US20090022663A1 - Antibodies and related molecules that bind to 161p2f10b proteins - Google Patents

Antibodies and related molecules that bind to 161p2f10b proteins Download PDF

Info

Publication number
US20090022663A1
US20090022663A1 US12/196,039 US19603908A US2009022663A1 US 20090022663 A1 US20090022663 A1 US 20090022663A1 US 19603908 A US19603908 A US 19603908A US 2009022663 A1 US2009022663 A1 US 2009022663A1
Authority
US
United States
Prior art keywords
161p2f10b
antibody
protein
seq
cancer
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.)
Abandoned
Application number
US12/196,039
Other languages
English (en)
Inventor
Aya Jakobovits
Steven B. Kanner
Pia M. Challita-Eid
Juan J. Perez-villar
Daulet Satpaev
Arthur B. Raitano
Robert Kendall Morrison
Karen Jane Meyrick Morrison
Xiao-Chi Jia
Jean Gudas
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
Priority claimed from US11/396,178 external-priority patent/US7427399B2/en
Application filed by Agensys Inc filed Critical Agensys Inc
Priority to US12/196,039 priority Critical patent/US20090022663A1/en
Assigned to AGENSYS, INC. reassignment AGENSYS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHALLITA-EID, PIA M., GUDAS, JEAN, JAKOBOVITS, AYA, JIA, XIAO-CHI, KANNER, STEVEN B., MORRISON, KAREN JANE MEYRICK, MORRISON, ROBERT KENDALL, PEREZ-VILLAR, JUAN J., RAITANO, ARTHUR B., SATPAEV, DAULET
Publication of US20090022663A1 publication Critical patent/US20090022663A1/en
Priority to US12/433,760 priority patent/US7811565B2/en
Priority to US12/757,935 priority patent/US8350009B2/en
Priority to US12/881,461 priority patent/US8236310B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the invention described herein relates to antibodies, as well as binding fragments thereof and molecules engineered therefrom, that bind proteins, termed 161P2F10B.
  • the invention further relates to diagnostic, prognostic, prophylactic and therapeutic methods and compositions useful in the treatment of cancers that express 161P2F10B.
  • 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.
  • carcinomas of the lung, prostate, breast, colon, pancreas, ovary, and bladder 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.
  • 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.
  • PSA serum prostate specific antigen
  • the LAPC Los Angeles Prostate Cancer
  • SCID severe combined immune deficient mice
  • 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.
  • Renal cell carcinoma 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.
  • 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 eight 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.
  • 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.
  • TUR transurethral resection of the bladder
  • 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.
  • 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.
  • 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.
  • lumpectomy local removal of the tumor
  • mastectomy surgical removal of the breast
  • radiation therapy chemotherapy
  • hormone therapy chemotherapy
  • 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.
  • DCIS ductal carcinoma in situ
  • 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).
  • the fallopian tubes salivary-oophorectomy
  • the uterus hematoma-oophorectomy
  • pancreatic 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.
  • mice are convenient for immunization and recognize most human antigens as foreign, mAbs against human targets with therapeutic potential have typically been of murine origin.
  • murine mAbs have inherent disadvantages as human therapeutics. They require more frequent dosing as mAbs have a shorter circulating half-life in humans than human antibodies.
  • HAMA human anti-mouse antibody
  • Such a HAMA response may result in allergic reaction and the rapid clearing of the murine antibody from the system thereby rendering the treatment by murine antibody useless.
  • attempts to create human immune systems within mice have been attempted.
  • the invention provides antibodies as well as binding fragments thereof and molecules engineered therefrom, that bind to 161P2F10B proteins and polypeptide fragments of 161P2F10B proteins.
  • the invention comprises 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.
  • there is a proviso that the entire nucleic acid sequence of FIG. 3 is not encoded and/or the entire amino acid sequence of FIG. 2 is not prepared.
  • the entire nucleic acid sequence of FIG. 3 is encoded and/or the entire amino acid sequence of FIG. 2 is prepared, either of which are in respective human unit dose forms.
  • the invention further provides methods for detecting the presence and status of 161P2F10B polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express 161P2F10B.
  • An embodiment of this invention provides methods for monitoring 161P2F10B 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 161P2F10B such as cancers of tissues listed in Table I, including therapies aimed at inhibiting the transcription, translation, processing or function of 161P2F10B as well as cancer vaccines.
  • the invention provides compositions, and methods comprising them, for treating a cancer that expresses 161P2F10B 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 161P2F10B.
  • the carrier is a uniquely human carrier.
  • the agent is a moiety that is immunoreactive with 161P2F10B 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.
  • the agent is a small molecule as defined herein.
  • FIG. 1 The cDNA (SEQ ID NO:1) and amino acid (SEQ ID NO:2) sequence of 161P2F10B variant 1 (also called “161P2F10B v.1” or “161P2F10B variant 1”) is shown in FIG. 1A .
  • the 3858 nucleotide sequence of 161P2F10B variant 1 is shown.
  • the start methionine is underlined.
  • the open reading frame extends from nucleic acid 44-2671 including the stop codon.
  • FIG. 1B The cDNA (SEQ ID NO:3) and amino acid (SEQ ID NO:4) sequence of 161P2F10B variant 2 (also called “161P2F10B v.2”) is shown in FIG. 1B .
  • the 3858 nucleotide sequence of 161P2F10B variant 2 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 44-2671 including the stop codon.
  • FIG. 1C The cDNA (SEQ ID NO:5) and amino acid (SEQ ID NO:6) sequence of 161P2F10B variant 3 (also called “161P2F10B v.3”) is shown in FIG. 1C .
  • the 3858 nucleotide sequence of 161P2F10B variant 3 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 44-2671 including the stop codon.
  • FIG. 1D The cDNA (SEQ ID NO:7) and amino acid (SEQ ID NO:8) sequence of 161P2F10B variant 4 (also called “161P2F10B v.4”) is shown in FIG. 1D .
  • the 3858 nucleotide sequence of 161P2F10B variant 4 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 44-2671 including the stop codon.
  • FIG. 1E The cDNA (SEQ ID NO:9) and amino acid (SEQ ID NO:10) sequence of 161P2F10B variant 5 (also called “161P2F10B v.5”) is shown in FIG. 1E .
  • the 3858 nucleotide sequence of 161P2F10B variant 5 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 44-2671 including the stop codon.
  • FIG. 1F The cDNA (SEQ ID NO:11) and amino acid (SEQ ID NO:12) sequence of 161P2F10B variant 6 (also called “161P2F10B v.6”) is shown in FIG. 1F .
  • the 3165 nucleotide sequence of 161P2F10B variant 6 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 84-2711 including the stop codon.
  • FIG. 1G The cDNA (SEQ ID NO:13) and amino acid (SEQ ID NO:14) sequence of 161P2F10B variant 7 (also called “161P2F10B v.7”) is shown in FIG. 1G .
  • the 3988 nucleotide sequence of 161P2F10B variant 7 is shown.
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 276-2801 including the stop codon.
  • FIG. 2 Nucleic Acid and Amino Acid sequences of 161P2F10b antibodies.
  • FIG. 2A The cDNA (SEQ ID NO:15) and amino acid (SEQ ID NO:16) sequence of H16-7.213 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2B The cDNA (SEQ ID NO:17) and amino acid (SEQ ID NO:18) sequence of H16-7.213 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2C The cDNA (SEQ ID NO:19) and amino acid (SEQ ID NO:20) sequence of H16-9.69 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2D The cDNA (SEQ ID NO:21) and amino acid (SEQ ID NO:22) sequence of H16-9.69 VL. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2E The cDNA (SEQ ID NO:23) and amino acid (SEQ ID NO:24) sequence of H16-1.52 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2F The cDNA (SEQ ID NO:25) and amino acid (SEQ ID NO:26) sequence of H16-1.52 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2G The cDNA (SEQ ID NO:27) and amino acid (SEQ ID NO:28) sequence of Ha16-1(1)23 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2H The cDNA (SEQ ID NO:29) and amino acid (SEQ ID NO:30) sequence of Ha16-1(1)23 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2I The cDNA (SEQ ID NO:31) and amino acid (SEQ ID NO:32) sequence of Ha16-9.44 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2J The cDNA (SEQ ID NO:33) and amino acid (SEQ ID NO:34) sequence of H16-9.44 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2K The cDNA (SEQ ID NO:35) and amino acid (SEQ ID NO:36) sequence of H16-1.67 VH.
  • FIG. 2L The cDNA (SEQ ID NO:37) and amino acid (SEQ ID NO:38) sequence of H16-1.67 VL. Underlined is the light chain constant region.
  • FIG. 2M The cDNA (SEQ ID NO:39) and amino acid (SEQ ID NO:40) sequence of Ha16-1(3,5)36 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2N The cDNA (SEQ ID NO:41) and amino acid (SEQ ID NO:42) sequence of Ha16-1(3,5)36 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2O The cDNA (SEQ ID NO:43) and amino acid (SEQ ID NO:44) sequence of H16-1.86 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2P The cDNA (SEQ ID NO:45) and amino acid (SEQ ID NO:46) sequence of H16-1.86 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2Q The cDNA (SEQ ID NO:47) and amino acid (SEQ ID NO:48) sequence of Ha16-9.10 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2R The cDNA (SEQ ID NO:49) and amino acid (SEQ ID NO:50) sequence of H16-9.10 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2S The cDNA (SEQ ID NO:51) and amino acid (SEQ ID NO:52) sequence of H16-9.33 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2T The cDNA (SEQ ID NO:53) and amino acid (SEQ ID NO:54) sequence of H16-9.33 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2U The cDNA (SEQ ID NO:55) and amino acid (SEQ ID NO:56) sequence of H16-1.68 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2V The cDNA (SEQ ID NO:57) and amino acid (SEQ ID NO:58) sequence of H16-1.68 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2W The cDNA (SEQ ID NO:59) and amino acid (SEQ ID NO:60) sequence of Ha16-1(1)11 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2X The cDNA (SEQ ID NO:61) and amino acid (SEQ ID NO:62) sequence of Ha16-1(1)11 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2Y The cDNA (SEQ ID NO:63) and amino acid (SEQ ID NO:64) sequence of Ha16-1(3,5)18 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2Z The cDNA (SEQ ID NO:65) and amino acid (SEQ ID NO:66) sequence of Ha16-1(3,5)18 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2 AA The cDNA (SEQ ID NO:67) and amino acid (SEQ ID NO:68) sequence of Ha16-1(2,4)4 VH.
  • FIG. 2 AB The cDNA (SEQ ID NO:69) and amino acid (SEQ ID NO:70) sequence of Ha16-1(2,4)4 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2 AC The cDNA (SEQ ID NO:71) and amino acid (SEQ ID NO:72) sequence of Ha16-1(3,5)56 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AD The cDNA (SEQ ID NO:73) and amino acid (SEQ ID NO:74) sequence of Ha16-1(3,5)56 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2 AE The cDNA (SEQ ID NO:75) and amino acid (SEQ ID NO:76) sequence of H16-1.93 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2 AF The cDNA (SEQ ID NO:77) and amino acid (SEQ ID NO:78) sequence of H16-1.93 VL. Double-underlined is the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AG The cDNA (SEQ ID NO:79) and amino acid (SEQ ID NO:80) sequence of H16-7.8 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2 AH The cDNA (SEQ ID NO:81) and amino acid (SEQ ID NO:82) sequence of H16-7.8 VL. Double-underlined is the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AI The cDNA (SEQ ID NO:83) and amino acid (SEQ ID NO:84) sequence of Ha16-1(3,5)27.1 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AJ The cDNA (SEQ ID NO:85) and amino acid (SEQ ID NO:86) sequence of Ha16-1(3,5)27 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2 AK The cDNA (SEQ ID NO:87) and amino acid (SEQ ID NO:88) sequence of H16-1.61 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AL The cDNA (SEQ ID NO:89) and amino acid (SEQ ID NO:90) sequence of H16-1.61 VL. Underlined is a portion of the light chain constant region.
  • FIG. 2 AM The cDNA (SEQ ID NO:91) and amino acid (SEQ ID NO:92) sequence of H16-1(3,5)5 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2 AN The cDNA (SEQ ID NO:93) and amino acid (SEQ ID NO:94) sequence of H16-1(3,5)5 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AO
  • FIG. 2 AP The cDNA (SEQ ID NO:97) and amino acid (SEQ ID NO:98) sequence of H16-7.200 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AQ The cDNA (SEQ ID NO:99) and amino acid (SEQ ID NO:100) sequence of Ha16-1(3,5)42 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2 AR The cDNA (SEQ ID NO:101) and amino acid (SEQ ID NO:102) sequence of Ha16-1(3,5)42 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AS The cDNA (SEQ ID NO:103) and amino acid (SEQ ID NO:104) sequence of H16-9.65 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 2 AT The cDNA (SEQ ID NO:105) and amino acid (SEQ ID NO:106) sequence of H16-9.65 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AU The cDNA (SEQ ID NO:107) and amino acid (SEQ ID NO:108) sequence of H16-1.29 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AV The cDNA (SEQ ID NO:109) and amino acid (SEQ ID NO:110) sequence of H16-3.4 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AW The cDNA (SEQ ID NO:111) and amino acid (SEQ ID NO:112) sequence of H16-1.92 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 2 AX The cDNA (SEQ ID NO:113) and amino acid (SEQ ID NO:114) sequence of Ha16-1(3,5)19 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AY The cDNA (SEQ ID NO:169) and amino acid (SEQ ID NO:170) sequence of Ha16-1(3,5)19 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AZ The cDNA (SEQ ID NO:171) and amino acid (SEQ ID NO:172) sequence of Ha16-1.80 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 2 AAA The cDNA (SEQ ID NO:173) and amino acid (SEQ ID NO:174) sequence of Ha16-1.80 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 Amino acid sequences of 161P2F10B antibodies.
  • FIG. 3A The amino acid sequence (SEQ ID NO:115) of H16-7.213 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3B The amino acid sequence (SEQ ID NO:116) of H16-7.213 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3C The amino acid sequence (SEQ ID NO:117) of H16-9.69 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3D The amino acid sequence (SEQ ID NO:118) of H16-9.69 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3E The amino acid sequence (SEQ ID NO:119) of H16-1.52 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3F The amino acid sequence (SEQ ID NO:120) of H16-1.52 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3G The amino acid sequence (SEQ ID NO:121) of Ha16-1(1)23 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3H The amino acid sequence (SEQ ID NO:122) of Ha16-1(1)23 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3I The amino acid sequence (SEQ ID NO:123) of H16-9.44 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3J The amino acid sequence (SEQ ID NO:124) of H16-9.44 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3K The amino acid sequence (SEQ ID NO:125) of H16-1.67 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3L The amino acid sequence (SEQ ID NO:126) of H16-1.67 VL. Underlined is the light chain constant region.
  • FIG. 3M The amino acid sequence (SEQ ID NO:127) of Ha16-1(3,5)36 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3N The amino acid sequence (SEQ ID NO:128) of Ha16-1(3,5)36 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3O The amino acid sequence (SEQ ID NO:129) of H16-1.86 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3P The amino acid sequence (SEQ ID NO:130) of H16-1.86 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3Q The amino acid sequence (SEQ ID NO:131) of H16-9.10 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3R The amino acid sequence (SEQ ID NO:132) of H16-9.10 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3S The amino acid sequence (SEQ ID NO:133) of H16-9.33 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3T The amino acid sequence (SEQ ID NO:134) of H16-9.33 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3U The amino acid sequence (SEQ ID NO:135) of H16-1.68 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3V The amino acid sequence (SEQ ID NO:136) of H16-1.68 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3W The amino acid sequence (SEQ ID NO:137) of Ha16-1(1)11 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3X The amino acid sequence (SEQ ID NO:138) of Ha16-1(1)11 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3Y The amino acid sequence (SEQ ID NO:139) of Ha16-1(3,5)18 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3Z The amino acid sequence (SEQ ID NO:140) of Ha16-1(3,5)18 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3 AA The amino acid sequence (SEQ ID NO:141) of Ha16-1(2,4)4 VH.
  • FIG. 3 AB The amino acid sequence (SEQ ID NO:142) of Ha16-1(2,4)4 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3 AC The amino acid sequence (SEQ ID NO:143) of Ha16-1(3,5)56 VH.
  • FIG. 3 AD The amino acid sequence (SEQ ID NO:144) of Ha16-1(3,5)56 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3 AE The amino acid sequence (SEQ ID NO:145) of H16-7.8 VH. Double-underlined is the leader sequence. Underlined is a portion of the light chain constant region.
  • FIG. 3 AF The amino acid sequence (SEQ ID NO:146) of H16-7.8 VL. Double-underlined is the leader sequence. Underlined is a portion of the light chain constant region.
  • FIG. 3 AG The amino acid sequence (SEQ ID NO:147) of H16-1.93 VH. Double-underlined is the leader sequence. Underlined is a portion of the light chain constant region.
  • FIG. 3 AH The amino acid sequence (SEQ ID NO:148) of H16-1.93 VL. Double-underlined is part of the leader sequence. Underlined is a portion of the light chain constant region.
  • FIG. 3 AI The amino acid sequence (SEQ ID NO:149) of Ha16-1(3,5)27.1 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 3 AJ The amino acid sequence (SEQ ID NO:150) of Ha16-1(3,5)27 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3 AK The amino acid sequence (SEQ ID NO:151) of H16-1.61 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3 AL The amino acid sequence (SEQ ID NO:152) of H16-1.61 VL. Underlined is a portion of the light chain constant region.
  • FIG. 3 The amino acid sequence (SEQ ID NO:153) of H16-1(3,5)5 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 3 AN The amino acid sequence (SEQ ID NO:154) of H16-1(3,5)5 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AO
  • FIG. 3 AP The amino acid sequence (SEQ ID NO:156) of H16-7.200 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AQ The amino acid sequence (SEQ ID NO:157) of Ha16-1(3,5)42 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 3 AR The amino acid sequence (SEQ ID NO:158) of Ha16-1(3,5)42 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AS The amino acid sequence (SEQ ID NO:159) of H16-9.65 VH. Double-underlined is the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 3 AT The amino acid sequence (SEQ ID NO:160) of H16-9.65 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AU The amino acid sequence (SEQ ID NO:161) of H16-1.29 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the heavy chain constant region.
  • FIG. 3 AV The amino acid sequence (SEQ ID NO:162) of H16-3.4 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3 AW The amino acid sequence (SEQ ID NO:163) of H16-1.92 VH. Underlined is a portion of the heavy chain constant region.
  • FIG. 3 AX The amino acid sequence (SEQ ID NO:164) of Ha16-1(3,5)19 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AY The amino acid sequence (SEQ ID NO:175) of Ha16-1(3,5)19 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AZ The amino acid sequence (SEQ ID NO:176) of Ha16-1.80 VL. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 3 AAA The amino acid sequence (SEQ ID NO:177) of Ha16-1.80 VH. Double-underlined is part of the leader sequence, and underlined is a portion of the light chain constant region.
  • FIG. 4 Alignment of 161P2F10B antibodies Heavy Chain Variable Region to germline V-D-J Sequences.
  • FIG. 4A Alignment of H16-7.213 Heavy Chain Variable Region to human germline VH1-2/D3-9/JH4
  • FIG. 4B Alignment of H16-9.69 Heavy Chain Variable Region to human germline VH1-8/D3-10/JH4
  • FIG. 4C Alignment of H16-1.52 Heavy Chain Variable Region to human germline VH3-23/6-19/JH4
  • FIG. 4D Alignment of Ha16-1(1)23 Heavy Chain Variable Region to human germline VH3-33/D3-10/JH6
  • FIG. 4E Alignment of H16-9.44 Heavy Chain Variable Region to human germline VH4-4/D3-22/JH4
  • FIG. 4F Alignment of H16-1.67 Heavy Chain Variable Region to human germline VH4-31/D3-10/JH6
  • FIG. 4G Alignment of Ha16-1(3,5)36 Heavy Chain Variable Region to human germline VH4-39/D6-19/JH4
  • FIG. 4H Alignment of H16-1.86 Heavy Chain Variable Region to human germline VH4-59/D1-26/JH6
  • FIG. 4I Alignment of H16-9.10 Heavy Chain Variable Region to human germline VH6-1/D6-19/JH5
  • FIG. 4J Alignment of H16-9.33 Heavy Chain Variable Region to human germline VH6-1/D6-19/JH5
  • FIG. 4K Alignment of Ha16-1(1)11 Heavy Chain Variable Region to human germline VH4-59/D4-23/JH6
  • FIG. 4L Alignment of Ha16-1(3,5)18 Heavy Chain Variable Region to human germline VH4-4/D4-17/JH6
  • FIG. 4M Alignment of Ha16-1(2,4)4 Heavy Chain Variable Region to human germline VH3-33/D1-26/JH6
  • FIG. 4N Alignment of Ha16-1(3,5)56 Heavy Chain Variable Region to human germline VH5-51/D1-26/JH6
  • FIG. 4O Alignment of H16-7.8 Heavy Chain Variable Region to human germline VH4-31/D5-12/JH6
  • FIG. 4P Alignment of H16-1.68 Heavy Chain Variable Region to human germline VH3-33/D3-3/JH6
  • FIG. 4Q Alignment of H16-1.93 Heavy Chain Variable Region to human germline VH1-18/D6-13/JH6
  • FIG. 4R Alignment of Ha16-1(3,5)27 Heavy Chain Variable Region to human germline VH5-51/D4-4/JH6
  • FIG. 4S Alignment of Ha16-1.61 Heavy Chain Variable Region to human germline VH3-33/D3-3/JH6
  • FIG. 4T Alignment of Ha16-1(3,5)5 Heavy Chain Variable Region to human germline VH5-51/D3-10/JH6
  • FIG. 4U Alignment of H16-7.200 Heavy Chain Variable Region to human germline VH4-31/D4-23/JH4
  • FIG. 4V Alignment of Ha16-1(3,5)42 Heavy Chain Variable Region to human germline VH5-51/D4-11/JH6
  • FIG. 4W Alignment of H16-9.65 Heavy Chain Variable Region to human germline VH3-33/D1-26/JH4
  • FIG. 4X Alignment of H16-1.29 Heavy Chain Variable Region to human germline VH1-2/D5-12/JH6
  • FIG. 4Y Alignment of H16-3.4 Heavy Chain Variable Region to human germline VH4-31/D5-12/JH4
  • FIG. 4Z Alignment of H16-1.92 Heavy Chain Variable Region to human germline VH4-39/D4-11/JH3
  • FIG. 5 Alignment of 161P2F10B antibodies Light Chain Variable Region to germline V-D-J Sequences.
  • FIG. 5A Alignment of H16-7.213 Light Chain Variable Region to human germline VK-O2/JK4.
  • FIG. 5B Alignment of H16-9.69 Light Chain Variable Region to human germline VK-B3/JK1.
  • FIG. 5C Alignment of H16-1.52 Light Chain Variable Region to human germline B3/JK2.
  • FIG. 5D Alignment of Ha16-1(1)23 Light Chain Variable Region to human germline V1-20/JL2.
  • FIG. 5E Alignment of H16-9.44 Light Chain Variable Region to human germline L1/JK3.
  • FIG. 5F Alignment of H16-1.67 Light Chain Variable Region to human germline O2/JK1.
  • FIG. 5G Alignment of Ha16-1(3, 5)36 Light Chain Variable Region to human germline V1-16/JL2.
  • FIG. 5H Alignment of H16-1.86 Light Chain Variable Region to human germline O2/JK1.
  • FIG. 5I Alignment of H16-9.10 Light Chain Variable Region to human germline L5/JK4.
  • FIG. 5J Alignment of H16-9.33 Light Chain Variable Region to human germline L5/JK4.
  • FIG. 5K Alignment of Ha16-1(1)11 Light Chain Variable Region to human germline O2/JK3.
  • FIG. 5L Alignment of Ha16-1(3, 5)18 Light Chain Variable Region to human germline V1-19/JL2.
  • FIG. 5M Alignment of Ha16-1(2, 4)4 Light Chain Variable Region to human germline V2-1/JL2.
  • FIG. 5N Alignment of Ha16-1(3, 5)56 Light Chain Variable Region to human germline V1-4/JL2.
  • FIG. 5O Alignment of H16-7.8 Light Chain Variable Region to human germline A26/JK1.
  • FIG. 5P Alignment of H16-1.68 Light Chain Variable Region to human germline O2/JK4.
  • FIG. 5Q Alignment of H16-1.93 Light Chain Variable Region to human germline A20/JK3.
  • FIG. 5R Alignment of Ha16-1(3, 5)27 Light Chain Variable Region to human germline V1-4/JL2.
  • FIG. 5S Alignment of H16-1.61 Light Chain Variable Region to human germline 012/JK3.
  • FIG. 5T Alignment of Ha16-1(3, 5)5 Light Chain Variable Region to human germline V1-4/JL2.
  • FIG. 5U Alignment of H16-7.200 Light Chain Variable Region to human germline A19/JK5.
  • FIG. 5V Alignment of Ha16-1(3, 5)42 Light Chain Variable Region to human germline V1-4/JL2.
  • FIG. 5W Alignment of H16-9.65 Light Chain Variable Region to human germline A19/JK5.
  • FIG. 5X Alignment of Ha16-1(3, 5)19 Light Chain Variable Region to human germline V1-4/JL2.
  • FIG. 6 161P2F10B MAbs bind to CAKI-161P2F10B cells by FACS. FACS analysis was performed by using CAKI-neo as a negative control. The results show that 161P2F10B mAbs specifically bind to human 161P2F10B on CAKI-161P2F10B cells.
  • FIG. 7 161P2F10B MAbs bind to UG-K3 cells by FACS. FACS analysis was performed by using CAKI-neo as a negative control. The results show that 161P2F10B mAbs specifically bind to human 161P2F10B on UG-K3 cells.
  • FIG. 8 161P2F10B MAb Epitope grouping using UG-K3 cells. Binding of each of biotinylated 161P2F10B MAbs on the UG-K3 cells were competed with excess amount of each of antibodies, biotinylated antibodies were detected by streptavidin-PE. The cells were analyzed using FACScan. MFI values from FACS were used for data analysis. A shown in the table, cells are highlighted to indicate self-competition (100% competition), the MFI value in these cells are background control for each biotinylated antibody. Additionally, cells with no color indicate that the two antibodies compete each other (low MFI), cells highlighted in gray (high MFI) indicate that the two antibodies bind to two distinct epitopes. The results show the antibodies that have the same binding pattern bind to the same epitope among the antibodies and that there are 16 epitope groups within the antibodies tested.
  • FIG. 9 Domain mapping of 161P2F10B MAbs by immunoprecipitation.
  • Tag5 expression constructs encoding either the full extracellular domain (ECD) of 161P2F10B (amino acids 46-875), the somatomedin-b-like domain (amino acids 46-157), the catalytic domain (amino acids 158-558), or the catalytic and nuclease domain (amino acids 158-875) were transfected into 293T cells and cellular lysates were made. These lysates were then used for immunoprecipitation with the indicated 161P2F10B MAbs or H3-1.5 control MAb (10 ⁇ g of MAb and 100-200 ⁇ g of cell lysate).
  • the lanes shown in the figure represent: lane 1. Vector, lane 2. pTag5 161P2F10b WT full length ECD, lane 3. pTag5 161P2F10b (46-157) somatomedin-b-like domain, lane 4. pTtag5 161P2F10b (158-558) catalytic domain, and lane 5. pTag5 161P2F10b (158-875) catalytic+nuclease domain. The results show that this technique enables mapping of MAbs to 161P2F10B to distinct regions of the extracellular domain.
  • FIG. 10 161P2F10B MAb domain mapping.
  • 161P2F10B MAb domain mapping Presented on the right is a summary table of selected 161P2F10B MAbs, their relative affinity as determined by BiaCore analysis on the full length ECD (amino acids 46-875), their epitope group as determined by a FACS based MAb competition assay, and their epitope domain as determined by immunoprecipitation assay using cell lysates containing either the full length ECD, the somatomedin-b-like domain (amino acids 46-157), the catalytic domain (amino acids 158-558), or the catalytic and nuclease domain (amino acids 158-875).
  • a schematic of the 161P2F10B protein and assignment of the MAbs groups is a schematic of the 161P2F10B protein and assignment of the MAbs groups.
  • the presence of MAbs within a group was inferred by a BiaCore competition assay in which the ability of MAbs to bind simultaneously to the 161P2F10B protein is analyzed. Inability to bind simultaneously suggests the MAbs share the same or an overlapping epitope. Simultaneous binding suggests that the MAbs employed belong in different epitope groups and recognize distinct or non-overlapping regions.
  • FIG. 11 Cross reactivity of human MAbs on mouse 161P2F10B.
  • 293T-mouse 161P2F10B and 293T-neo cells were used to test cross-reactivity of human mAbs with mouse 161P2F10B by FACS.
  • 50 ul of MAbs at 2 ug/ml were incubated with 293T-mouse 161P2F10B or 293T-neo cells (50,000 cells/100 ⁇ l).
  • Antibodies bound on the cells were detected using anti-hIgG-PE and analyzed on FACS. Data were analyzed using CellQuest Pro software.
  • the solid purple line is data from negative control cells.
  • the green line is from 161P2F10B-positive cells.
  • FIG. 12 MAbs to 161P2F10B mediate saporin dependent killing in KU-812 cells.
  • KU-812 cells are a CML cell line that expresses high levels of endogenous 161P2F10B.
  • KU-812 cells (3000 cells/well) were seeded into a 96 well plate on day 1. The following day an equal volume of medium containing 2 ⁇ concentration of the indicated primary antibody together with a 2-fold excess of anti-human (Hum-Zap) polyclonal antibody conjugated with saporin toxin (Advanced Targeting Systems, San Diego, Calif.) was added to each well. The cells were allowed to incubate for 5 days at 37 degrees C.
  • FIG. 13 161P2F10B MAbs inhibit the growth of renal cell carcinoma.
  • FIG. 14 161P2F10B MAbs inhibit the growth of UG-K3 cells in SCID mice.
  • FIG. 15 161P2F10B MAbs inhibit human renal cancer xenograft.
  • FIG. 16 161P2F10B MAbs inhibit the growth of human renal cancer SKRC-01 in SCID mice.
  • FIG. 17 161P2F10B MAbs inhibit the growth of human renal cancer UG-K3 in SCID mice.
  • FIG. 18 161P2F10B MAbs inhibit the growth of human renal cancer UG-K3 in SCID mice.
  • FIG. 19 Combined 161P2F10B MAbs inhibit the growth of human renal cancer Ug-K3 in SCID mice.
  • For 161P2F10B MAb treatment three MAbs at 200 ⁇ g each were pooled together at each dosing. Animals were treated twice weekly for a total of 7 doses until study day 27. Tumor growth was monitored using caliper measurements every 3 to 4 days as indicated. The results show that combination treatment with a cocktail of 161P2F10B MAbs statistically and significantly inhibited the growth of human renal cancer xenograft UG-K3 implanted subcutaneously in SCID mice (P ⁇ 0.05).
  • FIG. 20 Combination of 161P2F10B MAbs with Avastin® (bevacizumab).
  • FIG. 21 Validation of the 161P2F10B Qa siRNA duplex in HepG2 cells. 2 ⁇ 105 cells were plated in 6 well plates in DMEM plus 10% FBS O.N. and subsequently treated with 20 nM of each indicated siRNA duplex (CT1 or 161P2F10B Qa) complexed with 1 ⁇ g/ml Lipofectamine 2000 (Invitrogen) for 72 h at 37° C.
  • FIG. 22 RNAi silencing of 161P2F10B inhibits cell growth.
  • Cells endogenously expressing the 161P2F10B antigen (HepG2) or not expressing 161P2F10B (UMUC3) were treated with the indicated siRNA duplexes and concentrations.
  • Cells were prepared as stated in FIG. 22 and replated for a standard 3H-Thymidine incorporation assay (22A) or a colony growth assay (22B). Briefly, for the 3H-Thymidine incorporation assay, 2000 cells were replated in triplicate in DMEM plus 10% FBS, and 3H-Thymidine was added for 6 hours after which samples were harvested and incorporation of radioactivity was counted.
  • FIG. 23 161P2F10B silencing with RNAi inhibits cell migration and the Rho signal transduction pathway.
  • 2 ⁇ 105 cells were plated in replicate 6-well plates in DMEM plus 10% FBS O.N. and subsequently treated with 10 nM of each indicated siRNA duplex complexed with 1 ⁇ g/ml Lipofectamine 2000 for 72 h at 37° C.
  • Cells were harvested with 10 mM EDTA and replated in DMEM containing 0.1% FBS into the top chamber of collagen I pre-coated Boyden migration chambers (8 ⁇ m pore size).
  • Ten (10) percent FBS containing DMEM media was used as chemoattractant in the lower chamber.
  • FIG. 24 Relative expression and enzymatic activity of 161P2F10b mutants in recombinant Caki kidney cancer cells.
  • Caki kidney cancer cells were infected with retrovirus containing either wildtype 161P2F10B cDNA, or point mutant cDNAs encoding either a threonine to serine mutation (T/S) at amino acid 205, a threonine to alanine mutation (T/A) at amino acid 205, or a aspartic acid to glutamic acid mutation (D/E) at amino acid 80.
  • T/S threonine to serine mutation
  • T/A threonine to alanine mutation
  • D/E aspartic acid to glutamic acid mutation
  • Stably expressing cell lines were analyzed for 161P2F10B expression by flow cytometry with 97A6 (CD203c) MAb (25A) and for enzymatic activity with p-nTMP substrate (25B).
  • the results show that mutation of threonine 205 to aspartic acid or alanine abolishes the ability to cleave the substrate, demonstrating that threonine 205 is critical to the enzymatic activity of 161P2F10B.
  • FIG. 25 Over-expression of 161P2F10B suppresses ATP-induced intracellular Ca2+ oscillations in Caki-1 cells.
  • Fura-2-loaded Caki-1 cells overexpressing either wild-type (wt) 161P2F10B, a catalytically inactive mutant (T205A), or only a control neomycin resistance gene (neo) were tested for their intracellular Ca2+ mobilization response over time to the purine ligands ATP (25A), ATP ⁇ -S (non-hydrolsable ATP analog) (25B), AMP (25C), and adenosine (25D) using single cell fluorescent microscopic imaging.
  • FIG. 26 161P2F10B ECD induces HUVEC tube formation.
  • Primary human umbilical vein endothelial cells (HUVEC) were seeded in endothelial growth media (EGM) onto 200 ⁇ l of semi-solid Matrigel® in the presence of 0.1% FBS alone, 10% FBS alone, 0.1% FBS+161P2F10B ECD (1 ⁇ g/ml) or 0.1% FBS+control ECD (1 ⁇ g/ml). The cells were allowed to form tube networks for 7 hours and then photographed. The closed tube networks were quantitated in each well. The results indicate that the extracellular domain (ECD) of 161P2F10B induces the formation of tube networks in primary endothelial cells when plated on Matrigel®.
  • ECD extracellular domain
  • FIG. 27 Requirement of 161P2F10B phosphodiesterase activity for HUVEC tube formation.
  • Primary human umbilical vein endothelial cells (HUVEC) were seeded in endothelial growth media (EGM) onto 200 ⁇ l of semi-solid Matrigel® in the presence of 0.1% FBS alone, 10% FBS alone, 0.1% FBS+VEGF (50 ng/ml), 0.1% FBS+wild-type 161P2F10B WT ECD (0.1, 1 or 5 ⁇ g/ml), 0.1% FBS+non-catalytic mutant 161P2F10B D80E ECD (DE) (0.1, 1 or 5 ⁇ g/ml), or 0.1% FBS+catalytic mutant 161P2F10B T205A ECD (TA) (0.1, 1 or 5 ⁇ g/ml).
  • ECD endothelial growth media
  • the cells were allowed to form tube networks for 7 hours and then photographed.
  • the closed tube networks were quantitated in each well.
  • the results show that the phosphodiesterase activity of 161P2F10B is critical for the activity of the ECD in inducing the formation of tube networks in primary endothelial cells when plated on Matrigel®. Further, the RGD domain of the 161P2F10B ECD is also partially critical for the formation of tube networks in primary endothelial cells when plated on Matrigel®.
  • FIG. 28 161P2F10B MAbs inhibit HUVEC tube formation.
  • Primary human umbilical vein endothelial cells (HUVEC) were seeded in endothelial growth media (EGM) onto 200 ⁇ l of semi-solid Matrigel® in the presence of 0.1% FBS alone, 5% FBS alone, or 0.1% FBS+161P2F10B ECD (1 ⁇ g/ml) with or without MAbs to 161P2F10B (20 ⁇ g/ml).
  • the cells were allowed to form tube networks for 18 hours and then photographed.
  • the closed tube networks were quantitated in each well. Percent inhibition was calculated based on the control wells representing 100% tube formation.
  • the results indicate that MAbs to 161P2F10B inhibit the formation of tube networks in primary endothelial cells when plated on Matrigel®.
  • FIG. 29 161P2F10B ECD induces migration of HUVEC.
