatt 1140 tggctggttt ttcagtcctg gggtgcaggg tgcacctgta ctggaaaatt taggacctgg 1200 tttcattctt tgagtctcat gttcaagttg gttttaatgt tatgaagaca cttgggacgt 1260 aatcctgagg gcagctgggg ggaagaaagt ggtcactgga tggacttacc ctgtagcgag 1320 cccatgcatg gtttgttctc tgatcgtgca tgtgcttggc tctagaccca tgtaaccatg 1380 gtgaaggcca ctgggggatt cagttggcaa aggcatagtg ggcagaagaa tcttgaacaa 1440 ggagtccaga gcaggtcaag tctcctgata caggttgtga ctcatggttt ttgtctctgc 1500 ctgtagcagc tacaggtctg taaagcaagg ggagagtgat aaggaaagaa ctcacctttc 1560 tggggctctc tgacattaat gccacctccc atttgctttt tgcagacact gtcatctctc 1620 aagtacccat cttggagggt acggacccca catgagggtg aggctctctg cacactccag 1680 agtgaggact ttaataatct agtggactgt acatgttggg aggggaagag cggggtgccg 1740 agggtctgga gggagaagaa ttgactgccc cttttgctct tggagttaag cagaaatcta 1800 aagagaaggc aaagaatctt gccttcctgg cgtcatttcc tcctaccatc ccaggccatc 1860 atttatttat tacagccaac agactggcct ctttcttccc tttgactggg aatgggtcaa 1920 aggcggtgca ggaggaggat ctggtccaga taattcacaa gcagggtgca ttttcctσtc 1980 attattgaga actgtgagtg tttatcaaga aggcagagca ggagaagatg aaccagtctt 2040 cttcccctca ctacccagat ctctgcctgc caacaagccc cgtgttcacc ctggcaaaga 2100 gtctttacat tcagaccaag gagagtgtga ctccttctca gcactagcta gaaacctcaa 2160 gcccttgctt aagggccttt ttcagagaga cccaatgccc agaaggctag atgcgtgggg 2220 aggagccaca tacgagaaac tgcctccctg cttcgggtca gaacaagccc caggaagaaa 2280 gtatttcaaa caacaaggtg catctgcccc aacccatcca gcctgcatgt tggtgctgag 2340 aacagccttt tatggggctt gcactgagcc atgggcatgt ctgaacacaa caaggaagag 2400 gccagagcag caacagcacg caaagggttg atgggcattt cttttaagac agagcagaaa 2460 actcttagat actttgcgtc cttcctattt gactcagtct atgaaagcca ggttagcttg 2520 ctttcttcct ccctaaatcc tccatcctca tgaccaacaa agaaatagtt gaatcatttt 2580 ccaggcacat cttggggagg atgtggggcc attggaggct gtccttcctg gataagtctt 2640 taggagtgag aacaaggagt cttaccctcc tctgtccacc cacccccatg aatgggcctg 2700 gctccagcca ggagttgtgg tttttcctga gctcctcacc tatctcttct ggatttcaca 2760 ttggcaaacg gggttgcaaa gtgctcttcg tgctctttgg acagtgcctt gtggagagga 2820 atgcccatgc ccctgcattc caaggccttg gtaagcaagc tcagagtagc tggatttttc 2880 taaagcaatt gcagaacacc tgctttttct ttgtttcctc tagaaaggac caaccacacc 2940 gagctcagtt atggcacaca cagtgggacc tagacaaagg gagagggtga ccgacatccc 3000 aactagattt cagtggagtg aagttcagga ggcatggagc tgacaaccat gaggcctcgg 3060 cagccaccgc caccaccgcc gccgccacca ccgtagcagc agcagcagca gcagcagcag 3120 cagcagcagc agcaagagta actctgactt aggaatagag acagccagag agaaatgtga 3180 tcaatgaagg agacatctgg agtgtgcgtg cttcttcaga gggacgggtg atgggcagat 3240 tggaaaaagc accgcagatg ggaaccttaa tctttctttt ctaaaattga tgctatgaaa 3300 atttgcgttt tctgtaactt gtaaaaacta aaagttgctt gtctactgaa aaaaaaaaaa 3360 aaaaaaaaaa aaaaaaaaaa aaaa 3384
Table LV(A). Nucleotide sequence alignment of 121P1F1 v.l and 162P1E6 v.2
162P1E6V.1 i62PiE6v.2 CCTTGAAAΓGGGCTGAGΓCCCΓCTTGCTCACCCTTGACTTGGAAAAACCAGTΓTCTCTTT SO 5
162P1E6V.1
162P1E6V. TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120
0 162P1E6V.1
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180
162P1E6V.1 5 162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 2 0
162P1E6V.1
162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 0
162P1E6V.1
162P1E6 .2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 5
162P1E6V.1
162P1E6 .2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420
0 162P1ESV.1
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480
162P1E6V.1 GCCCCCACCTTTCCTCAG 18 5 162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540
******************
162P1E6V.1 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 78 162P1ESV.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 Q ************************************************************
162P1E6 .1 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 138 162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 ************************************************************ 5
162P1E6 .1 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 198 162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 ************************************************************ 0 1S2P1E6V.1 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 258 162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 ************************************************************
162P1E6 .1 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 318 5 162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 ************************************************************
162P1E6V.1 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 378 162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 Q ************************************************************
162P1E6V.1 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 438 162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 ************************************************************ 5
162P1E6V.1 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 498 162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 ************************************************************ 0 162P1E6V.1 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 558 162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 ************************************************************ 162P1E6V.1 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 618 162P1E6 .2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 ************************************************************
162P1E6 .1 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 678 162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 ************************************************************
162P1E6V.1 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 738 162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 ************************************************************
162P1E6 .1 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 798 162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 ************************************************************
162P1E6V.1 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 858 162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 ************************************************************
162P1E6V.1 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 918 162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 ************************************************************ 162P1E6V.1 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 978 162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 ************************************************************
162P1E6V.1 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1038 162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 ************************************************************
162P1E6V.1 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1098 162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 ************************************************************
162P1E6V.1 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1158 162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 ************************************************************
162P1E6V.1 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1218 162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 17 0 ************************************************************ 162P1E6V.1 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1278 152P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 ************************************************************
162P1E6V.1 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1338 162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 ************************************************************
162P1E6V.1 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1398 162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 ************************************************************
162P1E6V.1 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1458 162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 ************************************************************
162P1E6V.1 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 1518 162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 ************************************************************ 162P1E6V.1 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 1578 162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 ************************************************************
162P1E6V.1 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 1638 162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 ************************************************************ 162P1E6 .1 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 1698 162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 ************************************************************
162P1E6 .1 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 1758 162P1E6V .2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 ************************************************************
162P1E6V.1 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 1818 162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 ************************************************************
162P1E6V.1 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 1878 162P1E6 .2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 ************************************************************
162P1E6V.1 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 1938 162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 ************************************************************
162P1ESV.1 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 1998 162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 ************************************************************ 162P1E6V.1 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2058 162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 ************************************************************
162P1E6V.1 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2118 162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 ************************************************************
162P1E6V.1 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2178 162P1E6 .2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 ************************************************************
162P1E6V.1 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2238 162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 ************************************************************
162P1E6V.1 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2298 162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 ************************************************************ 162P1E6V.1 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2358 162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 ************************************************************
162P1E6V.1 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2418 162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 ************************************************************
162P1E6V.1 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 2478 162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 ************************************************************
162P1E6V.1 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 2538 162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 ************************************************************
162P1E6V.1 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 2598 162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 ************************************************************ 162P1E6V.1 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 2658 162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 ************************************************************
162P1E6V.1 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 2718 162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 ************************************************************ 162P1E6V.1 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 2778 162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 ************************************************************ 162P1E6V.1 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 2838 162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 ************************************************************
162P1E6V.1 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 2898 162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 ************************************************************
162P1E6V.1 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 2958 162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 ************************************************************
162P1E6V.1 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3018 162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 ************************************************************
162P1E6V.1 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3078 162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 ************************************************************ 162P1E6V.1 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3138 162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 ************************************************************
162P1E6V.1 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3198 162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 ************************************************************
162P1E6V.1 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3240 162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 ******************************************
Table LV(B). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.3
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.3 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6V.2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.3 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.3 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGC 24Q 162P1E6V.3 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 ************************************************************ 162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.3 CTTGGGCTCACTGCAGCCTCAACCTCCCAG 270
******************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.3
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6 .3 --AGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 328 **********************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.3 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 388 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.3 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 448 ************************************************************ 162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.3 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 498
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.3
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.3
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.3
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.3
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.3
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.3
162P1E6V .2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020
162P1E6V.3
162P1E6V.2 CAGTAAATΓCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC IO8O 162P1E6V.3
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.3
162P1E6V.2 GGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.3
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.3
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.3
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCG GCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.3
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.3
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTG GACTCATGGTTττTGTCTCTGC 1500 162P1E6V.3
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.3
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.3 162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.3
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.3
162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162P1E6V.3
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.3
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.3
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.3
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.3
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.3
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.3
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.3
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.3
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.3
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.3
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.3
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.3
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.3
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.3
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.3 162P1E6V .2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V .3
162P1E6V .2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.3
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.3
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.3
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.3
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.3
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.3
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.3
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.3
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.3
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.3
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.3 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 534
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.3 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 594 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.3 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 654 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.3 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 714 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.3 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 774 ************************************************************ 162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.3 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 834 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.3 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 876 ****************************************** Table LV(C). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.4
162P1E6 .2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.4 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6V.2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.4 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************ 162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6 .4 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6 .2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 162P1E6V.4 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 ************************************************************
162P1E6 .2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300
162P1E6V.4 CTTGGGCTCACTGCAGCCTCAACCTCCCAG 270
******************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.4
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.4 --AGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 328 **********************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.4 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 388 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.4 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 4 8 ************************************************************
162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.4 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 498
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.4
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.4
162P1E6V .2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780
162P1E6V.4
162P1E6V .2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.4
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.4
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.4
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.4
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.4 162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 11 0 162P1E6V.4
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.4
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.4
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.4
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.4
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.4
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V.4
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.4
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.4 ACACTGTCATCTCTC 513
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.4 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGG 550
*************************************
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.4
162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800
162P1E6V.4
162P1E6V .2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.4
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.4
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.4
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.4
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.4
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.4 162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.4
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.4
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.4
162P1E6V .2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.4
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.4
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.4
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.4
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.4
162P1E6V . 2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V .4
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.4
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.4
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.4
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.4
162P1E6V .2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V .4
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.4
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.4
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.4
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.4 162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.4
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.4
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3 20 162P1E6V.4 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 586
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.4 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 646 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.4 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 706 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.4 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 766 ************************************************************ 162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660
162P1E6V.4 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 826 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.4 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 886 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.4 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 928 ******************************************
Table LV(D). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.5 162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.5 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6V.2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.5 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.5 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 2 _0 162P1E6V.5 TΓTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 24O
************************************************************
162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.5 CTTGGGCTCACTGCAGCCTCAACCTCCCAG 270
******************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.5
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.5 --AGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 328
**********************************************************
162P1E6 .2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.5 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 388 ************************************************************ 162P1E6V .2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.5 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 448 ************************************************************
162P1E6V .2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.5 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 498
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCΓTAGGGGCAAAAGAAACATCCAAGATTΓGAAA 660 162P1E6V.5
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.5
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.5
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.5
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.5
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.5
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.5
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.5
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.5
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.5
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.5
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.5
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.5
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.5
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V.5
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.5 162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.5
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.5
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.5
162P1E6V .2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162P1E6V . 5
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.5
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.5
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.5
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.5
162P1E6V . 2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V .5
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.5
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.5
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCC&GGAAGAAA 2280 162P1E6V.5
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.5
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.5
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.5
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.5
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.5
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.5