  • Primary human umbilical vein endothelial cells (HUVEC) were grown in endothelial growth media (EGM) and labeled with the fluorescent dye Calcien AM. The cells were incubated with 0.1% BSA, 10% FBS, 0.1% BSA+VEGF (20 ng/ml), 0.1% BSA+161P2F10B ECD (1, 5 or 10 ⁇ g/ml) or 0.1% BSA+control ECD (1, 5 or 10 ug/ml) and then seeded onto the upper insert of Boyden chambers. The cells were allowed to migrate through the chambers for 20 hours and then quantitated using microscopy and MetaMorph software. The results show that the ECD of 161P2F10B induces the migration of endothelial cells in a dose-dependent manner.
  • FIG. 30 161P2F10B MAbs inhibit proliferation of SK-RC-01 (renal clear cell) cancer cells.
  • SK-RC-01 cells were incubated with 20 ⁇ g/ml MAbs and then grown for 3-5 days in culture with 10% FBS.
  • a 6-hour pulse of 3H-Thymidine was added to the cultures and the cells were then processed for incorporation of 3H-Thymidine.
  • the level of inhibition indicated was calculated using the control MAb as the maximum total counts incorporated.
  • the results show that the MAbs to 161P2F10B inhibit the proliferation of SK-RC-01 renal clear cell cancer cells (45-76%) relative to control MAb.
  • FIG. 31 Effect of 161P2F10B MAbs on kidney cancer cell proliferation, survival and apoptosis.
  • RFX-393 renal cancer cells were incubated with 20 ug/ml MAbs as indicated (or 5 ug/ml EGFR MAb M225) and then grown for 6 days in culture with 10% FBS.
  • a 6-hour pulse of 3H-Thymidine was added to the cultures and the cells were then processed for incorporation of 3H-Thymidine.
  • the level of inhibition indicated was calculated using the control MAb as the maximum total counts incorporated.
  • MTS survival assay
  • a small amount of the Solution Reagent from the kit was added directly to cell culture wells, followed by incubation for 1-4 hours, then recording absorbance at 490 nm with a 96-well plate reader.
  • the quantity of colored formazan product as measured by the amount of 490 nm absorbance is directly proportional to the mitochondrial activity and/or the number of living cells in culture.
  • the % inhibition was calculated relative to a control MAb.
  • the apoptosis assay the same cell lysates prepared for the MTS assay were used in a nucleosome release assay (cell death ELISA). The level of apoptosis is recorded as the % induction relative to control MAb.
  • the more potent MAbs affect all three assays to the highest degrees. The results indicate that the MAbs to 161P2F10B inhibit proliferation of RXF-393 renal cancer clear cells, reduce their cell survival capacity and induce the apoptotic program.
  • FIG. 32 161P2F10B MAbs dose-dependently inhibit HUVEC tube formation.
  • Primary human umbilical vein endothelial cells (HUVEC) were seeded in endothelial growth media (EGM) onto 200 ⁇ l of semi-solid Matrigel® in the presence of 0.1% FBS alone, 5% FBS alone, or 0.1% FBS+161P2F10B ECD (1 ⁇ g/ml) with or without MAbs to 161P2F10B (at the indicated concentrations).
  • the cells were allowed to form tube networks for 18 hours and then photographed.
  • the closed tube networks were quantitated in each well. Percent inhibition was calculated based on the control wells representing 100% tube formation.
  • the results indicate that MAbs to 161P2F10B dose-dependently inhibit the formation of tube networks in primary endothelial cells when plated on Matrigel®.
  • FIG. 33 161P2F10B MAbs inhibit HepG2 liver cancer cell migration.
  • FIG. 33A HepG2 liver cancer cells were grown in 10% FBS, labeled with Calcien AM fluorescent dye, and 2 ⁇ 10 4 cells were incubated with either control MAb or a pool of 161P2F10B MAbs (25 ⁇ g/ml each) and seeded onto the upper inserts of Boyden chambers in the absence or presence of 8 ng/ml Hepatocyte Growth Factor (HGF). The cells were allowed to migrate through the chambers for 24 hours and were then photographed and quantitated using the MetaMorph software.
  • HGF Hepatocyte Growth Factor
  • results show that MAbs to 161P2F10B inhibit the migration of HepG2 cells, and that treatment of the cells with Hepatocyte Growth Factor potentiates the cell migration which is sensitive to inhibition with MAbs to 161P2F10B.
  • the results are graphically displayed in FIG. 33B .
  • FIG. 34 161P2F10B MAbs inhibit A-704 renal clear cell migration.
  • FIG. 34A A-704 (renal clear cell) cancer cells were grown in 10% FBS, labeled with Calcein AM fluorescent dye, and 2 ⁇ 104 cells were incubated with either control MAb or a pool of 161P2F10B MAbs (25 ug/ml each) and seeded onto the upper inserts of Boyden chambers in the absence or presence of 8 ng/ml Hepatocyte Growth Factor (HGF). The cells were allowed to migrate through the chambers for 24 hours and were then photographed and quantitated using the MetaMorph software.
  • HGF Hepatocyte Growth Factor
  • results indicate that MAbs to 161P2F10B inhibit the migration of A-704 cells, and that treatment of the cells with Hepatocyte Growth Factor potentiates the cell migration which is sensitive to inhibition with MAbs to 161P2F10B.
  • the results are graphically displayed in FIG. 34B .
  • FIG. 35 161P2F10B MAbs inhibit A-704 renal clear cell invasion.
  • FIG. 35A A-704 (renal clear cell) cancer cells were grown in 10% FBS, labeled with Calcein AM fluorescent dye, and 2 ⁇ 104 cells were incubated with either control MAb or a pool of 161P2F10B MAbs (25 ug/ml each) and seeded onto the upper inserts of Boyden chambers coated with Matrigel® in the absence or presence of 8 ng/ml Hepatocyte Growth Factor (HGF). The cells were allowed to invade through the chambers for 44 hours and were then photographed and quantitated using the MetaMorph software.
  • HGF Hepatocyte Growth Factor
  • the results show that HGF stimulates the invasion of A-704 cells, and that the MAbs to 161P2F10B (H16-1.93) inhibits the cell invasion in both the HGF treated cells and the untreated cells.
  • the results demonstrate that MAbs to 161P2F10B inhibit the invasion of A-704 cells through Matrigel®, and that treatment of the cells with Hepatocyte Growth Factor potentiates the cell invasion which is sensitive to inhibition with MAbs to 161P2F10B.
  • the results are graphically displayed in FIG. 35B .
  • FIG. 36 161P2F10B dimerization on KU-812 cells.
  • KU-812 cells (2 ⁇ 105) were incubated with increasing concentrations of ethylene glycol bis[succinimidylsuccinate] (EGS) in PBS as indicated for 30 minutes at room temperature.
  • the cells were lysed in RIPA buffer (1% NP-40), subjected to 4-12% gradient non-reducing SDS-PAGE, and then Western blotted for 161P2F10B using a 1 ug/ml MAb mixture (H16-1.52, H16-1.68 and H16-1.92).
  • the results indicate that 161P2F10B is dimeric on the cell surface, and that this property may be required for full enzymatic activity and other functional activities of 161P2F10B when expressed on the surface of tumor cells.
  • FIG. 37 Detection of 161P2F10B protein in cancer patient specimens by IHC. Briefly, frozen tissues were cut into 6 micron sections and mounted on glass slides. The sections were dried for 2 hours at room temperature, fixed for 8 minutes in acetone and subsequently allowed to dry. Sections were then incubated in 161P2F10B antibody, M16-41(3)50, for 3 hours at room temperature. The slides were washed three times in buffer and further incubated with DAKO EnVision+TM peroxidase-conjugated goat anti-mouse immunoglobulin secondary antibody (DAKO Corporation, Carpenteria, Calif.) for 1 hour.
  • DAKO EnVision+TM peroxidase-conjugated goat anti-mouse immunoglobulin secondary antibody DAKO Corporation, Carpenteria, Calif.
  • RNAi and Therapeutic use of small interfering RNA are described in detail below.
  • Advanced cancer “locally advanced cancer”, “advanced disease” and “locally advanced disease” mean cancers that have extended through the relevant tissue capsule, and are meant to include stage C disease under the American Urological Association (AUA) system, stage C1-C2 disease under the Whitmore-Jewett system, and stage T3-T4 and N+disease under the TNM (tumor, node, metastasis) system.
  • AUA American Urological Association
  • TNM tumor, node, metastasis
  • “Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence 161P2F10B (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 161P2F10B.
  • 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.
  • analog refers to a molecule which is structurally similar or shares similar or corresponding attributes with another molecule (e.g. a 161P2F10B-related protein).
  • a 161P2F10B-related protein e.g. an analog of a 161P2F10B protein can be specifically bound by an antibody or T cell that specifically binds to 161P2F10B.
  • Antibody is used in the broadest sense unless clearly indicated otherwise. Therefore, an “antibody” can be naturally occurring or man-made such as monoclonal antibodies produced by conventional hybridoma technology.
  • Anti-161P2F10B 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.
  • the term “antibody” refers to any form of antibody or fragment thereof that specifically binds 161P2F10B and/or exhibits the desired biological activity and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they specifically bind 161P2F10B and/or exhibit the desired biological activity. Any specific antibody can be used in the methods and compositions provided herein.
  • the term “antibody” encompasses a molecule comprising at least one variable region from a light chain immunoglobulin molecule and at least one variable region from a heavy chain molecule that in combination form a specific binding site for the target antigen.
  • the antibody is an IgG antibody.
  • the antibody is a IgG1, IgG2, IgG3, or IgG4 antibody.
  • the antibodies useful in the present methods and compositions can be generated in cell culture, in phage, or in various animals, including but not limited to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats, monkeys, chimpanzees, apes. Therefore, in one embodiment, an antibody of the present invention is a mammalian antibody. Phage techniques can be used to isolate an initial antibody or to generate variants with altered specificity or avidity characteristics. Such techniques are routine and well known in the art.
  • the antibody is produced by recombinant means known in the art.
  • a recombinant antibody can be produced by transfecting a host cell with a vector comprising a DNA sequence encoding the antibody.
  • One or more vectors can be used to transfect the DNA sequence expressing at least one VL and one VH region in the host cell.
  • Exemplary descriptions of recombinant means of antibody generation and production include Delves, ANTIBODY PRODUCTION: ESSENTIAL TECHNIQUES (Wiley, 1997); Shephard, et al., MONOCLONAL ANTIBODIES (Oxford University Press, 2000); Goding, MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE (Academic Press, 1993); CURRENT PROTOCOLS IN IMMUNOLOGY (John Wiley & Sons, most recent edition).
  • An antibody of the present invention can be modified by recombinant means to increase greater efficacy of the antibody in mediating the desired function.
  • antibodies can be modified by substitutions using recombinant means.
  • the substitutions will be conservative substitutions.
  • at least one amino acid in the constant region of the antibody can be replaced with a different residue.
  • the modification in amino acids includes deletions, additions, substitutions of amino acids. In some cases, such changes are made to reduce undesired activities, e.g., complement-dependent cytotoxicity.
  • the antibodies are labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal.
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature.
  • These antibodies can be screened for binding to normal or defective 161P2F10B. See e.g., ANTIBODY ENGINEERING: A PRACTICAL APPROACH (Oxford University Press, 1996).
  • Suitable antibodies with the desired biologic activities can be identified the following in vitro assays including but not limited to: proliferation, migration, adhesion, soft agar growth, angiogenesis, cell-cell communication, apoptosis, transport, signal transduction, and the following in vivo assays such as the inhibition of tumor growth.
  • the antibodies provided herein can also be useful in diagnostic applications. As capture or non-neutralizing antibodies, they can be screened for the ability to bind to the specific antigen without inhibiting the receptor-binding or biological activity of the antigen. As neutralizing antibodies, the antibodies can be useful in competitive binding assays. They can also be used to quantify the 161P2F10B or its receptor.
  • 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-161P2F10B antibodies and clones thereof (including agonist, antagonist and neutralizing antibodies) and anti-161P2F10B antibody compositions with polyepitopic specificity.
  • the antibody of the present methods and compositions can be monoclonal or polyclonal.
  • An antibody can be in the form of an antigen binding antibody fragment including a Fab fragment, F(ab′)2 fragment, a single chain variable region, and the like. Fragments of intact molecules can be generated using methods well known in the art and include enzymatic digestion and recombinant means.
  • any form of the “antigen” can be used to generate an antibody that is specific for 161P2F10B.
  • the eliciting antigen may be a single epitope, multiple epitopes, or the entire protein alone or in combination with one or more immunogenicity enhancing agents known in the art.
  • the eliciting antigen may be an isolated full-length protein, a cell surface protein (e.g., immunizing with cells transfected with at least a portion of the antigen), or a soluble protein (e.g., immunizing with only the extracellular domain portion of the protein).
  • the antigen may be produced in a genetically modified cell.
  • the DNA encoding the antigen may genomic or non-genomic (e.g., cDNA) and encodes at least a portion of the extracellular domain.
  • portion refers to the minimal number of amino acids or nucleic acids, as appropriate, to constitute an immunogenic epitope of the antigen of interest.
  • Any genetic vectors suitable for transformation of the cells of interest may be employed, including but not limited to adenoviral vectors, plasmids, and non-viral vectors, such as cationic lipids.
  • the antibody of the methods and compositions herein specifically bind at least a portion of the extracellular domain of the 161P2F10B of interest.
  • bioactive agent refers to any synthetic or naturally occurring compound that binds the antigen and/or enhances or mediates a desired biological effect to enhance cell-killing toxins.
  • the binding fragments useful in the present invention are biologically active fragments.
  • biologically active refers to an antibody or antibody fragment that is capable of binding the desired the antigenic epitope and directly or indirectly exerting a biologic effect.
  • Direct effects include, but are not limited to the modulation, stimulation, and/or inhibition of a growth signal, the modulation, stimulation, and/or inhibition of an anti-apoptotic signal, the modulation, stimulation, and/or inhibition of an apoptotic or necrotic signal, modulation, stimulation, and/or inhibition the ADCC cascade, and modulation, stimulation, and/or inhibition the CDC cascade.
  • bispecific antibodies are also useful in the present methods and compositions.
  • the term “bispecific antibody” refers to an antibody, typically a monoclonal antibody, having binding specificities for at least two different antigenic epitopes.
  • the epitopes are from the same antigen.
  • the epitopes are from two different antigens.
  • Methods for making bispecific antibodies are known in the art. For example, bispecific antibodies can be produced recombinantly using the co-expression of two immunoglobulin heavy chain/light chain pairs. See, e.g., Milstein et al., Nature 305:537-39 (1983). Alternatively, bispecific antibodies can be prepared using chemical linkage.
  • Bispecific antibodies include bispecific antibody fragments. See, e.g., Hollinger, et al., Proc. Natl. Acad. Sci. U.S.A. 90:6444-48 (1993), Gruber, et al., J. Immunol. 152:5368 (1994).
  • the monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they specifically bind the target antigen and/or exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81: 6851-6855 (1984)).
  • chemotherapeutic Agent refers to all chemical compounds that are effective in inhibiting tumor growth.
  • Non-limiting examples of chemotherapeutic agents include alkylating agents; for example, nitrogen mustards, ethyleneimine compounds and alkyl sulphonates; antimetabolites; for example, folic acid, purine or pyrimidine antagonists; mitotic inhibitors; for example, vinca alkaloids and derivatives of podophyllotoxin, cytotoxic antibiotics, compounds that damage or interfere with DNA expression, and growth factor receptor antagonists.
  • chemotherapeutic agents include cytotoxic agents (as defined herein), antibodies, biological molecules and small molecules.
  • 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.”
  • a “combinatorial library” is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks” such as reagents.
  • a linear combinatorial chemical library such as a polypeptide (e.g., mutein) library, is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound).
  • Numerous chemical compounds are synthesized through such combinatorial mixing of chemical building blocks (Gallop et al., J. Med. Chem. 37(9): 1233-1251 (1994)).
  • combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Pat. No. 5,010,175, Furka, Pept. Prot. Res. 37:487-493 (1991), Houghton et al., Nature, 354:84-88 (1991)), peptoids (PCT Publication No WO 91/19735), encoded peptides (PCT Publication WO 93/20242), random bio-oligomers (PCT Publication WO 92/00091), benzodiazepines (U.S. Pat. No.
  • conservative substitution refers to substitutions of amino acids are known to those of skill in this art and may be made generally without altering the biological activity of the resulting molecule. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson, et al., MOLECULAR BIOLOGY OF THE GENE, The Benjamin/Cummings Pub. Co., p. 224 (4th Edition 1987)). Such exemplary substitutions are preferably made in accordance with those set forth in Table(s) III(a-b).
  • 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.
  • 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(a) herein; pages 13-15 “Biochemistry” 2nd ED.
  • 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.
  • cytotoxic agents include, but are not limited to auristatins, auristatin e, auromycins, maytansinoids, yttrium, bismuth, ricin, ricin A-chain, combrestatin, duocarmycins, dolostatins, doxorubicin, daunorubicin, taxol, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, Sap
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH-VL).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH-VL polypeptide chain
  • the “gene product” is used herein to indicate a peptide/protein or mRNA.
  • a “gene product of the invention” is sometimes referred to herein as a “cancer amino acid sequence”, “cancer protein”, “protein of a cancer listed in Table I”, a “cancer mRNA”, “mRNA of a cancer listed in Table I”, etc.
  • the cancer protein is encoded by a nucleic acid of FIG. 1 .
  • the cancer protein can be a fragment, or alternatively, be the full-length protein encoded by nucleic acids of FIG. 1 .
  • a cancer amino acid sequence is used to determine sequence identity or similarity.
  • the sequences are naturally occurring allelic variants of a protein encoded by a nucleic acid of FIG. 1 .
  • the sequences are sequence variants as further described herein.
  • Heteroconjugate antibodies are useful in the present methods and compositions.
  • the term “heteroconjugate antibody” refers to two covalently joined antibodies.
  • Such antibodies can be prepared using known methods in synthetic protein chemistry, including using crosslinking agents. See, e.g., U.S. Pat. No. 4,676,980.
  • “High throughput screening” assays for the presence, absence, quantification, or other properties of particular nucleic acids or protein products are well known to those of skill in the art.
  • binding assays and reporter gene assays are similarly well known.
  • U.S. Pat. No. 5,559,410 discloses high throughput screening methods for proteins
  • U.S. Pat. No. 5,585,639 discloses high throughput screening methods for nucleic acid binding (i.e., in arrays)
  • U.S. Pat. Nos. 5,576,220 and 5,541,061 disclose high throughput methods of screening for ligand/antibody binding.
  • high throughput screening systems are commercially available (see, e.g., Amersham Biosciences, Piscataway, N.J.; Zymark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio; Beckman Instruments, Inc. Fullerton, Calif.; Precision Systems, Inc., Natick, Mass.; etc.). These systems typically automate entire procedures, including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols for various high throughput systems. Thus, e.g., Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
  • 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.
  • the antibody provided herein is a “human antibody.”
  • the term “human antibody” refers to an antibody in which essentially the entire sequences of the light chain and heavy chain sequences, including the complementary determining regions (CDRs), are from human genes.
  • human monoclonal antibodies are prepared by the trioma technique, the human B-cell technique (see, e.g., Kozbor, et al., Immunol. Today 4: 72 (1983), EBV transformation technique (see, e.g., Cole et al. MONOCLONAL ANTIBODIES AND CANCER THERAPY 77-96 (1985)), or using phage display (see, e.g., Marks et al., J.
  • the human antibody is generated in a transgenic mouse.
  • Techniques for making such partially to fully human antibodies are known in the art and any such techniques can be used.
  • fully human antibody sequences are made in a transgenic mouse engineered to express human heavy and light chain antibody genes.
  • An exemplary description of preparing transgenic mice that produce human antibodies found in Application No. WO 02/43478 and U.S. Pat. No. 6,657,103 (Abgenix) and its progeny. B cells from transgenic mice that produce the desired antibody can then be fused to make hybridoma cell lines for continuous production of the antibody. See, e.g., U.S. Pat. Nos.
  • HLA Human Leukocyte Antigen
  • MHC Major Histocompatibility Complex
  • humanized antibody refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies are chimeric antibodies which contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. See e.g., Cabilly U.S. Pat. No.
  • hybridize used in the context of polynucleotides, are meant to refer to conventional hybridization conditions, preferably such as hybridization in 50% formamide/6 ⁇ SSC/0.1% SDS/100 ⁇ g/ml ssDNA, in which temperatures for hybridization are above 37 degrees C. and temperatures for washing in 0.1 ⁇ SSC/0.1% SDS are above 55 degrees C.
  • 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.
  • isolated peptides in accordance with the invention preferably do not contain materials normally associated with the peptides in their in situ environment.
  • 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 161P2F10B genes or that encode polypeptides other than 161P2F10B gene product or fragments thereof.
  • a skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated 161P2F10B polynucleotide.
  • a protein is said to be “isolated,” for example, when physical, mechanical or chemical methods are employed to remove the 161P2F10B proteins from cellular constituents that are normally associated with the protein.
  • a skilled artisan can readily employ standard purification methods to obtain an isolated 161P2F10B protein.
  • an isolated protein can be prepared by chemical means.
  • Suitable “labels” include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents teaching the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • the antibodies provided herein can be useful as the antigen-binding component of fluorobodies. See e.g., Zeytun et al., Nat. Biotechnol. 21:1473-79 (2003).
  • 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.
  • metalstatic cancer and “metastatic disease” mean cancers that have spread to regional lymph nodes or to distant sites, and are meant to include stage D disease under the AUA system and stage T ⁇ N ⁇ M+ under the TNM system.
  • modulator or “test compound” or “drug candidate” or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for the capacity to directly or indirectly alter the cancer phenotype or the expression of a cancer sequence, e.g., a nucleic acid or protein sequences, or effects of cancer sequences (e.g., signaling, gene expression, protein interaction, etc.)
  • a modulator will neutralize the effect of a cancer protein of the invention.
  • neutralize is meant that an activity of a protein is inhibited or blocked, along with the consequent effect on the cell.
  • a modulator will neutralize the effect of a gene, and its corresponding protein, of the invention by normalizing levels of said protein.
  • modulators alter expression profiles, or expression profile nucleic acids or proteins provided herein, or downstream effector pathways.
  • the modulator suppresses a cancer phenotype, e.g. to a normal tissue fingerprint.
  • a modulator induced a cancer phenotype.
  • a plurality of assay mixtures is run in parallel with different agent concentrations to obtain a differential response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.
  • Modulators, drug candidates or test compounds encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 100 and less than about 2,500 Daltons. Preferred small molecules are less than 2000, or less than 1500 or less than 1000 or less than 500 D.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Modulators also comprise biomolecules such as peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Particularly preferred are peptides.
  • One class of modulators are peptides, for example of from about five to about 35 amino acids, with from about five to about 20 amino acids being preferred, and from about 7 to about 15 being particularly preferred.
  • the cancer modulatory protein is soluble, includes a non-transmembrane region, and/or, has an N-terminal Cys to aid in solubility.
  • the C-terminus of the fragment is kept as a free acid and the N-terminus is a free amine to aid in coupling, i.e., to cysteine.
  • a cancer protein of the invention is conjugated to an immunogenic agent as discussed herein.
  • the cancer protein is conjugated to BSA.
  • the peptides of the invention e.g., of preferred lengths, can be linked to each other or to other amino acids to create a longer peptide/protein.
  • the modulatory peptides can be digests of naturally occurring proteins as is outlined above, random peptides, or “biased” random peptides.
  • peptide/protein-based modulators are antibodies, and fragments thereof, as defined herein.
  • Modulators of cancer can also be nucleic acids.
  • Nucleic acid modulating agents can be naturally occurring nucleic acids, random nucleic acids, or “biased” random nucleic acids. For example, digests of prokaryotic or eukaryotic genomes can be used in an approach analogous to that outlined above for proteins.
  • the term “monoclonal antibody”, as used herein, refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic epitope. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of antibodies directed against (or specific for) different epitopes. In one embodiment, the polyclonal antibody contains a plurality of monoclonal antibodies with different epitope specificities, affinities, or avidities within a single antigen that contains multiple antigenic epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature 256: 495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature 352: 624-628 (1991) and Marks et al., J. Mol.
  • These monoclonal antibodies will usually bind with at least a Kd of about 1 ⁇ M, more usually at least about 300 nM, typically at least about 30 nM, preferably at least about 10 nM, more preferably at least about 3 nM or better, usually determined by ELISA.
  • a “motif”, as in biological motif of a 161P2F10B-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.
  • 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. Frequently occurring motifs are set forth in Table V.
  • 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.
  • polynucleotide means a polymeric form of nucleotides of at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, and is meant to include single and double stranded forms of DNA and/or RNA. In the art, this term if often used interchangeably with “oligonucleotide”.
  • a polynucleotide can comprise a nucleotide sequence disclosed herein wherein thymidine (T), as shown for example in FIG. 1 , 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).
  • 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”.
  • 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.
  • 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.
  • the primary anchor residues of a peptide binds 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(a).
  • 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.
  • Radioisotopes include, but are not limited to the following (non-limiting exemplary uses are also set forth in Table IV(I)).
  • each nucleic acid and peptide consists of essentially random nucleotides and amino acids, respectively. These random peptides (or nucleic acids, discussed herein) can incorporate any nucleotide or amino acid at any position.
  • the synthetic process can be designed to generate randomized proteins or nucleic acids, to allow the formation of all or most of the possible combinations over the length of the sequence, thus forming a library of randomized candidate bioactive proteinaceous agents.
  • a library is “fully randomized,” with no sequence preferences or constants at any position.
  • the library is a “biased random” library. That is, some positions within the sequence either are held constant, or are selected from a limited number of possibilities.
  • the nucleotides or amino acid residues are randomized within a defined class, e.g., of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large) residues, towards the creation of nucleic acid binding domains, the creation of cysteines, for cross-linking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, etc., or to purines, etc.
  • a “recombinant” DNA or RNA molecule is a DNA or RNA molecule that has been subjected to molecular manipulation in vitro.
  • single-chain Fv or “scFv” or “single chain” antibody refers to antibody fragments comprising the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding.
  • Non-limiting examples of “small molecules” include compounds that bind or interact with 161P2F10B, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit 161P2F10B 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, 161P2F10B protein; are not found in naturally occurring metabolic pathways; and/or are more soluble in aqueous than non-aqueous solutions.
  • the term “specific” refers to the selective binding of the antibody to the target antigen epitope.
  • Antibodies can be tested for specificity of binding by comparing binding to appropriate antigen to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody binds to the appropriate antigen at least 2, 5, 7, and preferably 10 times more than to irrelevant antigen or antigen mixture then it is considered to be specific.
  • a specific antibody is one that only binds the 161P2F10B antigen, but does not bind to the irrelevent antigen.
  • a specific antibody is one that binds human 161P2F10B antigen but does not bind a non-human 161P2F10B antigen with 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or greater amino acid homology with the 161P2F10B antigen.
  • a specific antibody is one that binds human 161P2F10B antigen and binds murine 161P2F10B antigen, but with a higher degree of binding the human antigen.
  • a specific antibody is one that binds human 161P2F10B antigen and binds primate 161P2F10B antigen, but with a higher degree of binding the human antigen.
  • the specific antibody binds to human 161P2F10B antigen and any non-human 161P2F10B antigen, but with a higher degree of binding the human antigen or any combination thereof.
  • “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured nucleic acid sequences to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
  • “Stringent conditions” or “high stringency conditions”, as defined herein, are identified by, but not limited to, those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5 ⁇ SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 ⁇ Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1% SDS, and
  • Modely 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.
  • washing solution and hybridization conditions e.g., temperature, ionic strength and % SDS
  • An example of moderately stringent conditions is overnight incubation at 65° C.
  • HLA “supermotif” is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles. Overall phenotypic frequencies of HLA-supertypes in different ethnic populations are set forth in Table IV (f). The non-limiting constituents of various supertypes are as follows:
  • A2 A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*6802, A*6901, A*0207;
  • A3 A3, All, A31, A*3301, A*6801, A*0301, A*1101, A*3101;
  • B7 B7, B*3501-03, B*51, B*5301, B*5401, B*5501, B*5502, B*5601, B*6701, B*7801, B*0702, B*5101, B*5602;
  • B44 B*3701, B*4402, B*4403, B*60 (B*4001), B61 (B*4006);
  • A1 A*0102, A*2604, A*3601, A*4301, A*8001;
  • A24 A*24, A*30, A*2403, A*2404, A*3002, A*3003;
  • B27 B*1401-02, B*1503, B*1509, B*1510, B*1518, B*3801-02, B*3901, B*3902, B*3903-04, B*4801-02, B*7301, B*2701-08;
  • B58 B*1516, B*1517, B*5701, B*5702, B58;
  • B62 B*4601, B52, B*1501 (B62), B*1502 (B75), B*1513 (B77).
  • 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; as is readily appreciated in the art, full eradication of disease is a preferred out albeit not a requirement for a treatment act.
  • 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.
  • an HLA or cellular immune response “vaccine” is a composition that contains or encodes one or more peptides of the invention.
  • 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.
  • variant refers to a molecule that exhibits a variation from a described type or norm, such as a protein that has one or more different amino acid residues in the corresponding position(s) of a specifically described protein (e.g. the 161P2F10B protein shown in FIG. 1 .)
  • An analog is an example of a variant protein.
  • Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.
  • the “161P2F10B-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 161P2F10B proteins or fragments thereof, as well as fusion proteins of a 161P2F10B protein and a heterologous polypeptide are also included. Such 161P2F10B proteins are collectively referred to as the 161P2F10B-related proteins, the proteins of the invention, or 161P2F10B.
  • 161P2F10B-related protein refers to a polypeptide fragment or a 161P2F10B protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 330, 335, 339 or more amino acids.
  • One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a 161P2F10B gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a 161P2F10B-related protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a 161P2F10B gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a 161P2F10B gene, mRNA, or to a 161P2F10B encoding polynucleotide (collectively, “161P2F10B polynucleotides”).
  • T can also be U in FIG. 1 .
  • Embodiments of a 161P2F10B polynucleotide include: a 161P2F10B polynucleotide having the sequence shown in FIG. 1 , the nucleotide sequence of 161P2F10B as shown in FIG. 1 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 1 ; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 1 where T is U.
  • Polynucleotides encoding relatively long portions of a 161P2F10B protein are also within the scope of the invention.
  • 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 161P2F10B protein “or variant” shown in FIG. 1 or FIG. 3 can be generated by a variety of techniques well known in the art.
  • These polynucleotide fragments can include any portion of the 161P2F10B sequence as shown in FIG. 1 .
  • the polynucleotides of the preceding paragraphs have a number of different specific uses.
  • the human 161P2F10B gene maps to the chromosomal location set forth in the Example entitled “Chromosomal Mapping of 161P2F10B.”
  • polynucleotides that encode different regions of the 161P2F10B proteins are used to characterize cytogenetic abnormalities of this chromosomal locale, such as abnormalities that are identified as being associated with various cancers.
  • cytogenetic abnormalities of this chromosomal locale such as abnormalities that are identified as being associated with various cancers.
  • a variety of chromosomal abnormalities including rearrangements have been identified as frequent cytogenetic abnormalities in a number of different cancers (see e.g.