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.5 162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.5
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.5
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.5
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940
162P1E6V.5 AAAGGACCAACCACACC 515
*****************
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.5 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 575 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.5 AACTAG 581
******
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.5
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.5
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240
162P1E6V.5
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V .5
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.5
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.5 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 617
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.5 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 677 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.5 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 737 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.5 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 797 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.5 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 857 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.5 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 917 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.5 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 959 ****************************************** Table LV(E). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.6
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6 .6 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6 .2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6 .6 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V. CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V. CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 2 0 162P1E6V.6 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 2 _0
************************************************************ 162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300
162P1E6V.6 CTTGGGCTCACTGCAGCCTCAACCTCCCAG 270
******************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.6
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.6 --AGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 328 **********************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.6 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 388 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.6 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 448 ************************************************************ 162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.6 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 498
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.6
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.6
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.6
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.6
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.6
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.6
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 10 0 162P1E6V.6 162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.6
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.6
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.6
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.6
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.6
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.6
162P1E6V .2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440
162P1E6V .6
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V .6
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.6
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.6 ACACTGTCATCTCTC 513
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.6 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGG 550
************************************* 162P1E6V .2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740
162P1E6V .6
162P1E6V .2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162P1E6V .6
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.6
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.6
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.6
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.6
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.6 162P1E6V .2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 160 162P1E6V.6
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V .6
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.6
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.6
162P1E6V .2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V. 6
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.6
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520
162P1E6V.6
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V. 6
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.6
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.6
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.6
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.6
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.6
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V. 6 AAAGGACCAACCACACC 567
*****************
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.6 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 627 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.6 AACTAG 633
****** 162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.6
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.6 162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.6
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.6
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V .6
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3 20 162P1E6V.6 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 669
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.6 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 729 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.6 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 789 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.6 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 849 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.6 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 909 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.6 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 969 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.6 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 1011 ******************************************
Table LV(F). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.7
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.7 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60
************************************************************
162P1E6 .2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.7 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.7 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 162P1E6V.7 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 ************************************************************ 162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.7 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 ************************************************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.7 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360
************************************************************
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.7 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 20 ************************************************************ 162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.7 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.7 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 ************************************************************
162P1E6V . 2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V. 7 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 590
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.7
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.7
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.7
162P1E6V .2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840
162P1E6V .7
162P1E6V .2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.7
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.7
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.7
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.7
162P1E6V .2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140
162P1E6V.7
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V. 7
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.7
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.7
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.7
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.7
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V.7 162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.7
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.7 ACACTGTCATCTCTC 605
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V .7 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGG 642
*************************************
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.7
162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162P1E6V.7
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.7
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.7
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.7
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 20 0 162P1E6V.7
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.7
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.7
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.7
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.7
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.7
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.7
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.7
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.7
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.7 162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.7
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.7
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V .7
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.7
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.7
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.7
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.7
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.7
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.7
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.7
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.7
162P1E6V .2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300
162P1E6V .7
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V .7
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.7 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 678
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.7 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 738 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.7 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 798 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.7 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 858 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.7 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 918 ************************************************************ 162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.7 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 978 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.7 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 1020 ******************************************
Table LV(G). Nucleotide sequence alignment of121P1F1 v.2 and 162P1E6 v.8
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCΪTT 60 162P1E6V.8 CCTΓGAAATGGGCTGAGTCCCΓCTΓGCTCACCCTTGACTTGGAAAAACCAGΓTTCTCTTT S O
************************************************************
162P1E6V.2 TATTGTCΓGTTACTAATCTCTATΓCTAAAAAΓTCAGCΓCAATTCTCAACCATACTCCAAA 120 162P1E6V.8 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120
************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.8 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 162P1E6V.8 TTTTTTTTTTTTCTGACAGGGTCTCACT TGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240
************************************************************
1S2P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.8 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 ************************************************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.8 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 ************************************************************
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.8 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 20 ************************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.8 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.8 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 ************************************************************
162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.8 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 590
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.8
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.8