  • polynucleotides encoding specific regions of the 161P2F10B proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes 161P2F10B that may contribute to the malignant phenotype.
  • 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)).
  • 161P2F10B was shown to be highly expressed in prostate and other cancers, 161P2F10B polynucleotides are used in methods assessing the status of 161P2F10B gene products in normal versus cancerous tissues. Typically, polynucleotides that encode specific regions of the 161P2F10B 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 161P2F10B gene, such as regions containing one or more motifs.
  • perturbations such as deletions, insertions, point mutations, or alterations resulting in a loss of an antigen etc.
  • 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.
  • SSCP single-strand conformation polymorphism
  • 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 161P2F10B.
  • 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.
  • PNAs peptide nucleic acids
  • 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 161P2F10B 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., 161P2F10B. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988).
  • the 161P2F10B 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 are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom.
  • the S-oligos of the present invention can be prepared by treatment of the corresponding O-oligos with 3H-1,2-benzodithiol-3-one-1,1-dioxide, which is a sulfur transfer reagent. See, e.g., Iyer, R. P. et al., J. Org. Chem. 55:4693-4698 (1990); and Iyer, R. P. et al., J. Am. Chem. Soc. 112:1253-1254 (1990).
  • Additional 161P2F10B 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 161P2F10B 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 161P2F10B 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 161P2F10B mRNA and not to mRNA specifying other regulatory subunits of protein kinase.
  • 161P2F10B antisense oligonucleotides of the present invention are 15 to 30-mer fragments of the antisense DNA molecule that have a sequence that hybridizes to 161P2F10B mRNA.
  • 161P2F10B 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 161P2F10B.
  • the antisense molecules are modified to employ ribozymes in the inhibition of 161P2F10B expression, see, e.g., L. A. Couture & D. T. Stinchcomb; Trends Genet. 12: 510-515 (1996).
  • 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.
  • 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 161P2F10B polynucleotide in a sample and as a means for detecting a cell expressing a 161P2F10B protein.
  • probes include polypeptides comprising all or part of the human 161P2F10B cDNA sequence shown in FIG. 1 .
  • primer pairs capable of specifically amplifying 161P2F10B 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 161P2F10B mRNA.
  • the 161P2F10B 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 161P2F10B 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 161P2F10B polypeptides; as tools for modulating or inhibiting the expression of the 161P2F10B gene(s) and/or translation of the 161P2F10B transcript(s); and as therapeutic agents.
  • the present invention includes the use of any probe as described herein to identify and isolate a 161P2F10B or 161P2F10B related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence per se, which would comprise all or most of the sequences found in the probe used.
  • the 161P2F10B cDNA sequences described herein enable the isolation of other polynucleotides encoding 161P2F10B gene product(s), as well as the isolation of polynucleotides encoding 161P2F10B gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a 161P2F10B gene product as well as polynucleotides that encode analogs of 161P2F10B-related proteins.
  • Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a 161P2F10B gene are well known (see, for example, Sambrook, J.
  • a 161P2F10B gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with 161P2F10B DNA probes or primers.
  • BACs bacterial artificial chromosome libraries
  • YACs yeast artificial chromosome libraries
  • the invention also provides recombinant DNA or RNA molecules containing a 161P2F10B 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 161P2F10B polynucleotide, fragment, analog or homologue thereof within a suitable prokaryotic or eukaryotic host cell.
  • 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).
  • suitable mammalian cells include various prostate cancer cell lines such as DU145 and TsuPr1, other transfectable or transducible prostate cancer cell lines, primary cells (PrEC), as well as a number of mammalian cells routinely used for the expression of recombinant proteins (e.g., COS, CHO, 293, 293T cells). More particularly, a polynucleotide comprising the coding sequence of 161P2F10B or a fragment, analog or homolog thereof can be used to generate 161P2F10B proteins or fragments thereof using any number of host-vector systems routinely used and widely known in the art.
  • 161P2F10B 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 pSR tkneo (Muller et al., 1991, MCB 11:1785).
  • 161P2F10B can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPr1.
  • the host-vector systems of the invention are useful for the production of a 161P2F10B protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of 161P2F10B and 161P2F10B mutations or analogs.
  • Recombinant human 161P2F10B protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a 161P2F10B-related nucleotide.
  • 293T cells can be transfected with an expression plasmid encoding 161P2F10B or fragment, analog or homolog thereof, a 161P2F10B-related protein is expressed in the 293T cells, and the recombinant 161P2F10B protein is isolated using standard purification methods (e.g., affinity purification using anti-161P2F10B antibodies).
  • a 161P2F10B coding sequence is subcloned into the retroviral vector pSR ⁇ MSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPr1, 293 and rat-i in order to establish 161P2F10B expressing cell lines.
  • mammalian cell lines such as NIH 3T3, TsuPr1, 293 and rat-i
  • Various other expression systems well known in the art can also be employed.
  • Expression constructs encoding a leader peptide joined in frame to a 161P2F10B coding sequence can be used for the generation of a secreted form of recombinant 161P2F10B protein.
  • codon preferences for a specific species are calculated, for example, by utilizing codon usage tables available on the INTERNET such as at URL 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).
  • 161P2F10B-related proteins Another aspect of the present invention provides 161P2F10B-related proteins.
  • Specific embodiments of 161P2F10B proteins comprise a polypeptide having all or part of the amino acid sequence of human 161P2F10B as shown in FIG. 1 , preferably FIG. 1A .
  • embodiments of 161P2F10B proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of 161P2F10B shown in FIG. 1 .
  • Embodiments of a 161P2F10B polypeptide include: a 161P2F10B polypeptide having a sequence shown in FIG. 1 , a peptide encoded by a polynucleotide sequence of a 161P2F10B as shown in FIG. 1 wherein T is U; at least 10 contiguous nucleotides encoding a polypeptide having the sequence as shown in FIG. 1 ; or, at least 10 contiguous peptides encoded by a polynucleotide having the sequence as shown in FIG. 1 where T is U.
  • Proteins of the invention can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 conservative substitutions.
  • Embodiments of the invention disclosed herein include a wide variety of art-accepted variants or analogs of 161P2F10B proteins such as polypeptides having amino acid insertions, deletions and substitutions.
  • 161P2F10B variants can be made using methods known in the art such as site-directed mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl.
  • 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.
  • preferred scanning amino acids are relatively small, neutral amino acids.
  • 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.
  • 161P2F10B variants, analogs or homologs have the distinguishing attribute of having at least one epitope that is “cross reactive” with a 161P2F10B protein having an amino acid sequence of FIG. 1 .
  • cross reactive means that an antibody or T cell that specifically binds to a 161P2F10B variant also specifically binds to a 161P2F10B protein having an amino acid sequence set forth in FIG. 1 .
  • a polypeptide ceases to be a variant of a protein shown in FIG. 1 , when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting 161P2F10B protein.
  • 161P2F10B-related protein variants share 70%, 75%, 80%, 85%, 90%, 95% or more similarity with an amino acid sequence of FIG. 1 , or a fragment thereof.
  • Another specific class of 161P2F10B protein variants or analogs comprises one or more of the 161P2F10B biological motifs described herein or presently known in the art.
  • analogs of 161P2F10B fragments that have altered functional (e.g. immunogenic) properties relative to the starting fragment. It is to be appreciated that motifs now or which become part of the art are to be applied to the nucleic or amino acid sequences of FIG. 1 .
  • embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a 161P2F10B protein shown in FIG. 1 .
  • 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 161P2F10B protein shown in FIG. 1 .
  • 161P2F10B-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 161P2F10B-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a 161P2F10B protein (or variants, homologs or analogs thereof).
  • Additional illustrative embodiments of the invention disclosed herein include 161P2F10B polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a 161P2F10B polypeptide sequence set forth in FIG. 1 .
  • 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 such as BIMAS.
  • Table IV(h) sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu/). The columns of Table IV(h) 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 161P2F10B motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the 161P2F10B motifs discussed above are associated with growth dysregulation and because 161P2F10B is overexpressed in certain cancers (See, e.g., Table I).
  • Casein kinase II, cAMP and camp-dependent protein kinase, and Protein Kinase C are enzymes known to be associated with the development of the malignant phenotype (see e.g.
  • 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)).
  • proteins of the invention comprise one or more of the immunoreactive epitopes identified in accordance with art-accepted methods, such as the peptides previously disclosed.
  • CTL epitopes can be determined using specific algorithms to identify peptides within a 161P2F10B protein that are capable of optimally binding to specified HLA alleles (e.g., Table IV; EpimatrixTM and EpimerTM, Brown University; and BIMAS.)
  • HLA alleles e.g., Table IV; EpimatrixTM and EpimerTM, Brown University; and BIMAS.
  • 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.
  • 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.
  • epitopes in order to modulate immunogenicity. For example, one begins with an epitope that bears a CTL or HTL motif (see, e.g., the HLA Class I and HLA Class II motifs/supermotifs of Table IV).
  • the epitope is analoged by substituting out an amino acid at one of the specified positions, and replacing it with another amino acid specified for that position.
  • residues defined in Table IV one can substitute out a deleterious residue in favor of any other residue, such as a preferred residue; substitute a less-preferred residue with a preferred residue; or substitute an originally-occurring preferred residue with another preferred residue. Substitutions can occur at primary anchor positions or at other positions in a peptide; see, e.g., Table IV.
  • polypeptides comprising combinations of the different motifs set forth in Table(s) IV(a), IV(b), IV(c), IV(d), and IV(h), and/or, one or more of the predicted CTL epitopes of previously disclosed, 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 within the intervening sequences of the polypeptides.
  • 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).
  • 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.
  • 161P2F10B-related proteins are embodied in many forms, preferably in isolated form.
  • a purified 161P2F10B protein molecule will be substantially free of other proteins or molecules that impair the binding of 161P2F10B to antibody, T cell or other ligand. The nature and degree of isolation and purification will depend on the intended use.
  • Embodiments of a 161P2F10B-related proteins include purified 161P2F10B-related proteins and functional, soluble 161P2F10B-related proteins.
  • a functional, soluble 161P2F10B protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.
  • the invention also provides 161P2F10B proteins comprising biologically active fragments of a 161P2F10B amino acid sequence shown in FIG. 1 .
  • Such proteins exhibit properties of the starting 161P2F10B protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting 161P2F10B 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.
  • 161P2F10B-related polypeptides that contain particularly interesting structures can be predicted and/or identified using various analytical techniques well known in the art, including, for example, the methods of Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis, or based on immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-161P2F10B antibodies or T cells or in identifying cellular factors that bind to 161P2F10B. 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.
  • 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 161P2F10B protein that are capable of optimally binding to specified HLA alleles such as BIMAS and SYFPEITHI. Illustrating this, peptide epitopes from 161P2F10B that are presented in the context of human MHC Class I molecules, e.g., HLA-A1, A2, A3, A11, A24, B7 and B35 were predicted.
  • the complete amino acid sequence of the 161P2F10B protein and relevant portions of other variants i.e., for HLA Class I predictions 9 flanking residues on either side of a point mutation or exon juction, and for HLA Class II predictions 14 flanking residues on either side of a point mutation or exon junction corresponding to that variant, were entered into the HLA Peptide Motif Search algorithm found in the Bioinformatics and Molecular Analysis Section.
  • 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.
  • HLA class I binding peptides are 8-, 9-, 10 or 11-mers.
  • 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 161P2F10B predicted binding peptides have been shown.
  • 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.
  • every epitope predicted by the BIMAS site, EpimerTM and EpimatrixTM 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 are to be “applied” to a 161P2F10B protein in accordance with the invention.
  • “applied” means that a 161P2F10B 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 161P2F10B 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.
  • 161P2F10B 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 161P2F10B with a C-terminal 6 ⁇ His and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville Tenn.).
  • the Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted 161P2F10B protein in transfected cells.
  • the secreted HIS-tagged 161P2F10B in the culture media can be purified, e.g., using a nickel column using standard techniques.
  • Modifications of 161P2F10B-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 161P2F10B polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues of a 161P2F10B protein.
  • Another type of covalent modification of a 161P2F10B 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 161P2F10B comprises linking a 161P2F10B polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. No. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
  • PEG polyethylene glycol
  • polypropylene glycol polypropylene glycol
  • polyoxyalkylenes polyoxyalkylenes
  • the 161P2F10B-related proteins of the present invention can also be modified to form a chimeric molecule comprising 161P2F10B fused to another, heterologous polypeptide or amino acid sequence.
  • a chimeric molecule can be synthesized chemically or recombinantly.
  • a chimeric molecule can have a protein of the invention fused to another tumor-associated antigen or fragment thereof.
  • a protein in accordance with the invention can comprise a fusion of fragments of a 161P2F10B sequence (amino or nucleic acid) such that a molecule is created that is not, through its length, directly homologous to the amino or nucleic acid sequences shown in FIG. 1 .
  • Such a chimeric molecule can comprise multiples of the same subsequence of 161P2F10B.
  • a chimeric molecule can comprise a fusion of a 161P2F10B-related protein with a polyhistidine epitope tag, which provides an epitope to which immobilized nickel can selectively bind, with cytokines or with growth factors.
  • the epitope tag is generally placed at the amino- or carboxyl-terminus of a 161P2F10B protein.
  • the chimeric molecule can comprise a fusion of a 161P2F10B-related protein with an immunoglobulin or a particular region of an immunoglobulin.
  • the chimeric molecule also referred to as an “immunoadhesin”
  • 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 161P2F10B polypeptide in place of at least one variable region within an Ig molecule.
  • the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgG1 molecule.
  • the proteins of the invention have a number of different specific uses. As 161P2F10B is highly expressed in prostate and other cancers, 161P2F10B-related proteins are used in methods that assess the status of 161P2F10B gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a 161P2F10B 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).
  • perturbations such as deletions, insertions, point mutations etc.
  • Exemplary assays utilize antibodies or T cells targeting 161P2F10B-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a 161P2F10B 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.
  • 161P2F10B-related proteins that contain the amino acid residues of one or more of the biological motifs in a 161P2F10B protein are used to screen for factors that interact with that region of 161P2F10B.
  • 161P2F10B protein fragments/subsequences are particularly useful in generating and characterizing domain-specific antibodies (e.g., antibodies recognizing an extracellular or intracellular epitope of a 161P2F10B protein), for identifying agents or cellular factors that bind to 161P2F10B 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.
  • domain-specific antibodies e.g., antibodies recognizing an extracellular or intracellular epitope of a 161P2F10B protein
  • Proteins encoded by the 161P2F10B genes 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 161P2F10B gene product.
  • Antibodies raised against a 161P2F10B protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of 161P2F10B protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers.
  • 161P2F10B-related nucleic acids or proteins are also used in generating HTL or CTL responses.
  • Various immunological assays useful for the detection of 161P2F10B 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 161P2F10B-expressing cells (e.g., in radioscintigraphic imaging methods). 161P2F10B proteins are also particularly useful in generating cancer vaccines, as further described herein.
  • Another aspect of the invention provides antibodies that bind to 161P2F10B-related proteins.
  • Preferred antibodies specifically bind to a 161P2F10B-related protein and do not bind (or bind weakly) to peptides or proteins that are not 161P2F10B-related proteins under physiological conditions.
  • physiological conditions include: 1) phosphate buffered saline; 2) Tris-buffered saline containing 25 mM Tris and 150 mM NaCl; or normal saline (0.9% NaCl); 4) animal serum such as human serum; or, 5) a combination of any of 1) through 4); these reactions preferably taking place at pH 7.5, alternatively in a range of pH 7.0 to 8.0, or alternatively in a range of pH 6.5 to 8.5; also, these reactions taking place at a temperature between 4° C. to 37° C.
  • antibodies that bind 161P2F10B can bind 161P2F10B-related proteins such as the homologs or analogs thereof.
  • 161P2F10B 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 prostate and other cancers, to the extent 161P2F10B 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 161P2F10B is involved, such as advanced or metastatic prostate cancers or other advanced or metastatic cancers.
  • the invention also provides various immunological assays useful for the detection and quantification of 161P2F10B and mutant 161P2F10B-related proteins.
  • Such assays can comprise one or more 161P2F10B antibodies capable of recognizing and binding a 161P2F10B-related protein, as appropriate.
  • These assays are performed within various immunological assay formats well known in the art, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), and the like.
  • Immunological non-antibody assays of the invention also comprise T cell immunogenicity assays (inhibitory or stimulatory) as well as major histocompatibility complex (MHC) binding assays.
  • T cell immunogenicity assays inhibitory or stimulatory
  • MHC major histocompatibility complex
  • immunological imaging methods capable of detecting kidney cancer and other cancers expressing 161P2F10B are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled 161P2F10B antibodies.
  • assays are clinically useful in the detection, monitoring, and prognosis of 161P2F10B expressing cancers such as kidney cancer.
  • 161P2F10B antibodies are also used in methods for purifying a 161P2F10B-related protein and for isolating 161P2F10B homologues and related molecules.
  • a method of purifying a 161P2F10B-related protein comprises incubating a 161P2F10B antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a 161P2F10B-related protein under conditions that permit the 161P2F10B antibody to bind to the 161P2F10B-related protein; washing the solid matrix to eliminate impurities; and eluting the 161P2F10B-related protein from the coupled antibody.
  • Other uses of 161P2F10B antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a 161P2F10B protein.
  • antibodies can be prepared by immunizing a suitable mammalian host using a 161P2F10B-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)).
  • fusion proteins of 161P2F10B can also be used, such as a 161P2F10B GST-fusion protein.
  • a GST fusion protein comprising all or most of the amino acid sequence of FIG. 1 is produced, then used as an immunogen to generate appropriate antibodies.
  • a 161P2F10B-related protein is synthesized and used as an immunogen.
  • naked DNA immunization techniques known in the art are used (with or without purified 161P2F10B-related protein or 161P2F10B 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 161P2F10B protein as shown in FIG. 1 can be analyzed to select specific regions of the 161P2F10B protein for generating antibodies.
  • hydrophobicity and hydrophilicity analyses of a 161P2F10B amino acid sequence are used to identify hydrophilic regions in the 161P2F10B structure.
  • Regions of a 161P2F10B 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.
  • 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. Preferred methods for the generation of 161P2F10B 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.
  • 161P2F10B 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 161P2F10B-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 161P2F10B protein can also be produced in the context of chimeric or complementarity-determining region (CDR) grafted antibodies of multiple species origin. Humanized or human 161P2F10B antibodies can also be produced, and are preferred for use in therapeutic contexts.
  • CDR complementarity-determining region
  • Fully human 161P2F10B monoclonal antibodies can be generated using cloning technologies employing large human Ig gene combinatorial libraries (i.e., phage display) (Griffiths and Hoogenboom, Building an in vitro immune system: human antibodies from phage display libraries. In: Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man, Clark, M. (Ed.), Nottingham Academic, pp 45-64 (1993); Burton and Barbas, Human Antibodies from combinatorial libraries. Id., pp 65-82).
  • Fully human 161P2F10B monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application WO98/24893, Kucherlapati and Jakobovits et al., published Dec. 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 7(4): 607-614; U.S. Pat. Nos. 6,162,963 issued 19 Dec. 2000; 6,150,584 issued 12 Nov. 2000; and, 6,114,598 issued 5 Sep. 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.
  • 161P2F10B antibodies with a 161P2F10B-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 161P2F10B-related proteins, 161P2F10B-expressing cells or extracts thereof.
  • a 161P2F10B 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.
  • bi-specific antibodies specific for two or more 161P2F10B 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).
  • the invention provides for monoclonal antibodies identified as Ha16-1(3,5)18, Ha16-1(1)11, H16-1.93, H16-9.69 were sent (via Federal Express) to the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108 on 28-Mar.-2006 and assigned Accession numbers PTA-7452 and PTA-7450 and PTA-7449 and PTA-7451, respectively.
  • compositions of the invention induce a therapeutic or prophylactic immune responses in very broad segments of the world-wide population.
  • immunology-related technology For an understanding of the value and efficacy of compositions of the invention that induce cellular immune responses, a brief review of immunology-related technology is provided.
  • a complex of an HLA molecule and a peptidic antigen acts as the ligand recognized by HLA-restricted T cells (Buus, S. et al., Cell 47:1071, 1986; Babbitt, B. P. et al., Nature 317:359, 1985; Townsend, A. and Bodmer, H., Annu. Rev. Immunol. 7:601, 1989; Germain, R. N., Annu. Rev. Immunol. 11:403, 1993).
  • class I and class II allele-specific HLA binding motifs allows identification of regions within a protein that are correlated with binding to particular HLA antigen(s).
  • 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.
  • 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).
  • peptides in incomplete Freund's adjuvant are administered subcutaneously to HLA transgenic mice.
  • splenocytes are removed and cultured in vitro in the presence of test peptide for approximately one week.
  • Peptide-specific T cells are detected using, e.g., a 51Cr-release assay involving peptide sensitized target cells and target cells expressing endogenously generated antigen.
  • 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.
  • APC antigen presenting cells
  • T cell activity is detected using assays including 51Cr release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.
  • Nucleic acids that encode a 161P2F10B-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.
  • cDNA encoding 161P2F10B can be used to clone genomic DNA that encodes 161P2F10B. The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode 161P2F10B.
  • Methods for generating transgenic animals, particularly animals such as mice or rats have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 issued 12 Apr. 1988, and 4,870,009 issued 26 Sep. 1989.
  • particular cells would be targeted for 161P2F10B transgene incorporation with tissue-specific enhancers.
  • Transgenic animals that include a copy of a transgene encoding 161P2F10B can be used to examine the effect of increased expression of DNA that encodes 161P2F10B. 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.
  • non-human homologues of 161P2F10B can be used to construct a 161P2F10B “knock out” animal that has a defective or altered gene encoding 161P2F10B as a result of homologous recombination between the endogenous gene encoding 161P2F10B and altered genomic DNA encoding 161P2F10B introduced into an embryonic cell of the animal.
  • cDNA that encodes 161P2F10B can be used to clone genomic DNA encoding 161P2F10B in accordance with established techniques.
  • a portion of the genomic DNA encoding 161P2F10B can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration.
  • another gene such as a gene encoding a selectable marker that can be used to monitor integration.
  • several kilobases of unaltered flanking DNA are included in the vector (see, e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors).
  • the vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected (see, e.g., Li et al., Cell, 69:915 (1992)).
  • the selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras (see, e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152).
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal, and the embryo brought to term to create a “knock out” animal.
  • Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA.
  • Knock out animals can be characterized, for example, for their ability to defend against certain pathological conditions or for their development of pathological conditions due to absence of a 161P2F10B polypeptide.
  • Another aspect of the present invention relates to methods for detecting 161P2F10B polynucleotides and 161P2F10B-related proteins, as well as methods for identifying a cell that expresses 161P2F10B.
  • the expression profile of 161P2F10B makes it a diagnostic marker for metastasized disease. Accordingly, the status of 161P2F10B gene products provides information useful for predicting a variety of factors including susceptibility to advanced stage disease, rate of progression, and/or tumor aggressiveness.
  • the status of 161P2F10B 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.
  • the invention provides assays for the detection of 161P2F10B polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like.
  • Detectable 161P2F10B polynucleotides include, for example, a 161P2F10B gene or fragment thereof, 161P2F10B mRNA, alternative splice variant 161P2F10B mRNAs, and recombinant DNA or RNA molecules that contain a 161P2F10B polynucleotide.
  • a number of methods for amplifying and/or detecting the presence of 161P2F10B polynucleotides are well known in the art and can be employed in the practice of this aspect of the invention.
  • a method for detecting a 161P2F10B 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 161P2F10B polynucleotides as sense and antisense primers to amplify 161P2F10B cDNAs therein; and detecting the presence of the amplified 161P2F10B cDNA.
  • the sequence of the amplified 161P2F10B cDNA can be determined.
  • a method of detecting a 161P2F10B gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using 161P2F10B polynucleotides as sense and antisense primers; and detecting the presence of the amplified 161P2F10B gene.
  • Any number of appropriate sense and antisense probe combinations can be designed from a 161P2F10B nucleotide sequence (see, e.g., FIG. 1 ) and used for this purpose.
  • the invention also provides assays for detecting the presence of a 161P2F10B protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like.
  • Methods for detecting a 161P2F10B-related protein are also well known and include, for example, immunoprecipitation, immunohistochemical analysis, Western blot analysis, molecular binding assays, ELISA, ELIFA and the like.
  • a method of detecting the presence of a 161P2F10B-related protein in a biological sample comprises first contacting the sample with a 161P2F10B antibody, a 161P2F10B-reactive fragment thereof, or a recombinant protein containing an antigen-binding region of a 161P2F10B antibody; and then detecting the binding of 161P2F10B-related protein in the sample.
  • an assay for identifying a cell that expresses a 161P2F10B gene comprises detecting the presence of 161P2F10B mRNA in the cell.
  • Methods for the detection of particular mRNAs in cells include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled 161P2F10B riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for 161P2F10B, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
  • an assay for identifying a cell that expresses a 161P2F10B gene comprises detecting the presence of 161P2F10B-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 161P2F10B-related proteins and cells that express 161P2F10B-related proteins.
  • 161P2F10B expression analysis is also useful as a tool for identifying and evaluating agents that modulate 161P2F10B gene expression.
  • 161P2F10B expression is significantly upregulated in kidney cancer, and is expressed in cancers of the tissues listed in Table I.
  • Identification of a molecule or biological agent that inhibits 161P2F10B expression or over-expression in cancer cells is of therapeutic value.
  • such an agent can be identified by using a screen that quantifies 161P2F10B expression by RT-PCR, nucleic acid hybridization or antibody binding.
  • 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)).
  • examining a biological sample for evidence of dysregulated cell growth 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.
  • the status of 161P2F10B in a biological sample of interest can be compared, for example, to the status of 161P2F10B in a corresponding normal sample (e.g. a sample from that individual or alternatively another individual that is not affected by a pathology).
  • 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 161P2F10B in the biological sample provides evidence of dysregulated cellular growth.
  • a predetermined normative value such as a predetermined normal level of mRNA expression (see, e.g., Grever et al., J. Comp. Neurol. 1996 Dec. 9; 376(2): 306-14 and U.S. Pat. No. 5,837,501) to compare 161P2F10B status in a sample.
  • 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 161P2F10B expressing cells) as well as the level, and biological activity of expressed gene products (such as 161P2F10B mRNA, polynucleotides and polypeptides).
  • an alteration in the status of 161P2F10B comprises a change in the location of 161P2F10B and/or 161P2F10B expressing cells and/or an increase in 161P2F10B mRNA and/or protein expression.
  • 161P2F10B 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 161P2F10B 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).
  • the status of 161P2F10B 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 161P2F10B gene), Northern analysis and/or PCR analysis of 161P2F10B mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of 161P2F10B 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 161P2F10B proteins and/or associations of 161P2F10B proteins with polypeptide binding partners).
  • genomic Southern analysis to examine, for example perturbations in a 161P2F10B gene
  • Northern analysis and/or PCR analysis of 161P2F10B mRNA to examine, for example alterations in the polynucleotide sequences or expression levels of 161P2F10B
  • Detectable 161P2F10B polynucleotides include, for example, a 161P2F10B gene or fragment thereof, 161P2F10B mRNA, alternative splice variants, 161P2F10B mRNAs, and recombinant DNA or RNA molecules containing a 161P2F10B polynucleotide.
  • the expression profile of 161P2F10B makes it a diagnostic marker for local and/or metastasized disease, and provides information on the growth or oncogenic potential of a biological sample.
  • the status of 161P2F10B provides information useful for predicting susceptibility to particular disease stages, progression, and/or tumor aggressiveness.
  • the invention provides methods and assays for determining 161P2F10B status and diagnosing cancers that express 161P2F10B, such as cancers of the tissues listed in Table I.
  • 161P2F10B mRNA is so highly expressed in kidney and other cancers relative to normal kidney tissue
  • assays that evaluate the levels of 161P2F10B mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with 161P2F10B dysregulation, and can provide prognostic information useful in defining appropriate therapeutic options.
  • the expression status of 161P2F10B 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 161P2F10B 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.
  • the status of 161P2F10B in a biological sample can be examined by a number of well-known procedures in the art.
  • the status of 161P2F10B 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 161P2F10B expressing cells (e.g. those that express 161P2F10B mRNAs or proteins).
  • This examination can provide evidence of dysregulated cellular growth, for example, when 161P2F10B-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 161P2F10B in a biological sample are often associated with dysregulated cellular growth.
  • 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).
  • 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 1):474-8).
  • the invention provides methods for monitoring 161P2F10B gene products by determining the status of 161P2F10B 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 161P2F10B gene products in a corresponding normal sample.
  • the presence of aberrant 161P2F10B 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.
  • the invention provides assays useful in determining the presence of cancer in an individual, comprising detecting a significant increase in 161P2F10B 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 161P2F10B mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I.
  • the presence of significant 161P2F10B 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 161P2F10B mRNA or express it at lower levels.
  • 161P2F10B status is determined at the protein level rather than at the nucleic acid level.
  • a method comprises determining the level of 161P2F10B protein expressed by cells in a test tissue sample and comparing the level so determined to the level of 161P2F10B expressed in a corresponding normal sample.
  • the presence of 161P2F10B protein is evaluated, for example, using immunohistochemical methods.
  • 161P2F10B antibodies or binding partners capable of detecting 161P2F10B protein expression are used in a variety of assay formats well known in the art for this purpose.
  • 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).
  • a mutation in the sequence of 161P2F10B may be indicative of the presence or promotion of a tumor.
  • Such assays therefore have diagnostic and predictive value where a mutation in 161P2F10B indicates a potential loss of function or increase in tumor growth.
  • 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 161P2F10B gene products are observed by the Northern, Southern, Western, PCR and DNA sequencing protocols discussed herein.
  • other methods for observing perturbations in nucleotide and amino acid sequences such as single strand conformation polymorphism analysis are well known in the art (see, e.g., U.S. Pat. Nos. 5,382,510 issued 7 Sep. 1999, and 5,952,170 issued 17 Jan. 1995).
  • methylation status of a 161P2F10B 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.
  • 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)).
  • methylation-sensitive restriction enzymes that cannot cleave sequences that contain methylated CpG sites to assess the methylation status of CpG islands.
  • MSP methylation specific PCR
  • MSP methylation specific PCR
  • 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 161P2F10B.
  • 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.
  • 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 161P2F10B expression.
  • the presence of RT-PCR amplifiable 161P2F10B mRNA provides an indication of the presence of cancer.
  • RT-PCR assays are well known in the art. RT-PCR detection assays for tumor cells in peripheral blood are currently being evaluated for use in the diagnosis and management of a number of human solid tumors. In the prostate cancer field, these include RT-PCR assays for the detection of cells expressing PSA and PSM (Verkaik et al., 1997, Urol. Res. 25:373-384; Ghossein et al., 1995, J. Clin. Oncol. 13:1195-2000; Heston et al., 1995, Clin. Chem. 41:1687-1688).
  • a further aspect of the invention is an assessment of the susceptibility that an individual has for developing cancer.
  • a method for predicting susceptibility to cancer comprises detecting 161P2F10B mRNA or 161P2F10B protein in a tissue sample, its presence indicating susceptibility to cancer, wherein the degree of 161P2F10B mRNA expression correlates to the degree of susceptibility.
  • the presence of 161P2F10B in prostate or other tissue is examined, with the presence of 161P2F10B in the sample providing an indication of prostate cancer susceptibility (or the emergence or existence of a prostate tumor).
  • the presence of one or more perturbations in 161P2F10B gene products in the sample is an indication of cancer susceptibility (or the emergence or existence of a tumor).
  • a method for gauging aggressiveness of a tumor comprises determining the level of 161P2F10B mRNA or 161P2F10B protein expressed by tumor cells, comparing the level so determined to the level of 161P2F10B mRNA or 161P2F10B protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of 161P2F10B mRNA or 161P2F10B protein expression in the tumor sample relative to the normal sample indicates the degree of aggressiveness.
  • aggressiveness of a tumor is evaluated by determining the extent to which 161P2F10B is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors.
  • Another embodiment is the evaluation of the integrity of 161P2F10B 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.
  • methods for observing the progression of a malignancy in an individual over time comprise determining the level of 161P2F10B mRNA or 161P2F10B protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of 161P2F10B mRNA or 161P2F10B protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of 161P2F10B mRNA or 161P2F10B protein expression in the tumor sample over time provides information on the progression of the cancer.
  • the progression of a cancer is evaluated by determining 161P2F10B expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity 161P2F10B 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.
  • Another embodiment of the invention is directed to methods for observing a coincidence between the expression of 161P2F10B gene and 161P2F10B gene products (or perturbations in 161P2F10B gene and 161P2F10B gene products) and a factor that is associated with malignancy, as a means for diagnosing and prognosticating the status of a tissue sample.
  • factors associated with malignancy can be utilized, such as the expression of genes associated with malignancy as well as gross cytological observations (see, e.g., Bocking et al., 1984, Anal. Quant. Cytol.
  • Standard methods for the detection and quantification of 161P2F10B mRNA include in situ hybridization using labeled 161P2F10B riboprobes, Northern blot and related techniques using 161P2F10B polynucleotide probes, RT-PCR analysis using primers specific for 161P2F10B, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like.
  • semi-quantitative RT-PCR is used to detect and quantify 161P2F10B mRNA expression.
  • Any number of primers capable of amplifying 161P2F10B can be used for this purpose, including but not limited to the various primer sets specifically described herein.
  • polyclonal or monoclonal antibodies specifically reactive with the wild-type 161P2F10B protein can be used in an immunohistochemical assay of biopsied tissue.
  • the 161P2F10B protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with 161P2F10B, as well as pathways activated by 161P2F10B via any one of a variety of art accepted protocols.
  • one can utilize one of the so-called interaction trap systems also referred to as the “two-hybrid assay”.
  • molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed.
  • Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. Nos.
  • peptide libraries can be screen peptide libraries to identify molecules that interact with 161P2F10B protein sequences.
  • peptides that bind to 161P2F10B 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 161P2F10B protein(s).
  • peptides having a wide variety of uses 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 161P2F10B protein sequences are disclosed for example in U.S. Pat. Nos. 5,723,286 issued 3 Mar. 1998 and 5,733,731 issued 31 Mar. 1998.
  • 161P2F10B protein-protein interactions mediated by 161P2F10B. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun. 1999, 261:646-51). 161P2F10B protein can be immunoprecipitated from 161P2F10B-expressing cell lines using anti-161P2F10B antibodies. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of 161P2F10B and a His-tag (vectors mentioned above). The immunoprecipitated complex can be examined for protein association by procedures such as Western blotting, 35S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
  • Small molecules and ligands that interact with 161P2F10B 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 161P2F10B'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.
  • small molecules that modulate 161P2F10B-related ion channel, protein pump, or cell communication functions are identified and used to treat patients that have a cancer that expresses 161P2F10B (see, e.g., Hille, B., Ionic Channels of Excitable Membranes 2nd Ed., Sinauer Assoc., Sunderland, Mass., 1992).
  • ligands that regulate 161P2F10B function can be identified based on their ability to bind 161P2F10B and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued 27 Jul.
  • hybrid ligands in which at least one ligand is a small molecule.
  • cells engineered to express a fusion protein of 161P2F10B 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 161P2F10B.
  • An embodiment of this invention comprises a method of screening for a molecule that interacts with a 161P2F10B amino acid sequence shown in FIG. 1 , comprising the steps of contacting a population of molecules with a 161P2F10B amino acid sequence, allowing the population of molecules and the 161P2F10B amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the 161P2F10B amino acid sequence, and then separating molecules that do not interact with the 161P2F10B amino acid sequence from molecules that do.
  • the method further comprises purifying, characterizing and identifying a molecule that interacts with the 161P2F10B amino acid sequence. The identified molecule can be used to modulate a function performed by 161P2F10B.
  • the 161P2F10B amino acid sequence is contacted with a library of peptides.
  • 161P2F10B as a protein that is normally expressed in a restricted set of tissues, but which is also expressed in cancers such as those listed in Table I, opens a number of therapeutic approaches to the treatment of such cancers.
  • targeted antitumor therapies have been useful even when the targeted protein is expressed on normal tissues, even vital normal organ tissues.
  • a vital organ is one that is necessary to sustain life, such as the heart or colon.
  • a non-vital organ is one that can be removed whereupon the individual is still able to survive. Examples of non-vital organs are ovary, breast, and prostate.
  • Herceptin® is an FDA approved pharmaceutical that consists of an antibody which is immunoreactive with the protein variously known as HER2, HER2/neu, and erb-b-2. It is marketed by Genentech and has been a commercially successful antitumor agent. Herceptin® sales reached almost $400 million in 2002. Herceptin® is a treatment for HER2 positive metastatic breast cancer. However, the expression of HER2 is not limited to such tumors. The same protein is expressed in a number of normal tissues. In particular, it is known that HER2/neu is present in normal kidney and heart, thus these tissues are present in all human recipients of Herceptin.
  • HER2/neu in normal kidney is also confirmed by Latif, Z., et al., B.J.U. International (2002) 89:5-9.
  • this article which evaluated whether renal cell carcinoma should be a preferred indication for anti-HER2 antibodies such as Herceptin
  • both protein and mRNA are produced in benign renal tissues.
  • HER2/neu protein was strongly overexpressed in benign renal tissue.
  • Herceptin is a very useful, FDA approved, and commercially successful drug.
  • the effect of Herceptin on cardiac tissue i.e., “cardiotoxicity,” has merely been a side effect to treatment.
  • cardiac tissue i.e., “cardiotoxicity”
  • significant cardiotoxicity occurred in a very low percentage of patients.
  • To minimize cariotoxicity there is a more stringent entry requirement for the treatment with HER2/neu.
  • Factors such as predisposition to heart condition are evaluated before treatment can occur.
  • kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever.
  • kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever.
  • cardiac tissue has manifested any appreciable side effect at all.
  • EGFR epidermal growth factor receptor
  • Erbitux InClone
  • a target protein in normal tissue does not defeat the utility of a targeting agent for the protein as a therapeutic for certain tumors in which the protein is also overexpressed.
  • expression in vital organs is not in and of itself detrimental.
  • organs regarded as dispensible such as the prostate and ovary, can be removed without affecting mortality.
  • some vital organs are not affected by normal organ expression because of an immunoprivilege.
  • Immunoprivileged organs are organs that are protected from blood by a blood-organ barrier and thus are not accessible to immunotherapy. Examples of immunoprivileged organs are the brain and testis.
  • therapeutic approaches that inhibit the activity of a 161P2F10B protein are useful for patients suffering from a cancer that expresses 161P2F10B.
  • These therapeutic approaches generally fall into three classes.
  • the first class modulates 161P2F10B function as it relates to tumor cell growth leading to inhibition or retardation of tumor cell growth or inducing its killing.
  • the second class comprises various methods for inhibiting the binding or association of a 161P2F10B protein with its binding partner or with other proteins.
  • the third class comprises a variety of methods for inhibiting the transcription of a 161P2F10B gene or translation of 161P2F10B mRNA.
  • the invention provides cancer vaccines comprising a 161P2F10B-related protein or 161P2F10B-related nucleic acid.
  • cancer vaccines prevent and/or treat 161P2F10B-expressing cancers with minimal or no effects on non-target tissues.
  • the use of a tumor antigen in a vaccine that generates cell-mediated humoral immune responses as anti-cancer therapy is well known in the art and has been employed in prostate cancer using human PSMA and rodent PAP immunogens (Hodge et al., 1995, Int. J. Cancer 63:231-237; Fong et al., 1997, J. Immunol. 159:3113-3117).
  • Such methods can be readily practiced by employing a 161P2F10B-related protein, or a 161P2F10B-encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the 161P2F10B immunogen (which typically comprises a number of T-cell epitopes or antibody).
  • 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(1):66-78; Maruyama et al., Cancer Immunol Immunother 2000 June 49(3):123-32) Briefly, such methods of generating an immune response (e.g.
  • cell-mediated and/or humoral in a mammal, comprise the steps of: exposing the mammal's immune system to an immunoreactive epitope (e.g. an epitope present in a 161P2F10B protein shown in FIG. 1 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).
  • an immunoreactive epitope e.g. an epitope present in a 161P2F10B protein shown in FIG. 1 or analog or homolog thereof
  • 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.
  • lipopeptides e.g., Vitiello, A. et al., J. Clin. Invest. 95:341, 1995
  • PLG poly(DL-lactide-co-glycolide)
  • Toxin-targeted delivery technologies also known as receptor mediated targeting, such as those of Avant Immunotherapeutics, Inc. (Needham, Mass.) may also be used.
  • the vaccine and antibody 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.
  • CTL epitopes can be determined using specific algorithms to identify peptides within 161P2F10B protein that bind corresponding HLA alleles (e.g., Brown University, BIMAS, and SYFPEITHI.
  • a 161P2F10B immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables previously disclosed 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)).
  • 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.
  • 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.
  • 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.
  • 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 161P2F10B protein) so that an immune response is generated.
  • a protein e.g. a 161P2F10B protein
  • a typical embodiment consists of a method for generating an immune response to 161P2F10B in a host, by contacting the host with a sufficient amount of at least one 161P2F10B B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the 161P2F10B B cell or cytotoxic T-cell epitope or analog thereof.
  • a specific embodiment consists of a method of generating an immune response against a 161P2F10B-related protein or a man-made multiepitopic peptide comprising: administering 161P2F10B immunogen (e.g.
  • a 161P2F10B protein or a peptide fragment thereof, a 161P2F10B fusion protein or analog etc. in a vaccine preparation to a human or another mammal.
  • vaccine preparations further contain a suitable adjuvant (see, e.g., U.S. Pat. No. 6,146,635) or a universal helper epitope such as a PADRETM peptide (Epimmune Inc., San Diego, Calif.; see, e.g., Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res.
  • An alternative method comprises generating an immune response in an individual against a 161P2F10B 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 161P2F10B immunogen, the DNA sequence operatively linked to regulatory sequences which control the expression of the DNA sequence; wherein the DNA molecule is taken up by cells, the DNA sequence is expressed in the cells and an immune response is generated against the immunogen (see, e.g., U.S. Pat. No. 5,962,428).
  • a genetic vaccine facilitator such as anionic lipids; saponins; lectins; estrogenic compounds; hydroxylated lower alkyls; dimethyl sulfoxide; and urea is also administered.
  • an antiidiotypic antibody can be administered that mimics 161P2F10B, in order to generate a response to the target antigen.
  • 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 161P2F10B.
  • Constructs comprising DNA encoding a 161P2F10B-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 161P2F10B protein/immunogen.
  • a vaccine comprises a 161P2F10B-related protein.
  • 161P2F10B-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a 161P2F10B protein.
  • Various prophylactic and therapeutic genetic immunization techniques known in the art can be used. Nucleic acid-based delivery is described, for instance, in Wolff et. al., Science 247:1465 (1990) as well as U.S. Pat. Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; WO 98/04720.
  • 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. Pat. No. 5,922,687).
  • proteins of the invention can be expressed via viral or bacterial vectors.
  • viral gene delivery systems that can be used in the practice of the invention include, but are not limited to, vaccinia, fowlpox, canarypox, adenovirus, influenza, poliovirus, adeno-associated virus, lentivirus, and Sindbis virus (see, e.g., Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Inst. 87:982-990 (1995)).
  • Non-viral delivery systems can also be employed by introducing naked DNA encoding a 161P2F10B-related protein into the patient (e.g., intramuscularly or intradermally) to induce an anti-tumor response.
  • Vaccinia virus is used, for example, as a vector to express nucleotide sequences that encode the peptides of the invention. Upon introduction into a host, the recombinant vaccinia virus expresses the protein immunogenic peptide, and thereby elicits a host immune response.
  • Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848.
  • Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al., Nature 351:456-460 (1991).
  • 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 an
  • gene delivery systems are used to deliver a 161P2F10B-related nucleic acid molecule.
  • the full-length human 161P2F10B cDNA is employed.
  • 161P2F10B nucleic acid molecules encoding specific cytotoxic T lymphocyte (CTL) and/or antibody epitopes are employed.
  • APCs antigen presenting cells
  • DC dendritic cells
  • DRCs antigen presenting cells
  • DRCs dendritic cells
  • MHC class I and II molecules MHC class I and II molecules
  • B7 co-stimulator B7 co-stimulator
  • IL-12 IL-12
  • PSMA prostate-specific membrane antigen
  • dendritic cells can be used to present 161P2F10B peptides to T cells in the context of MHC class I or II molecules.
  • autologous dendritic cells are pulsed with 161P2F10B peptides capable of binding to MHC class I and/or class II molecules.
  • dendritic cells are pulsed with the complete 161P2F10B protein.
  • Yet another embodiment involves engineering the overexpression of a 161P2F10B 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.
  • Cells that express 161P2F10B can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.
  • 161P2F10B 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).
  • 161P2F10B is expressed by cancer cells of various lineages relative to corresponding normal cells, systemic administration of 161P2F10B-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 161P2F10B are useful to treat 161P2F10B-expressing cancers systemically, either as conjugates with a toxin or therapeutic agent, or as naked antibodies capable of inhibiting cell proliferation or function.
  • 161P2F10B antibodies can be introduced into a patient such that the antibody binds to 161P2F10B 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 161P2F10B, inhibition of ligand binding or signal transduction pathways, modulation of tumor cell differentiation, alteration of tumor angiogenesis factor profiles, and/or apoptosis.
  • antibodies can be used to specifically target and bind immunogenic molecules such as an immunogenic region of a 161P2F10B sequence shown in FIG. 1 .
  • skilled artisans understand that it is routine to conjugate antibodies to cytotoxic agents (see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)).
  • cytotoxic agents see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)
  • the cytotoxic agent When cytotoxic and/or therapeutic agents are delivered directly to cells, such as by conjugating them to antibodies specific for a molecule expressed by that cell (e.g. 161P2F10B), the cytotoxic agent will exert its known biological effect (i.e. cytotoxicity) on those cells.
  • compositions and methods for using antibody-cytotoxic agent conjugates to kill cells are known in the art.
  • 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-161P2F10B antibody) that binds to a marker (e.g. 161P2F10B) expressed, accessible to binding or localized on the cell surfaces.
  • a targeting agent e.g. an anti-161P2F10B antibody
  • a marker e.g. 161P2F10B
  • a typical embodiment is a method of delivering a cytotoxic and/or therapeutic agent to a cell expressing 161P2F10B, comprising conjugating the cytotoxic agent to an antibody that immunospecifically binds to a 161P2F10B 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-161P2F10B 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.
  • 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)
  • breast cancer Shepard et al., 1991, J. Clin. Immunol. 11:117-127.
  • Some therapeutic approaches involve conjugation of naked antibody to a toxin or radioisotope, such as the conjugation of Y91 or I131 to anti-CD20 antibodies (e.g., ZevalinTM, IDEC Pharmaceuticals Corp.
  • antibodies and other therapeutic agents such as HerceptinTM (trastuzu MAb) with paclitaxel (Genentech, Inc.).
  • the antibodies can be conjugated to a therapeutic agent.
  • 161P2F10B antibodies can be administered in conjunction with radiation, chemotherapy or hormone ablation.
  • antibodies can be conjugated to a toxin such as calicheamicin (e.g., MylotargTM, Wyeth-Ayerst, Madison, N.J., a recombinant humanized IgG4 kappa antibody conjugated to antitumor antibiotic calicheamicin) or a maytansinoid (e.g., taxane-based Tumor-Activated Prodrug, TAP, platform, ImmunoGen, Cambridge, Mass., also see e.g., U.S. Pat. No. 5,416,064) or Auristatin E (Nat. Biotechnol. 2003 July; 21(7):778-84. (Seattle Genetics)).
  • calicheamicin e.g., MylotargTM, Wyeth-Ayerst, Madison, N.J., a recombinant humanized IgG4 kappa antibody conjugated to antitumor antibiotic calicheamicin
  • a maytansinoid e
  • 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.
  • antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment.
  • 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.
  • 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 161P2F10B expression, preferably using immunohistochemical assessments of tumor tissue, quantitative 161P2F10B imaging, or other techniques that reliably indicate the presence and degree of 161P2F10B 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-161P2F10B 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.
  • anti-161P2F10B monoclonal antibodies MAbs
  • ADCC antibody-dependent cell cytotoxicity
  • anti-161P2F10B MAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express 161P2F10B.
  • 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-161P2F10B 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.
  • 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 161P2F10B antigen with high affinity but exhibit low or no antigenicity in the patient.
  • Therapeutic methods of the invention contemplate the administration of single anti-161P2F10B MAbs as well as combinations, or cocktails, of different MAbs (i.e. 161P2F10B MAbs or Mabs that bind another protein).
  • Such MAb cocktails can have certain advantages inasmuch as they contain MAbs that target different epitopes, exploit different effector mechanisms or combine directly cytotoxic MAbs with MAbs that rely on immune effector functionality. Such MAbs in combination can exhibit synergistic therapeutic effects.
  • 161P2F10B MAbs can be administered concomitantly with other therapeutic modalities, including but not limited to various chemotherapeutic and biologic agents, androgen-blockers, immune modulators (e.g., IL-2, GM-CSF), surgery or radiation.
  • the 161P2F10B MAbs are administered in their “naked” or unconjugated form, or can have a therapeutic agent(s) conjugated to them.
  • 161P2F10B Mab 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 161P2F10B Mab preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range, including but not limited to, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight.
  • IV intravenous injection
  • doses in the range of 10-1000 mg MAb per week are effective and well tolerated.
  • 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 MAb preparation represents an acceptable dosing regimen.
  • 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.
  • 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 MAbs used, the degree of 161P2F10B expression in the patient, the extent of circulating shed 161P2F10B 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.
  • patients should be evaluated for the levels of 161P2F10B in a given sample (e.g. the levels of circulating 161P2F10B antigen and/or 161P2F10B expressing cells) in order to assist in the determination of the most effective dosing regimen, etc.
  • levels of 161P2F10B in a given sample e.g. the levels of circulating 161P2F10B antigen and/or 161P2F10B expressing cells
  • Such evaluations are also used for monitoring purposes throughout therapy, and are useful to gauge therapeutic success in combination with the evaluation of other parameters (for example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).
  • Anti-idiotypic anti-161P2F10B antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a 161P2F10B-related protein.
  • the generation of anti-idiotypic antibodies is well known in the art; this methodology can readily be adapted to generate anti-idiotypic anti-161P2F10B antibodies that mimic an epitope on a 161P2F10B-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 anti-idiotypic antibody can be used in cancer vaccine strategies.
  • An object of the present invention is to provide 161P2F10B Mabs, which inhibit or retard the growth of tumor cells expressing 161P2F10B.
  • a further object of this invention is to provide methods to inhibit angiogenesis and other biological functions and thereby reduce tumor growth in mammals, preferably humans, using such 161P2F10B Mabs, and in particular using such 161P2F10B Mabs combined with other drugs or immunologically active treatments, including but not limited to: Avastin® (bevacizumab), Sutent® (sunitinib malate), Nexavar® (Sorafinib tosylate), Taxotere® (docetaxel), Interleukin-2 (a.k.a. Proleukin®, IL-2, or Aldesleukin), or Interferon Alpha (Interferon-Alpha-2a, or Interferon-Alpha-2b) and others in the art known to treat renal and other cancers.
  • tumors including human tumors
  • 161P2F10B antibodies in conjunction with chemotherapeutic agents or radiation or combinations thereof.
  • the inhibition of tumor growth by a 161P2F10B antibody is enhanced more than expected when combined with chemotherapeutic agents or radiation or combinations thereof.
  • Synergy may be shown, for example, by greater inhibition of tumor growth with combined treatment than would be expected from a treatment of only 161P2F10B antibodies or the additive effect of treatment with a 161P2F10B antibody and a chemotherapeutic agent or radiation.
  • synergy is demonstrated by remission of the cancer where remission is not expected from treatment either from a naked 161P2F10B antibody or with treatment using an additive combination of a 161P2F10B antibody and a chemotherapeutic agent or radiation.
  • the method for inhibiting growth of tumor cells using a 161P2F10B antibody and a combination of chemotherapy or radiation or both comprises administering the 161P2F10B antibody before, during, or after commencing chemotherapy or radiation therapy, as well as any combination thereof (i.e. before and during, before and after, during and after, or before, during, and after commencing the chemotherapy and/or radiation therapy).
  • the 161P2F10B antibody is typically administered between 1 and 60 days, preferably between 3 and 40 days, more preferably between 5 and 12 days before commencing radiation therapy and/or chemotherapy.
  • the method is performed in a manner that will provide the most efficacious treatment and ultimately prolong the life of the patient.
  • chemotherapeutic agents can be accomplished in a variety of ways including systemically by the parenteral and enteral routes.
  • the 161P2F10B antibody and the chemotherapeutic agent are administered as separate molecules.
  • the 161P2F10B antibody is attached, for example, by conjugation, to a chemotherapeutic agent. (See the Example entitled “Human Clinical Trials for the Treatment and Diagnosis of Human Carcinomas through use of 161P2F10B Mabs”).
  • chemotherapeutic agents or chemotherapy include cisplatin, dacarbazine (DTIC), dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU), doxorubicin (adriamycin), daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel (taxol), docetaxel (taxotere), aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, floxuridine, fludarabine, hydroxyurea, ifosfamide, interferon alpha, leuprolide, megestrol, melphalan, mercaptopurine, plicamycin,
  • the source of radiation used in combination with a 161P2F10B Mab, can be either external or internal to the patient being treated.
  • the therapy is known as external beam radiation therapy (EBRT).
  • EBRT external beam radiation therapy
  • BT brachytherapy
  • the above described therapeutic regimens may be further combined with additional cancer treating agents and/or regimes, for example additional chemotherapy, cancer vaccines, signal transduction inhibitors, agents useful in treating abnormal cell growth or cancer, antibodies (e.g. Anti-CTLA-4 antibodies as described in WO/2005/092380 (Pfizer)) or other ligands that inhibit tumor growth by binding to IGF-1R, and cytokines.
  • additional chemotherapy e.g., cancer vaccines, signal transduction inhibitors, agents useful in treating abnormal cell growth or cancer
  • antibodies e.g. Anti-CTLA-4 antibodies as described in WO/2005/092380 (Pfizer)
  • other ligands that inhibit tumor growth by binding to IGF-1R, and cytokines.
  • chemotherapeutic agents described above may be used.
  • growth factor inhibitors for example anti-estrogens such as Nolvadex (tamoxifen) or, anti-androgens such as Casodex (4′-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3-′-(trifluoromethyl)propionanilide) may be used.
  • anti-hormones for example anti-estrogens such as Nolvadex (tamoxifen) or, anti-androgens such as Casodex (4′-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3-′-(trifluoromethyl)propionanilide) may be used.
  • cancer vaccines may be, without limitation, those comprised of cancer-associated antigens (e.g. BAGE, carcinoembryonic antigen (CEA), EBV, GAGE, gp100 (including gp100:209-217 and gp100:280-288, among others), HBV, HER-2/neu, HPV, HCV, MAGE, mammaglobin, MART-1/Melan-A, Mucin-1, NY-ESO-1, proteinase-3, PSA, RAGE, TRP-1, TRP-2, Tyrosinase (e.g., Tyrosinase: 368-376), WT-1), GM-CSF DNA and cell-based vaccines, dendritic cell vaccines, recombinant viral (e.g.
  • cancer-associated antigens e.g. BAGE, carcinoembryonic antigen (CEA), EBV, GAGE, gp100 (including gp100:209-217 and gp100:280
  • vaccinia virus vaccinia virus
  • HSP heat shock protein
  • useful vaccines also include tumor vaccines, such as those formed of melanoma cells, and can be autologous or allogeneic.
  • the vaccines may be, e.g., peptide, DNA or cell-based.
  • Vaccines may be administered prior to, or subsequent to, stem cell transplantation, and when chemotherapy is part of the regimen, a vaccine may be administered prior to chemotherapy.
  • the antibody of the invention may also be administered prior to chemotherapy.
  • Vaccine may also be administered after stem cell transplantation and in certain embodiments concomitantly with the antibody.
  • the above described treatments may also be used with signal transduction inhibitors, such as agents that can inhibit EGFR (epidermal growth factor receptor) responses, such as EGFR antibodies, EGF antibodies, and molecules that are EGFR inhibitors; VEGF (vascular endothelial growth factor) inhibitors, such as VEGF receptors and molecules that can inhibit VEGF; and erbB2 receptor inhibitors, such as organic molecules or antibodies that bind to the erbB2 receptor.
  • signal transduction inhibitors such as agents that can inhibit EGFR (epidermal growth factor receptor) responses, such as EGFR antibodies, EGF antibodies, and molecules that are EGFR inhibitors; VEGF (vascular endothelial growth factor) inhibitors, such as VEGF receptors and molecules that can inhibit VEGF; and erbB2 receptor inhibitors, such as organic molecules or antibodies that bind to the erbB2 receptor.
  • EGFR inhibitors are described in, for example in WO 95/19970 (published Jul. 27, 1995), WO 98/14451 (published Apr. 9, 1998), WO 98/02434 (published Jan. 22, 1998), and U.S. Pat. No. 5,747,498 (issued May 5, 1998), and such substances can be used in the present invention as described herein.
  • EGFR-inhibiting agents include, but are not limited to, the monoclonal antibodies ERBITUX (ImClone Systems Incorporated of New York, N.Y.), and ABX-EGF (Abgenix Inc.
  • VEGF inhibitors for example SU-5416 and SU-6668 (Sugen Inc. of South San Francisco, Calif.), can also be employed in combination with the antibody.
  • VEGF inhibitors are described for example in WO 99/24440 (published May 20, 1999), PCT International Application PCT/IB99/00797 (filed May 3, 1999), in WO 95/21613 (published Aug. 17, 1995), WO 99/61422 (published Dec. 2, 1999), U.S. Pat. No. 5,834,504 (issued Nov. 10, 1998), WO 98/50356 (published Nov. 12, 1998), U.S. Pat. No. 5,883,113 (issued Mar. 16, 1999), U.S. Pat. No. 5,886,020 (issued Mar.
  • ErbB2 receptor inhibitors such as GW-282974 (Glaxo Wellcome), and the monoclonal antibodies AR-209 (Aronex Pharmaceuticals Inc. of The Woodlands, Tex.) and 2B-1 (Chiron), can furthermore be combined with the antibody, for example those indicated in WO 98/02434 (published Jan. 22, 1998), WO 99/35146 (published Jul. 15, 1999), WO 99/35132 (published Jul. 15, 1999), WO 98/02437 (published Jan. 22, 1998), WO 97/13760 (published Apr. 17, 1997), WO 95/19970 (published Jul. 27, 1995), U.S. Pat. No. 5,587,458 (issued Dec. 24, 1996), and U.S. Pat.
  • ErbB2 receptor inhibitors useful in the present invention are also described in EP1029853 (published Aug. 23, 2000) and in WO 00/44728, (published Aug. 3, 2000).
  • the erbB2 receptor inhibitor compounds and substance described in the aforementioned PCT applications, U.S. patents, and U.S. provisional applications, as well as other compounds and substances that inhibit the erbB2 receptor, can be used with the antibody in accordance with the present invention.
  • the present treatment regimens may also be combined with antibodies or other ligands that inhibit tumor growth by binding to IGF-1R (insulin-like growth factor 1 receptor).
  • IGF-1R insulin-like growth factor 1 receptor
  • Specific anti-IGF-1R antibodies that can be used in the present invention include those described in PCT application PCT/US01/51113, filed Dec. 20, 2001 and published as WO02/053596.