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780
162P1E6V. 8
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.8
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.8 162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.8
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.8
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V .8
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.8
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.8
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTΓGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.8
162P1E6V .2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320
162P1E6V. 8
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.8
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.8
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V.8
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.8
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620
162P1E6V.8 ACACTGTCATCTCTC 605
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.8 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGG 642
*************************************
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.8
162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162P1E6V.8
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V.8
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.8
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.8 162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.8
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.8
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.8
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.8
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.8
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.8
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400
162P1E6V .8
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V .8
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.8
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.8
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.8
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.8
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.8
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.8
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.8
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.8 AAAGGACCAACCACACC 659
*****************
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.8 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 719 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.8 AACTAG 725
****** 162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.8
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.8
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V. 8
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.8
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.8
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.8 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 761
************************************ 162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480
162P1E6V. 8 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 821 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V. 8 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 881 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.8 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 941 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.8 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 1001 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.8 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 1061 ************************************************************ 162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.8 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 1103 ******************************************
Table LV(H). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.9
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.9 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6V.2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.9 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************ 162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.9 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 162P1E6V.9 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 ************************************************************
162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.9 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 ************************************************************ 162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.9 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 ************************************************************
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.9 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 ************************************************************
162P1E6V .2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 80 162P1E6V .9 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.9 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 5 0 ************************************************************
162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.9 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 590
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.9
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.9
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.9
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.9
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.9
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V.9
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.9
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.9
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.9
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.9
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.9
162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.9
162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.9 162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.9
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162P1E6V.9
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.9
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.9 ACACTGTCATCTCTC 605
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.9 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 665 ************************************************************
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.9 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 725 ************************************************************ 162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800
162P1E6V .9 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 785 ************************************************************
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 162P1E6V .9 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 845 ************************************************************
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.9 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 905 ************************************************************
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6V.9 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCAΪTTTCCTCTC 965 ************************************************************
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.9 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 1025 ************************************************************ 162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100
162P1E6V.9 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 1085 ************************************************************
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.9 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 1145 ************************************************************
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.9 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 1205 ************************************************************
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.9 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 1265 ************************************************************
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.9 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 1325 ************************************************************ 162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.9 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 1385 ************************************************************
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2 60 162P1E6V.9 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 1445 ************************************************************ 162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.9 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 1505 ************************************************************
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.9 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 1565 ************************************************************
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCΓΓCCΓGGATAAGTCTT 2640 162P1E6V.9 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 1625
************************************************************
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 700 162P1E6V.9 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 1685 ************************************************************
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.9 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 1745 ************************************************************
162P1E6V.2 TTGGCAAACGGGGTΓGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 28 0 162P1E6V.9 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 1805 ************************************************************
162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.9 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 1865 ************************************************************
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.9 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 1925 ************************************************************
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.9 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 1985 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.9 AACTAG 1991
******
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.9
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.9
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.9
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.9
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.9
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.9 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 2027
************************************ 162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.9 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 2087 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.9 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 2147 ************************************************************ 162P1E6 .2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6 .9 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 2207 ************************************************************ 162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.9 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 2267 ************************************************************
162P1E6V.2 AAAAΓΓGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.9 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 2327 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.9 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 2369 ******************************************
Table LV(I). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.10
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162P1E6V.10 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 ************************************************************
162P1E6V.2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6V.10 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162P1E6V.10 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 162P1E6V.10 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGGCAGGAGTGCAGTGGCTCAAT 240 ************************************************************
162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162P1E6V.10 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 ************************************************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.10 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 ************************************************************
162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162P1E6V.10 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 ************************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162P1E6V.10 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162P1E6V.10 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 ************************************************************
162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162P1E6V.10 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGG 590
**************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162P1E6V.10
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162P1E6V.10