  • the treatment regimens described herein may be combined with anti-angiogenesis agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors, can be used in conjunction with the antibody in the method of the invention.
  • anti-angiogenesis agents such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors
  • MMP-2 matrix-metalloproteinase 2
  • MMP-9 matrix-metalloproteinase 9
  • COX-II cyclooxygenase II
  • 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.
  • 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.
  • 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)).
  • CpG cytosine-phosphorothiolated-guanine-containing
  • the immune system of the host 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 161P2F10B antigen, or derives at least some therapeutic benefit when the antigen was tumor-associated.
  • class I peptide components may be desirable to combine with components that induce or facilitate neutralizing antibody and or helper T cell responses directed to the target antigen.
  • a preferred embodiment of such a composition comprises class I and class II epitopes in accordance with the invention.
  • An alternative embodiment of such a composition comprises a class I and/or class II epitope in accordance with the invention, along with a cross reactive HTL epitope such as PADRETM (Epimmune, San Diego, Calif.) molecule (described e.g., in U.S. Pat. No. 5,736,142).
  • a vaccine of the invention can also include antigen-presenting cells (APC), such as dendritic cells (DC), as a vehicle to present peptides of the invention.
  • APC antigen-presenting cells
  • DC dendritic cells
  • Vaccine compositions can be created in vitro, following dendritic cell mobilization and harvesting, whereby loading of dendritic cells occurs in vitro.
  • 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.
  • 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.
  • Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance.
  • this includes 3-4 epitopes that come from at least one tumor associated antigen (TAA).
  • TAA tumor associated antigen
  • 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.
  • 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.
  • 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.
  • 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.
  • a general objective is to provide the greatest number of epitopes per sequence.
  • 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.
  • 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.
  • 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.
  • potential peptide epitopes can also be selected on the basis of their conservancy.
  • 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.
  • 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.
  • a multi-epitope DNA plasmid encoding supermotif- and/or motif-bearing epitopes derived 161P2F10B, the PADRETM universal helper T cell epitope or multiple HTL epitopes from 161P2F10B, 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.
  • 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.
  • additional elements can be incorporated into the minigene design.
  • 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.
  • 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.
  • a promoter with a down-stream cloning site for minigene insertion a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g., ampicillin or kanamycin resistance).
  • Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.
  • 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.
  • mRNA stabilization sequences and sequences for replication in mammalian cells may also be considered for increasing minigene expression.
  • 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.
  • immunostimulatory sequences 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.
  • 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.
  • 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 (PADRETM, Epimmune, San Diego, Calif.).
  • Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction.
  • immunosuppressive molecules e.g. TGF- ⁇
  • TGF- ⁇ immunosuppressive molecules
  • Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli , followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, Calif.). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.
  • Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as “naked DNA,” is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be desirable. A variety of methods have been described, and new techniques may become available.
  • Cationic lipids, glycolipids, and fusogenic liposomes can also be used in the formulation (see, e.g., as described by WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Felgner, et al., Proc. Nat'l Acad. Sci. USA 84:7413 (1987).
  • peptides and compounds referred to collectively as protective, interactive, non-condensing compounds 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.
  • 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).
  • FACS fluorescence activated cell sorting
  • HTL epitopes 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.
  • 51Cr chromium-51
  • 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.
  • nucleic acids can be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253.
  • particles comprised solely of DNA are administered.
  • DNA can be adhered to particles, such as gold particles.
  • Minigenes can also be delivered using other bacterial or viral delivery systems well known in the art, e.g., an expression construct encoding epitopes of the invention can be incorporated into a viral vector such as vaccinia.
  • 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.
  • 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.
  • a CTL peptide can be directly linked to a T helper peptide, often CTL epitope/HTL epitope conjugates are linked by a spacer molecule.
  • the spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions.
  • the spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids.
  • the optionally present spacer need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues and sometimes 10 or more residues.
  • the CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide.
  • the amino terminus of either the immunogenic peptide or the T helper peptide may be acylated.
  • HTL peptide epitopes can also be modified to alter their biological properties. For example, they can be modified to include D-amino acids to increase their resistance to proteases and thus extend their serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase their biological activity.
  • a T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.
  • 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.
  • palmitic acid residues can be attached to the ⁇ - and ⁇ -amino groups of a lysine residue and then linked, e.g., via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic peptide.
  • the lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant, e.g., incomplete Freund's adjuvant.
  • a particularly effective immunogenic composition comprises palmitic acid attached to ⁇ - and ⁇ -amino groups of Lys, which is attached via linkage, e.g., Ser-Ser, to the amino terminus of the immunogenic peptide.
  • E. coli lipoproteins such as tripalmitoyl-5-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 prime specifically an immune response to the target antigen.
  • P3CSS tripalmitoyl-5-glycerylcysteinlyseryl-serine
  • two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses.
  • 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 ProgenipoietinTM (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.
  • a vaccine comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces.
  • the DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to 161P2F10B.
  • 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.
  • HTL helper T cell
  • a vaccine in accordance with the invention is used to treat a cancer which expresses or overexpresses 161P2F10B.
  • Antigenic 161P2F10B-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.
  • APC antigen-presenting cells
  • the cells After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cell (e.g., a tumor cell).
  • CTL destroy
  • HTL facilitate destruction
  • Transfected dendritic cells may also be used as antigen presenting cells.
  • compositions of the invention are typically used to treat and/or prevent a cancer that expresses or overexpresses 161P2F10B.
  • peptide and/or nucleic acid compositions are administered to a patient in an amount sufficient to elicit an effective B cell, CTL and/or HTL response to the antigen and to cure or at least partially arrest or slow symptoms and/or complications.
  • An amount adequate to accomplish this is defined as “therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the particular composition administered, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician.
  • the immunogenic peptides of the invention are generally administered to an individual already bearing a tumor that expresses 161P2F10B.
  • 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.
  • administration should generally begin at the first diagnosis of 161P2F10B-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 161P2F10B, a vaccine comprising 161P2F10B-specific CTL may be more efficacious in killing tumor cells in patient with advanced disease than alternative embodiments.
  • 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 ⁇ g and the higher value is about 10,000; 20,000; 30,000; or 50,000 ⁇ g.
  • Dosage values for a human typically range from about 500 ⁇ g to about 50,000 ⁇ g per 70 kilogram patient.
  • Boosting dosages of between about 1.0 ⁇ g to about 50,000 ⁇ g of peptide pursuant to a boosting regimen over weeks to months may be administered depending upon the patient's response and condition as determined by measuring the specific activity of CTL and HTL obtained from the patient's blood. Administration should continue until at least clinical symptoms or laboratory tests indicate that the neoplasia, has been eliminated or reduced and for a period thereafter.
  • the dosages, routes of administration, and dose schedules are adjusted in accordance with methodologies known in the art.
  • the peptides and compositions of the present invention are employed in serious disease states, that is, life-threatening or potentially life threatening situations.
  • life-threatening or potentially life threatening situations in certain embodiments, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these peptide compositions relative to these stated dosage amounts.
  • the vaccine compositions of the invention can also be used purely as prophylactic agents.
  • 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 ⁇ g and the higher value is about 10,000; 20,000; 30,000; or 50,000 ⁇ g.
  • Dosage values for a human typically range from about 500 ⁇ g to about 50,000 ⁇ g per 70 kilogram patient. This is followed by boosting dosages of between about 1.0 ⁇ g to about 50,000 ⁇ g 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.
  • compositions for therapeutic treatment are intended for parenteral, topical, oral, nasal, intrathecal, or local (e.g. as a cream or topical ointment) administration.
  • the pharmaceutical compositions are administered parentally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly.
  • compositions for parenteral administration which comprise a solution of the immunogenic peptides dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • 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.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • auxiliary substances 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.
  • concentration of peptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • a human unit dose form of a composition is typically included in a pharmaceutical composition that comprises a human unit dose of an acceptable carrier, in one embodiment an aqueous carrier, and is administered in a volume/quantity that is known by those of skill in the art to be used for administration of such compositions to humans (see, e.g., Remington's Pharmaceutical Sciences, 17th Edition, A. Gennaro, Editor, Mack Publishing Co., Easton, Pa., 1985).
  • a peptide dose for initial immunization can be from about 1 to about 50,000 ⁇ g, generally 100-5,000 ⁇ g, for a 70 kg patient.
  • an initial immunization may be performed using an expression vector in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites.
  • the nucleic acid (0.1 to 1000 ⁇ g) can also be administered using a gene gun.
  • a booster dose is then administered.
  • the booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5 ⁇ 10 9 pfu.
  • a treatment generally involves repeated administration of the anti-161P2F10B 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.
  • IV intravenous injection
  • doses in the range of 10-500 mg MAb per week are effective and well tolerated.
  • 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-161P2F10B MAb preparation represents an acceptable dosing regimen.
  • 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 161P2F10B expression in the patient, the extent of circulating shed 161P2F10B antigen, the desired steady-state concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.
  • Non-limiting preferred human unit doses are, for example, 500 ⁇ g-1 mg, 1 mg-50 mg, 50 mg-100 mg, 100 mg-200 mg, 200 mg-300 mg, 400 mg-500 mg, 500 mg-600 mg, 600 mg-700 mg, 700 mg-800 mg, 800 mg-900 mg, 900 mg-1 g, or 1 mg-700 mg.
  • the dose is in a range of 2-5 mg/kg body weight, e.g., with follow on weekly doses of 1-3 mg/kg; 0.5 mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg/kg body weight followed, e.g., in two, three or four weeks by weekly doses; 0.5-10 mg/kg body weight, e.g., followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400 mg m 2 of body area weekly; 1-600 mg m 2 of body area weekly; 225-400 mg m 2 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.
  • human unit dose forms of polynucleotides comprise a suitable dosage range or effective amount that provides any therapeutic effect.
  • a therapeutic effect depends on a number of factors, including the sequence of the polynucleotide, molecular weight of the polynucleotide and route of administration. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like.
  • a dosage range may be selected from, for example, an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg.
  • 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.
  • 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.
  • parenteral routes of administration may require higher doses of polynucleotide compared to more direct application to the nucleotide to diseased tissue, as do polynucleotides of increasing length.
  • human unit dose forms of T-cells comprise a suitable dosage range or effective amount that provides any therapeutic effect.
  • 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 10 4 cells to about 10 6 cells, about 10 6 cells to about 10 8 cells, about 10 8 to about 10 11 cells, or about 10 8 to about 5 ⁇ 10 10 cells.
  • a dose may also about 10 6 cells/m 2 to about 10 10 cells/m 2 , or about 10 6 cells/m 2 to about 10 8 cells/m 2 .
  • 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.
  • the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a receptor prevalent among lymphoid cells (such as monoclonal antibodies which bind to the CD45 antigen) or with other therapeutic or immunogenic compositions.
  • a molecule which binds to a receptor prevalent among lymphoid cells such as monoclonal antibodies which bind to the CD45 antigen
  • liposomes either filled or decorated with a desired peptide of the invention can be directed to the site of lymphoid cells, where the liposomes then deliver the peptide compositions.
  • Liposomes for use in accordance with the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol.
  • lipids are 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. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.
  • a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells.
  • a liposome suspension containing a peptide may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.
  • nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • 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%.
  • 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.
  • the surfactant may constitute about 0.1%-20% by weight of the composition, preferably about 0.25-5%.
  • the balance of the composition is ordinarily propellant.
  • a carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery.
  • 161P2F10B 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 161P2F10B in normal tissues, and patient specimens”).
  • 161P2F10B 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)).
  • PSA prostate associated antigen PSA
  • 161P2F10B polynucleotides and polypeptides as well as 161P2F10B polynucleotide probes and anti-161P2F10B antibodies used to identify the presence of these molecules
  • 161P2F10B polynucleotide probes and anti-161P2F10B antibodies used to identify the presence of these molecules allows skilled artisans to utilize these molecules in methods that are analogous to those used, for example, in a variety of diagnostic assays directed to examining conditions associated with cancer.
  • Typical embodiments of diagnostic methods which utilize the 161P2F10B 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.)
  • 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.
  • the 161P2F10B polynucleotides described herein can be utilized in the same way to detect 161P2F10B overexpression or the metastasis of kidney and other cancers expressing this gene.
  • 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 161P2F10B polypeptides described herein can be utilized to generate antibodies for use in detecting 161P2F10B overexpression or the metastasis of kidney cells and cells of other cancers expressing this gene.
  • 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 161P2F10B polynucleotides and/or polypeptides can be used to provide evidence of metastasis.
  • tissue that does not normally contain 161P2F10B-expressing cells is found to contain 161P2F10B-expressing cells this finding is indicative of metastasis.
  • 161P2F10B 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 161P2F10B or express 161P2F10B at a different level are found to express 161P2F10B or have an increased expression of 161P2F10B (see, e.g., the 161P2F10B expression in the cancers listed in Table I and in patient samples etc. shown in the accompanying Figures).
  • 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 161P2F10B).
  • the use of immunohistochemistry to identify the presence of a 161P2F10B polypeptide within a tissue section can indicate an altered state of certain cells within that tissue. It is well understood in the art that the ability of an antibody to localize to a polypeptide that is expressed in cancer cells is a way of diagnosing presence of disease, disease stage, progression and/or tumor aggressiveness. Such an antibody can also detect an altered distribution of the polypeptide within the cancer cells, as compared to corresponding non-malignant tissue.
  • the 161P2F10B polypeptide and immunogenic compositions are also useful in view of the phenomena of altered subcellular protein localization in disease states. Alteration of cells from normal to diseased state causes changes in cellular morphology and is often associated with changes in subcellular protein localization/distribution. For example, cell membrane proteins that are expressed in a polarized manner in normal cells can be altered in disease, resulting in distribution of the protein in a non-polar manner over the whole cell surface.
  • normal breast epithelium is either negative for Her2 protein or exhibits only a basolateral distribution whereas malignant cells can express the protein over the whole cell surface (De Potter, et al, International Journal of Cancer, 44; 969-974 (1989): McCormick, et al, 117; 935-943 (2002)).
  • distribution of the protein may be altered from a surface only localization to include diffuse cytoplasmic expression in the diseased state. Such an example can be seen with MUC1 (Diaz, et al, The Breast Journal, 7: 40-45 (2001)).
  • the 161P2F10B protein and immune responses related thereto are very useful. Use of the 161P2F10B compositions allows those skilled in the art to make important diagnostic and therapeutic decisions.
  • Immunohistochemical reagents specific to 161P2F10B are also useful to detect metastases of tumors expressing 161P2F10B when the polypeptide appears in tissues where 161P2F10B is not normally produced.
  • 161P2F10B polypeptides and antibodies resulting from immune responses thereto are useful in a variety of important contexts such as diagnostic, prognostic, preventative and/or therapeutic purposes known to those skilled in the art.
  • 161P2F10B-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of 161P2F10B.
  • the amino acid or nucleic acid sequence of FIG. 1 or fragments of either, can be used to generate an immune response to a 161P2F10B antigen.
  • Antibodies or other molecules that react with 161P2F10B can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.
  • the invention includes various methods and compositions for inhibiting the binding of 161P2F10B to its binding partner or its association with other protein(s) as well as methods for inhibiting 161P2F10B function.
  • a recombinant vector that encodes single chain antibodies that specifically bind to 161P2F10B are introduced into 161P2F10B expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti-161P2F10B antibody is expressed intracellularly, binds to 161P2F10B protein, and thereby inhibits its function.
  • Methods for engineering such intracellular single chain antibodies are well known.
  • intracellular antibodies also known as “intrabodies”, are specifically targeted to a particular compartment within the cell, providing control over where the inhibitory activity of the treatment is focused. This technology has been successfully applied in the art (for review, see Richardson and Marasco, 1995, TIBTECH vol. 13).
  • Intrabodies have been shown to virtually eliminate the expression of otherwise abundant cell surface receptors (see, e.g., Richardson et al., 1995, Proc. Natl. Acad. Sci. USA 92: 3137-3141; Beerli et al., 1994, J. Biol. Chem. 289: 23931-23936; Deshane et al., 1994, Gene Ther. 1: 332-337).
  • Single chain antibodies comprise the variable domains of the heavy and light chain joined by a flexible linker polypeptide, and are expressed as a single polypeptide.
  • 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 target precisely the intrabody to the desired intracellular compartment.
  • 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.
  • cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.
  • intrabodies are used to capture 161P2F10B in the nucleus, thereby preventing its activity within the nucleus.
  • Nuclear targeting signals are engineered into such 161P2F10B intrabodies in order to achieve the desired targeting.
  • Such 161P2F10B intrabodies are designed to bind specifically to a particular 161P2F10B domain.
  • cytosolic intrabodies that specifically bind to a 161P2F10B protein are used to prevent 161P2F10B from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing 161P2F10B from forming transcription complexes with other factors).
  • recombinant molecules bind to 161P2F10B and thereby inhibit 161P2F10B function.
  • these recombinant molecules prevent or inhibit 161P2F10B from accessing/binding to its binding partner(s) or associating with other protein(s).
  • Such recombinant molecules can, for example, contain the reactive part(s) of a 161P2F10B specific antibody molecule.
  • the 161P2F10B binding domain of a 161P2F10B binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two 161P2F10B ligand binding domains linked to the Fc portion of a human IgG, such as human IgG 1 .
  • a human IgG such as human IgG 1
  • Such IgG portion can contain, for example, the CH 2 and CH 3 domains and the hinge region, but not the CH 1 domain.
  • dimeric fusion proteins are administered in soluble form to patients suffering from a cancer associated with the expression of 161P2F10B, whereby the dimeric fusion protein specifically binds to 161P2F10B and blocks 161P2F10B interaction with a binding partner.
  • Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.
  • the present invention also comprises various methods and compositions for inhibiting the transcription of the 161P2F10B gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of 161P2F10B mRNA into protein.
  • a method of inhibiting the transcription of the 161P2F10B gene comprises contacting the 161P2F10B gene with a 161P2F10B antisense polynucleotide.
  • a method of inhibiting 161P2F10B mRNA translation comprises contacting a 161P2F10B mRNA with an antisense polynucleotide.
  • a 161P2F10B specific ribozyme is used to cleave a 161P2F10B message, thereby inhibiting translation.
  • Such antisense and ribozyme based methods can also be directed to the regulatory regions of the 161P2F10B gene, such as 161P2F10B promoter and/or enhancer elements.
  • proteins capable of inhibiting a 161P2F10B gene transcription factor are used to inhibit 161P2F10B 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.
  • 161P2F10B factors that inhibit the transcription of 161P2F10B by interfering with 161P2F10B transcriptional activation are also useful to treat cancers expressing 161P2F10B.
  • factors that interfere with 161P2F10B processing are useful to treat cancers that express 161P2F10B. Cancer treatment methods utilizing such factors are also within the scope of the invention.
  • Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing 161P2F10B (i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 161P2F10B inhibitory molecules).
  • 161P2F10B i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 161P2F10B inhibitory molecules.
  • a number of gene therapy approaches are known in the art.
  • Recombinant vectors encoding 161P2F10B antisense polynucleotides, ribozymes, factors capable of interfering with 161P2F10B 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 can be evaluated using various in vitro and in vivo assay systems.
  • In vitro assays that evaluate therapeutic activity include cell growth assays, soft agar assays and other assays indicative of tumor promoting activity, binding assays capable of determining the extent to which a therapeutic composition will inhibit the binding of 161P2F10B to a binding partner, etc.
  • a 161P2F10B therapeutic composition can be evaluated in a suitable animal model.
  • xenogenic kidney 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).
  • PCT Patent Application WO98/16628 and U.S. Pat. No. 6,107,540 describe various xenograft models of human prostate cancer capable of recapitulating the development of primary tumors, micrometastasis, and the formation of osteoblastic metastases characteristic of late stage disease. Efficacy can be predicted using assays that measure inhibition of tumor formation, tumor regression or metastasis, and the like.
  • 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.
  • Suitable carriers include any material that when combined with the therapeutic composition retains the anti-tumor function of the therapeutic composition and is generally non-reactive with the patient's immune system. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like (see, generally, Remington's Pharmaceutical Sciences 16th Edition, A. Osal., Ed., 1980).
  • Therapeutic formulations can be solubilized and administered via any route capable of delivering the therapeutic composition to the tumor site.
  • Potentially effective routes of administration include, but are not limited to, intravenous, parenteral, intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like.
  • a preferred formulation for intravenous injection comprises the therapeutic composition in a solution of preserved bacteriostatic water, sterile unpreserved water, and/or diluted in polyvinylchloride or polyethylene bags containing 0.9% sterile Sodium Chloride for Injection, USP.
  • Therapeutic protein preparations can be lyophilized and stored as sterile powders, preferably under vacuum, and then reconstituted in bacteriostatic water (containing for example, benzyl alcohol preservative) or in sterile water prior to injection.
  • Dosages and administration protocols for the treatment of cancers using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.
  • screening is performed to identify modulators that induce or suppress a particular expression profile, suppress or induce specific pathways, preferably generating the associated phenotype thereby.
  • having identified differentially expressed genes important in a particular state screens are performed to identify modulators that alter expression of individual genes, either increase or decrease.
  • screening is performed to identify modulators that alter a biological function of the expression product of a differentially expressed gene. Again, having identified the importance of a gene in a particular state, screens are performed to identify agents that bind and/or modulate the biological activity of the gene product.
  • screens are done for genes that are induced in response to a candidate agent.
  • identifying a modulator one that suppresses a cancer expression pattern leading to a normal expression pattern, or a modulator of a cancer gene that leads to expression of the gene as in normal tissue
  • a screen is performed to identify genes that are specifically modulated in response to the agent. Comparing expression profiles between normal tissue and agent-treated cancer tissue reveals genes that are not expressed in normal tissue or cancer tissue, but are expressed in agent treated tissue, and vice versa.
  • agent-specific sequences are identified and used by methods described herein for cancer genes or proteins. In particular these sequences and the proteins they encode are used in marking or identifying agent-treated cells.
  • antibodies are raised against the agent-induced proteins and used to target novel therapeutics to the treated cancer tissue sample.
  • Proteins, nucleic acids, and antibodies of the invention are used in screening assays.
  • the cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing these sequences are used in screening assays, such as evaluating the effect of drug candidates on a “gene expression profile,” expression profile of polypeptides or alteration of biological function.
  • the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent (e.g., Davis, G F, et al, J Biol Screen 7:69 (2002); Zlokarnik, et al., Science 279:84-8 (1998); Heid, Genome Res 6:986-94, 1996).
  • the cancer proteins, antibodies, nucleic acids, modified proteins and cells containing the native or modified cancer proteins or genes are used in screening assays. That is, the present invention comprises methods for screening for compositions which modulate the cancer phenotype or a physiological function of a cancer protein of the invention. This is done on a gene itself or by evaluating the effect of drug candidates on a “gene expression profile” or biological function. In one embodiment, expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring after treatment with a candidate agent, see Zlokarnik, supra.
  • assays are executed directed to the genes and proteins of the invention. Assays are run on an individual nucleic acid or protein level. That is, having identified a particular gene as up regulated in cancer, test compounds are screened for the ability to modulate gene expression or for binding to the cancer protein of the invention. “Modulation” in this context includes an increase or a decrease in gene expression. The preferred amount of modulation will depend on the original change of the gene expression in normal versus tissue undergoing cancer, with changes of at least 10%, preferably 50%, more preferably 100-300%, and in some embodiments 300-1000% or greater.
  • a gene exhibits a 4-fold increase in cancer tissue compared to normal tissue, a decrease of about four-fold is often desired; similarly, a 10-fold decrease in cancer tissue compared to normal tissue a target value of a 10-fold increase in expression by the test compound is often desired.
  • Modulators that exacerbate the type of gene expression seen in cancer are also useful, e.g., as an upregulated target in further analyses.
  • the amount of gene expression is monitored using nucleic acid probes and the quantification of gene expression levels, or, alternatively, a gene product itself is monitored, e.g., through the use of antibodies to the cancer protein and standard immunoassays. Proteomics and separation techniques also allow for quantification of expression.
  • gene expression monitoring i.e., an expression profile
  • Such profiles will typically involve one or more of the genes of FIG. 1 .
  • cancer nucleic acid probes are attached to biochips to detect and quantify cancer sequences in a particular cell.
  • PCR can be used.
  • a series e.g., wells of a microtiter plate, can be used with dispensed primers in desired wells. A PCR reaction can then be performed and analyzed for each well.
  • Expression monitoring is performed to identify compounds that modify the expression of one or more cancer-associated sequences, e.g., a polynucleotide sequence set out in FIG. 1 .
  • a test modulator is added to the cells prior to analysis.
  • screens are also provided to identify agents that modulate cancer, modulate cancer proteins of the invention, bind to a cancer protein of the invention, or interfere with the binding of a cancer protein of the invention and an antibody or other binding partner.
  • high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such “combinatorial chemical libraries” are then screened in one or more assays to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds,” as compounds for screening, or as therapeutics.
  • combinatorial libraries of potential modulators are screened for an ability to bind to a cancer polypeptide or to modulate activity.
  • new chemical entities with useful properties are generated by identifying a chemical compound (called a “lead compound”) with some desirable property or activity, e.g., inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds.
  • HTS high throughput screening
  • gene expression monitoring is conveniently used to test candidate modulators (e.g., protein, nucleic acid or small molecule).
  • candidate modulators e.g., protein, nucleic acid or small molecule.
  • the target sequence is prepared using known techniques. For example, a sample is treated to lyse the cells, using known lysis buffers, electroporation, etc., with purification and/or amplification such as PCR performed as appropriate. For example, an in vitro transcription with labels covalently attached to the nucleotides is performed. Generally, the nucleic acids are labeled with biotin-FITC or PE, or with cy3 or cy5.
  • the target sequence can be labeled with, e.g., a fluorescent, a chemiluminescent, a chemical, or a radioactive signal, to provide a means of detecting the target sequence's specific binding to a probe.
  • the label also can be an enzyme, such as alkaline phosphatase or horseradish peroxidase, which when provided with an appropriate substrate produces a product that is detected.
  • the label is a labeled compound or small molecule, such as an enzyme inhibitor, that binds but is not catalyzed or altered by the enzyme.
  • the label also can be a moiety or compound, such as, an epitope tag or biotin which specifically binds to streptavidin.
  • the streptavidin is labeled as described above, thereby, providing a detectable signal for the bound target sequence. Unbound labeled streptavidin is typically removed prior to analysis.
  • these assays can be direct hybridization assays or can comprise “sandwich assays”, which include the use of multiple probes, as is generally outlined in U.S. Pat. Nos. 5,681,702; 5,597,909; 5,545,730; 5,594,117; 5,591,584; 5,571,670; 5,580,731; 5,571,670; 5,591,584; 5,624,802; 5,635,352; 5,594,118; 5,359,100; 5,124, 246; and 5,681,697.
  • the target nucleic acid is prepared as outlined above, and then added to the biochip comprising a plurality of nucleic acid probes, under conditions that allow the formation of a hybridization complex.
  • hybridization conditions are used in the present invention, including high, moderate and low stringency conditions as outlined above.
  • the assays are generally run under stringency conditions which allow formation of the label probe hybridization complex only in the presence of target.
  • Stringency can be controlled by altering a step parameter that is a thermodynamic variable, including, but not limited to, temperature, formamide concentration, salt concentration, chaotropic salt concentration pH, organic solvent concentration, etc. These parameters may also be used to control non-specific binding, as is generally outlined in U.S. Pat. No. 5,681,697. Thus, it can be desirable to perform certain steps at higher stringency conditions to reduce non-specific binding.