162P1E6V.2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162P1E6V.10 162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.10
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162P1E6V.10
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162P1E6V .10
162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162P1E6V.10
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162P1E6V.10
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.10
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200
162P1E6V.10
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162P1E6V.10
162P1E6V .2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162P1E6V.10
162P1E6V .2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162P1E6V.10
162P1E6V.2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.10
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500
162P1E6V .10
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V.10
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162P1E6V.10 ACACTGTCATCTCTC 605
***************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162P1E6V.10 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 665 ************************************************************
162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V.10 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 725 ************************************************************ 162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 1'62P1E6V.10 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 785 ************************************************************
162P1E6V.2 AAGAGAAGGCAAAGAATCΓΓGCCTTCCΓGGCGTCATTTCCTCCΓACCATCCCAGGCCATC i860 162P1E6V.10 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 845 ************* *********************************************** 162P1E6 .2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162P1E6V.10 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 905 ************************************************************
162P1E6 .2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162P1E6 .10 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 965 ************************************************************
162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162P1E6V.10 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 1025 ************************************************************
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162P1E6V.10 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 1085 ************************************************************
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.10 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA ____
************************************************************
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162P1E6V.10 GCCCTTGCTTAAGGGCCTTTΪTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 1205 ************************************************************
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162P1E6V.10 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 1265 ************************************************************
162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162P1E6V.10 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 1325 ************************************************************
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162P1E6V.10 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 1385 ************************************************************
162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162P1E6V.10 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 1445 ************************************************************
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162P1E6V.10 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 1505 ************************************************************ 162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162P1E6V.10 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 1565 ************************************************************
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162P1E6V.10 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 1625 ************************************************************
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 162P1E6V.10 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 1685 ************************************************************
162P1E6V.2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162P1E6V.10 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 1745 ************************************************************
162P1E6V.2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 162P1E6V.10 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 1805 ************************************************************ 162P1E6V.2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162P1E6V.10 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 1865 ************************************************************
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162P1E6V.10 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 1925 ************************************************************ 162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162P1E6V.10 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 1985 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162P1E6V.10 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 2045 ************************************************************ '
162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120 162P1E6V.10 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 2105 ************************************************************
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162P1E6V.10 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 2165 ************************************************************
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162P1E6V.10 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 2225 ************************************************************
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162P1E6V.10 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 2285 ************************************************************
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162P1E6V.10 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 2345 ************************************************************
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.10 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 2 05 ************************************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.10 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 2465 ************************************************************
162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162P1E6V.10 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 2525 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162P1E6V.10 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 2585 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162P1E6V.10 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 2645 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.10 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 2705
************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162P1E6V.10 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 2747 ******************************************
Table LV(J). Nucleotide sequence alignment of 121P1F1 v.2 and 162P1E6 v.ll
162P1E6V.2 CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60 162PlE6v.ll CCTTGAAATGGGCTGAGTCCCTCTTGCTCACCCTTGACTTGGAAAAACCAGTTTCTCTTT 60
************************************************************
162P1E6 .2 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 162P1E6 .11 TATTGTCTGTTACTAATCTCTATTCTAAAAATTCAGCTCAATTCTCAACCATACTCCAAA 120 ************************************************************
162P1E6V.2 CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 162PlE6v.ll CTCTCTCTTTTCCAGCTACCTTTACTCCCTCTCCTTCAATTCCACTTTCCTCTGCTTACT 180 ************************************************************
162P1E6V.2 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCC 24Q 162P1E6V.11 TTTTTTTTTTTTCTGACAGGGTCTCACTTTGTCGCCCGGG 240 ************************************************************ 162P1E6V.2 CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 162PlE6v.ll CTTGGGCTCACTGCAGCCTCAACCTCCCAGGTTCAAGCGATTCTCCTGCCTCAGCCCCTC 300 ************************************************************
162P1E6V.2 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 162P1E6V.11 AAGTAGCTGGGACTACAAGCGCACACCACCACGCCTGACTAATTTTTTGTATTTTTTTGT 360 ************************************************************ 162P1E6V.2 AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 162PlE6v.ll AGAGGCGGGGTTTCACCATGTTGCCCAGACTGGTCTTGAACTCCTGAGCTTAAGCAATCC 420 ************************************************************
162P1E6V.2 ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 480 162PlE6v.ll ACCTGCCTCGGCCTCCCAAAGTGTTGGGATCACAGGCGTGAGCCACCGCATCCGGCCTCA 80 ************************************************************
162P1E6V.2 TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 162PlE6v.ll TGTTCTTTTTCATTAAAGAGAGAAATCAACTATTCAGGACCGGCCCCCACCTTTCCTCAG 540 ************************************************************
162P1E6V.2 GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 162PlE6v.ll GAGTCATTTCTGTTCCGCACAGGCCTGCTGAACTGGGTGCTTTATATAGGGTAAGTGTTT 600 ************************************************************
162P1E6V.2 CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 162PlE6v.ll CTCATTTTTTGTTCCCTGTCCTCAAGCCTTAGGGGCAAAAGAAACATCCAAGATTTGAAA 660 ************************************************************
162P1E6V.2 TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 162PlE6v.ll TTTCTTTTCTTCTTCTCATCTGCATGGCTGTAGCCATCTCTCTGTTCTGCATTATCTTAT 720 ************************************************************
162P1E6V..2 GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 162PlE6v.ll GACAAAAAAAAAAAATTCTTATTTTGAAGCAAACTCAAAGCTAGGTCCTGATGTCTCAAG 780 ************************************************************
162P1E6V.2 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 162P1E6V.11 GCACAGGTACTCGTACTTAAAGGTGAGTCTGAAATCTGTGGATTTGGGGAACTTTGGAAA 840 ************************************************************
162P1E6V.2 AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 162PlE6v.ll AACAAAGATGAGTGGCTAGATCAGGGGGCTCATTGGGCAGGAAGAGGAGACTGGAAAATG 900 ************************************************************
162P1E6V.2 CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 162PlE6v.ll CCATATTCACTGCAAGTCAATTATCAACTTCCTCCAAGGCTAAAATAGCTGAACCTGCTG 960 ************************************************************ 162P1E6V.2 CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 162PlE6v.ll CATTTTAAACCAATCCTCAGCCACTTTGGTGTTTTCTCAAGGATTTCCAGGGATCCCAGG 1020 ************************************************************
162P1E6V.2 CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 162PlE6v.ll CAGTAAATTCTGCTGATAATAGGAATTGGTGTGATAAGGTGGGTGCTGAGCAGTTTAAGC 1080 ************************************************************
162P1E6V.2 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 162P1E6V.11 ACCAAGATTGTAGCTCTGTCTGGTTTTGTGGAGATTTACTCAACTAGAAGAACAGAGATT 1140 ************************************************************
162P1E6V.2 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 162P1E6V.11 TGGCTGGTTTTTCAGTCCTGGGGTGCAGGGTGCACCTGTACTGGAAAATTTAGGACCTGG 1200 ************************************************************
162P1E6V.2 TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 162PlE6v.ll TTTCATTCTTTGAGTCTCATGTTCAAGTTGGTTTTAATGTTATGAAGACACTTGGGACGT 1260 ************************************************************ 162P1E6V.2 AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 162PlE6v.ll AATCCTGAGGGCAGCTGGGGGGAAGAAAGTGGTCACTGGATGGACTTACCCTGTAGCGAG 1320 ************************************************************ 162P1E6V.2 CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 162PlE6v.ll CCCATGCATGGTTTGTTCTCTGATCGTGCATGTGCTTGGCTCTAGACCCATGTAACCATG 1380 ************************************************************
162P1E6V .2 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 162P1E6V.11 GTGAAGGCCACTGGGGGATTCAGTTGGCAAAGGCATAGTGGGCAGAAGAATCTTGAACAA 1440 ************************************************************
162P1E6V.2 GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 162PlE6v.ll GGAGTCCAGAGCAGGTCAAGTCTCCTGATACAGGTTGTGACTCATGGTTTTTGTCTCTGC 1500 ************************************************************
162P1E6V.2 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 162P1E6V .11 CTGTAGCAGCTACAGGTCTGTAAAGCAAGGGGAGAGTGATAAGGAAAGAACTCACCTTTC 1560 ************************************************************
162P1E6V.2 TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 162PlE6v.ll TGGGGCTCTCTGACATTAATGCCACCTCCCATTTGCTTTTTGCAGACACTGTCATCTCTC 1620 ************************************************************
162P1E6V.2 AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 162PlE6v.ll AAGTACCCATCTTGGAGGGTACGGACCCCACATGAGGGTGAGGCTCTCTGCACACTCCAG 1680 ************************************************************ 162P1E6V.2 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 162P1E6V .11 AGTGAGGACTTTAATAATCTAGTGGACTGTACATGTTGGGAGGGGAAGAGCGGGGTGCCG 1740 ************************************************************
162P1E6V.2 AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 162PlE6v.ll AGGGTCTGGAGGGAGAAGAATTGACTGCCCCTTTTGCTCTTGGAGTTAAGCAGAAATCTA 1800 ************************************************************
162P1E6V.2 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860
162P1E6V .11 AAGAGAAGGCAAAGAATCTTGCCTTCCTGGCGTCATTTCCTCCTACCATCCCAGGCCATC 1860 ************************************************************
162P1E6V.2 ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 162PlE6v.ll ATTTATTTATTACAGCCAACAGACTGGCCTCTTTCTTCCCTTTGACTGGGAATGGGTCAA 1920 ************************************************************
162P1E6V.2 AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 162PlE6v.ll AGGCGGTGCAGGAGGAGGATCTGGTCCAGATAATTCACAAGCAGGGTGCATTTTCCTCTC 1980 ************************************************************ 162P1E6V.2 ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 162PlE6v.ll ATTATTGAGAACTGTGAGTGTTTATCAAGAAGGCAGAGCAGGAGAAGATGAACCAGTCTT 2040 ************************************************************
162P1E6V.2 CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 162PlE6v.ll CTTCCCCTCACTACCCAGATCTCTGCCTGCCAACAAGCCCCGTGTTCACCCTGGCAAAGA 2100 ************************************************************
162P1E6V.2 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 162P1E6V.11 GTCTTTACATTCAGACCAAGGAGAGTGTGACTCCTTCTCAGCACTAGCTAGAAACCTCAA 2160 ************************************************************
162P1E6V.2 GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 162PlE6v.ll GCCCTTGCTTAAGGGCCTTTTTCAGAGAGACCCAATGCCCAGAAGGCTAGATGCGTGGGG 2220 ************************************************************
162P1E6V.2 AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 162PlE6v.ll AGGAGCCACATACGAGAAACTGCCTCCCTGCTTCGGGTCAGAACAAGCCCCAGGAAGAAA 2280 ************************************************************ 162P1E6V.2 GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 162PlE6v.ll GTATTTCAAACAACAAGGTGCATCTGCCCCAACCCATCCAGCCTGCATGTTGGTGCTGAG 2340 ************************************************************
162P1E6V.2 AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2400 162PlE6v.ll AACAGCCTTTTATGGGGCTTGCACTGAGCCATGGGCATGTCTGAACACAACAAGGAAGAG 2 00 ************************************************************ 162P1E6V.2 GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 162PlE6v.ll GCCAGAGCAGCAACAGCACGCAAAGGGTTGATGGGCATTTCTTTTAAGACAGAGCAGAAA 2460 ************************************************************
162P1E6V.2 ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 2520 162PlE6v.ll ACTCTTAGATACTTTGCGTCCTTCCTATTTGACTCAGTCTATGAAAGCCAGGTTAGCTTG 520 ************************************************************
162P1E6V.2 CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 162PlE6v.ll CTTTCTTCCTCCCTAAATCCTCCATCCTCATGACCAACAAAGAAATAGTTGAATCATTTT 2580 ************************************************************
162P1E6V.2 CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 162PlE6v. ll CCAGGCACATCTTGGGGAGGATGTGGGGCCATTGGAGGCTGTCCTTCCTGGATAAGTCTT 2640 ************************************************************
162P1E6V.2 TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 700 162PlE6v. ll TAGGAGTGAGAACAAGGAGTCTTACCCTCCTCTGTCCACCCACCCCCATGAATGGGCCTG 2700 ************************************************************
162P1E6V .2 GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 162PlE6v . ll GCTCCAGCCAGGAGTTGTGGTTTTTCCTGAGCTCCTCACCTATCTCTTCTGGATTTCACA 2760 ************************************************************ 162P1E6V .2 TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820
162PlE6v. ll TTGGCAAACGGGGTTGCAAAGTGCTCTTCGTGCTCTTTGGACAGTGCCTTGTGGAGAGGA 2820 ************************************************************
162P1E6V .2 ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 162PlE6v. ll ATGCCCATGCCCCTGCATTCCAAGGCCTTGGTAAGCAAGCTCAGAGTAGCTGGATTTTTC 2880 ************************************************************
162P1E6V.2 TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 162PlE6v.ll TAAAGCAATTGCAGAACACCTGCTTTTTCTTTGTTTCCTCTAGAAAGGACCAACCACACC 2940 ************************************************************
162P1E6V.2 GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 162PlE6v.ll GAGCTCAGTTATGGCACACACAGTGGGACCTAGACAAAGGGAGAGGGTGACCGACATCCC 3000 ************************************************************
162P1E6V.2 AACTAGGTAAACACAGAGGAGGTTCCACATGGACTTATCTGGGTGGCTGTTTTGAAAACG 3060 162PlE6v.ll AACTAG 3006
****** 162P1E6V.2 AGAAACAGTCAAGAGTCCCTGGCCCCACAGACCCACCTCCCCAACTCAGCACTGTCTGTC 3120
162PlE6v.ll
162P1E6V.2 TGTGCAGCAGGTGCAAGGACGTGTTGAACTAGCTCTCTGCAGCCTCCTTGGAGGATGTGA 3180 162PlE6v. ll
162P1E6V.2 TCCTATGGGAGGGGTAGGAGTATTCAGGTCCTTGACATCTCCCAAATGTGTGATTCCGGG 3240 162PlE6v.ll
162P1E6V.2 ATGCCAAAGGCCTTTGGCCAGGTAATGCAGTGTCTACAGGCTGAGGTTGACATGCATCCC 3300 162PlE6v.ll
162P1E6V.2 CACCCTCTGAGAAAAAGATCCTCAGACAATCCATGTGCTTCTCTTGTCCTTCATTCCACC 3360 162PlE6v.ll
162P1E6V.2 GGAGTCTGTCTCATACCCAACCAGATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3420 162P1E6V.11 ATTTCAGTGGAGTGAAGTTCAGGAGGCATGGAGCTG 3042
************************************
162P1E6V.2 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3480 162P1E6V.11 ACAACCATGAGGCCTCGGCAGCCACCGCCACCACCGCCGCCGCCACCACCGTAGCAGCAG 3102 ************************************************************ 162P1E6V.2 CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3540 162PlE6v.ll CAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAAGAGTAACTCTGACTTAGGAATAGAGAC 3162 ************************************************************
162P1E6V.2 AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3600 162PlE6v.ll AGCCAGAGAGAAATGTGATCAATGAAGGAGACATCTGGAGTGTGCGTGCTTCTTCAGAGG 3222 ************************************************************
162P1E6V.2 GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3660 162PlE6v.ll GACGGGTGATGGGCAGATTGGAAAAAGCACCGCAGATGGGAACCTTAATCTTTCTTTTCT 3282 ************************************************************
162P1E6V.2 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3720 162P1E6V.11 AAAATTGATGCTATGAAAATTTGCGTTTTCTGTAACTTGTAAAAACTAAAAGTTGCTTGT 3342 ************************************************************
162P1E6V.2 CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3762 162PlE6v.ll CTACTGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 3384 ******************************************
Table LVI(A). Peptide sequences of protein coded by 162P1E6 v.2
MTNKEIVESF SRHILGRMWG HWRLSFLDKS LGVRTRSLTL LCPPTPMNGP GSSQELWFFL 60
SSSPISΞGFH IGKRGCKVLF V FGQCLVER NAHAPAFQGL GKQAQSSWIF LKQLQNTCFF 120
FVSSRKDQPH RAQLWHTQWD LDKGRG 146
Table LVI(B). Peptide sequences of protein coded by 162P1E6 v.3
LKWAESLLLT LDLEKPVSLL LSVTNLYSKN SAQFSTILQT LSFPATFTPS PSIPLSSAYF 60
FFFSDRVSLC RPGRSAVAQS WAHCSLNLPE AGFHHVAQTG LELLSLSNPP ASASQSVGIT 120 GVSHRIRPHV LFH 133
Table LVI(C). Peptide sequences of protein coded by 162P1E6 v.4
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRTLS SLKYPSWRVR TPHEDFSGVK 60 FRRHGADNHE ASAATATTAA ATTVAAAAAA AAAAAAARVT LT 102
Table LVI(D). Peptide sequences of protein coded by 162P1E6 v.5
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRKGP TTPSSVMAHT VGPRQRERVT 60 DIPTRFQWSE VQEAWS 76
Table LVI(E). Peptide sequences of protein coded by 162P1E6 v.6
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRTLS SLKYPSWRVR TPHEERTNHT 60 ELSYGTHSGT 70
Table LVI(F). Peptide sequences of protein coded by 162P1E6 v.7
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRTLS SLKYPSWRVR TPHEDFSGVK 60 FRRHGADNHE ASAATATTAA ATTVAAAAAA AAAAAAARVT LT 102
Table LVI(G). Peptide sequences of protein coded by 162P1E6 v.8
MFFFIKERNQ LFRTGPHLSS GVISVPHRPA ELGALYRTLS SLKYPSWRVR TPHEERTNHT 60 ELSYGTHSGT 70
Table LVI(H). Peptide sequences of protein coded by 162P1E6 v.9
MTNKEIVESF SRHILGRMWG HWRLSFLDKS LGVRTRSLTL LCPPTPMNGP GSSQELWFFL 60 SSSPISSGFH IGKRGCKVLF VLFGQCLVER NAHAPAFQGL GKQAQSSWIF LKQLQNTCFF 120
FVSSRKDQPH RAQLWHTQWD LDKGRG 146
Table LVI(I). Peptide sequences of protein coded by 162P1E6 v.10 MTNKEIVESF SRHILGRMWG HWRLSFLDKS LGVRTRSLTL LCPPTPMNGP GSSQELWFFL 60
SSSPISSGFH IGKRGCKVLF VLFGQCLVER NAHAPAFQGL GKQAQSSWIF LKQLQNTCFF 120
FVSSRKDQPH RAQLWHTQWD LDKGRG 146
Table LVI(J). Peptide sequences of protein coded by 162P1E6 v.ll
MTNKEIVESF SRHILGRMWG HWRLSFLDKS LGVRTRSLTL LCPPTPMNGP GSSQELWFFL 60
SSSPISSGFH IGKRGCKVLF VLFGQCLVER NAHAPAFQGL GKQAQSSWIF LKQLQNTCFF 120
FVSSRKDQPH RAQLWHTQWD LDKGRG 146 Table LVII(A). Amino acid sequence alignment of 121P1F1 v.l and 162P1E6 v.2
162P1E6V.1 MTNKEIVESFSRHILGRrWGHWRLSFLDKSLGTOTRSLTLLCPPTP NGPGSSQELWFFL 60 162ple6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 ************************************************************
162P1E6V.1 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 162ple6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 ************************************************************
162P1E6V.1 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 **************************
Table LVII(B). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.3
162ple6V.2 -MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFF 59 162ple6V.3 LKWAESLLLTLDLEKPVSLLLSVTNLYSKNSAQFSTILQTLSFPATFTPSPSIPLSSAYF 60 :.:: ::. . : : .:* . * ** *.* .*. . . :*
162ple6V.2 LSSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCF 119 162ple6V.3 FFFSDRVSLCRPGRSAVAQSWAHCSLNLPEAGFHHVAQTGLELLSLS--NPPASASQSVG 118 * * . *. . * * * * ** * *
162ple6V.2 FFVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.3 ITGVSHRIRPH--VLFH 133
Table LVH(C). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.4
162ple6V.2 -MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFF 59
162ple6V.4 MFFFIKERNQLFRTGPHLSSGVISVPHRPAELGALYRTLSSLKYPS WRV 49
: : :.: * * :.. .**. *;*; * *; * ,
162ple6V.2 LSSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCF 119 162ple6V.4 RTPHEDFSGVKFRRHGADNHEASAATATTAAATTVAAAAAAAAAAAAARVTLT 102
162ple6V.2 FFVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.4
Table LVH(D). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.5
162ple6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 162plβ6V.5 MF--FFIKERNQLFRTGPHLSSGVISVPHRPAELGA LY 36
162ple6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 162ple6V.5 RKGPTTP SSVMAHTVGPRQRERVTDIP TRFQWS EVQ 72
162plβ6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.5 ESMS 76
Table LVII(E). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.6
162ple6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 162ple6V.6 MFFFI 5
. ** .
162ple6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120
162ple6V.6 KER--NQLFRTGPH LSSGVIS-VPHRP--AELG ALYRTLSSLK 43
. . * . * . . . . * * ** • • * * . 162ple6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG- 146 162ple6V.6 YPSWRVRTPHEERTNHTELSYGTHSGT 70 . * * ** _ . **: . .. * Table LVII(F). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.7
5 162ple6V.2 -MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFF 59
162ple6V.7 MFFFIKERNQLFRTGPHLSSGVISVPHRPAELGALYRTLSSLKYPS WRV 49
: : :.: * * :.. .**. *:*: * *: * .
162ple6V.2 LSSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCF 119 10 162ple6V.7 RTPHEDFSGVKFRRHGADNHEASAATATTAAATTVAAAAAAAAAAAAARVTLT 102
**
162ple6V.2 FFVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.7
15
Table LVπ(G). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.8
20 162ple6V.2 MTNKEIVESFSRH1LGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60
162ple6V.8 FFFI 5
. . ** .
162ple6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVER AHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120
25 162ple6V.8 KER- -NQLFRTGPH LSSGVIS-VPHRP--AELG ALYRTLSSLK 43
. . * : * : : . . : : . . * * ** : : * . . * :
162ple6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG- 146 162ple6V.8 YPSWRVRTPHEERTNHTELSYGTHSGT 70 30 : ** **. : **: . .. *
Table LVH(H). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.9
35 162P1E6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 162ple6V.9 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 ************************************************************
162P1E6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 40 162ple6V.9 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120
************************************************************
162P1E6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.9 FVSSRKDQPHRAQLWHTQWDLDKGRG 146
45 **************************
Table LVH(I). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.10
50 162P1E6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 162ple6V.10 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 ************************************************************
162P1E6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 55 162ple6V.10 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSW1FLKQLQNTCFF 120
************************************************************
162P1E6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.10 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 o0 **************************
Table LVH(J). Amino acid sequence alignment of 121P1F1 v.2 and 162P1E6 v.ll
65 162P1E6V.2 MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTPMNGPGSSQELWFFL 60 162ple6V.ll MTNKEIVESFSRHILGRMWGHWRLSFLDKSLGVRTRSLTLLCPPTP NGPGSSQELWFFL 60 ************************************************************
162P1E6V.2 SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120 70 162ple6V.ll SSSPISSGFHIGKRGCKVLFVLFGQCLVERNAHAPAFQGLGKQAQSSWIFLKQLQNTCFF 120
************************************************************ 162P1E6V.2 FVSSRKDQPHRAQLWHTQWDLDKGRG 146 162ple6V.ll FVSSRKDQPHRAQLWHTQWDLDKGRG 146
**************************

Claims (51)

CLAIMS:
1. A composition comprising: a substance that a) modulates the status of a protein of Figure 2 (SEQ ID NOS: ), or b) a molecule that is modulated by a protein of Figure 2, whereby the status of a cell that expresses a protein of Figure 2 is modulated.
2. A composition of claim 1, further comprising a physiologically acceptable carrier.
3. A pharmaceutical composition that comprises the composition of claim 1 in a human unit dose form.
4. A composition of claim 1 wherein the substance comprises an antibody or f agment thereof that specifically binds to a protein that is related to a protein of Figure 2.
5. An antibody or fragment thereof of claim 4, which is monoclonal.
6. An antibody of claim 4, which is a human antibody, a humanized antibody or a chimeric antibody.
7. A non-human transgenic animal that produces an antibody of claim 4.
8. A hybridoma that produces an antibody of claim 5.
9. A method of delivering a cytotoxic agent or a diagnostic agent to a cell that expresses a protein of Figure 2 (SEQ ID NOS: ), said method comprising: providing the cytotoxic agent or the diagnostic agent conjugated to an antibody or fragment thereof of claim 4; and, exposing the cell to the antibody-agent or fragment-agent conjugate.
10. A composition of claim 1 wherein the substance comprises a polynucleotide that encodes an antibody or fragment thereof, either of which immunospecifically bind to a protein of Figure 2.
11. A composition of claim 1 wherein the substance comprises a protein related to a protein of
Figure 2.
12. A protein of claim 11 that is at least 90% homologous to an entire amino acid sequence shown in Figure 2 (SEQ ID NOS: ).
13. A composition of claim 1 wherein the substance comprises: a) a peptide of eight, nine, ten, or eleven contiguous amino acids of a protein of Figure 2; b) a peptide of Tables V to XVIII (SEQ ID NOS: ); c) a peptide of Tables XXII to XLVII (SEQ ID NOS: ); or, d) a peptide of Tables XLVIII to LI (SEQ ID NOS: ).
14. A composition of claim 1 wherein the substance comprises a CTL polypeptide or an analog thereof, from the amino acid sequence of a protein of Figure 2 (SEQ ID NOS: ).
15. A composition of claim 14 further limited by a proviso that the epitope is not an entire amino acid sequence of Figure 2 (SEQ ID NOS:).
16. A composition of claim 14 wherein the substance comprises a CTL polypeptide set forth in Tables V to XVIII (SEQ ID NOS: ).
17. A composition of claim 16 further limited by a proviso that the polypeptide is not an entire amino acid sequence of a protein of Figure 2 (SEQ ID NOS: ).
18. A composition of claim 1 wherein the substance comprises an antibody polypeptide epitope from an amino acid sequence of Figure 2 (SEQ ID NOS: ).
19. A composition of claim 18 further limited by a proviso that the epitope is not an entire amino acid sequence of Figure 2 (SEQ ID NOS: ).
20. A composition of claim 18 wherein the antibody epitope comprises a peptide region of at least 5 amino acids of Figure 2 (SEQ ID NOS: ) in any whole number increment up to the end of said peptide, wherein the epitope comprises an amino acid position selected from: a) an amino acid position having a value greater than 0.5 in the Hydrophilicity profile of Figure 5, b) an amino acid position having a value less than 0.5 in the Hydropathicity profile of Figure 6; c) an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profile of Figure 7; d) an amino acid position having a value greater than 0.5 in the Average Flexibility profile of Figure 8; e) an amino acid position having a value greater than 0.5 in the Beta-turn profile of Figure 9; f) a combination of at least two of a) through e); g) a combination of at least three of a) through e); h) a combination of at least four of a) through e); or i) a combination of five of a) through e).
21. A composition of claim 20 further limited by a proviso that the epitope is not an entire amino acid sequence of Figure 2 (SEQ ID NOS: ).
22. A polynucleotide that encodes a protein of claim 11.
23. A polynucleotide of claim 22 that comprises a nucleic acid molecule set forth in Figure 2.
24. A polynucleotide of claim 22 further limited by a proviso that the encoded protein is not an entire amino acid sequence of Figure 2 (SEQ ID NOS: ).
25. A polynucleotide of claim 22 wherein T is substituted with U.
26. A composition of claim 1 wherein the substance comprises a polynucleotide that comprises a coding sequence of a nucleic acid sequence of Figure 2 (SEQ ID NOS: ).
27. A polynucleotide of claim 22 that further comprises an additional nucleotide sequence that encodes an additional protein of claim 11.
28. A composition comprising a polynucleotide that is fully complementary to a polynucleotide of claim 22.
29. A composition comprising a polynucleotide that is fiilly complementary to a polynucleotide of claim 25.
30. A composition comprising a polynucleotide that is fully complementary to a polynucleotide of claim 27.
31. A composition of claim 1 wherein the substance comprises a) a ribozyme that cleaves a polynucleotide having a 162P1E6 coding sequence, or b) a nucleic acid molecule that encodes the ribozyme; and, a physiologically acceptable carrier.
32. A composition of claim 1 wherein the substance comprises human T cells, wherein said T cells specifically recognize a 162P1E6 peptide subsequence in the context of a particular HLA molecule.
33. A method of inhibiting growth of cancer cells that express a protein of Figure 2, the method comprising: administering to the cells the composition of claim 1.
34. A method of claim 33 of inhibiting growth of cancer cells that express a protein of Figure 2, the method comprising steps of: administering to said cells an antibody or fragment thereof, either of which specifically bind to a 162PlE6-related protein.
35. A method of claim 33 of inhibiting growth of cancer cells that express a protein of Figure 2, the method comprismg steps of: administering to said cells a 162PlE6-related protein.
36. A method of claim 33 of inhibiting growth of cancer cells that express a protein of Figure 2, the method comprising steps of: administering to said cells a polynucleotide comprising a coding sequence for a 162PlE6-related protein or comprising a polynucleotide complementary to a coding sequence for a 162PlE6-related protein.
37. A method of claim 33 of inhibiting growth of cancer cells that express a protein of Figure 2, the method comprising steps of: administering to said cells a ribozyme that cleaves a polynucleotide that encodes a protein of Figure 2.
38. A method of claim 33 of inhibiting growth of cancer cells that express a protein of Figure 2 and a particular HLA molecule, the method comprising steps of: administering human T cells to said cancer cells, wherein said T cells specifically recognize a peptide subsequence of a protein of Figure 2 while the subsequence is in the context of the particular HLA molecule.
39. A method of claim 33, the method comprising steps of: administering a vector that delivers a nucleotide that encodes a single chain monoclonal antibody, whereby the encoded single chain antibody is expressed intracellularly within cancer cells that express a protein of Figure 2.
40. A method of generating a mammalian immune response directed to a protein of Figure 2, the method comprising: exposing cells of the mammal's immune system to a portion of a) a 162PlE6-related protein and/or b) a nucleotide sequence that encodes said protehi, whereby an immune response is generated to said protein.
41. A method of generating an immune response of claim 40, said method comprising: providing a 162PlE6-related protein that comprises at least one T cell or at least one B cell epitope; and, contacting the epitope with a mammalian immune system T cell or B cell respectively, whereby the T cell or B cell is activated.
42. A method of claim 41 wherein the immune system cell is a B cell, whereby the induced B cell generates antibodies that specifically bind to the 162PlE6-related protein.
43. A method of claim 41 wherein the immune system cell is a T cell that is a cytotoxic T cell (CTL), whereby the activated CTL kills an autologous cell that expresses the 162PlE6-related protein.
44. A method of claim 41 wherein the immune system cell is a T cell that is a helper T cell (HTL), whereby the activated HTL secretes cytokines that facilitate the cytotoxic activity of a cytotoxic T cell (CTL) or the antibody-producing activity of a B cell.
45. A method for detecting, in a sample, the presence of a 162PlE6-related protein or a 162P1E6- related polynucleotide, comprising steps of: contacting the sample with a substance of claim 1 that specifically binds to the 162PlE6-related protein or to the 162PlE6-related polynucleotide, respectively; and, determining that there is a complex of the substance with the 162PlE6-related protein or the substance with the 162PlE6-related polynucleotide, respectively.
46. A method of claim 45 for detecting the presence of a 162PlE6-related protein in a sample comprising steps of: contacting the sample with an antibody or fragment thereof either of which specifically bind to the
162PlE6-related protein; and, determining that there is a complex of the antibody or fragment thereof and the 162PlE6-related protein.
47. A method of claim 45 further comprising a step of: taking the sample from a patient who has or who is suspected of having cancer.
48. A method of claim 45 for detecting the presence of a protein of Figure 2 mRNA in a sample comprising: producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using 162P1E6 polynucleotides as sense and antisense primers, wherein the 162P1E6 polynucleotides used as the sense and antisense primers serve to amplify a 162P1E6 cDNA; and, detecting the presence of the amplified 162P1E6 cDNA.
49. A method of claim 45 for monitoring one or more 162P1E6 gene products in a biological sample from a patient who has or who is suspected of having cancer, the method comprising: determining the status of one or more 162P1E6 gene products expressed by cells in a tissue sample from an individual; comparing the status so determined to the status of one or more 162P1E6 gene products in a corresponding normal sample; and, identifying the presence of one or more aberrant gene products of 162P1E6 in the sample relative to the normal sample.
50. The method of claim 49 fiirther comprising a step of determining if there are one or more elevated gene products of a 162P1E6 mRNA or a 162P1E6 protein, whereby the presence of one or more elevated gene products in the test sample relative to the normal tissue sample indicates the presence or status of a cancer.
51. A method of claim 50 wherein the cancer occurs in a tissue set forth in Table I.
AU2002305169A 2001-04-10 2002-04-09 Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer Ceased AU2002305169C1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2007237282A AU2007237282B2 (en) 2001-04-10 2007-11-30 Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US28311201P 2001-04-10 2001-04-10
US60/283,112 2001-04-10
US28663001P 2001-04-25 2001-04-25
US60/286,630 2001-04-25
PCT/US2002/011544 WO2002083916A2 (en) 2001-04-10 2002-04-09 Nucleic acid and corresponding protein entitled 162p1e6 useful in treatment and detection of cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2007237282A Division AU2007237282B2 (en) 2001-04-10 2007-11-30 Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer

Publications (3)

Publication Number Publication Date
AU2002305169A1 true AU2002305169A1 (en) 2003-04-17
AU2002305169B2 AU2002305169B2 (en) 2007-09-13
AU2002305169C1 AU2002305169C1 (en) 2008-05-01

Family

ID=26961881

Family Applications (4)

Application Number Title Priority Date Filing Date
AU2002258626A Ceased AU2002258626B2 (en) 2001-04-10 2002-03-25 Nucleid acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer
AU2002305169A Ceased AU2002305169C1 (en) 2001-04-10 2002-04-09 Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer
AU2002258785A Abandoned AU2002258785A1 (en) 2001-04-10 2002-04-09 Nucleic acid and corresponding protein entitled 161p5c5 useful in treatment and detection of cancer
AU2007201445A Ceased AU2007201445B2 (en) 2001-04-10 2007-03-30 Nucleic acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2002258626A Ceased AU2002258626B2 (en) 2001-04-10 2002-03-25 Nucleid acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer

Family Applications After (2)

Application Number Title Priority Date Filing Date
AU2002258785A Abandoned AU2002258785A1 (en) 2001-04-10 2002-04-09 Nucleic acid and corresponding protein entitled 161p5c5 useful in treatment and detection of cancer
AU2007201445A Ceased AU2007201445B2 (en) 2001-04-10 2007-03-30 Nucleic acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer

Country Status (6)

Country Link
US (8) US20040003418A1 (en)
EP (3) EP1383922A4 (en)
AU (4) AU2002258626B2 (en)
CA (3) CA2442993C (en)
IL (2) IL158293A0 (en)
WO (3) WO2002083928A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1383922A4 (en) 2001-04-10 2005-03-30 Agensys Inc Nucleid acid and corresponding protein entitled 158p3d2 useful in treatment and detection of cancer
US7811575B2 (en) * 2001-04-10 2010-10-12 Agensys, Inc. Nucleic acids and corresponding proteins entitled 158P3D2 useful in treatment and detection of cancer
AR044058A1 (en) * 2003-04-22 2005-08-24 Avanir Pharmaceuticals PEPTIDIC MEDIATORS OF COLESTEROL INVEST TRANSPORTATION FOR THE TREATMENT OF HYPERCHOLESTEROLEMIA
GB0409748D0 (en) * 2004-04-30 2004-06-09 Chiron Srl Lactoferrin cleavage
EP1824877A1 (en) * 2004-11-19 2007-08-29 Agensys, Inc. Nucleic acids corresponding proteins entitled 158p3d2 useful in treatment and detection of cancer
US8449512B2 (en) 2010-04-09 2013-05-28 Davinci Biomedical Research Products Inc. Stoma stabilitating device and method
AU2015299753A1 (en) * 2014-08-04 2017-02-23 Christopher James CONNELL A well system
WO2016036352A1 (en) 2014-09-03 2016-03-10 Hewlett-Packard Development Company, L.P. Presentation of a digital image of an object
US20220025025A1 (en) * 2018-10-09 2022-01-27 Ibex Biosciences, Llc Antibodies directed to filamin-a and therapeutic uses thereof

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2153480A1 (en) 1993-11-12 1995-06-01 Kenichi Matsubara Gene signature
US5599677A (en) * 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
DE69841894D1 (en) * 1997-11-21 2010-10-21 Merck Serono Biodevelopment Sa Outer membrane polypeptide of Chlamydia pneumoniae and fragments thereof and their use, in particular for the diagnosis, prevention and treatment of an infection
KR20010086224A (en) 1998-01-23 2001-09-10 추후제출 Meth1 and meth2 polynucleotides and polypeptides
DE19813839A1 (en) 1998-03-20 1999-09-23 Metagen Gesellschaft Fuer Genomforschung Mbh Human nucleic acid sequences and protein products from tumor breast tissue, useful for breast cancer therapy
AU3371199A (en) 1998-04-09 1999-11-01 Whitehead Institute For Biomedical Research Biallelic markers
AU3896699A (en) * 1998-05-07 1999-11-23 Regents Of The University Of California, The Use of neglected target tissue antigens in modulation of immune responses
KR20030000011A (en) 1999-05-25 2003-01-03 휴먼 게놈 사이언시즈, 인크. Meth1 and meth2 polynucleotides and polypeptides
WO2001009317A1 (en) * 1999-07-29 2001-02-08 Helix Research Institute Stomach cancer-associated gene
EP1074617A3 (en) 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
EP1265582A2 (en) 1999-09-29 2002-12-18 Human Genome Sciences, Inc. Colon and colon cancer associated polynucleotides and polypeptides
CN1296967A (en) * 1999-11-23 2001-05-30 上海博容基因开发有限公司 Polypeptide-human deaf related gene 14 and polynucleotide for coding said polypeptide
WO2001042467A2 (en) 1999-12-08 2001-06-14 Millennium Pharmaceuticals, Inc. Genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
WO2001051628A2 (en) 2000-01-14 2001-07-19 Millennium Pharmaceuticals, Inc. Genes compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
WO2001055325A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
US20020132767A1 (en) * 2000-01-31 2002-09-19 Rosen Craig A. Nucleic acids, proteins, and antibodies
EP1290217A2 (en) 2000-02-04 2003-03-12 Aeomica, Inc. Methods and apparatus for predicting, confirming, and displaying functional information derived from genomic sequence
US20030165831A1 (en) * 2000-03-21 2003-09-04 John Lee Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of ovarian cancer
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
CA2411404A1 (en) 2000-05-26 2001-12-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
AU2001270082A1 (en) 2000-06-21 2002-01-02 Hitachi Chemical Co. Ltd. Gene markers for lung cancer
US20030165843A1 (en) 2000-07-28 2003-09-04 Avi Shoshan Oligonucleotide library for detecting RNA transcripts and splice variants that populate a transcriptome
CA2427858A1 (en) * 2000-11-03 2002-05-10 University Of Vermont And State Agricultural College Compositions for inhibiting grb7
AU2002254482A1 (en) * 2001-03-19 2002-10-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of kidney cancer
EP1383922A4 (en) 2001-04-10 2005-03-30 Agensys Inc Nucleid acid and corresponding protein entitled 158p3d2 useful in treatment and detection of cancer
US20040076955A1 (en) * 2001-07-03 2004-04-22 Eos Biotechnology, Inc. Methods of diagnosis of bladder cancer, compositions and methods of screening for modulators of bladder cancer
JP2003135075A (en) * 2001-11-05 2003-05-13 Research Association For Biotechnology NEW FULL-LENGTH cDNA

Similar Documents

Publication Publication Date Title
US7928196B2 (en) Nucleic acid and corresponding protein entitled 125P5C8 useful in treatment and detection of cancer
US7879570B2 (en) Nucleic acid and corresponding protein entitled 184P1E2 useful in treatment and detection of cancer
US7923543B1 (en) Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer
WO2002014361A2 (en) NUCLEIC ACIDS AND CORRESPONDING PROTEINS ENTITLED 83P2H3 AND CaTrF2E11 USEFUL IN TREATMENT AND DETECTION OF CANCER
AU2002305169A1 (en) Nucleic acid and corresponding protein entitled 162P1E6 useful in treatment and detection of cancer
AU2002258626A1 (en) Nucleid acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer
EP2311863A1 (en) Nucleic acid and corresponding protein entitled 121P1F1 useful in treatment and detection of cancer
US8647826B2 (en) Nucleic acid and corresponding protein entitled 125P5C8 useful in treatment and detection of cancer
AU2002324842A1 (en) 205P1B5 in treatment and detection of cancer
WO2003020954A2 (en) 205p1b5 in treatment and detection of cancer
EP1790662A1 (en) Nucleic acid and corresponding protein entitled 121P2A3 useful in treatment and detection of cancer
AU2002307250A1 (en) Nucleic acid and corresponding protein entitled 121P2A3 useful in treatment and detection of cancer
WO2003085081A2 (en) Nucleic acid and corresponding protein entitled 238p1b2 useful in treatment and detection of cancer