  • the reactions outlined herein can be accomplished in a variety of ways. Components of the reaction can be added simultaneously, or sequentially, in different orders, with preferred embodiments outlined below.
  • the reaction may include a variety of other reagents. These include salts, buffers, neutral proteins, e.g. albumin, detergents, etc. which can be used to facilitate optimal hybridization and detection, and/or reduce nonspecific or background interactions. Reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may also be used as appropriate, depending on the sample preparation methods and purity of the target.
  • the assay data are analyzed to determine the expression levels of individual genes, and changes in expression levels as between states, forming a gene expression profile.
  • the invention provides methods to identify or screen for a compound that modulates the activity of a cancer-related gene or protein of the invention.
  • the methods comprise adding a test compound, as defined above, to a cell comprising a cancer protein of the invention.
  • the cells contain a recombinant nucleic acid that encodes a cancer protein of the invention.
  • a library of candidate agents is tested on a plurality of cells.
  • the assays are evaluated in the presence or absence or previous or subsequent exposure of physiological signals, e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e., cell-cell contacts).
  • physiological signals e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e., cell-cell contacts).
  • the determinations are made at different stages of the cell cycle process. In this way, compounds that modulate genes or proteins of the invention are identified. Compounds with pharmacological activity are able to enhance or interfere with the activity of the cancer protein of the invention. Once identified, similar structures are evaluated to identify critical structural features of the compound.
  • a method of modulating (e.g., inhibiting) cancer cell division comprises administration of a cancer modulator.
  • a method of modulating (e.g., inhibiting) cancer is provided; the method comprises administration of a cancer modulator.
  • methods of treating cells or individuals with cancer are provided; the method comprises administration of a cancer modulator.
  • a method for modulating the status of a cell that expresses a gene of the invention comprises such art-accepted parameters such as growth, proliferation, survival, function, apoptosis, senescence, location, enzymatic activity, signal transduction, etc. of a cell.
  • a cancer inhibitor is an antibody as discussed above.
  • the cancer inhibitor is an antisense molecule.
  • a variety of cell growth, proliferation, and metastasis assays are known to those of skill in the art, as described herein.
  • the assays to identify suitable modulators are amenable to high throughput screening. Preferred assays thus detect enhancement or inhibition of cancer gene transcription, inhibition or enhancement of polypeptide expression, and inhibition or enhancement of polypeptide activity.
  • modulators evaluated in high throughput screening methods are proteins, often naturally occurring proteins or fragments of naturally occurring proteins.
  • proteins e.g., cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, are used.
  • libraries of proteins are made for screening in the methods of the invention.
  • Particularly preferred in this embodiment are libraries of bacterial, fungal, viral, and mammalian proteins, with the latter being preferred, and human proteins being especially preferred.
  • Particularly useful test compound will be directed to the class of proteins to which the target belongs, e.g., substrates for enzymes, or ligands and receptors.
  • Normal cells require a solid substrate to attach and grow. When cells are transformed, they lose this phenotype and grow detached from the substrate. For example, transformed cells can grow in stirred suspension culture or suspended in semi-solid media, such as semi-solid or soft agar. The transformed cells, when transfected with tumor suppressor genes, can regenerate normal phenotype and once again require a solid substrate to attach to and grow. Soft agar growth or colony formation in assays are used to identify modulators of cancer sequences, which when expressed in host cells, inhibit abnormal cellular proliferation and transformation. A modulator reduces or eliminates the host cells' ability to grow suspended in solid or semisolid media, such as agar.
  • Normal cells typically grow in a flat and organized pattern in cell culture until they touch other cells. When the cells touch one another, they are contact inhibited and stop growing. Transformed cells, however, are not contact inhibited and continue to grow to high densities in disorganized foci. Thus, transformed cells grow to a higher saturation density than corresponding normal cells. This is detected morphologically by the formation of a disoriented monolayer of cells or cells in foci.
  • labeling index with (3H)-thymidine at saturation density is used to measure density limitation of growth, similarly an MTT or Alamar blue assay will reveal proliferation capacity of cells and the ability of modulators to affect same. See Freshney (1994), supra.
  • Transformed cells when transfected with tumor suppressor genes, can regenerate a normal phenotype and become contact inhibited and would grow to a lower density.
  • labeling index with 3H)-thymidine at saturation density is a preferred method of measuring density limitation of growth.
  • Transformed host cells are transfected with a cancer-associated sequence and are grown for 24 hours at saturation density in non-limiting medium conditions.
  • the percentage of cells labeling with (3H)-thymidine is determined by incorporated cpm.
  • a modulator reduces or eliminates contact independent growth, and returns the cells to a normal phenotype.
  • Transformed cells have lower serum dependence than their normal counterparts (see, e.g., Temin, J. Natl. Cancer Inst. 37:167-175 (1966); Eagle et al., J. Exp. Med 131:836-879 (1970)); Freshney, supra. This is in part due to release of various growth factors by the transformed cells.
  • the degree of growth factor or serum dependence of transformed host cells can be compared with that of control. For example, growth factor or serum dependence of a cell is monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
  • Tumor cells release an increased amount of certain factors (hereinafter “tumor specific markers”) than their normal counterparts.
  • plasminogen activator PA
  • PA plasminogen activator
  • Tumor Angiogenesis Factor TAF
  • TAF Tumor Angiogenesis Factor
  • the degree of invasiveness into Matrigel or an extracellular matrix constituent can be used as an assay to identify and characterize compounds that modulate cancer associated sequences.
  • Tumor cells exhibit a positive correlation between malignancy and invasiveness of cells into Matrigel or some other extracellular matrix constituent.
  • tumorigenic cells are typically used as host cells. Expression of a tumor suppressor gene in these host cells would decrease invasiveness of the host cells. Techniques described in Cancer Res. 1999; 59:6010; Freshney (1994), supra, can be used. Briefly, the level of invasion of host cells is measured by using filters coated with Matrigel or some other extracellular matrix constituent.
  • Penetration into the gel, or through to the distal side of the filter, is rated as invasiveness, and rated histologically by number of cells and distance moved, or by prelabeling the cells with 1251 and counting the radioactivity on the distal side of the filter or bottom of the dish. See, e.g., Freshney (1984), supra.
  • Transgenic organisms are prepared in a variety of art-accepted ways. For example, knock-out transgenic organisms, e.g., mammals such as mice, are made, in which a cancer gene is disrupted or in which a cancer gene is inserted. Knock-out transgenic mice are made by insertion of a marker gene or other heterologous gene into the endogenous cancer gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting the endogenous cancer gene with a mutated version of the cancer gene, or by mutating the endogenous cancer gene, e.g., by exposure to carcinogens.
  • transgenic chimeric animals e.g., mice
  • a DNA construct is introduced into the nuclei of embryonic stem cells.
  • Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells some of which are derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, e.g., Capecchi et al., Science 244:1288 (1989)).
  • Chimeric mice can be derived according to U.S. Pat. No. 6,365,797, issued 2 Apr. 2002; U.S. Pat. No.
  • various immune-suppressed or immune-deficient host animals can be used.
  • a genetically athymic “nude” mouse see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)
  • SCID mouse see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)
  • SCID mouse e.g., a thymectornized mouse
  • an irradiated mouse see, e.g., Bradley et al., Br. J. Cancer 38:263 (1978); Selby et al., Br. J. Cancer 41:52 (1980)
  • Transplantable tumor cells typically about 10 6 cells injected into isogenic hosts produce invasive tumors in a high proportion of cases, while normal cells of similar origin will not.
  • cells expressing cancer-associated sequences are injected subcutaneously or orthotopically. Mice are then separated into groups, including control groups and treated experimental groups) e.g. treated with a modulator). After a suitable length of time, preferably 4-8 weeks, tumor growth is measured (e.g., by volume or by its two largest dimensions, or weight) and compared to the control. Tumors that have statistically significant reduction (using, e.g., Student's T test) are said to have inhibited growth.
  • Assays to identify compounds with modulating activity can be performed in vitro.
  • a cancer polypeptide is first contacted with a potential modulator and incubated for a suitable amount of time, e.g., from 0.5 to 48 hours.
  • the cancer polypeptide levels are determined in vitro by measuring the level of protein or mRNA.
  • the level of protein is measured using immunoassays such as Western blotting, ELISA and the like with an antibody that selectively binds to the cancer polypeptide or a fragment thereof.
  • amplification e.g., using PCR, LCR, or hybridization assays, e.g., Northern hybridization, RNAse protection, dot blotting, are preferred.
  • the level of protein or mRNA is detected using directly or indirectly labeled detection agents, e.g., fluorescently or radioactively labeled nucleic acids, radioactively or enzymatically labeled antibodies, and the like, as described herein.
  • a reporter gene system can be devised using a cancer protein promoter operably linked to a reporter gene such as luciferase, green fluorescent protein, CAT, or P-gal.
  • the reporter construct is typically transfected into a cell. After treatment with a potential modulator, the amount of reporter gene transcription, translation, or activity is measured according to standard techniques known to those of skill in the art (Davis G F, supra; Gonzalez, J. & Negulescu, P. Curr. Opin. Biotechnol. 1998: 9:624).
  • in vitro screens are done on individual genes and gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of the expression of the gene or the gene product itself is performed.
  • screening for modulators of expression of specific gene(s) is performed. Typically, the expression of only one or a few genes is evaluated. In another embodiment, screens are designed to first find compounds that bind to differentially expressed proteins. These compounds are then evaluated for the ability to modulate differentially expressed activity. Moreover, once initial candidate compounds are identified, variants can be further screened to better evaluate structure activity relationships.
  • a purified or isolated gene product of the invention is generally used.
  • antibodies are generated to a protein of the invention, and immunoassays are run to determine the amount and/or location of protein.
  • cells comprising the cancer proteins are used in the assays.
  • the methods comprise combining a cancer protein of the invention and a candidate compound such as a ligand, and determining the binding of the compound to the cancer protein of the invention.
  • a cancer protein of the invention utilizes the human cancer protein; animal models of human disease of can also be developed and used.
  • other analogous mammalian proteins also can be used as appreciated by those of skill in the art.
  • variant or derivative cancer proteins are used.
  • the cancer protein of the invention, or the ligand is non-diffusibly bound to an insoluble support.
  • the support can, e.g., be one having isolated sample receiving areas (a microtiter plate, an array, etc.).
  • the insoluble supports can be made of any composition to which the compositions can be bound, is readily separated from soluble material, and is otherwise compatible with the overall method of screening.
  • the surface of such supports can be solid or porous and of any convenient shape.
  • suitable insoluble supports include microtiter plates, arrays, membranes and beads. These are typically made of glass, plastic (e.g., polystyrene), polysaccharide, nylon, nitrocellulose, or TeflonTM, etc. Microtiter plates and arrays are especially convenient because a large number of assays can be carried out simultaneously, using small amounts of reagents and samples. The particular manner of binding of the composition to the support is not crucial so long as it is compatible with the reagents and overall methods of the invention, maintains the activity of the composition and is nondiffusable.
  • Preferred methods of binding include the use of antibodies which do not sterically block either the ligand binding site or activation sequence when attaching the protein to the support, direct binding to “sticky” or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the surface, etc. Following binding of the protein or ligand/binding agent to the support, excess unbound material is removed by washing. The sample receiving areas may then be blocked through incubation with bovine serum albumin (BSA), casein or other innocuous protein or other moiety.
  • BSA bovine serum albumin
  • Binding agents include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc.
  • assays to identify agents that have a low toxicity for human cells.
  • a wide variety of assays can be used for this purpose, including proliferation assays, cAMP assays, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
  • a determination of binding of the test compound (ligand, binding agent, modulator, etc.) to a cancer protein of the invention can be done in a number of ways.
  • the test compound can be labeled, and binding determined directly, e.g., by attaching all or a portion of the cancer protein of the invention to a solid support, adding a labeled candidate compound (e.g., a fluorescent label), washing off excess reagent, and determining whether the label is present on the solid support.
  • a labeled candidate compound e.g., a fluorescent label
  • only one of the components is labeled, e.g., a protein of the invention or ligands labeled.
  • more than one component is labeled with different labels, e.g., I 125 , for the proteins and a fluorophor for the compound.
  • Proximity reagents e.g., quenching or energy transfer reagents are also useful.
  • the binding of the “test compound” is determined by competitive binding assay with a “competitor.”
  • the competitor is a binding moiety that binds to the target molecule (e.g., a cancer protein of the invention). Competitors include compounds such as antibodies, peptides, binding partners, ligands, etc. Under certain circumstances, the competitive binding between the test compound and the competitor displaces the test compound.
  • the test compound is labeled. Either the test compound, the competitor, or both, is added to the protein for a time sufficient to allow binding. Incubations are performed at a temperature that facilitates optimal activity, typically between four and 40° C.
  • Incubation periods are typically optimized, e.g., to facilitate rapid high throughput screening; typically between zero and one hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
  • the competitor is added first, followed by the test compound.
  • Displacement of the competitor is an indication that the test compound is binding to the cancer protein and thus is capable of binding to, and potentially modulating, the activity of the cancer protein.
  • either component can be labeled.
  • the presence of label in the post-test compound wash solution indicates displacement by the test compound.
  • the presence of the label on the support indicates displacement.
  • the test compound is added first, with incubation and washing, followed by the competitor.
  • the absence of binding by the competitor indicates that the test compound binds to the cancer protein with higher affinity than the competitor.
  • the presence of the label on the support, coupled with a lack of competitor binding indicates that the test compound binds to and thus potentially modulates the cancer protein of the invention.
  • the competitive binding methods comprise differential screening to identity agents that are capable of modulating the activity of the cancer proteins of the invention.
  • the methods comprise combining a cancer protein and a competitor in a first sample.
  • a second sample comprises a test compound, the cancer protein, and a competitor.
  • the binding of the competitor is determined for both samples, and a change, or difference in binding between the two samples indicates the presence of an agent capable of binding to the cancer protein and potentially modulating its activity. That is, if the binding of the competitor is different in the second sample relative to the first sample, the agent is capable of binding to the cancer protein.
  • differential screening is used to identify drug candidates that bind to the native cancer protein, but cannot bind to modified cancer proteins.
  • the structure of the cancer protein is modeled and used in rational drug design to synthesize agents that interact with that site, agents which generally do not bind to site-modified proteins.
  • drug candidates that affect the activity of a native cancer protein are also identified by screening drugs for the ability to either enhance or reduce the activity of such proteins.
  • Positive controls and negative controls can be used in the assays.
  • control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of all samples occurs for a time sufficient to allow for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radiolabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
  • reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., can be used. The mixture of components is added in an order that provides for the requisite binding.
  • Polynucleotide modulators of cancer can be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand-binding molecule, as described in WO 91/04753.
  • Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors.
  • conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell.
  • a polynucleotide modulator of cancer can be introduced into a cell containing the target nucleic acid sequence, e.g., by formation of a polynucleotide-lipid complex, as described in WO 90/10448. It is understood that the use of antisense molecules or knock out and knock in models may also be used in screening assays as discussed above, in addition to methods of treatment.
  • the activity of a cancer-associated protein is down-regulated, or entirely inhibited, by the use of antisense polynucleotide or inhibitory small nuclear RNA (snRNA), i.e., a nucleic acid complementary to, and which can preferably hybridize specifically to, a coding mRNA nucleic acid sequence, e.g., a cancer protein of the invention, mRNA, or a subsequence thereof. Binding of the antisense polynucleotide to the mRNA reduces the translation and/or stability of the mRNA.
  • snRNA inhibitory small nuclear RNA
  • antisense polynucleotides can comprise naturally occurring nucleotides, or synthetic species formed from naturally occurring subunits or their close homologs. Antisense polynucleotides may also have altered sugar moieties or inter-sugar linkages. Exemplary among these are the phosphorothioate and other sulfur containing species which are known for use in the art. Analogs are comprised by this invention so long as they function effectively to hybridize with nucleotides of the invention. See, e.g., Isis Pharmaceuticals, Carlsbad, Calif.; Sequitor, Inc., Natick, Mass.
  • antisense polynucleotides can readily be synthesized using recombinant means, or can be synthesized in vitro. Equipment for such synthesis is sold by several vendors, including Applied Biosystems. The preparation of other oligonucleotides such as phosphorothioates and alkylated derivatives is also well known to those of skill in the art.
  • Antisense molecules as used herein include antisense or sense oligonucleotides.
  • Sense oligonucleotides can, e.g., be employed to block transcription by binding to the anti-sense strand.
  • the antisense and sense oligonucleotide comprise a single stranded nucleic acid sequence (either RNA or DNA) capable of binding to target mRNA (sense) or DNA (antisense) sequences for cancer molecules.
  • Antisense or sense oligonucleotides, according to the present invention comprise a fragment generally at least about 12 nucleotides, preferably from about 12 to 30 nucleotides.
  • ribozymes can be used to target and inhibit transcription of cancer-associated nucleotide sequences.
  • a ribozyme is an RNA molecule that catalytically cleaves other RNA molecules.
  • Different kinds of ribozymes have been described, including group I ribozymes, hammerhead ribozymes, hairpin ribozymes, RNase P, and axhead ribozymes (see, e.g., Castanotto et al., Adv. in Pharmacology 25: 289-317 (1994) for a general review of the properties of different ribozymes).
  • hairpin ribozymes are described, e.g., in Hampel et al., Nucl. Acids Res. 18:299-304 (1990); European Patent Publication No. 0360257; U.S. Pat. No. 5,254,678.
  • Methods of preparing are well known to those of skill in the art (see, e.g., WO 94/26877; Ojwang et al., Proc. Natl. Acad. Sci. USA 90:6340-6344 (1993); Yamada et al., Human Gene Therapy 1:39-45 (1994); Leavitt et al., Proc. Natl. Acad Sci. USA 92:699-703 (1995); Leavitt et al., Human Gene Therapy 5: 1151-120 (1994); and Yamada et al., Virology 205: 121-126 (1994)).
  • a test compound is administered to a population of cancer cells, which have an associated cancer expression profile.
  • administration or “contacting” herein is meant that the modulator is added to the cells in such a manner as to allow the modulator to act upon the cell, whether by uptake and intracellular action, or by action at the cell surface.
  • a nucleic acid encoding a proteinaceous agent i.e., a peptide
  • a viral construct such as an adenoviral or retroviral construct
  • expression of the peptide agent is accomplished, e.g., PCT US97/01019.
  • Regulatable gene therapy systems can also be used.
  • the cells are washed if desired and are allowed to incubate under preferably physiological conditions for some period.
  • the cells are then harvested and a new gene expression profile is generated.
  • cancer tissue is screened for agents that modulate, e.g., induce or suppress, the cancer phenotype.
  • a change in at least one gene, preferably many, of the expression profile indicates that the agent has an effect on cancer activity.
  • altering a biological function or a signaling pathway is indicative of modulator activity.
  • screens are done to assess genes or gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of either the expression of the gene or the gene product itself is performed.
  • Measurements of cancer polypeptide activity, or of the cancer phenotype are performed using a variety of assays. For example, the effects of modulators upon the function of a cancer polypeptide(s) are measured by examining parameters described above. A physiological change that affects activity is used to assess the influence of a test compound on the polypeptides of this invention.
  • a variety of effects can be assesses such as, in the case of a cancer associated with solid tumors, tumor growth, tumor metastasis, neovascularization, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., by Northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as cGNIP.
  • the invention provides methods for identifying cells containing variant cancer genes, e.g., determining the presence of, all or part, the sequence of at least one endogenous cancer gene in a cell. This is accomplished using any number of sequencing techniques.
  • the invention comprises methods of identifying the cancer genotype of an individual, e.g., determining all or part of the sequence of at least one gene of the invention in the individual. This is generally done in at least one tissue of the individual, e.g., a tissue set forth in Table I, and may include the evaluation of a number of tissues or different samples of the same tissue.
  • the method may include comparing the sequence of the sequenced gene to a known cancer gene, i.e., a wild-type gene to determine the presence of family members, homologies, mutations or variants.
  • the sequence of all or part of the gene can then be compared to the sequence of a known cancer gene to determine if any differences exist. This is done using any number of known homology programs, such as BLAST, Bestfit, etc.
  • the presence of a difference in the sequence between the cancer gene of the patient and the known cancer gene correlates with a disease state or a propensity for a disease state, as outlined herein.
  • the cancer genes are used as probes to determine the number of copies of the cancer gene in the genome.
  • the cancer genes are used as probes to determine the chromosomal localization of the cancer genes.
  • Information such as chromosomal localization finds use in providing a diagnosis or prognosis in particular when chromosomal abnormalities such as translocations, and the like are identified in the cancer gene locus.
  • RNAi AND THERAPEUTIC USE OF SMALL INTERFERING RNA siRNAs
  • the present invention is also directed towards siRNA oligonucleotides, particularly double stranded RNAs encompassing at least a fragment of the 161P2F10B coding region or 5′′ UTR regions, or complement, or any antisense oligonucleotide specific to the 161P2F10B sequence.
  • siRNA oligonucleotides are used to elucidate a function of 161P2F10B, or are used to screen for or evaluate modulators of 161P2F10B function or expression.
  • gene expression of 161P2F10B is reduced by using siRNA transfection and results in significantly diminished proliferative capacity of transformed cancer cells that endogenously express the antigen; cells treated with specific 161P2F10B siRNAs show reduced survival as measured, e.g., by a metabolic readout of cell viability, correlating to the reduced proliferative capacity.
  • 161P2F10B siRNA compositions comprise siRNA (double stranded RNA) that correspond to the nucleic acid ORF sequence of the 161P2F10B protein or subsequences thereof; these subsequences are generally 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 or more than 35 contiguous RNA nucleotides in length and contain sequences that are complementary and non-complementary to at least a portion of the mRNA coding sequence
  • the subsequences are 19-25 nucleotides in length, most preferably 21-23 nucleotides in length.
  • RNA interference is a novel approach to silencing genes in vitro and in vivo, thus small double stranded RNAs (siRNAs) are valuable therapeutic agents.
  • siRNAs small double stranded RNAs
  • the power of siRNAs to silence specific gene activities has now been brought to animal models of disease and is used in humans as well. For example, hydrodynamic infusion of a solution of siRNA into a mouse with a siRNA against a particular target has been proven to be therapeutically effective.
  • siRNAs small interfering RNAs
  • This work provided the first in vivo evidence that infusion of siRNAs into an animal could alleviate disease.
  • the authors gave mice injections of siRNA designed to silence the FAS protein (a cell death receptor that when over-activated during inflammatory response induces hepatocytes and other cells to die). The next day, the animals were given an antibody specific to Fas.
  • mice died of acute liver failure within a few days, while over 80% of the siRNA-treated mice remained free from serious disease and survived. About 80% to 90% of their liver cells incorporated the naked siRNA oligonucleotides. Furthermore, the RNA molecules functioned for 10 days before losing effect after 3 weeks.
  • siRNA is delivered by efficient systems that induce long-lasting RNAi activity.
  • a major caveat for clinical use is delivering siRNAs to the appropriate cells. Hepatocytes seem to be particularly receptive to exogenous RNA.
  • targets located in the liver are attractive because liver is an organ that can be readily targeted by nucleic acid molecules and viral vectors. However, other tissue and organs targets are preferred as well.
  • Formulations of siRNAs with compounds that promote transit across cell membranes are used to improve administration of siRNAs in therapy.
  • Chemically modified synthetic siRNA, that are resistant to nucleases and have serum stability have concomitant enhanced duration of RNAi effects, are an additional embodiment.
  • siRNA technology is a therapeutic for human malignancy by delivery of siRNA molecules directed to 161P2F10B to individuals with the cancers, such as those listed in Table 1.
  • Such administration of siRNAs leads to reduced growth of cancer cells expressing 161P2F10B, and provides an anti-tumor therapy, lessening the morbidity and/or mortality associated with malignancy.
  • kits are within the scope of the invention.
  • 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, along with a label or insert comprising instructions for use, such as a use described herein.
  • the container(s) can comprise a probe that is or can be detectably labeled.
  • probe can be an antibody or polynucleotide specific for a protein or a gene or message of the invention, respectively.
  • kits can also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence.
  • Kits can comprise a container comprising a reporter, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, fluorescent, or radioisotope label; such a reporter can be used with, e.g., a nucleic acid or antibody.
  • the kit can include all or part of the amino acid sequences in FIG. 1 , FIG. 2 , or FIG. 3 or analogs thereof, or a nucleic acid molecule that encodes such amino acid sequences.
  • the kit of the invention will typically comprise the container described above and one or more other containers associated therewith that comprise materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • a label can be present on or with the container to indicate that the composition is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic or laboratory application, and can also indicate directions for either in vivo or in vitro use, such as those described herein. Directions and or other information can also be included on an insert(s) or label(s) which is included with or on the kit.
  • the label can be on or associated with the container.
  • a label a can be on a container when letters, numbers or other characters forming the label are molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • the label can indicate that the composition is used for diagnosing, treating, prophylaxing or prognosing a condition, such as a neoplasia of a tissue set forth in Table I.
  • an article(s) of manufacture containing compositions such as amino acid sequence(s), small molecule(s), nucleic acid sequence(s), and/or antibody(s), e.g., materials useful for the diagnosis, prognosis, prophylaxis and/or treatment of neoplasias of tissues such as those set forth in Table I.
  • the article of manufacture typically comprises at least one container and at least one label. Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers can be formed from a variety of materials such as glass, metal or plastic.
  • the container can hold amino acid sequence(s), small molecule(s), nucleic acid sequence(s), cell population(s) and/or antibody(s).
  • the container holds a polynucleotide for use in examining the mRNA expression profile of a cell, together with reagents used for this purpose.
  • a container comprises an antibody, binding fragment thereof or specific binding protein for use in evaluating protein expression of 161P2F10B in cells and tissues, or for relevant laboratory, prognostic, diagnostic, prophylactic and therapeutic purposes; indications and/or directions for such uses can be included on or with such container, as can reagents and other compositions or tools used for these purposes.
  • a container comprises materials for eliciting a cellular or humoral immune response, together with associated indications and/or directions.
  • a container comprises materials for adoptive immunotherapy, such as cytotoxic T cells (CTL) or helper T cells (HTL), together with associated indications and/or directions; reagents and other compositions or tools used for such purpose can also be included.
  • CTL cytotoxic T cells
  • HTL helper T cells
  • the container can alternatively hold a composition that is effective for treating, diagnosis, prognosing or prophylaxing a condition and can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agents in the composition can be an antibody capable of specifically binding 161P2F10B and modulating the function of 161P2F10B.
  • the article of manufacture can further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and/or dextrose solution. It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and/or dextrose solution.
  • It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
  • First strand cDNA was generated from normal stomach, normal brain, normal heart, normal liver, normal skeletal muscle, normal testis, normal prostate, normal bladder, normal kidney, normal colon, normal lung, normal pancreas, and a pool of cancer specimens from prostate cancer patients, bladder cancer patients, kidney cancer patients, colon cancer patients, lung cancer patients, pancreas cancer patients, a pool of prostate cancer xenografts (LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI), and a pool of 2 patient prostate metastasis to lymph node.
  • Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 161P2F10B, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software.
  • 161P2F10B Expression of 161P2F10B in a panel of kidney cancer clear cell carcinoma, kidney cancer papillary carcinoma, and in uterus patient cancer specimens has been shown previously.
  • First strand cDNA was prepared from the patient specimens. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 161P2F10B, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software. Expression was recorded as absent, low, medium or strong. Results show expression of 161P2F10B in 94.7% of clear cell renal carcinoma, 62.5% of papillary renal cell carcinoma, and in 61.5% of uterus cancer.
  • 161P2F10B is a therapeutic target and a diagnostic marker for human cancers.
  • Transcript variants are variants of mature mRNA from the same gene, which arise by alternative transcription or alternative splicing.
  • Alternative transcripts are transcripts from the same gene but start transcription at different points.
  • Splice variants are mRNA variants spliced differently from the same transcript.
  • 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 transcript.
  • Transcript variants can code for similar or different proteins with the same or a similar function or can encode proteins with different functions, and can be expressed in the same tissue at the same time, or in different tissues at the same time, or in the same tissue at different times, or in different tissues at different times. Proteins encoded by transcript variants can have similar or different cellular or extracellular localizations, e.g., secreted versus intracellular.
  • Transcript variants are identified by a variety of art-accepted methods. For example, alternative transcripts and splice variants are identified by full-length cloning experiment, or by use of full-length transcript and EST sequences. First, all human ESTs were grouped into clusters which show direct or indirect identity with each other. Second, ESTs in the same cluster were further grouped into sub-clusters and assembled into a consensus sequence. The 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. 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.
  • 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 and GenScan.
  • FgenesH A. Salamov and V. Solovyev, “Ab initio gene finding in Drosophila genomic DNA,” Genome Research. 2000 April; 10(4):516-22
  • Grail and GenScan GenScan.
  • 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.
  • PCR-based Validation Wellmann S, et al., Specific reverse transcription-PCR quantification of vascular endothelial growth factor (VEGF) splice variants by LightCycler technology, Clin Chem. 2001 April; 47(4):654-60; Jia, H. P., et al., Discovery of new human beta-defensins using a genomics-based approach, Gene. 2001 Jan. 24; 263(1-2):211-8.
  • VEGF vascular endothelial growth factor
  • genomic regions are modulated in cancers.
  • the alternative transcripts or splice variants of the gene are modulated as well.
  • 161P2F10B has a particular expression profile related to cancer.
  • Alternative transcripts and splice variants of 161P2F10B are also involved in cancers in the same or different tissues, thus serving as tumor-associated markers/antigens.
  • 161P2F10B amino acid and nucleic acid sequences are set forth on a variant by variant basis in FIG. 1 .
  • a Single Nucleotide Polymorphism is a single base pair variation in a nucleotide sequence at a specific location.
  • SNP Single Nucleotide Polymorphism
  • Genotype refers to the specific base pair sequence of one or more locations in the genome of an individual.
  • Haplotype refers to the base pair sequence of more than one location on the same DNA molecule (or the same chromosome in higher organisms), often in the context of one gene or in the context of several tightly linked genes.
  • 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.
  • SNPs and/or combinations of alleles have many applications, including diagnosis of inherited diseases, determination of drug reactions and dosage, identification of genes responsible for diseases, and analysis of the genetic relationship between individuals (P. Nowotny, J. M. Kwon and A. M. Goate, “SNP analysis to dissect human traits,” Curr. Opin. Neurobiol. 2001 October; 11(5):637-641; M. Pirmohamed and B. K. Park, “Genetic susceptibility to adverse drug reactions,” Trends Pharmacol. Sci. 2001 June; 22(6):298-305; J. H. Riley, C.
  • SNPs are identified by a variety of art-accepted methods (P. Bean, “The promising voyage of SNP target discovery,” Am. Clin. Lab. 2001 October-November; 20(9):18-20; K. M. Weiss, “In search of human variation,” Genome Res. 1998 July; 8(7):691-697; M. M. She, “Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies,” Clin. Chem. 2001 February; 47(2):164-172).
  • 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).
  • RFLP restriction fragment length polymorphism
  • DGGE denaturing gradient gel electrophoresis
  • SNPs can also be discovered by direct sequencing of DNA samples pooled from different individuals or by comparing sequences from different DNA samples. With the rapid accumulation of sequence data in public and private databases, one can discover SNPs by comparing sequences using computer programs (Z. Gu, L. Hillier and P. Y. Kwok, “Single nucleotide polymorphism hunting in cyberspace,” Hum. Mutat. 1998; 12(4):221-225). SNPs can be verified and genotype or haplotype of an individual can be determined by a variety of methods including direct sequencing and high throughput microarrays (P. Y. Kwok, “Methods for genotyping single nucleotide polymorphisms,” Annu. Rev. Genomics Hum. Genet.
  • transcripts or proteins with alternative alleles were designated as variants 161P2F10B v.2, v.3, v.4, and v.5, respectively.
  • Alleles of the SNPs though discussed separately, can occur in different combinations (haplotypes) and in any one of the transcript variants (such as 161P2F10B v.7) that contains the sequence context of the SNPs.
  • 161P2F10B amino acid and nucleic acid sequences are set forth on a variant by variant basis in FIG. 1 .
  • 161P2F10B and 161P2F10B variants are cloned into any one of a variety of expression vectors known in the art.
  • One or more of the following regions of 161P2F10B variants are expressed: the full length sequence presented in FIG. 1 , 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 161P2F10B, variants, or analogs thereof.
  • pCRII To generate 161P2F10B sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad Calif.) are generated encoding either all or fragments of the 161P2F10B cDNA.
  • the pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of 161P2F10B RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of 161P2F10B at the RNA level.
  • Transcribed 161P2F10B RNA representing the cDNA amino acid coding region of the 161P2F10B gene is used in in vitro translation systems such as the TnTTM Coupled Reticulolysate System (Promega, Corp., Madison, Wis.) to synthesize 161P2F10B protein.
  • TnTTM Coupled Reticulolysate System Promega, Corp., Madison, Wis.
  • pGEX Constructs To generate recombinant 161P2F10B proteins in bacteria that are fused to the Glutathione S-transferase (GST) protein, all or parts of the 161P2F10B cDNA protein coding sequence are cloned into the pGEX family of GST-fusion vectors (Amersham Pharmacia Biotech, Piscataway, N.J.). These constructs allow controlled expression of recombinant 161P2F10B protein sequences with GST fused at the amino-terminus and a six histidine epitope (6 ⁇ His) at the carboxyl-terminus.
  • GST Glutathione S-transferase
  • the GST and 6 ⁇ His tags permit purification of the recombinant fusion protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-GST and anti-His antibodies.
  • the 6 ⁇ His tag is generated by adding 6 histidine codons to the cloning primer at the 3′ end, e.g., of the open reading frame (ORF).
  • a proteolytic cleavage site such as the PreScissionTM recognition site in pGEX-6P-1, may be employed such that it permits cleavage of the GST tag from 161P2F10B-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 161P2F10B proteins that are fused to maltose-binding protein (MBP), all or parts of the 161P2F10B cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, Mass.). These constructs allow controlled expression of recombinant 161P2F10B protein sequences with MBP fused at the amino-terminus and a 6 ⁇ His epitope tag at the carboxyl-terminus.
  • MBP maltose-binding protein
  • the MBP and 6 ⁇ His tags permit purification of the recombinant protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-MBP and anti-His antibodies.
  • the 6 ⁇ His epitope tag is generated by adding 6 histidine codons to the 3′ cloning primer.
  • a Factor Xa recognition site permits cleavage of the pMAL tag from 161P2F10B.
  • the pMAL-c2X and pMAL-p2X vectors are optimized to express the recombinant protein in the cytoplasm or periplasm respectively. Periplasm expression enhances folding of proteins with disulfide bonds.
  • pET Constructs To express 161P2F10B in bacterial cells, all or parts of the 161P2F10B cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, Wis.). These vectors allow tightly controlled expression of recombinant 161P2F10B protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6 ⁇ His and S-TagTM 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 161P2F10B protein are expressed as amino-terminal fusions to NusA.
  • pESC Constructs To express 161P2F10B in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the 161P2F10B cDNA protein coding sequence are cloned into the pESC family of vectors each of which contain 1 of 4 selectable markers, HIS3, TRP1, LEU2, and URA3 (Stratagene, La Jolla, Calif.). These vectors allow controlled expression from the same plasmid of up to 2 different genes or cloned sequences containing either FlagTM or Myc epitope tags in the same yeast cell. This system is useful to confirm protein-protein interactions of 161P2F10B. 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 161P2F10B in the yeast species Saccharomyces pombe , all or parts of the 161P2F10B cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a 161P2F10B 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 FlagTM epitope tag allows detection of the recombinant protein with anti-FlagTM antibody.
  • 161P2F10B in eukaryotic cells, the full or partial length 161P2F10B cDNA sequences, or variants thereof, can be cloned into any one of a variety of expression vectors known in the art.
  • One or more of the following regions of 161P2F10B are expressed in these constructs, amino acids 1 to 875, 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 161P2F10B v.1, 161P2F10B variants, or analogs thereof.
  • constructs can be transfected into any one of a wide variety of mammalian cells such as 293T cells.
  • Transfected 293T cell lysates can be probed with the anti-161P2F10B polyclonal serum, described herein.
  • pcDNA4/HisMax Constructs To express 161P2F10B in mammalian cells, a 161P2F10B ORF, or portions thereof, of 161P2F10B are cloned into pcDNA4/HisMax Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter and the SP16 translational enhancer. The recombinant protein has XpressTM and six histidine (6 ⁇ His) epitopes fused to the amino-terminus.
  • CMV cytomegalovirus
  • SP16 translational enhancer The recombinant protein has XpressTM and six histidine (6 ⁇ His) epitopes fused to the amino-terminus.
  • the pcDNA4/HisMax vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen.
  • BGH bovine growth hormone
  • the Zeocin resistance gene allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • pcDNA3.1/MycHis Constructs To express 161P2F10B in mammalian cells, a 161P2F10B ORF, or portions thereof, of 161P2F10B with a consensus Kozak translation initiation site was cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the myc epitope and 6 ⁇ His epitope fused to the carboxyl-terminus.
  • CMV cytomegalovirus
  • 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.
  • BGH bovine growth hormone
  • the Neomycin resistance gene can be used, as it allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • pcDNA3.1/CT-GFP-TOPO Construct To express 161P2F10B in mammalian cells and to allow detection of the recombinant proteins using fluorescence, a 161P2F10B 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.
  • CMV cytomegalovirus
  • the pcDNA3.1CT-GFP-TOPO vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen.
  • BGH bovine growth hormone
  • the Neomycin resistance gene allows for selection of mammalian cells that express the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli .
  • Additional constructs with an amino-terminal GFP fusion are made in pcDNA3.1/NT-GFP-TOPO spanning the entire length of a 161P2F10B protein.
  • PAPtag A 161P2F10B ORF, or portions thereof, is cloned into pAPtag-5 (GenHunter Corp. Nashville, Tenn.). This construct generates an alkaline phosphatase fusion at the carboxyl-terminus of a 161P2F10B protein while fusing the IgGK signal sequence to the amino-terminus. Constructs are also generated in which alkaline phosphatase with an amino-terminal IgGK signal sequence is fused to the amino-terminus of a 161P2F10B protein.
  • the resulting recombinant 161P2F10B 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 161P2F10B proteins.
  • Protein expression is driven from the CMV promoter and the recombinant proteins also contain myc and 6 ⁇ His epitopes fused at the carboxyl-terminus that facilitates detection and purification.
  • the Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the recombinant protein and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • ptag5 A 161P2F10B ORF, or portions thereof, was cloned into pTag-5.
  • This vector is similar to pAPtag but without the alkaline phosphatase fusion.
  • This construct generates 161P2F10B protein with an amino-terminal IgGK signal sequence and myc and 6 ⁇ His epitope tags at the carboxyl-terminus that facilitate detection and affinity purification.
  • the resulting recombinant 161P2F10B protein is optimized for secretion into the media of transfected mammalian cells, and is used as immunogen or ligand to identify proteins such as ligands or receptors that interact with the 161P2F10B proteins. Protein expression is driven from the CMV promoter.
  • the Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • PsecFc A 161P2F10B ORF, or portions thereof, was cloned into psecFc.
  • the psecFc vector was assembled by cloning the human immunoglobulin G1 (IgG) Fc (hinge, CH2, CH3 regions) into pSecTag2 (Invitrogen, California). This construct generates an IgG1 Fc fusion at the carboxyl-terminus of the 161P2F10B proteins, while fusing the IgGK signal sequence to N-terminus. 161P2F10B fusions utilizing the murine IgG1 Fc region are also used.
  • IgG human immunoglobulin G1
  • the resulting recombinant 161P2F10B 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 161P2F10B 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.
  • pSR ⁇ Constructs To generate mammalian cell lines that express 161P2F10B constitutively, 161P2F10B ORF, or portions thereof, of 161P2F10B were cloned into pSR ⁇ constructs. Amphotropic and ecotropic retroviruses were generated by transfection of pSR ⁇ constructs into the 293T-10A1 packaging line or co-transfection of pSR ⁇ 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, 161P2F10B, into the host cell-lines.
  • Protein expression is driven from a long terminal repeat (LTR).
  • LTR long terminal repeat
  • the Neomycin resistance gene present in the vector allows for selection of mammalian cells that express the protein, and the ampicillin resistance gene and ColE1 origin permit selection and maintenance of the plasmid in E. coli .
  • the retroviral vectors can thereafter be used for infection and generation of various cell lines using, for example, PC3, NIH 3T3, TsuPr1, 293 or rat-I cells.
  • Additional pSR ⁇ constructs are made that fuse an epitope tag such as the FLAGTM tag to the carboxyl-terminus of 161P2F10B sequences to allow detection using anti-Flag antibodies.
  • the FLAGTM sequence 5′ GAT TAC AAG GAT GAC GAC GAT AAG 3′ (SEQ ID NO: 165) is added to cloning primer at the 3′ end of the ORF.
  • Additional pSR ⁇ constructs are made to produce both amino-terminal and carboxyl-terminal GFP and myc/6 ⁇ His fusion proteins of the full-length 161P2F10B proteins.
  • Additional Viral Vectors Additional constructs are made for viral-mediated delivery and expression of 161P2F10B.
  • High virus titer leading to high level expression of 161P2F10B is achieved in viral delivery systems such as adenoviral vectors and herpes amplicon vectors.
  • a 161P2F10B coding sequences or fragments thereof are amplified by PCR and subcloned into the AdEasy shuttle vector (Stratagene). Recombination and virus packaging are performed according to the manufacturer's instructions to generate adenoviral vectors.
  • 161P2F10B coding sequences or fragments thereof are cloned into the HSV-1 vector (Imgenex) to generate herpes viral vectors.
  • the viral vectors are thereafter used for infection of various cell lines such as PC3, NIH 3T3, 293 or rat-I cells.
  • coding sequences of 161P2F10B, 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 161P2F10B. These vectors are thereafter used to control expression of 161P2F10B in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • 161P2F10B ORF To generate recombinant 161P2F10B proteins in a baculovirus expression system, 161P2F10B ORF, or portions thereof, are cloned into the baculovirus transfer vector pBlueBac 4.5 (Invitrogen), which provides a His-tag at the N-terminus.
  • pBlueBac-161P2F10B is co-transfected with helper plasmid pBac-N-Blue (Invitrogen) into SF9 ( Spodoptera frugiperda ) insect cells to generate recombinant baculovirus (see Invitrogen instruction manual for details). Baculovirus is then collected from cell supernatant and purified by plaque assay.
  • Recombinant 161P2F10B protein is then generated by infection of HighFive insect cells (Invitrogen) with purified baculovirus.
  • Recombinant 161P2F10B protein can be detected using anti-161P2F10B or anti-His-tag antibody.
  • 161P2F10B protein can be purified and used in various cell-based assays or as immunogen to generate polyclonal and monoclonal antibodies specific for 161P2F10B.
  • 161P2F10B Mouse and monkey orthologs of 161P2F10B were cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the myc epitope and 6 ⁇ His epitope fused to the carboxyl-terminus. These vectors allow expression of 161P2F10B orthologs to assay cross-reactivity of monoclonal anti-human 161P2F10B antibodies.
  • CMV cytomegalovirus
  • Mouse and monkey orthologs of 161P2F10B were also cloned into pSR ⁇ constructs.
  • the pSR ⁇ constructs allow for the generation of mammalian cell lines that express 161P2F10B orthologs constitutively. Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the myc epitope and 6 ⁇ His epitope fused to the carboxyl-terminus.
  • CMV cytomegalovirus
  • These vectors allow expression of 161P2F10B orthologs to assay cross-reactivity of monoclonal anti-human 161P2F10B antibodies and to study functional activity of 161P2F10B orthologs.
  • Amphotropic and ecotropic retroviruses were generated by transfection of pSR ⁇ constructs into the 293T-10A1 packaging line or co-transfection of pSR ⁇ 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, 161P2F10B ortholog, into the host cell-lines.
  • Amino acid profiles of 161P2F10B and 161P2F10B variants were found accessing the ProtScale website on the World Wide Web at on the ExPasy molecular biology server.
  • Hydrophilicity, Hydropathicity, and Percentage Accessible Residues 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 and Beta-turn 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 161P2F10B variant proteins indicated, e.g., by the profiles described above are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-161P2F10B 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 161P2F10B protein variants listed in FIG. 1 of which the amino acid profiles can be inferred because the variant contains sequence that is the same as a variant depicted.
  • peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of FIG. 1 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profile; a peptide region of at least 5 amino acids of FIG. 1 in any whole number increment that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile; a peptide region of at least 5 amino acids of FIG. 1 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profiles; a peptide region of at least 5 amino acids of FIG.
  • 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 161P2F10B and 161P2F10B variants 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 (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C. and Deléage G., accessed from the ExPasy molecular biology server located on the World Wide Web.
  • NPS@ Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C. and Deléage G.
  • the analysis indicates that 161P2F10B variant 1 is composed of 31.31% alpha helix, 11.31% extended strand, and 57.37% random coil.
  • Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant.
  • the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections.
  • computer algorithms are employed in design of immunogens that, based on amino acid sequence analysis contain characteristics of being antigenic and available for recognition by the immune system of the immunized host (see the Example entitled “Antigenicity Profiles and Secondary Structure”). Such regions would be predicted to be hydrophilic, flexible, in beta-turn conformations, and be exposed on the surface of the protein.
  • recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta-turn regions of 161P2F10B protein variants are used as antigens to generate polyclonal antibodies in New Zealand White rabbits or monoclonal antibodies as described in the Example entitled “Generation of 161P2F10B Monoclonal Antibodies (MAbs)”.
  • such regions include, but are not limited to, amino acids 43-93, 100-134, 211-246, 567-492, 500-517 and amino acids 810-870.
  • 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 161P2F10B 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).
  • 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 161P2F10B in Eukaryotic Systems”), and retain post-translational modifications such as glycosylations found in native protein.
  • amino acids 45-875 are cloned into the Tag5 mammalian secretion vector.
  • the recombinant protein was purified by metal chelate chromatography from tissue culture supernatants of 293T cells stably expressing the recombinant vector.
  • the purified Tag5 161P2F10B protein was then used as immunogen.
  • adjuvants include, but are not limited to, complete Freund's adjuvant (CFA) and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • CFA complete Freund's adjuvant
  • MPL-TDM adjuvant monophosphoryl Lipid A, synthetic trehalose dicorynomycolate
  • rabbits are initially immunized subcutaneously with up to 200 ⁇ g, typically 100-200 ⁇ g, 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 ⁇ g, typically 100-200 ⁇ g, of the immunogen in incomplete Freund's adjuvant (IFA). Test bleeds are taken approximately 7-10 days following each immunization and used to monitor the titer of the antiserum by ELISA.
  • CFA complete Freund's adjuvant
  • the respective full-length 161P2F10B variant cDNA is cloned into pcDNA 3.1 myc-his expression vector (Invitrogen, see the Example entitled “Production of Recombinant 161P2F10B in Eukaryotic Systems”). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-variant serum and with anti-His antibody (Santa Cruz Biotechnologies) to determine specific reactivity to denatured variant protein using the Western blot technique.
  • the immune serum is tested by fluorescence microscopy, flow cytometry and immunoprecipitation against 293T and other recombinant 161P2F10B variant-expressing cells to determine specific recognition of native protein.
  • Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express 161P2F10B are also carried out to test reactivity and specificity.
  • Anti-serum from the Tag5 161P2F10B immunized rabbit is affinity purified by passage over a column of GST protein covalently coupled to AffiGel matrix (BioRad).
  • the antiserum is then affinity purified by passage over a column composed of a MBP-161P2F10B fusion protein 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.
  • therapeutic Monoclonal Antibodies (“MAbs”) to 161P2F10B and 161P2F10B variants comprise those that react with epitopes specific for each protein or specific to sequences in common between the variants that would bind, internalize, disrupt or modulate the biological function of 161P2F10B or 161P2F10B variants, for example, those that would disrupt the interaction with ligands, substrates, and binding partners.
  • Immunogens for generation of such MAbs include those designed to encode or contain the extracellular domain or the entire 161P2F10B protein sequence, regions predicted to contain functional motifs, and regions of the 161P2F10B protein variants predicted to be antigenic from computer analysis of the amino acid sequence.
  • Immunogens include peptides and recombinant proteins such as tag5-161P2F10B a mammalian expressed purified His tagged protein.
  • cells engineered through retroviral transduction to express high levels of 161P2F10B variant 1, such as RAT1-161P2F10B are used to immunize mice.
  • mice are first immunized in the foot pad (FP) with, typically, 5-50 ⁇ g of protein immunogen or between 10 6 and 10 7 161P2F10B-expressing cells mixed in a suitable adjuvant.
  • suitable adjuvants for FP immunizations are TiterMax (Sigma) for the initial FP injection and alum gel for subsequent immunizations.
  • mice are immunized twice a week until the time they are sacrificed. Upon sacrifice, lymph nodes are removed and their B-cells are harvested for electro-cell fusion.
  • test bleeds are taken to monitor the titer and specificity of the immune response.
  • appropriate reactivity and specificity are obtained as determined by ELISA, Western blotting, immunoprecipitation, fluorescence microscopy or flow cytometric analyses, fusion and hybridoma generation are then carried out using electrocell fusion (BTX, ECM2000).
  • the invention provides for monoclonal antibodies designated: Ha16-1(3,5)18, Ha16-1(2,4)4, Ha16-1(3,5)56, Ha16-1(1)11, H16-1.68, H16-1.93, H16-7.8, H16-9.10, H16-9.44, H16-9.69, Ha16-1(1)23, Ha16-1(3,5)36, H16-1.52, H16-1.67, H16-1.86, H16-7.213, H16-9.33, Ha16-1(3,5)27.1, H16-1.61.1, H16-1(3,5)5, H16-7.200, Ha16-1(3,5)42, H16-9.65, H16-1.29.1.1, H16-3.4, H16-1.92.1.1, Ha16-1(3,5)19, and H16-1.80.
  • the antibodies listed above were shown to react and bind with cell surface 161P2F10B by flow cytometry or immobilized 161P2F10B by ELISA.
  • MAbs to 161P2F10B were generated using XenoMouse Technology® wherein the murine heavy and kappa light chain loci have been inactivated and a majority of the human heavy and kappa light chain immunoglobulin loci have been inserted.
  • MAbs designated Ha16-1(3,5)18, Ha16-1(2,4)4, Ha16-1(3,5)56, Ha16-1(1)11, Ha16-1(1)23, Ha16-1(3,5)36, Ha16-1(3,5)27.1, Ha16-1(3,5)42, and Ha16-1(3,5)19 were generated from immunization of human gamma 1 producing XenoMice with RAT1-161P2F10B cells.
  • MAbs designated H16-1.68, H16-1.93, H16-1.52, H16-1.67, H16-1.86, H16-1.61.1, H16-1(3,5)5, H16-1.29.1.1, H16-1.80, and H16-1.92.1.1 were generated from immunization of human gamma 1 producing XenoMice with purified tag5-161P2F10B.
  • MAbs designated H16-7.8, H16-9.10, H16-9.44, H16-9.69, H16-7.213, H16-9.33, and H16-7.200 were generated from immunization with human gamma 2 producing XenoMice with tag5-161P2F10B.
  • the 161P2F10B MAbs Ha16-1(3,5)18, Ha16-1(2,4)4, Ha16-1(3,5)56, Ha16-1(1)11, H16-1.68, H16-1.93, H16-7.8, H16-9.10, H16-9.44, H16-9.69, Ha16-1(1)23, Ha16-1(3,5)36, H16-1.52, H16-1.67, H16-1.86, H16-7.213, H16-9.33, Ha16-1(3,5)27.1, H16-1.61.1, H16-1(3,5)5, H16-7.200, Ha16-1(3,5)42, H16-9.65, H16-1.29.1.1, H16-3.4, H16-1.92.1.1, and Ha16-1(3,5)19 specifically bind to recombinant 161P2F10B expressing cells and endogenous cell surface 161P2F10B expressed in cancer xenograft cells ( FIG. 6 and FIG. 7 ).
  • the antibodies designated Ha16-1(3,5)18, Ha16-1(1)11, H16-1.93, H16-9.69 were sent (via Federal Express) to the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108 on 28 Mar. 2006 and assigned Accession numbers PTA-7452 and PTA-7450 and PTA-7449 and PTA-7451, respectively.
  • DNA coding sequences for 161P2F10B MAbs Ha16-1(3,5)18, Ha16-1(2,4)4, Ha16-1(3,5)56, Ha16-1(1)11, H16-1.68, H16-1.93, H16-7.8, H16-9.10, H16-9.44, H16-9.69, Ha16-1(1)23, Ha16-1(3,5)36, H16-1.52, H16-1.67, H16-1.86, H16-7.213, H16-9.33, Ha16-1(3,5)27.1, H16-1.61.1, H16-1(3,5)5, H16-7.200, Ha16-1(3,5)42, H16-9.65, H16-1.29.1.1, H16-3.4, H16-1.92.1.1, Ha16-1(3,5)19, and H16-1.80 were determined after isolating mRNA from the respective hybridoma cells with Trizol reagent (Life Technologies, Gibco BRL).
  • First strand cDNAs was generated from total RNA with oligo (dT)12-18 priming using the Gibco BRL Superscript Preamplification system.
  • First strand cDNA was amplified using human immunoglobulin variable heavy chain primers, and human immunoglobulin variable light chain primers. PCR products were cloned into the pCRScript vector (Stratagene). Several clones were sequenced and the variable heavy and light chain regions determined.
  • FIG. 2 and FIG. 3 The nucleic acid and amino acid sequences of the variable heavy and light chain regions are listed in FIG. 2 and FIG. 3 . Alignment of 161P2F10B antibodies to germline V-D-J Sequences is set forth in FIG. 4A-FIG . 4 Z and FIG. 5A-FIG . 5 X.
  • Antibodies generated using the procedures set forth in the example entitled “Generation of 161P2F10B Monoclonal Antibodies (MAbs)” were screened, identified, and characterized using a combination of assays including ELISA, FACS, affinity ranking by Surface Plasmon Resonance (BIAcore) (“SPR”), epitope grouping, affinity to recombinant 161P2F10B, and 161P2F10B expressed on the cell surface.
  • assays including ELISA, FACS, affinity ranking by Surface Plasmon Resonance (BIAcore) (“SPR”)
  • SPR Surface Plasmon Resonance
  • the protocol is as follows: 50 ⁇ l/well of hybridoma supernatant (neat) or purified antibodies (in serial dilutions) are added to 96-well FACS plates and mixed with 161P2F10B-expressing cells (endogenous or recombinant, 50,000 cells/well). The mixture is incubated at 4° C. for two hours. At the end of incubation, the cells are washed with FACS Buffer and incubated with 100 ⁇ l of detection antibody (anti-hIgG-PE) for 45 minutes at 4° C. At the end of incubation, the cells are washed with FACS Buffer, fixed with Formaldehyde and analyzed using FACScan. Data are analyzed using CellQuest Pro software.
  • Positive hybridomas identified from primary screens are transferred to 24-well plates and supernatants collected for confirmatory screens.
  • Confirmatory screens included FACS analysis on Caki-161P2F10B/Caki-neo, HepG2 (human liver cancer cell line), KU812 (human promyeolcytic cell line), SKRC-01 (human renal tumor cell line), and ELISA using Tag5-161P2F10B.
  • 161P2F10B MAbs were screened by ELISA to determine antibody isotype.
  • the protocol used is as follows, ELISA plates were coated with Tag5-161P2F10B-ECD or anti-hIgG antibody. Several sets of testing antibodies were added on the plates and incubated for 1 hour. After washing the plates to wash out unbound antibodies, bound antibodies were detected by the following HRP conjugated detection antibodies: anti-hIgG1, anti-hIgG2, anti-hIgK, and anti-hIgL.
  • SPR allows identification and real time characterization of the kinetics and affinity of protein-protein interactions and therefore is a useful technique in the selection and characterization of MAbs to target antigens of interest.
  • SPR analysis is employed to screen and characterize hybridoma supernatants and purified MAbs to 161P2F10B.
  • Hybridoma screening for MAbs to 161P2F10B by SPR biosensor are performed as follows: 50 ⁇ l/well of hybridoma supernatant (neat) diluted to 1.5-2 ⁇ g/ml with the running buffer (HBS-P, 10 ug/ml BSA) are added to 96-well plates (BIAcore) and MAbs (20 ⁇ l) are captured on goat-anti-human Fc ⁇ pAbs covalently immobilized on the surface of the CM5 sensor chip. Three (3) MAbs containing hybridoma supernatants are tested per run (cycle) on channels 2, 3 and 4 of the flow cell, where channel 1 is reserved as reference for non-specific binding.
  • 60 ⁇ l of running buffer is injected over the chip surface at the flowrate of 20 ⁇ l/min to serve as reference for drift in captured MAb baseline.
  • Sixty microliters (60 ⁇ l) of the purified recombinant 161P2F10B at 150 nM is then injected over the chip surface at the same flowrate of 20 ⁇ l/min to measure antigen binding.
  • Each cycle of antigen binding to MAbs are followed by surface regeneration with injection of 100 mM phosphoric acid (for 1 min) to strip the surface of any captured MAb.
  • the affinities are calculated from the association and dissociation rate constants. As is apparent to one of ordinary skill in the art, slow dissociation rates generally indicate higher overall affinity for MAbs.
  • the preliminary affinity data and dissociation rates are used as a basis of the selection criteria for therapeutic MAbs to 161P2F10B.
  • 161P2F10B antibodies were grouped according to epitope by evaluating their binding pattern on UG-K3 (human renal cancer cell line), or KU812 cells.
  • UG-K3 human renal cancer cell line
  • KU812 cells were biotinylated; then each of the biotinylated antibodies were incubated with KU812 in the presence of excess (100 ⁇ ) amount of non-biotinylated antibodies at 4° C. for 1 hour.
  • excess amount of antibodies will compete with biotinylated antibodies if they bind to the same epitope.
  • cells were washed and incubated with Streptavidin-PE for 45 min at 4° C.
  • the cells were analyzed using FACS. MFI values were obtained using CellQuest Pro software and were used for data analysis. As shown in FIG. 8 , twenty-five (25) 161P2F10B MAbs were epitope grouped using UG-K3 cells. Cells are highlighted to indicate self-competition (100% competition), the MFI value in these cells are background control for each biotinylated antibody. Additionally, cells with no color indicate that the two antibodies compete each other (low MFI), cells highlighted in gray (high MFI) indicate that the two antibodies bind to two distinct epitopes. The results show the antibodies that have the same binding pattern bind to the same epitope among the antibodies and that there are 16 epitope groups within the antibodies tested.
  • Tag5 expression constructs encoding either the full extracellular domain (ECD) of 161P2F10B (amino acids 46-875), the somatomedin-b-like domain (amino acids 46-157), the catalytic domain (amino acids 158-558), or the catalytic and nuclease domain (amino acids 158-875) were transfected into 293T cells and cellular lysates were made.
  • ECD extracellular domain
  • lysates were then used for immunoprecipitation with the indicated 161P2F10B MAbs or control MAb.
  • Western blotting of the immunoprecipitates was then performed using an anti-His polyclonal polyclonal MAb that recognizes the His epitope tag present on each recombinant protein.
  • the specific molecular weight band of each recombinant protein was identified by straight Western blotting of the lysates.
  • the domain containing the binding epitope of each MAb is determined through identification of the pattern of recombinant proteins immunoprecipitated by each MAb.
  • MAbs that bind to the full length ECD and to the somatomedin-b-like domain map to the somatomedin-b-like domain.
  • MAbs that bind to the full length ECD and the catalytic domain, but not the catalytic+nuclease domain map to the catalytic domain.
  • MAbs that bind the full length ECD and to the catalytic+nuclease domain, but not to the catalytic domain map to the nuclease domain.
  • Such analysis is presented in FIG. 9 .
  • Such data when combined with SPR competition data is useful in grouping together MAbs that bind to similar or overlapping epitopes as presented in FIG. 10 .
  • a panel of 33 human 161P2F10B MAbs were tested for their binding affinity to 161P2F10B on several cell lines (HepG2, KU812, SKRC-01, and RXF393) which express 161P2F10B. Briefly, twenty-three (23) serial 1:2 dilutions of purified antibodies were incubated with 161P2F10B expressing cells (50,000 cells per well) overnight at 4° C. at a final concentration of 167 nM to 0.01 pM. At the end of the incubation, cells were washed and incubated with anti-hIgG-PE detection antibody for 45 min at 4° C. After washing the unbound detection antibodies, the cells were analyzed by FACS. MFI values of each point were obtained using CellQuest Pro software and were used for the affinity calculation using Graphpad Prism software and the one site binding (hyperbola) equation. A summary of affinity values of thirteen (13) antibodies are set forth in Table VI.
  • each purified human MAb is captured onto a CM5 sensor chip surface. On average approximately 150 RUs of each MAb is captured in every cycle. A series of 5-6 dilutions of recombinant 161P2F10B ranging from 1 nM to 100 nM is injected over such surface to generate binding curves (sensograms) that are globally fit to a 1:1 interaction model using CLAMP software (Myszka and Morton, 1998).
US12/196,039 2005-03-31 2008-08-21 Antibodies and related molecules that bind to 161p2f10b proteins Abandoned US20090022663A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/196,039 US20090022663A1 (en) 2005-03-31 2008-08-21 Antibodies and related molecules that bind to 161p2f10b proteins
US12/433,760 US7811565B2 (en) 2005-03-31 2009-04-30 Antibodies and related molecules that bind to 161P2F10B proteins
US12/757,935 US8350009B2 (en) 2005-03-31 2010-04-09 Antibodies and related molecules that bind to 161P2F10B proteins
US12/881,461 US8236310B2 (en) 2005-03-31 2010-09-14 Antibodies and related molecules that bind to 161P2F10B proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US66758805P 2005-03-31 2005-03-31
US70097505P 2005-07-20 2005-07-20
US11/396,178 US7427399B2 (en) 2001-11-07 2006-03-31 Antibodies and related molecules that bind to 161P2F10B proteins
US12/196,039 US20090022663A1 (en) 2005-03-31 2008-08-21 Antibodies and related molecules that bind to 161p2f10b proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/396,178 Division US7427399B2 (en) 2001-11-07 2006-03-31 Antibodies and related molecules that bind to 161P2F10B proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/433,760 Continuation US7811565B2 (en) 2005-03-31 2009-04-30 Antibodies and related molecules that bind to 161P2F10B proteins

Publications (1)

Publication Number Publication Date
US20090022663A1 true US20090022663A1 (en) 2009-01-22

Family

ID=37054217

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/196,039 Abandoned US20090022663A1 (en) 2005-03-31 2008-08-21 Antibodies and related molecules that bind to 161p2f10b proteins
US12/433,760 Active US7811565B2 (en) 2005-03-31 2009-04-30 Antibodies and related molecules that bind to 161P2F10B proteins
US12/881,461 Active US8236310B2 (en) 2005-03-31 2010-09-14 Antibodies and related molecules that bind to 161P2F10B proteins

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/433,760 Active US7811565B2 (en) 2005-03-31 2009-04-30 Antibodies and related molecules that bind to 161P2F10B proteins
US12/881,461 Active US8236310B2 (en) 2005-03-31 2010-09-14 Antibodies and related molecules that bind to 161P2F10B proteins

Country Status (13)

Country Link
US (3) US20090022663A1 (de)
EP (3) EP2444099A1 (de)
JP (3) JP4324636B2 (de)
KR (1) KR20080003390A (de)
AU (1) AU2006230563B8 (de)
BR (1) BRPI0609655A2 (de)
CA (1) CA2603093A1 (de)
HK (1) HK1253391A1 (de)
IL (1) IL186385A0 (de)
NO (1) NO20075103L (de)
NZ (1) NZ562453A (de)
RU (1) RU2413735C2 (de)
WO (1) WO2006105488A2 (de)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110217321A1 (en) * 2010-02-08 2011-09-08 Michael Torgov Antibody drug conjugates (adc) that bind to 161p2f10b proteins
CN114173875A (zh) * 2019-03-01 2022-03-11 Xencor股份有限公司 结合enpp3和cd3的异二聚抗体

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL161268A0 (en) 2001-11-07 2004-09-27 Agensys Inc Nucleic acid and corresponding protein entitled 161p2f10b and pharmaceutical and diagnostic compositions containing the same
BRPI0609655A2 (pt) * 2005-03-31 2010-03-16 Agensys Inc anticorpos e moléculas relacionadas que ligam-se às proteìnas 161p2f10b
US8350009B2 (en) * 2005-03-31 2013-01-08 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
WO2008029807A1 (fr) * 2006-09-06 2008-03-13 Japan Science And Technology Agency Nouveau polypeptide ayant une cytotoxicité contre le cancer, procédé pour cribler le polypeptide et utilisation du polypeptide
EP2547965B1 (de) * 2010-03-15 2020-11-04 Klatu Networks Systeme und verfahren zur überwachung, beeinflussung des status und optimierung der effizienz von kühlsystemen
SG185027A1 (en) * 2010-05-03 2012-11-29 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
TW202344270A (zh) 2010-09-29 2023-11-16 美商艾澤西公司 與191p4d12蛋白結合之抗體藥物共軛物(adc)
US9353150B2 (en) 2012-12-04 2016-05-31 Massachusetts Institute Of Technology Substituted pyrazino[1′,2′:1 ,5]pyrrolo[2,3-b]-indole-1,4-diones for cancer treatment
WO2017197045A1 (en) 2016-05-11 2017-11-16 Movassaghi Mohammad Convergent and enantioselective total synthesis of communesin analogs
WO2018209239A1 (en) 2017-05-11 2018-11-15 Massachusetts Institute Of Technology Potent agelastatin derivatives as modulators for cancer invasion and metastasis
US10640508B2 (en) 2017-10-13 2020-05-05 Massachusetts Institute Of Technology Diazene directed modular synthesis of compounds with quaternary carbon centers
US11535634B2 (en) 2019-06-05 2022-12-27 Massachusetts Institute Of Technology Compounds, conjugates, and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines and uses thereof
WO2023236099A1 (en) * 2022-06-08 2023-12-14 Zhejiang Shimai Pharmaceutical Co., Ltd. Antibodies against enpp3 and uses thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5001225A (en) * 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
US6323321B1 (en) * 1997-03-05 2001-11-27 Eberhard-Karls-Universitat Tubingen Universitatsklinikum Antibody 97A6
US6342219B1 (en) * 1999-04-28 2002-01-29 Board Of Regents, The University Of Texas System Antibody compositions for selectively inhibiting VEGF
US20020137139A1 (en) * 1999-01-12 2002-09-26 Byatt John C Nucleic acid and other molecules associated with lactation and muscle and fat deposition
US20030165505A1 (en) * 2001-11-07 2003-09-04 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20030206905A1 (en) * 2001-11-07 2003-11-06 Aya Jakobovits Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20050055733A1 (en) * 2001-07-06 2005-03-10 Zairen Sun Small intestine and colon genes
US20070031335A1 (en) * 2001-04-10 2007-02-08 Aya Jakobovits Nucleic acids and corresponding proteins useful in the detection and treatment of various cancers

Family Cites Families (137)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154598B (nl) 1970-11-10 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van laagmoleculire verbindingen en van eiwitten die deze verbindingen specifiek kunnen binden, alsmede testverpakking.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4722848A (en) 1982-12-08 1988-02-02 Health Research, Incorporated Method for immunizing animals with synthetically modified vaccinia virus
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
EP0206448B1 (de) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hämoglobin, das an ein Poly(alkenylenoxid) gebunden ist
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
DE3883899T3 (de) 1987-03-18 1999-04-22 Sb2 Inc Geänderte antikörper.
US5124246A (en) 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5359100A (en) 1987-10-15 1994-10-25 Chiron Corporation Bifunctional blocked phosphoramidites useful in making nucleic acid mutimers
GB8724885D0 (en) 1987-10-23 1987-11-25 Binns M M Fowlpox virus promotors
US5254678A (en) 1987-12-15 1993-10-19 Gene Shears Pty. Limited Ribozymes
US5010175A (en) 1988-05-02 1991-04-23 The Regents Of The University Of California General method for producing and selecting peptides with specific properties
CA1340323C (en) 1988-09-20 1999-01-19 Arnold E. Hampel Rna catalyst for cleaving specific rna sequences
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
ATE104857T1 (de) 1989-03-07 1994-05-15 Genentech Inc Kovalente konjugate von lipiden und oligonukleotiden.
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
WO1991004753A1 (en) 1989-10-02 1991-04-18 Cetus Corporation Conjugates of antisense oligonucleotides and therapeutic uses thereof
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
ATE157012T1 (de) 1989-11-03 1997-09-15 Univ Vanderbilt Verfahren zur in vivo-verabreichung von funktionsfähigen fremden genen
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
ATE356869T1 (de) 1990-01-12 2007-04-15 Amgen Fremont Inc Bildung von xenogenen antikörpern
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6657103B1 (en) 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5204253A (en) 1990-05-29 1993-04-20 E. I. Du Pont De Nemours And Company Method and apparatus for introducing biological substances into living cells
IE66205B1 (en) 1990-06-14 1995-12-13 Paul A Bartlett Polypeptide analogs
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
JP3179481B2 (ja) 1990-06-27 2001-06-25 プリンストン ユニバーシティ 突然変異体p53検出用プローブ
US5650489A (en) 1990-07-02 1997-07-22 The Arizona Board Of Regents Random bio-oligomer library, a method of synthesis thereof, and a method of use thereof
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ES2108048T3 (es) 1990-08-29 1997-12-16 Genpharm Int Produccion y utilizacion de animales inferiores transgenicos capaces de producir anticuerpos heterologos.
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5733731A (en) 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
ATE207080T1 (de) 1991-11-25 2001-11-15 Enzon Inc Multivalente antigen-bindende proteine
DE69334255D1 (de) 1992-02-06 2009-02-12 Novartis Vaccines & Diagnostic Marker für Krebs und biosynthetisches Bindeprotein dafür
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5541061A (en) 1992-04-29 1996-07-30 Affymax Technologies N.V. Methods for screening factorial chemical libraries
JP3285355B2 (ja) 1992-06-04 2002-05-27 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア invivo遺伝子治療のための方法及び組成物
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US6177401B1 (en) 1992-11-13 2001-01-23 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften Use of organic compounds for the inhibition of Flk-1 mediated vasculogenesis and angiogenesis
US5559410A (en) 1992-12-17 1996-09-24 Valeo Systemes D'essuyage Device for controlling the function of an electric starting motor for a windshield wiper of a rear window of an automotive vehicle and for supplying an electric motor particularly to activate the windshield
US5576220A (en) 1993-02-19 1996-11-19 Arris Pharmaceutical Corporation Thin film HPMP matrix systems and methods for constructing and displaying ligands
CN1127527A (zh) 1993-05-17 1996-07-24 加利福尼亚大学董事会 人免疫缺陷型病毒感染和艾滋病的核酶基因治疗方法
US5837501A (en) 1993-07-09 1998-11-17 Akzo Nobel N.V. Nucleic acid quantitation by co-amplification of target with multiple internal controls
US5679647A (en) 1993-08-26 1997-10-21 The Regents Of The University Of California Methods and devices for immunizing a host against tumor-associated antigens through administration of naked polynucleotides which encode tumor-associated antigenic peptides
US5804566A (en) 1993-08-26 1998-09-08 The Regents Of The University Of California Methods and devices for immunizing a host through administration of naked polynucleotides with encode allergenic peptides
DK0735893T3 (da) 1993-09-14 2009-03-09 Pharmexa Inc PAN DR-bindende peptider til styrkelse af immunsvaret
US5681697A (en) 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor
CH686982A5 (fr) 1993-12-16 1996-08-15 Maurice Stroun Méthode pour le diagnostic de cancers.
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
IL112248A0 (en) 1994-01-25 1995-03-30 Warner Lambert Co Tricyclic heteroaromatic compounds and pharmaceutical compositions containing them
US5593853A (en) 1994-02-09 1997-01-14 Martek Corporation Generation and screening of synthetic drug libraries
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5739118A (en) 1994-04-01 1998-04-14 Apollon, Inc. Compositions and methods for delivery of genetic material
US5525735A (en) 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
JP4064452B2 (ja) 1994-07-20 2008-03-19 ザ ジェネラル ホスピタル コーポレーション 蛋白質相互作用を検出するための相互作用補足系
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5681702A (en) 1994-08-30 1997-10-28 Chiron Corporation Reduction of nonspecific hybridization by using novel base-pairing schemes
US5736524A (en) 1994-11-14 1998-04-07 Merck & Co.,. Inc. Polynucleotide tuberculosis vaccine
US5962428A (en) 1995-03-30 1999-10-05 Apollon, Inc. Compositions and methods for delivery of genetic material
DE69636866D1 (en) 1995-04-11 2007-03-15 Gen Hospital Corp Reverse "two-hybrid"-systeme
US5922687A (en) 1995-05-04 1999-07-13 Board Of Trustees Of The Leland Stanford Junior University Intracellular delivery of nucleic acids using pressure
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
EP2258726A1 (de) 1995-06-14 2010-12-08 The Regents of the University of California Menschliche Antikörper mit hoher Affinität gegen c-erbB-2
US5585639A (en) 1995-07-27 1996-12-17 Hewlett-Packard Company Optical scanning apparatus
US5569588A (en) 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
GB9520822D0 (en) 1995-10-11 1995-12-13 Wellcome Found Therapeutically active compounds
GB9624482D0 (en) 1995-12-18 1997-01-15 Zeneca Phaema S A Chemical compounds
CU22559A1 (es) 1996-01-17 1999-05-03 Ct Ingenieria Genetica Biotech Sistema de expresión de antígenos heterologos en e. coli como proteínas de fusión
US6153380A (en) 1996-01-23 2000-11-28 Rigel Pharmaceuticals, Inc. Methods for screening for transdominant intracellular effector peptides and RNA molecules
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
JP2001504799A (ja) 1996-03-11 2001-04-10 エピミューン インコーポレイテッド Hla分子への結合アフィニティーが増加したペプチド
WO1997041255A1 (en) 1996-04-26 1997-11-06 Massachusetts Institute Of Technology Three-hybrid screening assay
PT912559E (pt) 1996-07-13 2003-03-31 Glaxo Group Ltd Compostos heterociclicos fundidos como inibidores de proteina tirosina quinase
ID19430A (id) 1996-07-13 1998-07-09 Glaxo Group Ltd Senyawa senyawa heterosiklik
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
EP0960204A4 (de) 1996-07-26 2002-01-23 Sloan Kettering Inst Cancer Methoden und reagenzien zur genetischen immunisierung
US5925523A (en) 1996-08-23 1999-07-20 President & Fellows Of Harvard College Intraction trap assay, reagents and uses thereof
ID18494A (id) 1996-10-02 1998-04-16 Novartis Ag Turunan pirazola leburan dan proses pembuatannya
US6365797B1 (en) 1997-10-15 2002-04-02 The Regents Of The University Of California Mice models of human prostate cancer progression
AU4818997A (en) 1996-10-15 1998-05-11 Regents Of The University Of California, The Animal models of human prostate cancer progression
US5846722A (en) 1996-10-16 1998-12-08 Terrapin Technologies, Inc. System to detect small molecule/peptide interaction
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
EP0984930B1 (de) 1997-05-07 2005-04-06 Sugen, Inc. 2-indolinonderivate als modulatoren der proteinkinase-ativität
CA2291709A1 (en) 1997-05-30 1998-12-03 Merck & Co., Inc. Novel angiogenesis inhibitors
JP4959049B2 (ja) 1997-08-22 2012-06-20 アストラゼネカ・ユーケイ・リミテッド 血管新生阻害剤としてのオキシインドリルキナゾリン誘導体
CA2303830A1 (en) 1997-09-26 1999-04-08 Merck & Co., Inc. Novel angiogenesis inhibitors
CZ20001709A3 (cs) 1997-11-11 2001-12-12 Pfizer Products Inc. Deriváty thienopyrimidu a thienopyridinu, farmaceutické kompozice a způsoby léčení na jejich bázi
GB9800569D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
GB9800575D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
AU4004699A (en) 1998-05-15 1999-12-06 Quark Biotech, Inc. Mechanical stress induced genes, expression products therefrom, and uses thereof
CA2314156C (en) 1998-05-29 2010-05-25 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
UA60365C2 (uk) 1998-06-04 2003-10-15 Пфайзер Продактс Інк. Похідні ізотіазолу, спосіб їх одержання, фармацевтична композиція та спосіб лікування гіперпроліферативного захворювання у ссавця
AU1208100A (en) 1998-10-15 2000-05-01 Genetics Institute Inc. Secreted expressed sequence tags (sests)
UA71945C2 (en) 1999-01-27 2005-01-17 Pfizer Prod Inc Substituted bicyclic derivatives being used as anticancer agents
JP3270834B2 (ja) 1999-01-27 2002-04-02 ファイザー・プロダクツ・インク 抗がん剤として有用なヘテロ芳香族二環式誘導体
JP3759412B2 (ja) 1999-05-19 2006-03-22 エベルハルト‐カルルス‐ウニベアジテート、テュービンゲン、ウニベアジテートスクリニクム 好塩基球および/もしくはマスト細胞、ならびに/またはこれらの前駆体細胞、ならびに/またはこれらの表面構造の検出および/または定量および/または単離のための抗体の使用
WO2001057251A2 (en) 2000-02-04 2001-08-09 Aeomica, Inc. Methods and apparatus for predicting, confirming, and displaying functional information derived from genomic sequence
AU2001241541A1 (en) 2000-02-17 2001-08-27 Millennium Predictive Medicine, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of human prostate cancer
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
KR100857943B1 (ko) 2000-11-30 2008-09-09 메다렉스, 인코포레이티드 인간 항체의 제조를 위한 형질전환 트랜스염색체 설치류
NZ569856A (en) 2001-01-05 2010-03-26 Pfizer Antibodies to insulin-like growth factor 1 receptor
WO2002079411A2 (en) 2001-03-29 2002-10-10 Van Andel Institute Microarray gene expression profiling in clear cell renal cell carcinoma: prognosis and drug target identification
WO2003016475A2 (en) 2001-08-14 2003-02-27 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
DE10160921B4 (de) * 2001-12-06 2005-09-22 Eberhard-Karls-Universität Tübingen Universitätsklinikum Verfahren zum Nachweis von Allergien und Verwendung eines Antikörpers zum Nachweis von Allergien
US7662387B2 (en) 2003-02-20 2010-02-16 Seattle Genetics Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
EP1732600A2 (de) 2004-03-26 2006-12-20 Pfizer Products Incorporated Verwendungen von anti-ctla-4-antikörpern
BRPI0609655A2 (pt) 2005-03-31 2010-03-16 Agensys Inc anticorpos e moléculas relacionadas que ligam-se às proteìnas 161p2f10b
JO3421B1 (ar) 2011-06-20 2019-10-20 H Lundbeck As طريقة لإعطاء 1-((1ار.3س)-6-كلورو-3-فينيل-اندان-1-ايل)-1.2.2-ترايميثيل- بيبيرازين واملاجها لمعالجة انفصام الشخصية
CN104010774B (zh) 2011-09-15 2017-10-13 康富真信息技术股份有限公司 用于自动生成机器人程序的系统和方法

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5001225A (en) * 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
US6323321B1 (en) * 1997-03-05 2001-11-27 Eberhard-Karls-Universitat Tubingen Universitatsklinikum Antibody 97A6
US20020137139A1 (en) * 1999-01-12 2002-09-26 Byatt John C Nucleic acid and other molecules associated with lactation and muscle and fat deposition
US6342219B1 (en) * 1999-04-28 2002-01-29 Board Of Regents, The University Of Texas System Antibody compositions for selectively inhibiting VEGF
US20070031335A1 (en) * 2001-04-10 2007-02-08 Aya Jakobovits Nucleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20050265924A1 (en) * 2001-04-10 2005-12-01 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20050055733A1 (en) * 2001-07-06 2005-03-10 Zairen Sun Small intestine and colon genes
US20030165505A1 (en) * 2001-11-07 2003-09-04 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20030206905A1 (en) * 2001-11-07 2003-11-06 Aya Jakobovits Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20050265921A1 (en) * 2001-11-07 2005-12-01 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20060002993A1 (en) * 2001-11-07 2006-01-05 Challita-Eid Pia M Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US7067130B2 (en) * 2001-11-07 2006-06-27 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20060275211A1 (en) * 2001-11-07 2006-12-07 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
US20070004913A1 (en) * 2001-11-07 2007-01-04 Challita-Eid Pia M Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20030191073A1 (en) * 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US7226594B2 (en) * 2001-11-07 2007-06-05 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US7405290B2 (en) * 2001-11-07 2008-07-29 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110217321A1 (en) * 2010-02-08 2011-09-08 Michael Torgov Antibody drug conjugates (adc) that bind to 161p2f10b proteins
US8609092B2 (en) * 2010-02-08 2013-12-17 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 161P2F10B proteins
US20140194601A1 (en) * 2010-02-08 2014-07-10 Agensys, Inc. Antibody drug conjugates (adc) that bind to 161p2f10b proteins
US9308278B2 (en) * 2010-02-08 2016-04-12 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 161P2F10B proteins
CN114173875A (zh) * 2019-03-01 2022-03-11 Xencor股份有限公司 结合enpp3和cd3的异二聚抗体

Also Published As

Publication number Publication date
RU2413735C2 (ru) 2011-03-10
JP5480921B2 (ja) 2014-04-23
WO2006105488A2 (en) 2006-10-05
BRPI0609655A2 (pt) 2010-03-16
EP3300739A2 (de) 2018-04-04
EP1863848A4 (de) 2009-09-23
EP3300739A3 (de) 2018-07-18
AU2006230563A2 (en) 2010-02-11
HK1253391A1 (zh) 2019-06-14
EP1863848A2 (de) 2007-12-12
KR20080003390A (ko) 2008-01-07
US20090214556A1 (en) 2009-08-27
WO2006105488A3 (en) 2009-04-16
NZ562453A (en) 2010-04-30
AU2006230563B2 (en) 2010-06-03
JP2012090642A (ja) 2012-05-17
EP2444099A1 (de) 2012-04-25
AU2006230563A1 (en) 2006-10-05
NO20075103L (no) 2007-12-28
JP2008538182A (ja) 2008-10-16
US20110020353A1 (en) 2011-01-27
US8236310B2 (en) 2012-08-07
IL186385A0 (en) 2011-08-01
CA2603093A1 (en) 2006-10-05
JP2009171982A (ja) 2009-08-06
AU2006230563B8 (en) 2010-06-17
JP4324636B2 (ja) 2009-09-02
US7811565B2 (en) 2010-10-12
RU2007140311A (ru) 2009-05-10

Similar Documents

Publication Publication Date Title
US7427399B2 (en) Antibodies and related molecules that bind to 161P2F10B proteins
US8278424B2 (en) Antibodies that bind to PSCA proteins for diagnosis of cancer
US7595379B2 (en) Antibodies and related molecules that bind to PSCA proteins
EP2185574B1 (de) An 24p4c12-proteine bindende antikörper und zugehörige moleküle
US8236310B2 (en) Antibodies and related molecules that bind to 161P2F10B proteins
US7541442B2 (en) Antibodies and related molecules that bind to PSCA proteins
US8039597B2 (en) Antibodies and related molecules that bind to 24P4C12 proteins
CA2567449C (en) Antibodies and related molecules that bind to psca proteins
US20090068098A1 (en) Antibodies and related molecules that bind to 58p1d12 proteins
AU2006237616B2 (en) Antibodies and related molecules that bind to PSCA proteins
CA2742088A1 (en) Antibodies that bind psca proteins for diagnosis of cancer
US8350009B2 (en) Antibodies and related molecules that bind to 161P2F10B proteins
US20100047166A1 (en) Antibodies and related molecules that bind to 58p1d12 proteins
WO2010022279A1 (en) Antibodies and related molecules that bind to 58p1d12 proteins

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGENSYS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JAKOBOVITS, AYA;KANNER, STEVEN B.;CHALLITA-EID, PIA M.;AND OTHERS;REEL/FRAME:021429/0794

Effective date: 20060328

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION