WO2008039874A2 - Cancer stem cell antigen vaccines and methods - Google Patents

Cancer stem cell antigen vaccines and methods Download PDF

Info

Publication number
WO2008039874A2
WO2008039874A2 PCT/US2007/079600 US2007079600W WO2008039874A2 WO 2008039874 A2 WO2008039874 A2 WO 2008039874A2 US 2007079600 W US2007079600 W US 2007079600W WO 2008039874 A2 WO2008039874 A2 WO 2008039874A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cancer stem
stem cell
cancer
neural
Prior art date
Application number
PCT/US2007/079600
Other languages
French (fr)
Other versions
WO2008039874A3 (en
Inventor
John S. Yu
Original Assignee
Cedars-Sinai Medical Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cedars-Sinai Medical Center filed Critical Cedars-Sinai Medical Center
Priority to CA2700573A priority Critical patent/CA2700573C/en
Priority to EP07843269.7A priority patent/EP2084267B1/en
Publication of WO2008039874A2 publication Critical patent/WO2008039874A2/en
Publication of WO2008039874A3 publication Critical patent/WO2008039874A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001128CD44 not IgG
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001129Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001148Regulators of development
    • A61K39/00115Apoptosis related proteins, e.g. survivin or livin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001154Enzymes
    • A61K39/001162Kinases, e.g. Raf or Src
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001154Enzymes
    • A61K39/001163Phosphatases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464428CD44 not IgG
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464429Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464448Regulators of development
    • A61K39/46445Apoptosis related proteins, e.g. survivin or livin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464454Enzymes
    • A61K39/464462Kinases, e.g. Raf or Src
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464499Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0695Stem cells; Progenitor cells; Precursor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration

Abstract

Method of stimulating an immune response (e.g., to treat cancer) include administering to a patient a composition including dendritic cells that present cancer stem cell antigens. Compositions including cancer stem cell antigens are also provided herein.

Description

CANCER STEM CELL ANTIGEN VACCINES AND METHODS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of priority to U.S. Provisional Patent Application Serial No. 60/826,955, filed on September 26, 2006, the entire contents of which are incorporated herein by reference.
BACKGROUND
Over the past 30 years, a wealth of information has been generated concerning the in vivo and in vitro properties of brain tumors and rodent models of brain tumor. The 9L gliosarcoma, which was generated from inbred Fisher rats, is a widely used syngeneic rat model for brain tumors. Originally produced by N-mcthyl-nitrosourea mutagenesis in Fisher rats, the tumor was cloned and designated 9L gliosarcoma because of its dual appearance of a glioblastoma and a sarcoma (Benda et al., J. Neurosurg., 34:310-323, 1971; Schmidek ct al., J. Neurosurg., 34:335-340, 1971). The tumor could be proliferated under in vivo and in vitro conditions, making it a useful candidate as a glioma tumor model. The 9L gliosarcoma model clinically mimics rapidly growing and fatal intracerebral tumors, making it the most widely used rat brain tumor model.
Stem cells have been defined as multipotent, self-renewing cells with the potential to differentiate into multiple cell types. Systems have been developed to identify the first neural stem cells in a defined media, whereby striatal embryonic progenitors could be harvested and grown in culture as undifferentiated neurospheres (clonally derived aggregates of cells derived from a single stem cell) under the influence of the mitogens EGF and bFGF. Many of these cells expressed nestin (an intermediate filament found in neuroepithelial stem cells), but not markers for the more differentiated principal cell types of the CNEJ — neuronal and glial cells. However, when grown on coverslips treated with poly-L-ornithine, a substrate that allows cellular adhesion, many of the cells within the neurospheres differentiated into neurons and astrocytes with discontinued nestin expression. The isolated striatal cells fulfilled the critical features expected from neural stem cells: an unlimited capacity for self-renewal and capacity to differentiate into the principal mature neural cells (Pottcn et al., Development, 110:1001-1020, 1990; Lee et al., Nat. Neurosci., 8:723-729, 2005; Marie et al., J. Neurosci., 23:240-251, 2003; Weissman et al., Annu. Rev. Cell. Dcv. Biol., 17:387-403, 2001 ; Seaberg et al., Trends Neurosci., 26:125-131, 2003; Reya et al., Nature, 414:105-111, 2001). Under similar circumstances "cancer stem cells" appear to have the same characteristics of self-renewal and multipotentcy.
Malignant brain tumors carry a poor prognosis even in the midst of surgical, radio-, and chemotherapy. With the poor prognosis of brain tumors amidst the available therapeutic treatments, there exists a significant need for more effective therapies to treat such tumors.
SUMMARY
This invention is based, inter alia, on the discovery that vaccines based on cancer stem cell antigens are exceptionally useful for therapy of cancer. Immunization of animals with dendritic cells pulsed with antigens from isolated cancer stem cells provided a significant survival benefit as compared to immunization with dendritic cells pulsed with differentiated tumor cells. Cancer stem cells were found to express major histocompatibility (MHC), indicating that they can display antigens. Further, proteins differentially expressed in cancer stem cells as compared to differentiated tumor cells were identified. These proteins can be useful in providing antigenic compositions for treatment of cancers (e.g., neural cancers such as gliomas).
Accordingly, this application provides methods and compositions for cancer vaccines that target cancer stem cells. Cancer stem cells are important in tumor maintenance, proliferation, and resistance to chemotherapy and radiation therapy. Thus, the new vaccines that target cancer stem cells provide for greater therapeutic and/or prophylactic effect, particularly in cancers that are resistant to conventional treatments. In one aspect, this application provides methods of treating cancer (e.g., neural cancer) in a patient that include administering to the patient a composition that includes antigen presenting cells (e.g., dendritic cells) that present cancer stem cell antigens (e.g., neural stem cell antigens). In another aspect, this application provides methods of treating cancer (e.g., neural cancer) in a patient that include the steps of: obtaining a population of antigen presenting cells (e.g., dendritic cells); contacting the antigen presenting cells with a cancer stem cell antigen composition (e.g., a neural cancer stem cell antigen composition) under conditions such that the antigen presenting cells present cancer stem cell antigens (e.g., neural cancer stem cell antigens); and administering to a patient a composition that includes the antigen presenting cells. In a further aspect, this application provides methods of inducing or stimulating an immune response in a patient, and methods of generating antibodies specific for cancer stem cell antigens, that include administering to the patient a composition that includes antigen presenting cells (e.g., dendritic cells) that present cancer stem cell antigens (e.g., neural stem cell antigens). In another aspect, this application provides methods of inducing or stimulating an immune response in a patient, and methods of generating antibodies specific for cancer stem cell antigens, by obtaining a population of antigen presenting cells (e.g., dendritic cells); contacting the dendritic cells with a cancer stem cell antigen composition (e.g., a neural cancer stem cell antigen composition) under conditions such that the antigen presenting cells present cancer stem cell antigens (e,g., neural cancer stem cell antigens); and administering to a patient a composition that includes the antigen presenting cells.
In a further aspect, this application provides methods of preparing a cancer vaccine (e.g., a neural cancer vaccine), that include the steps of: obtaining a population of antigen presenting cells (e.g., dendritic cells), and contacting the antigen presenting cells with a cancer stem cell antigen composition (e.g., a neural cancer stem cell antigen composition) under conditions such that the antigen presenting cells present cancer stem cell antigens (e.g., neural cancer stem cell antigens), thus preparing a cancer vaccine. In some embodiments, the methods further include administering the vaccine to a patient.
In another aspect, this application provides methods of preparing a cell vaccine for treating a cancer (e.g., a neural cancer) by obtaining mononuclear cells from a subject; culturing the mononuclear cells in vitro under conditions in which mononuclear cells differentiate into antigen presenting cells; isolating cancer stem cells (e.g., neural cancer stem cells) from the same or different subject; preparing a cancer stem cell antigen composition (e.g., a neural cancer stem cell antigen composition) from the cancer stem cells; and culturing the antigen presenting cells in the presence of the cancer stem cell antigen composition, thus preparing a cell vaccine. In some embodiments, the methods further include administering the vaccine to a patient.
In some embodiments of any of the above aspects, the antigen presenting cells are autologous to the subject or patient. In some embodiments, the antigen presenting cells are allogeneic to the subject or patient.
In some embodiments of any of the above aspects, the cancer stem cell antigen composition is a lysate of cancer stem cells (e.g., neural stem cells). In other embodiments, the cancer stem cell antigen composition is an acid eluate of cancer stem cells (e.g., neural cancer stem cells).
In some embodiments of any of the above aspects, the neural cancer stem cell antigen composition is obtained from a brain tumor (e.g., a glioma). In some embodiments of any of the above aspects, the cancer stem cells express CD133. In some embodiments of any of the above aspects, the cancer stem cell antigen composition includes one or more isolated peptides of CD133, CD90, CD44, CXCR4, Nestin, Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK)5 GLIl, PTCHl, BmM, phosphoscrine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, cIAP2, NAIP, or survivin. In some embodiments, the peptides are synthetic. In another aspect, this application provides kits for preparing a cell vaccine for inducing an immune response or treating a cancer (e.g., a brain cancer) that include one or more isolated peptides of CD133, CD90, CD44, CXCR4, Nestin, Musashi-l (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, cIAP2, NAlP, or survivin.
In a further aspect, this application provides compositions (e.g., immunomodulatory compositions) that include antigen presenting cells (e.g., dendritic cells) that present cancer stem cell antigens (e.g., neural cancer stem cell antigens). In some embodiments, the cancer stem cell antigens include peptides of one or more of CD133, CD90, CD44, CXCR4, Nestin, Musashi-I (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, cIAP2, NAIP, or survivin. In some embodiments, the compositions are produced by methods described herein. In another aspect, this application provides for the use of compositions (e.g., immunomodulatory compositions) that include antigen presenting cells (e.g., dendritic cells) that present cancer stem cell antigens (e.g., neural cancer stem cell antigens) in the preparation of a medicament for modulating an immune response or treating cancer in a subject. In some embodiments, the cancer stem cell antigens include peptides of one or more of CD 133, CD90, CD44, CXCR4, Nestin, Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoscrine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, cIAP2, NAIP, or survivin. In certain embodiments, the compositions are produced by methods described herein. This application also provides immunogenic compositions that include, or encode cancer stem cell antigens, and methods of using the compositions. For example, preparations of cancer stem cell antigens, for use as cancer vaccines (e.g., peptide vaccines, DNA vaccines) are provided.
"Beneficial results" may include, but are in no way limited to, lessening or alleviating the severity of a disease condition, inhibiting the disease condition from worsening, improving symptoms of a disease condition, and prolonging a patient's life or life expectancy.
"Cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, breast cancer, colon cancer, lung cancer, prostate cancer, hepatocellular cancer, gastric cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, cancer of the urinary tract, thyroid cancer, renal cancer, carcinoma, melanoma, head and neck cancer, and brain cancer; including, but not limited to, gliomas, glioblastomas, glioblastoma multiforme (GBM), oligodendrogliomas, primitive neuroectodermal tumors, low, mid and high grade astrocytomas, ependymomas (e.g., myxopapillary ependymoma papillary ependymoma, subependymoma, anaplastic ependymoma), oligodendrogliomas, medulloblastomas, meningiomas, pituitary adenomas, neuroblastomas, and craniopharyngiomas.
"Conditions" and "disease conditions," as used herein may include, but are in no way limited to any form of neoplastic cell growth and proliferation, whether malignant or benign, pre-cancerous and cancerous cells and tissues; in particular, gliomas, glioblastomas, glioblastoma multiforme (GBM), oligodendrogliomas, primitive neuroectodermal tumors, low, mid and high grade astrocytomas, ependymomas (e.g., myxopapillary ependymoma papillary ependymoma, subependymoma, anaplastic ependymoma), oligodendrogliomas, medulloblastomas, meningiomas, pituitary adenomas, neuroblastomas, and craniopharyngiomas.
"Mammal" as used herein refers to any member of the class Mammalia, including, without limitation, humans and nonhuman primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs, and the like. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be included within the scope of this term. The terms "patient" and "subject" are used herein interchangeably, and cover mammals, including humans.
"Pathology" of cancer includes all phenomena that compromise the well-being of the patient. This includes, without limitation, abnormal or uncontrollable cell growth, metastasis, interference with the normal functioning of neighboring cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of inflammatory or immunological response, neoplasia, premalignancy, malignancy, invasion of surrounding or distant tissues or organs, such as lymph nodes, etc.
"Stem-like" or "stem," as used herein refers to cells that are able to self renew from a single clone, differentiate into terminal cell types, and be serially transplantable in immunodeficient animals.
"Treatment" and "treating," as used herein refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder even if the treatment is ultimately unsuccessful. Those in need of treatment include those already diagnosed as having the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. For example, in tumor (e.g., cancer) treatment, a therapeutic agent may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutic agents or by the subject's own immune system.
"Tumor," as used herein refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference herein in their entirety, hi case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Other features and advantages of the invention will be apparent from the following detailed description, and from the claims. BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a schematic diagram of an exemplary process for vaccination of a patient with tumor antigen-pulsed dendritic cells. Monocytes are isolated from a patient and used to generate immature dendritic cells. After excision of a tumor or a portion of the tumor, brain tumor stem-like cells are isolated and used to prepare antigens for vaccination. The dendritic cells are pulsed with the antigens derived from the brain tumor stem-like cells. The mature antigen-presenting dendritic cells are then used to vaccinate the patient, providing an increased immune response against the tumor or residual tumor cells.
Figure 2 is a schematic diagram depicting an exemplary method of isolation and generation of tumor neurospheres.
Figures 3A-3F are a set of histograms depicting FACS analysis of neurospheres differentiated for 2 weeks. Neurospheres differentiated for 2 weeks show (3B) 96% of cells positive for the astrocyte marker GFAP; (3C) 88% of cells positive for the NSC marker nestin; (3D) 29% of cells positive for the neuronal marker MAP2; (3E) 0% of cells positive for the neuronal marker beta-tubulin III; (3F) 6% of cells positive for the oligodendrocyte marker myelin/oligodendroctye. The samples were compared to a negative control as shown (3A).
Figure 4Λ is a graph depicting volume of tumors induced by neurospheres (n = 4) and monolayer cells (n = 4) at day 18. Tumors induced by neurospheres were significantly larger (p < 0.02) than those from the monolayer group.
Figure 4B is a line chart depicting survival of rats with tumors induced by neurospheres (n = 9) and monolayer cells (n = 9). On average, animals in the neurosphere group died at a significantly (p < 0.02) earlier time point compared to those in the monolayer group. Figures 5A-5E are a set of micrographs of tumors formed by implantation of neurospheres. (5A) neurospheres formed high grade gliomas with necrosis as seen on H&E; (5B) large and well circumferential tumors grew as evidenced in non-stained sections; (5C) large and well circumferential tumors grew as evidenced in sections stained for the nuclear marker DAPl; (5D) & (5E) comparison of non-tumor area (5D) and tumor area (5E) stained for reticulin reveals the high levels of reticulin formed in tumor engulfed regions, showing the histological sarcomatous component of the gliosarcoma. Scale bar = 3000 μm (5A) Scale bar = 1250 μm (5B, 5C); Scale bar = 250 μm (5D, 5E). Figures 6A-6B are line graphs depicting chemorcsistance of neurospheres and monolayer cells to the indicated concentrations of carboplatin (6A) and Temodar (6B). Each value represents the mean of three independent trials. Neurospheres were significantly more resistant (p < 0.05) to both chemotherapeutic agents as compared to monolayer cells. Figure 7 is a bar graph depicting proliferation rates of neurospheres and monolayer cells in serum-containing media for 2 days. The WST-I proliferation assay was used to measure absorbance, which directly correlates to the cell number. The monolayer cells proliferated at a significantly (p > 0.05) greater rate than the neurospherc cells. Figures 8A-8C are micrographs depicting primary culture of adult human glioblastoma cells. (8A) Neurosphere-like tumor cells were found in glioblastoma primary cell culture in F-t2/DMEM 10% FBS medium. Scale bar = 200 μm. (8B) Neurosphere derived from a single isolated CDl 33+ cell cultured in serum free medium with EGF/FGF. Scale bar = 200 μm. (8C) CD 133 expression on a neurosphere derived from a single isolated CDl 33+ cell in serum free medium with EGF/FGF. Staining indicates CD 133 expression. Scale bar = 50 μm.
Figures 9A-9C are histograms depicting CDl 33 protein expression on primary cultured tumor cells. Tumor cells were cultured in F-12/DMEM 10% FBS medium for 3-4 passages and stained with specific mAb to CDl 33 and isotype control-matched mAb. Results are given as the percentage of CD133 positive cells in the total population. In the histograms, the thick lines represent staining with CD 133 mAb, and the thin lines represent the isotype control-matched mAb. (9A) tumor cells from patient No. 1049; (9B) tumor cells from patient No. 377; (9C) tumor cells from patient No. 66.
Figures 10A-10D are line graphs depicting drug sensitivity of CD133 positive cancer stem cells derived from patient No. 66. Tumor cells of No.66 were cultured in FBS/F-12/DMEM medium for 4 passages. Both CD 133 positive and negative rumor cells were collected by FACS sorting. IxIO4 cells/well were plated in 96- well plate and treated with the indicated concentrations of VP- 16, Taxol, temozolomide, and carboplatin for 48 hours in FBS/F-12/ DMEM medium. * indicates p < 0.05 compared to autologous CD 133 negative cells. Data are representative of two independent experiments. (10A) VP16; (1 OB) Taxol; (1 OC) temozolomide; (1 OD) carboplatin.
Figures 1 IA-11C are a set of line graphs depicting drug sensitivity of CD133 positive cancer stem cells derived from patient Nos. 377 and 1049. Tumor cells of patient
No.377 and No. 1049 were cultured in FBS/F-12/DMEM medium for 3 passages. Both CDl 33 positive and negative tumor cells were collected by FACS sorting. IxIO4 cells/well were plated in 96-well plate and treated with the indicated concentrations of carboplatin (1 IA and 11C) or VP-16 (1 IB) for 48 hours in FBS/F-12/DMAEM medium. * indicates p < 0.05 compared to autologous CD 133 negative cells. Data are representative of two independent experiments. Figure 12 is a bar graph depicting CD 133 mRNA expression in primary (P) and recurrent (R) tumor tissue. Total RNA was extracted from both primary and recurrent tumor tissue derived from five patients, and CDl 33 mRNA expression was measured by real-time qPCR. The relative CDl 33 mRNA level of recurrent tumor was presented as the fold increase compared to that of autologous primary tumor tissue. Data are representative of two independent experiments.
Figure 13 is a line graph depicting survival of 9L tumor-bearing rats vaccinated with dendritic cells pulsed with antigens from 9L adherent cells (9L-AC), 9L daughter cells (9L-DC), 9L neurospheres (9L-NS), and dendritic cells alone (CONTROL). Kaplan- Meier survival curve showed that the 9L-DC group had significantly longer survival than the other groups (p = 0.0015).
Figure 14 is a bar graph depicting IFNγ production of splcnocytes harvested from tumor bearing rats that were re-stimulated and by either re-exposure to a 9L tumor cell target (Target) or to no target (NO Target). The re-stimulated splenocytes from NS vaccinated rats released high level of IFNγ when re-exposed to NS, whereas splenocytes from rats vaccinated with AC or DtC had no detectable IFNγ in response to re-exposure to AC or DC.
Figures 15A and 15B are a pair of micrographs of tumor slices from rats vaccinated with dendritic cells pulsed with daughter cells (17A) and neurospheres (17B) and stained with anti-CD4 antibody. Greater infiltration of CD4+ cells was observed in the tumors of rats vaccinated with neurosphere-pulsed dendritic cells. (Magnification *40).
Figure 16 is a structural diagram of CD133 depicting extracellular, intracellular, and transmembrane regions of CD 133.
Figures 17A and 17B are cell plots and histograms depicting expression of isotype control (17A) and MHC class I expression (17B) in human cancer stem cells. Figures 18A and 18B are cell plots and histograms depicting expression of isotype control (18A) and MHC class I expression (18B) in human neural stem cells. DETAILED DESCRIPTION
The present application describes compositions useful as vaccines for treating cancer (e.g., neural cancers, e.g., gliomas) that include dendritic cells pulsed with antigens obtained from cancer stem cells (e.g., neurospheres); methods of producing vaccines that include dendritic cells pulsed with antigens obtained from cancer stem cells; methods of treating cancer with vaccines that include dendritic cells pulsed with antigens obtained from cancer stem cells; and kits for treating cancer that include dendritic cells pulsed with antigens obtained from cancer stem cells.
Greater tumor infiltration of cytotoxic T cells was observed in animals vaccinated against cancer stem cell antigens, and a stronger response against tumor cells was observed in T cells isolated from animals vaccinated with cancer stem cell antigens, than in responses in which antigens were not prepared from stem cell enriched cell populations. Cancer stem cells obtained from brain tumors were capable of self-renewal and proliferation, and could recapitulate the tumor when injected into rats. Isolated cancer stem cells formed more aggressive tumors as compared to differentiated tumor cells in vitro, and the cancer stem cells showed a higher resistance to chemotherapeutic agents. Similarly, CD133-positive cancer stem cells were obtained from human tumors. These cells were similarly resistant to chemotherapeutic agents and CD133-positive cells were found at a higher level in patients in whom tumors had recurred following resection.
Vaccination Methods
Described herein are methods of vaccinating a subject, e.g., to treat cancer (e.g., neural cancer, e.g., gliomas) with antigen-presenting cells ("APC"), e.g., dendritic cells ("DC"), that include antigens from cancer stem cells, e.g., presented on the surface of the antigen-presenting cells. Dendritic cells (e.g., autologous or allogeneic dendritic cells) are contacted with cancer stem cell antigens as a cell lysate, acid elution, cell extract, partially purified antigens, purified antigens, isolated antigens, partially purified peptides, purified peptides, isolated peptides, synthetic peptides, or a combination of two or more of the above. The antigen-presenting cells are then administered to a subject in need of cancer vaccination (e.g., a subject diagnosed with or at risk for cancer) to treat the cancer. Figure 1 is a schematic diagram of an exemplary process for vaccination of a patient with cancer stem cell antigen-pulsed dendritic cells. Cancer Stem Cells
The "cancer stem cell" hypothesis proposes that only a small portion of a tumor is represented by the "cancer stem cell," which allows the tumor to proliferate and self renew, and eventually differentiate into the phenotypically diverse and heterogeneous tumor cell population (Bjerkvig et al., Nat. Rev. Cancer, 5:899-904, 2005). Cancer stem cells can be isolated from any type of cancers, e.g., leukemias (Bonnet and Dick, Nat. Med., 3:730-737, 1997), breast cancers (Al-Hajj et al., Proc. Natl. Acad. Sci. USA, 100:3983-88, 2003), colon cancers (O'Brien et al., Nature, 445:106-110, 2007), and brain cancers (Singh et al., Nature, 432:396-401, 2004; Hemmati et al., Proc. Natl. Acad. Sci. USA, 100:15178-83, 2003; Singh et al., Cancer Res., 63:5821-28, 2003; Sanai et ah, N. Engl. J. Med., 353:811-822, 2005; Turrici ct al., MoI. Cancer, 3:25, 2004). Cancer stem cells are characterized by their ability to self-renew and proliferate, and recapitulate through differentiation the tumor from which it is isolated. Additionally, neural cancer stem cells form clonally derived neurospheres in culture.
Cancer stem cells can be isolated by dissociating tumor cells and culturing them under conditions that promote proliferation of stem cells (e.g., conditions that inhibit differentiation of the stem cells). Methods and conditions for isolating stem cells are known in the art. Exemplary methods and conditions can be found in U.S. Patent No. 5,589,376, U.S. Patent No. 5,643,741, U.S. Patent No. 5,650,299, U.S. Patent No. 5,824,489, U.S. Patent No. 5,849,553, U.S. Patent No. 5,928,947, U.S. Patent No. 5,981,708, U.S. Patent No. 6,337,184, U.S. Patent No. 6,645,763, U.S. Patent No. 6,800,790, U.S. Patent No. 6,875,607, U.S. Patent No. 6,984,522, U.S. Patent No. 7,109,03, U.S. Patent No. 7,115,267, and U.S. Patent No. 7,115,360.
Additionally, cancer stem cells can be identified or isolated (e.g., isolated from non-stem tumor cells) on the basis of expression (e.g., nucleic acid or protein expression) of molecular markers, e.g., molecular markers described in the U.S. patents referred to in the above paragraph. Exemplary molecular markers include CD 133, BmM, Notch, Sonic hedgehog, and Wnt. Additionally, exemplary molecular markers of neural cancer stem cells include CD90, CD44, CXCR4, Nestin, Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl , PTCHl , Bmi-1 , phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl , cIAP2, NAlP, and survivin. Isolation or identification of cancer stem cells can be performed by standard means, e.g., cell sorting (e.g., fluorescence activated cell sorting (FACS) or magnetic cell sorting (MACS)).
Antigens Antigenic peptides useful for loading DCs for vaccination are peptides that stimulate a T cell mediated immune response (e.g., a cytotoxic T cell response) by presentation to T cells on MHC molecules. Useful antigenic peptides and proteins include those derived from cancer stem cells (e.g., neural cancer stem cells, CDl 33+ tumor cells, or neurospheres derived from tumors). In some embodiments, the cancer stem cell antigens are presented as a lysale of the cancer stem cells. In other embodiments, the cancer stem cell antigens are obtained by acid elution of peptides presented on MHC molecules of the cancer stem cells, hi an exemplary method, cancer stem cells are washed with an isotonic solution (e.g., Hank's buffered saline solution) to remove media components. The cells are then treated with acid (e.g., citrate phosphate buffer, pH 3.2) to dissociate peptides from surface MHCs, and the cells removed from the solution containing the soluble peptides. The acid-eluted cancer stem cell peptide antigens can be further purified (e.g., on a Cl 8 column) and frozen for storage prior to use.
Specific antigens that can be used in the methods described herein include portions of the amino acid sequences of CD133, CD90, CD44, CXCR4, Nestin, Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl , PTCHl , Bmi-1 , phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, CΪAP2, NAIP, and survivin that bind to MHC molecules and are presented to T cells. Peptides that bind to MHC class I molecules are generally 8-10 amino acids in length. Peptides that bind to MHC class II molecules are generally 13 amino acids or longer (e.g., 13-17 amino acids long).
CDl 33 is a 120 kϋa, five-transmembranc domain glycoprotein expressed on neural and hematopoietic stem and progenitor cells (Yin et al., Blood, 90:5002-12, 1997). Table 1 provides an amino acid sequence of CDl 33 (also available in GenBank under accession number NP_006008.1, GI:5174387), The structure of CDl 33 includes an extracellular N- terminus, two short intracellular loops, two large extracellular loops, and an intracellular C-terminus (FIG. 18). Exemplary CD133 T cell epitopes include 8-10 or 13-20 contiguous amino acid residues of amino acid residues 325-350 of SEQ ID NO:53. An alternately spliced version of CDI33 is described in Yu et al., J. Biol. Chem., 277:20711-16, 2002. CD90 is a cell surface glycoprotein found on T cells and neurons. Table 1 provides an amino acid sequence of CD90 (also available in GenBank under accession number NP_006279.2, GI:19923362).
CD44 is a cell surface glycoprotein that may be involved in matrix adhesion. Table 1 provides an amino acid sequence of CD44 (also available in GenBank under accession number NP 000601.3, GI:48255935). Several isoforms of CD44 are produced, primarily by alternative splicing (see Marhaba et al., J. MoI. Histol., 35:211-31, 2004; Zoeller, Cancer Immunol. Immunother., 53:567-79, 2004).
CXCR4 is a chemokine receptor that has been found to be expressed in breast cancers (Muller et al., Nature, 410:50-56, 2001). Table 1 provides an amino acid sequence of CXCR4 (also available in GenBank under accession number NPJ)Ol 008540.1 , GI:56790927). An alternative spliced variant is available in GenBank under accession number NP_003458.1 , GI:4503175.
Nestin is an intermediate filament protein expressed in neural progenitor cells (Dahlstrand et al., J. Cell Sci., 103:589-597, 1992). Table 1 provides an amino acid sequence of Nestin (also available in GenBank under accession number NP_006608.1 , Gl:38176300).
Musashi-1 (Msil) is an RNA-binding protein expressed in neural progenitor cells (Good et al., Genomics, 52:382-384, 1998; Siddall et al., Proc. Nat. Acad. Sci. USA, 103:84-8407, 2006; Okano et al., Exp. Cell Res., 306:349-356, 2005). Table 1 provides an amino acid sequence of Musashi-1 (also available in GenBank under accession number NP_002433.1, GI:4505255).
Maternal embryonic leucine zipper kinase (MELK) is a protein kinase expressed in multiple cancers (Gray et al., Cancer Res., 65:9751-61, 2005). Table 1 provides an amino acid sequence of MELK (also available in GenBank under accession number NP_055606.1, GI:7661974).
GLIl is a zinc-finger transcription factor upregulated in cancers, including gliomas (Kinder et al., Science, 236:70-73, 1987; Kinzler et al., Nature, 332:371-374, 1988; Kasper et al., Eur. J. Cancer, 42:437-445, 2006). Table 1 provides an amino acid sequence of GLIl (also available in GenBank under accession number NP_005260.1, GI:4885279). PTCH 1 is a transmembrane protein that is believed to function as a tumor suppressor (Katoh et al., Cancer Biol. Ther., 4:1050-54, 2005). Table 1 provides an amino acid sequence of PTCHl (also available in GenBank under accession number NP_000255.2, GI: 134254446). Five isoforms of PTCHl are produced by alternative splicing (Nagao et al., Genomics, 85:462-71, 2005).
Bmi-1 is a polycomb ring finger protein involved in proliferation of progenitor cells (Lessard et al., Nature, 423:255-260, 2003; Park ct al., Nature, 423:302-305, 2003; Molofsky et al., Nature, 425:962-967, 2003). Bmi-1 can play a role in the malignant transformation of the HOX A9/MEIS-induced murine leukemia model (Lessard et al., Nature, 423:255-260, 2003) as well as in tumors of neural origin (van Lohuizen et al., Nature, 353:353-355, 1991). Table 1 provides an amino acid sequence of Bmi-1 (also available in GenBank under accession number NP_005171.4, GI:27883842). Exemplary Bmi-1 T cell epitopes include TLQDIVYKL (SEQ ID NO:86), CLPSPSTPV (SEQ ID NO.-87), VRYLETSKY (SEQ ID NO:88), KRYLRCPAΛ (SEQ ID NO:89),
YEEEPLKDY (SEQ ID NO:90), and KEEVNDKRY (SEQ ID NO:91) (Steele et al., Br. J. Cancer 95:1202-11, 2006).
Phosphoserinc phosphatase (PSP) is an enzyme that catalyzes the hydrolysis of O- phosphoserine. Table 1 provides an amino acid sequence of PSP (also available in GenBank under accession number NP_004568.2, GI:46249388).
Snail is a zinc- finger transcription factor and anti-apoptotic protein (Vega et al., Genes Dcv., 18:1131-1143, 2004). Table 1 provides an amino acid sequence of Snail (also available in GenBank under accession number NP_005976.2, GI: 18765741 ).
OCT4 is a POU homeodomain-containing transcription factor expressed in piuripotent cells (Nichols et al., Cell, 95:379-391, 1998). Table 1 provides an amino acid sequence of OCT4 (also available in GenBank under accession number NP_002692.2, GI:42560248). An alternate isoform of OCT4 is available in GenBank under accession number NP_976034.3, GI:11623549],
BCRPl is an ATP-binding cassette (ABC) transporter protein involved in multidrug resistance of tumors (Doyle et al., Proc. Nat. Acad. Sci. USA, 95: 1566570, 1998). Table 1 provides an amino acid sequence of BCRPl (also available in GenBank under accession number NP_004818.2, GI:62526033).
MGMT is an 0-6-methylguanine-DNA methyltransferase DNA -mismatch repair protein that can provide resistance to some methylating and chloroethylating agents, such as temozolomide (Rabik et al., Cancer Treat. Rev., 32:261-276, 2006; Cai et al., Cancer Res., 65:3319-27, 2005). Table 1 provides an amino acid sequence of MGMT (also available in GenBank under accession number NP_002403.1, GI:4505177). BCL-2 is a mitochondrial anti-apoptotic protein correlated with chemotherapy resistant cancers and decreased overall survival (Campos et al., Blood, 81 :3091-3096, 1993). Table 1 provides an amino acid sequence of BCL-2 (also available in GenBank under accession number NP_000624.2, GI:72198189). An alternatively spliced isoform of BCL-2 is available in GenBank under accession number NP_000648.2, Gi:72198346. FLIP is an anti-apoptotic protein (Irmler et al., Nature, 388:190-195, 1997). Table
1 provides an amino acid sequence of FLIP (also available in GenBank under accession number NP_003870.3, GI:21361769).
BCL-XL is an anti-apoptotic protein related to BCL-2, which may be involved in chemoresistancc (Boise et al., Cell, 74:597-608, 1993; Andreeff et al., Leukemia, 13:1881- 92, 1999). Table 1 provides an amino acid sequence of BCL-XL (also available in GenBank under accession number NP_612815.1, GI:20336335). Exemplary T cell epitopes of BCL-XL include Bcl-xLl 18-126 (TAYQSFEQV; SEQ ID NO:92), BcI- xLl 73-182 (YLNDHLEPWI; SEQ ID NO:93), and Bcl-xL169-178 (WMATYLNDHL; SEQ ID NO:94) (Andersen et al., J. Immunol., 175:2709-14, 2005). XIAP is a member of the inhibitor of apoptosis protein (IAP) family (Deveraux et al., Nature, 388:300-304, 1997). Table 1 provides an amino acid sequence of XIAP (also available in GenBank under accession number NP_001158.2, GI:32528299). cIAPl is a member of the IAP family of apoptosis inhibitors (Rothe et al., Cell, 83: 1243-1252, 1995; Liston et al., Nature, 379:349-353, 1996). Table 1 provides an amino acid sequence of cIAP 1 (also available in GenBank under accession number NP_001157.1 , GI:4502141). cIAP2 is a member of the IAP family of apoptosis inhibitors (Liston ct al., Nature, 379:349-353, 1996). Table 1 provides an amino acid sequence of cIAP2 (also available in GenBank under accession number NP_031490.1, GI:6680696). NAIP is a member of the IAP family of apoptosis inhibitors (Roy et al., Cell,
80:167-178, 1995). Table 1 provides an amino acid sequence of NAIP (also available in GenBank under accession number NP_004527.2, GI: 119393878). An alternatively spliced isoform of NAIP is available in GenBank under accession number NP_075043.1, GI: 119393876. Survivin is a member of the IAP family of apoptosis inhibitors (Li et al., Nature,
396:580-584, 1998). Table 1 provides an amino acid sequence of NAIP (also available in GenBank under accession number NP_001159.2, GI:59859878). Alternatively spliced isoforms of survivin have been identified (see Wheatley et al., Int. Rev. Cytol., 247:35-88, 2005; Noton et al., J. Biol. Chem., 281 : 1286-95, 2006; and Taubert et al., Oncogene, 24:5258-61, 2005). Sequences of exemplary alternative isoforms are available in GenBank under accession numbers NP_001012270.1, GI:59859880, and NP_001012271.1, GI:59859882. Exemplary T cell epitopes of survivin include Sur20-28 (STFKNWPFL; SEQ ID NO:95), Sur96-104 (LTLGEFLKL; SEQ ID NO:96), Surl33- 141 (RAIEQLAAM; SEQ ID NO:97), and Surl26-135 (ETAKKVRRAI; SEQ ID NO:98) (Bachinsky et al., Cancer hnmun., 5:6, 2005). Other exemplary T cell epitopes of survivin include Sur92-101 (QFEELTLGEF; SEQ ID NO:99), Sur54-δ2 (LAQCFFCFK; SEQ ID NO:100), Surl 12-120 (KIAKETNNK; SEQ ID NO:101), Sur53-62 (DLAQCFFCFK; SEQ ID NO:102), Surl 12-121 (KIAKETNNKK; SEQ ID NO:103), Surl8-28 (RISTFKNWPFL; SEQ ID NO: 104), Sur86-96 (FLSVKKQFEEL; SEQ ID NO: 105), and the modified peptides Sur92T2 (QTEELTLGEF; SEQ ID NO: 106), Sur93T2 (FTELTLGEF; SEQ ID NO:107), Sur93S2 (FSELTLGEF; SEQ ID NO:108), Sur38Y9 (MAEAGFIHY; SEQ ID NO:109), Sur47Yl 0 (PTENEPDLAY; SEQ ID NO:110), Sur5K9 (TLPPAWQPK; SEQ ID NO:111), Sur54L2 (LLQCFFCFK; SEQ ID NO:112), and Surl8K10 (RISTFKNWPK; SEQ ID NO:113) (Reker et al., Cancer Biol. Ther., 3:173- 179, 2004). Other exemplary T cell epitopes of survivin include ELTLGEFLKL (SEQ ID NO:114) and TLPPAWQPFL (SEQ ID NO:115) (Schmitz et al., Cancer Res. 60:4845- 4849, 2000). Additional survivin epitopes are described in Siegel et al., Br. J. Haematol., 122:911-914, 2003. Table 1. Sequences of Human Antigens
Antigen Amino acid sequence
CDl33 MALVTiGSLLLLGLCGNSFSGGQPSSTDAPKAWNYELPATNYETQDSHKAGPIGILFELVHI FLYWQPRDFPEOTLRKFLQKAYESKIDYDKPETVILGLKIVYYEAGIILCCVLGLLFIIL MPLVGYFFCMCRCCNKCGGEMHQRQKENGPFLRKCFAISLLVICIIISIGIFYGFVΛNHQV RTRIKRSRKLADSNFKDLRTLLNETPEQIKYILAQYNTTKDKAFTDLNSINSVLGGGILDR LRPNIIPVLDEIKSMATAIKETKEALENMNSTLKSLHQQSTQLSSSLTSVKTSLRSSLNDP LCLWIPSSETCNSIRLSLSQLNSNPELRQLPPVDAELDNVNNVLRTDLDGLVQQGYQSLND IPDRVQRQTTa^VAGIKRVIXNSIGSDIDNOTQRLPIQDILSAFSVYVNNTESYIHRNLPTL EEYDSYWWIX]IGLVICSLLTLIVTFYYLGLLCGVCGYDRI^IATPTTRGCVSNTGGVFLMVGVG LSFLFCWILMIIWLTFVFGANVEKLICEPYTSKELFRVLDTPYLLNEDWEYYLSGKLFNK SIOJKLTFEQWSDCKKITOGTYGTLHLQNSFWISEHI^INEHTGSISSELESLKVNLNIFLL GAAGRKNLQDFAACGIDRMNYDSYI-AQTGKSPAGVNLLSFAYDLEAKANSLPPGNLRNSLK RDAQTIKTIHQQRVLPIEQSLSTLYQSVKILQRTGNGLLERVTRILASLDFAQNFITNNTS SVIIEETKKYGRTIIGYFEHYLQWIEFSISEKVASCKPVATΛLDTAVDVFLCSYIIDPLNL FWFGIGKATVFLLPALIFAVKI-AKYYRRrøSEDVYDDVETIPMKNMENGNNGYHKDHVYGI HNPVMTSPSQH (SEQ ID NO: 53)
CD90 MNLAISIALLLTVLQVSRGQKVTSLTACLVDQSLRLDCRHENTSSSPIQYEFSLTRETKKH VLFGTVGVPEHTYRSRTNFTSKYNMKVLYLSAFTSKDEGTYTCALHHSGHSPPISSQNVTV LRDKLVKCEGISLLAQNTSWLLLLLLSLSLLQATDFMSL (SEQ ID NO: 54)
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
T cell epitopes can be identified by a number of different methods. Naturally processed MHC epitopes can be identified by mass spectrophotomctric analysis of peptides eluted from antigen-loaded APC (e.g., APC that have taken up antigen, or that have been engineered to produce the protein intracellularly). After incubation at 37 0C, cells are lysed in detergent and the MHC protein is purified (e.g., by affinity chromatography). Treatment of the purified MHC with a suitable chemical medium (e.g., under acidic conditions) results in the elution of peptides from the MHC. This pool of peptides is separated and the profile compared with peptides from control APC treated in the same way. The peaks unique to the protein expressing/fed cells are analyzed (for example by mass spectrometry) and the peptide fragments identified. This protocol identifies peptides generated from a particular antigen by antigen processing.
Alternatively, epitopes can be identified by screening a synthetic library of peptides that overlap and span the length of the antigen in an in vitro assay. For example, peptides that are 9 amino acids in length and which overlap by 5 amino acids may be used. The peptides are tested in an antigen presentation system that includes antigen presenting cells and T cells. T cell activation in the presence of APCs presenting the peptide can be measured (e.g., by measuring T cell proliferation or cytokine production) and compared to controls, to determine whether a particular epitope is recognized by the T cells.
T cell epitopes can be predicted in silico, e.g., using the methods described in Parker et al., J. Immunol., 152:163, 1994 and Rammensec ct al., Immunogenet., 50:213- 219, 1999.
Antigenic peptides can be obtained by chemical synthesis using a commercially available automated peptide synthesizer. Synthetic peptides can be precipitated and further purified, for example by high performance liquid chromatography (HPLC). Alternatively, isolated peptides can be obtained by purification and/or recombinant methods using host cell and vector expression systems.
Preparation of Antigen Presenting Cells
Antigen presenting cells (APC), such as DCs, suitable for administration to subjects (e.g., glioma patients) can be isolated or obtained from any tissue in which such cells are found, or may be otherwise cultured and provided using standard techniques. Methods of preparing antigen presenting cells are well-known to those of skill in the art. Mature dendritic cells are typically identified as having the following cell surface marker phenotype: MAC3~, CD80+, CD86\ CD40low, CD54+, MHC Class I and MHC Class II, and arc capable of FITC-dextran uptake.
APCs (e.g., DCs) can be found, by way of example, in the bone marrow or PBMCs of a mammal, in the spleen of a mammal or in the skin of a mammal (i.e., Langerhan's cells, which possess certain qualities similar to that of DC, may be found in the skin). For instance, bone marrow can be harvested from a mammal and cultured in a medium that promotes the growth of DCs. GM-CSF, IL-4 and/or other cytokines (e.g., TNF-α), growth factors and supplements may be included in this medium.
After a suitable amount of time in culture in medium containing appropriate cytokines (e.g., time suitable to expand and differentiate the DCs into mature DCs, e.g., 2, 4, 6, 8, 10, 12, or 15 days), clusters of DCs are cultured in the presence of a sufficient number of antigens of interest (e.g., in the presence of cancer stem cell lysate, acid eluted peptides of cancer stem cells, peptides of CDl 33, CD90, CD44, CXCR4, Nestin, Musashi- 1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAP 1 , cIAP2, NAIP, or survivin, or a combination of two or more of the above antigens) and harvested for use in a cancer vaccine. For example, peptide antigens can be added to the culture medium at a concentration of about 1.0 to 50, e.g., 5, 10, 15, 20, or 30 μg/ml (per antigen).
Alternately, or in combination, antigens can be transgenically expressed in DCs, e.g., by transfection of nucleic acids encoding one or more of the antigens or portions of antigens.
In one exemplary method, APCs are isolated from a subject (e.g., a human) according to the following exemplary procedure. Mononuclear cells are isolated from blood using leukapheresis (e.g., using a COBE Spectra Apheresis System). The mononuclear cells are allowed to become adherent by incubation in tissue culture flasks for 2 hours at 37 0C. Nonadherent cells are removed by washing. Adherent cells are cultured in medium supplemented with granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4) for five days. On day five, TNF-α is added to the culture medium for another 3-4 days. On day 8 or 9, cells are harvested and washed, and incubated with peptide antigens for 16-20 hours on a tissue rotator. Peptide antigens are added to the cultures at a concentration of ~ 10 μg/ml (per antigen). Various other methods can be used to isolate the APCs, as would be recognized by one of skill in the art. DCs occur in low numbers in all tissues in which they reside, making isolation and enrichment of DCs a requirement. Any of a number of procedures entailing repetitive density gradient separation, fluorescence activated cell sorting techniques, positive selection, negative selection or a combination thereof are routinely used to obtain enriched populations or isolated DCs. Guidance on such methods for isolating DCs can be found in O'Doherty et al., J. Exp. Med., 178:1067-78, 1993; Young and Steinman, J. Exp. Med., 171:1315-32, 1990; Freudenthal and Steinman, Proc. Nat. Acad. Sci. USA, 57:7698-7702, 1990; Macatonia et al., 67:285-289, 1989; Markowicz and Engleman, J. Clin. Invest., 85:955-961, 1990; Mehta-Damani et al., J. Immunol., 153:996- 1003, 1994; and Thomas et al., J. Immunol., 151:6840-6852, 1993. One method for isolating DCs from human peripheral blood is described in U.S. Patent No. 5,643,786. Methods of producing DCs from embryonic stem cells are described in U.S. Patent No. 7,247,480. Administration of Cancer Vaccines The APC-based cancer vaccine may be delivered to a recipient by any suitable delivery route, which can include injection, infusion, inoculation, direct surgical delivery, or any combination thereof. In some embodiments, the cancer vaccine is administered to a human in the deltoid region or axillary region. In some embodiments, the vaccine is administered to a subject locally to the site of a tumor, within the tumor, or to an area from which a tumor has been surgically resected.
An appropriate carrier for administering the cells may be selected by one of skill in the art by routine techniques. For example, the pharmaceutical carrier can be a buffered saline solution, e.g., cell culture media.
The quantity of APC appropriate for administration to a patient as a cancer vaccine to effect the methods of the present invention and the most convenient route of such administration may be based upon a variety of factors, as may the formulation of the vaccine itself. Some of these factors include the physical characteristics of the patient (e.g., age, weight, and sex), the physical characteristics of the tumor (e.g., location, size, rate of growth, and accessibility), and the extent to which other therapeutic methodologies (e.g., chemotherapy, and beam radiation therapy) are being implemented in connection with an overall treatment regimen. Notwithstanding the variety of factors one should consider in implementing the methods of the present invention to treat a disease condition, a mammal can be administered with from about 105 to about 109 APC (e.g., 107 APC) in from about 0.05 mL to about 5 mL solution (e.g., saline) in a single administration. Additional administrations can be carried out, depending upon the above-described and other factors, such as the severity of tumor pathology. In one embodiment, from about one to about five administrations of about 106 APC is performed at two-week intervals.
DC vaccination can be accompanied by other treatments. For example, a patient receiving DC vaccination may also be receiving chemotherapy, radiation, and/or surgical therapy concurrently. Methods of treating cancer using DC vaccination in conjunction with chemotherapy are described in Wheeler et al., U.S. Pat. Pub. No. 2007/0020297. In some embodiments, a patient receiving DC vaccination has already received chemotherapy, radiation, and/or surgical treatment for the cancer. In one embodiment, a patient receiving DC vaccination is treated with a COX-2 inhibitor, as described in Yu and Akasaki, WO 2005/037995. Pharmaceutical Compositions
In various embodiments, the present invention provides pharmaceutical compositions including a pharmaceutically acceptable excipient along with a therapeutically effective amount of the inventive vaccine comprising dendritic cells pulsed with cancer stem cell antigens as described herein. "Pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients may be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
In various embodiments, the pharmaceutical compositions according to the invention may be formulated for delivery via any route of administration. "Route of administration" may refer to any administration pathway known in the art, including but not limited to aerosol, nasal, transmucosal, transdermal, or parenteral. "Parenteral" refers to a route of administration that is generally associated with injection, including intraorbital, infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intrapulmonary, intraspinal, intrasternal, intrathecal, intrauterine, intravenous, subarachnoid, subcapsular, subcutaneous, transmucosal, or transtracheal. Via the parenteral route, the compositions may be in the form of solutions or suspensions for infusion or for injection, or as lyophilized powders.
The pharmaceutical compositions according to the invention can also contain any pharmaceutically acceptable carrier. "Pharmaceutically acceptable carrier" as used herein refers to a pharmaceutically acceptable material, composition, or vehicle that is involved in carrying or transporting a compound of interest from one tissue, organ, or portion of the body to another tissue, organ, or portion of the body. For example, the carrier may be a liquid or solid filler, diluent, excipient, solvent, or encapsulating material, or a combination thereof. Each component of the carrier must be "pharmaceutically acceptable" in that it must be compatible with the other ingredients of the formulation. It must also be suitable for use in contact with any tissues or organs with which it may come in contact, meaning that it must not carry a risk of toxicity, irritation, allergic response, immunogenicity, or any other complication that excessively outweighs its therapeutic benefits.
The pharmaceutical compositions according to the invention may be delivered in a therapeutically effective amount. The precise therapeutically effective amount is that amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given subject. This amount will vary depending upon a variety of factors, including but not limited to the characteristics of the therapeutic compound (including activity, pharmacokinetics, pharmacodynamics, and bioavailability), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), the nature of the pharmaceutically acceptable carrier or carriers in the formulation, and the route of administration. One skilled in the clinical and pharmacological arts will be able to determine a therapeutically effective amount through routine experimentation, for instance, by monitoring a subject's response to administration of a compound and adjusting the dosage accordingly. For additional guidance, see Remington: The Science and Practice of Pharmacy (Gennaro ed. 20th edition, Williams & Wilkins PA, USA) (2000).
Kits
Kits to treat cancer are also contemplated. The kits are useful for practicing the inventive method of treating cancer with a vaccine comprising dendritic cells pulsed with cancer stem cell antigens as described herein. The kit is an assemblage of materials or components, including at least one of the inventive compositions. Thus, in some embodiments the kit contains a composition including a vaccine comprising dendritic cells pulsed with cancer stem cell antigens as described herein. The exact nature of the components configured in the inventive kit depends on its intended purpose. For example, some embodiments are configured for the purpose of treating a particular cancer. In one embodiment, the kit is configured for the purpose of treating brain tumors. In one particular embodiment, the brain tumor is a glioma. In another embodiment, the brain tumor is GBM. In one embodiment, the kit is configured particularly for the purpose of treating mammalian subjects. In another embodiment, the kit is configured particularly for the purpose of treating human subjects. In further embodiments, the kit is configured for veterinary applications, treating subjects such as, but not limited to, farm animals, domestic animals, and laboratory animals. Instructions for use may be included in the kit. "Instructions for use" typically include a tangible expression describing the technique to be employed in using the components of the kit to effect a desired outcome, such as induction of an immune response against a tumor, to treat a cancer. For example, the instructions may comprise instructions to administer a vaccine comprising dendritic cells pulsed with cancer stem cell antigens to the patient.
Optionally, the kit also contains other useful components, such as, diluents, buffers, pharmaceutically acceptable carriers, syringes, catheters, applicators, pipetting or measuring tools, or other useful paraphernalia as will be readily recognized by those of skill in the art. The materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility. For example the components can be in dissolved, dehydrated, or lyophilized form; they can be provided at room, refrigerated or frozen temperatures. The components are typically contained in suitable packaging material(s). As employed herein, the phrase "packaging material" refers to one or more physical structures used to house the contents of the kit, such as inventive compositions and the like. The packaging material is constructed by well known methods, preferably to provide a sterile, contaminant-free environment. The packaging materials employed in the kit are those customarily utilized in cancer treatments or in vaccinations. As used herein, the term "package" refers to a suitable solid matrix or material such as glass, plastic, paper, foil, and the like, capable of holding the individual kit components. Thus, for example, a package can be a glass vial used to contain suitable quantities of an inventive composition containing for example, a vaccine comprising dendritic cells pulsed with cancer stem cell antigens as described herein. The packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components. EXAMPLES
The following examples are provided to better illustrate the claimed invention and are not to be interpreted as limiting the scope of the invention. To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention. One skilled in the art may develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention.
Example 1. The 9L Gliosarcoma Cell Line Contains Self-Renewing Cells
To determine if a population of self-renewing stem cells exist within the phenotypically heterogeneous 9L gliosarcoma tumor, the cells were grown as monolayers in the presence of 10% FBS and subsequently grew them in serum free media containing mitogens. A schematic diagram of culturing of tumor stem cells is presented in Fig. 2. 9L gliosarcomas were resuspended in Dulbecco's modified Eagle's medium/F-12 medium containing 10% fetal bovine serum (FBS) and plated at a density of 1 x 106 live cells per 75 cm2 flask. The cells attached and grew as monolayers and were passaged upon conflucncy. Spheres were derived by placing the 9L gliosarcomas cells grown as monolayers into a defined serum-free NSC medium (Reynolds et al., J. Neurosci., 12: 4565-74, 1992; Reynolds et al., Science, 255:1707-10, 1992) consisting of Dulbecco's modified Eagle's medium/F-12 medium supplemented with 20 ng/mL of both epidermal growth factor (EGF; Peprotech, Rocky Hill, NJ) and basic fibroblast growth factor (bFGF; Peprotech, Rocky Hill, NJ). Cells were fed every 2 days by adding fresh NSC media supplemented with growth factors. After primary spheres formed and reached 10-200 cells per sphere, the cells were harvested, dissociated into single cells using trypsin and EDTA (GIBCO BRL) and mechanical pipetting, strained through a cell strainer, and plated at a clonal density of 1,000 cells/mL in neurosphere-conditioned medium to generate clonally derived subspheres (Geschwind et al., Neuron, 29:325-339, 2001; Groszer et al., Science, 294: 2186-89, 2001). The cells were fed every 2 days by adding fresh NSC media supplemented with mitogens. Cells and subsequent spheres were observed daily for 18 days, and passaged into fresh media. Subspheres ranging from approximately 15 cells to 40 cells were evident after 18 days and displayed the self-renewing and proliferative capacity of the 9L spheres.
Subtle differences were observed in surface adhesion properties between the two cell populations when using different coating solutions. It was found that poly-L-lysine proved less effective in binding to neurospheres than did laminin. Even after 2 days, only a small portion of the cells adhered (~ 5%). However, when ncurospheres were allowed to adhere to chamber slides coated with laminin, nearly all the cells (~ 70%) adhered within the first 3 hours. As a consequence, poly-L-lysine was not used as an adhesive substrate in further studies. A similar trend was observed when comparing adhesion of monolayers to chamber slides coated with either poly-L-lysine or laminin — cellular morphology was more apparent in monolayers grown on laminin coated chamber slides within a 6 hour time period. Furthermore, a distinct morphological characteristic was apparent in the differentiated ncurospheres, where arms of differentiated cells originating from one neurosphere homed to arms formed by another nearby neurosphere (FIG. 8A). These examples of extracellular adhesion and cellular homing can be used to distinguish cancer stem-like cells (CSLCs) from non-CSLCs within a tumor.
Example 2. 9L Neurospheres Express NSC Markers and Can Generate Both Neuronal and Glial Cells in Culture
To determine the expression of markers for stem cells, neurospheres were immunostained for NSC markers nestin and Sox2. Neurospheres were also stained for the lineage markers for astrocytes, GFAP, neurons, beta-tubulin IH and MAP2, and oligodendrocytes, myelin/oligodendrocyte. Cells in the outer region of the neurosphere labeled for nestin, while cells throughout the neurosphere were labeled for Sox2. A large number of cells within the tumor spheres were also found to be positive for the lineage marker GFAP, while relatively few cells expressed the neuronal lineage markers β-tubulin III , MAP2, and myelin/oligodendrocyte.
To test whether spheres have multipotent capabilities and produce progenies of different lineages, spheres were seeded into chamber slides (Lab-Tekll, Nalge Nunc International) for differentiation assay. The cells were grown for 14 days in medium devoid of growth factors bFGF and EGF but permissive for differentiation, and processed for immunocytochemistry as described below. The medium included Dulbecco's modified Eagle's medium/F-12 medium containing 10% fetal bovine serum (FBS).
To examine the expression of NSC markers and lineage markers, immuncyto- and immunohistochemical staining was performed. For staining of differentiated spheres, spheres and 9L monolayers, cells growing in chamber slides were fixed with 4% paraformaldehyde for 15 minutes at 4 0C, treated with 5% NHS (normal horse serum)/0.1% Triton-X, and then stained with the following antibodies: rabbit anti-nestin
(1:200, Chemicon), rabbit anti-Sox2 (1:1,000, Chemicon), rabbit anti-MAP2 (1 :1,000, Sigma), mouse anti-β-tubulin HI (1 :200, Chemicon), rabbit anti-GFAP (1 : 1000, Chemicon), mouse anti-myelin/oligodendrocyte (1:1,000, Chemicon). The primary antibodies were detected with Cy3 or FITC-conjugated anti-mouse or anti-rabbit IgG antibody (1 :200, Jackson Immuno Research). The cells were counterstained with 4',6- diamidino-2-phenylindole (DAPI; Vector Laboratories) to identify all nuclei. The stained sections were examined and photographed using a QED cell scanner program and Nikon Eclipse TE2000-E microscope, and analyzed using Image J (NIH). For immunostaining of spheres, the spheres where allowed to adhere to prccoated (with laminin) chamber slides for 3 hours before fixation, while monolayers were allowed to adhere overnight in non- fixed chamber slides. Cells stained positive for the NSC markers nestin and Sox2, as well as the lineage markers for astrocytes, GFAP, neurons, MAP2, and oligodendrocytes, myelin/oligodendrocyte. However, no cells were labeled with the neuronal marker beta- tubulin III. FACS analysis showed similar results to those derived via immunocytochemistry, which showed differentiated cells labeled for nestin (88%; FIG. 3C), Sox2 (24%), GFAP (96%; FIG. 3B), MAP2 (29%; FIG. 3D), and myelin/oligodendrocyte (6%; FIG. 3F), but not for β-rubulin III (0%; FIG. 3E). For FACS analysis, cells were fixed with 4% paraformaldehyde for 15 minutes at 4 0C, treated with 5% NHS (normal horse serum)/0.1% Triton™-X, and then stained with the following antibodies: rabbit anti-nestin (1:1,000, Chemicon), rabbit anti-Sox2 (1:1,000, Chemicon), rabbit anti-MAP2 (1:1,000, Sigma), mouse anti-β-tubulin III (1 :500, Chemicon), rabbit anti-GFAP (1:1000, Chemicon), and mouse anti-myelin/oligodendrocyte (1 :1,000, Chemicon), The primary antibodies were detected with FITC-conjugated anti-mouse or anti-rabbit IgG antibody (1:200, Jackson Immuno Research) using 104 cells in a FACSVantage™ fluorescence activated cell sorter (Becton Dickinson). These results indicate that these cancer stem-like cells (CSLCs), unlike normal stem cells, differentiate into aberrant cells that are positive for multiple differentiation markers, notably GFAP and MAP2. Such a dual nature is apparent in the majority of cells differentiated from neurospheres. Furthermore, many cells still remained highly positive for nestin and Sox2. The expression pattern of the differentiated progeny was similar in profile to that of primary cultured tumor cells from which the spheres had originally been isolated and predominantly differentiated into GFAP and MAP2 positive cells that recapitulated the parental tumor phenotype. Additionally, the level of labeling for the NSC markers nestin and Sox2 still remained high, even after 14 days of differentiation. The staining pattern for the monolayer population was similar to neurospheres differentiated for 14 days, except for the expression of Sox2, which was greater in neurospheres differentiated for 14 days. These results reveal that 9L spheres are multipotent for the three neural cell types and differentiate into cells found in the original tumor from which they were obtained.
Example 3. The Aggressiveness of 9L Cells in vivo is Reliant on the Neurosphere Cancer Stem-Like Cells
To deteπninc if 9L cells grown as monolayer or neurospheres differ in their ability to grow as a tumor after implantation 5,000 cells from both populations of cells were injected into rats and survival and tumor volume were assayed. Fisher F344 rats 6-8 weeks old (Harlan Sprague-Dawlcy, Indianapolis, IN) were anesthetized with i.p. ketamine and xylazine, and stercotactically implanted in the right striatum (from bregma in mm: anterior-posterior +1 mm; medial-lateral -3 mm; dorsal- ventral -5 mm) with cither isolated 9L sphere cells containing the luciferase gene (5,000 cells) or non-sphere-forming monolayer cells containing the luciferase gene (5,000 sells) in 4μL of 1.2% methylcellulose/PBS (Rehemtulla et al., Neoplasia, 2:491-495, 2000). Rats were portioned to either the tumor volume group (n = 10) or the survival group (n = 18), with control rats (n = 6) receiving 4μL of 1.2% methylcellulose/PBS only.
Because half of each group consisted of either animals implanted with spheres or monolayers, tumor aggression could be determined. Animals in the tumor volume group were sacrificed 18 days after tumor implantation. Tumor volume was assessed by using the formula for an ellipsoid, (length x width x height)/2 (Advani et al., Cancer Res., 59:2055-58, 1999), with the height and the width of the tumor being approximately equal because of the well-defined circumference of the tumors generated by the 9L gliosarcoma (see Sibenaller et al., Neurosurg. Focus, 19:E1, 2005). A significantly (P < 0.02) greater tumor volume was observed in the rats implanted with neurosphere cells as compared to rats implanted with monolayer cells (FIG. 4A),
Animals in the survival group were followed for survival and euthanized via CO2 asphyxiation when terminal neurological signs developed (e.g., inability to access food, water, seizure activity, weakness, and paralysis) or if animals exceeded a survival period of 40 days. Following euthanization, brains were harvested and frozen in 2-methylbutane (Sigma), cooled to -200C, and stored at -800C until sectioning. For H&E staining, 20 μm coronal brain sections described above were mounted on slide and stained with Harris hematoxylin for 2 minutes and then counterstained with alcoholic eosin. Reticulin stains were performed on 12 μm coronal brain sections.
Similar to the results seen in the tumor size groups, rats implanted with neurospheres on average had shorter survival times than rats implanted with monolayer cells (FIG. 4B). Six rats in the neurosphere group had large tumors that lead to terminal neurological symptoms, whereas 3 showed evidence of small tumors after 40 days of survival as seen by H&E staining of brain sections. In contrast, only 4 rats had tumors large enough to create terminal neurological symptoms in the monolayer group, whereas 5 showed evidence of small tumor or engraftment after 40 days of survival as determined by H&E staining. Furthermore, the rats in the monolayer group which developed terminal neurological signs of tumor did so at a later time (36 days compared to 29 days in the neurosphere group), which was significant when analyzed using the Kaplan-Meier test (P < 0.02).
To determine the establishment of tumor in vivo, 150 mg/kg of the luciferase substrate D-luciferin (Biosynth, International, Inc., Naperville, IL) was administered i.p. to animals in the survival group, and luciferase scans were taken 15 minutes later. Luciferase scans generated at 14 days post implantation demonstrated a greater proportion of animals with tumor burden in the neurosphere group compared to the monolayer group, even though a higher expression level of luciferase in vitro was observed in monolayer cells as compared to neurosphere cells. To determine if the 9L neurospheres could recapitulate the 9L sarcoma in vivo, histological analysis of the tumors was performed. The tumor-cell-implanted rat brains were cut with a cryostat into 20 μm coronal sections and fixed in 4% paraformaldehyde, washed with PBS, and air dried. To characterize the brain tissue by immunohistochcmistry, free-floating sections were blocked with 5% NHS (normal horse serum) for 30 minutes at room temperature and then stained with the following antibodies: rabbit anti-nestin (1 :200, Chemicon), rabbit anti-Sox2 (1 : 1 ,00O5 Chemicon), rabbit anti- MAP2 (1:1,000, Sigma), mouse anti-β-tubulin III (1 :200, Chemicon), rabbit anti-GFAP (1:1000, Chemicon), mouse anti-myelin/oligodcndrocyte (1:1000; Chemicon). The primary antibodies were detected as in Example 1. The cells were counterstained with 4',6-diamidino-2-phenylindole (DAPl; Vector Laboratories) to identify all nuclei. The stained sections were examined and photographed using the Zeiss Axiovision 3.1 program in conjunction with Zeiss Axioskop™ 2 microscope, and analyzed using Image J (NIH). Tumors from the neurosphere cell population were large and well-circumferential, and showed cells positive for the NSC marker ncstin, as well as cells positive for the lineage markers GFAP, β-tubulin III, and myelin/oligodendrocytes. Most of the labeling within the tumor volume was directed against GFAP, and a lesser degree of labeling was observed for β-tubulin III, myelin/oligodendrocyte, and ncstin. A significant portion (> 75%) of the tumor volume stained positive for reticulin, consistent with a sarcomatous component. Additionally, H&E staining revealed a high grade glioma with necrosis consistent with a glioblastoma, displaying the dual nature of the gliosarcoma. Neurospheres formed high grade gliomas with necrosis as seen on H&E (FIG. 5A). The tumors were large and well circumferential as evidenced in non-stained sections (FIG. 5B) and stained for the nuclear marker DAPI (FIG. 5D). A comparison of non-tumor area (FIG. 5D) with tumor area (FIG. 5E) stained for reticulin revealed high levels of reticulin in tumor engulfed regions, showing the histological sarcomatous component of the gliosarcoma. The staining patterns suggest that 9L neurospheres recapitulated the original tumor by differentiating into both neural and glial lineages in vivo.
Example 4. Proliferation Rate and Drug Sensitivity of 9L Neurospheres
To compare the differential proliferation rates and resistance to chemotherapeutic agents, 2,000 healthy 9L sphere and monolayer cells were exposed to either Dulbecco's modified Eagle's medium/F-12 medium containing 10% fetal bovine serum (FBS), or 100 μM stock solution of Temozolamide or Carboplatin dissolved in PBS at concentrations of 1,000 μM, 500 μM, 250 μM, and 125 μM for 2 days. The viability of the cells was scored by measurement of the absorption of formazan dye (the amount of formazan dye formed directly correlates to the number of metabolically active cells) using the cellular proliferation assay WST-I (Roche Molecular Biochemicals, Mannheim, Germany). Formazan was measured with the use of a microplate reader (Tecan) and spectrophotometer set at a wavelength of 440 run and a reference wavelength of 890 nm. Cellular viability was determined by exposing cells to WST-I for 4 hours, and calculating the percentage of viable cells. Proliferation was also assessed by using manual cell counting after 7 days in culture, with an initial cellular concentration of 100,000 cells/mL in a 25 mm2 flask.
The neurospheres demonstrated significantly (P < 0.05) greater resistance to temozolamide (FIG. 6B) and carboplatin (FIG. 6A) when compared to the 9L cells grown as a monolayer under the same conditions. When the cells were not treated with the chemotherapeutic agents and simply grown in medium, there was a significantly (P < 0.05) greater increase in cell number in the monolayer group by a factor of 1.48 when using the WST-I proliferation assay when the cells were not treated with the chemotherapeutic agents. The results of the proliferation assays are depicted in FIG. 7, which is a bar graph that shows a greater increase in cells in the monolayer group. A similar trend in the untreated cells was also observed using the manual cell count method, which showed a greater increase in monolayers by a factor of 1.35. This example demonstrates that neurosphere cells have a greater resistance to chemotherapeutic agents.
Example 5. Isolation of Human CD133-Positive Cancer Stem Cells Glioblastoma specimens were obtained from patients (with informed consent) via the Brain Tumor Registry and were reviewed and released by a pathologist in the operating room. Independent pathologists classified the tumors by type and grade in accordance with the WHO histological grading of central nervous system tumors. IRB certified technicians processed the glioma tissues under sterile conditions in a laminar flow hood. Tumor cells were cultured in the following complete medium: Ham's F-12/DMEM with high glucose (Irvine scientific, Santa Ana, CA), 10 mM HEPES (Invitrogen, Carlsbad, CA), 0.1 mg/ml Gentamicin (Invitrogen) and 10% heat-inactivated FBS (Irvine Scientific, Santa Ana, CA). The cultured cells were maintained for 3-4 passages. Floating ncurosphere-like cells were obtained that were capable of forming new spheres in medium containing FBS for 3-4 passages.
Three adult glioblastoma primary tumor cell lines (Nos. 1049, 377, and 66) were derived by the above method and analyzed by FACS for CD 133 expression. Tumor cells were collected and stained with anti-CD133 antibody (mouse monoclonal IgGl; 1: 10; Milteny Biotec) or IgGl isotype control antibody (BD Pharmingen, San Diego, CA). After PE-anti-mouse IgGl (BD Pharmingen) staining for 30 minutes, CDl 33 staining was analyzed by flow cytometry using a FACSCalibur™ fluorescence activated cell sorter (Becton Dickinson, San Jose, CA). CD133 expression was observed in 10.2% (No. 66; FIG. 9C), 27% (No. 1049, FIG. 9A) and 69.7% (No. 377; FIG. 9B) of the total population examined. To investigate the capacity of self-renewal and clonogenic potential of CD 133+ cell, a single isolated CDl 33 positive cancer stem cell was isolated by DAKOcytomation™ (DAKO, Carpinteria, CA) sorting and cultured in a defined serum- free NSC medium (Kabos et al., Exp. Neurol, 178:288-293, 2002) containing 20 ng/ml of basic fibroblast growth factor (bFGF, Peprotcch, Rocky Hill, NJ), 20 ng/ml of epidermal growth factor (EGF, Peprotech) and 20 ng/ml leukemia inhibitory factor (LIF, Chemicon, Temecula, CA). Single isolated CDl 33 positive cancer stem cells were able to form neurospheres (FIG. 8A), and were demonstrated to have the capacity for self-renewal and clonogenic potential (FIG. 8B), and sustained expression of CD 133 in serum-free medium containing EGF/FGF (FIG. 8C).
Example 6. Human CD 133 Positive Tumor Cells Express Markers Associated With Neural Precursors
To determine the expression of other genes in CD133 positive cells, CD133 positive cells and CD133 negative cells were obtained by FACS sorting as described in Example 5 and real-time PCR was used to analyze some markers associated with neural precursors in these two populations. Total RNA was extracted from the isolated CD 133 positive and CDl 33 negative cells using an RNA4PCR™ kit (Ambion, Austin, TX) according to the manufacturer's protocol. For cDNA synthesis, ~1 μg total RNA was reverse-transcribed into cDNA using Oligo dT primer and iScript™ cDNA synthesis kit reverse transcriptase. cDNA was stored at -20 0C. Gene expression was quantified by real-time quantitative RT-PCR using QuantiTect™ SYBR Green dye (Qiagen, Valencia, CA). DNA amplification was carried out using Icycler™ (BIO-RAD, Hercules, CA), and the detection was performed by measuring the binding of the fluorescence dye SYBR Green I to double-stranded DNA. All the primer sets were obtained from Qiagen (see Table 1).
Table 2. Oligonucleotide primers sequences used for SYBR Green real-time PCR
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Quantification of target gene mRNA as compared to an internal control (beta-actin) was performed by following a ΔOr method. An amplification plot that had been the plot of fluorescence signal vs. cycle number was drawn. The difference (ΔOr) between the mean values in the duplicated samples of target gene and those of beta-actin were calculated by Microsoft Excel and the relative quantified value (RQV) was expressed as 2" ΛCT. The relative expression of each gene was compared to autologous CD133 negative cells. The results of the QT-PCT analysis arc presented in Table 3.
Figure imgf000038_0002
Figure imgf000039_0001
CD90, CD44, CXCR4, Ncstin, Musashi-1 (Msil), and maternal embryonic leucine zipper kinase (MELK) mRNA expression on CD 133 positive cancer stem cells was upregulated by an average of 15.6, 5.7, 337.8, 2.14, 84, and 1351 fold, respectively, compared to the levels found on autologous CDl 33 negative rumor cells. mRNA levels for GLIl and PTCHl were upregulated an average of 46 and 16 times, respectively, in CD133 positive cells, as compared to CD133 negative cells. Furthermore, Bmi-1, phosphoserinc phosphatase (PSP), SHH, OCT4 and Snail mRNA were expressed in CD 133 positive cells derived from the three cell lines; none of the five genes were detectable on CD 133 negative cells. Additionally, anti-apoptotic genes were also upregulated (see Example 8).
Example 7. CDl 33 Positive Cancer Stem Cells Are Resistant to Chemotherapeutic Agents
To determine whether CD133 positive cancer stem cells were resistant to chemotherapeutic agents, the WST-I Cell Proliferation Assay was used to examine the drug sensitivity of CD133 positive cells and CDl 33 negative cells (both collected by FACS sorting from the three glioblastoma patients' primary cultured tumor cells as described above). CDl 33 positive and negative cells were exposed to conventional chemotherapeutic agents, temozolomide, carboplatin, VP- 16 or taxol at various concentrations, for up to 48 hours in 10% FBS/F-12/DMEM culture medium. Temozotomide was supplied by the Schering-Plough Research Institute (Kenilworth, NJ) and was dissolved in DMSO (Sigma Chemical Co., St Louis, MO) at 100 mM stock solution. Carboplatin, ctoposidc (VP-16) and paclitaxel (Taxol) were obtained from Sigma-Aldrich (St. Louis, MO). CD 133 positive cells isolated from No. 66 showed dramatic drug resistance to the above four agents including temozolomidc (FIG. 1 OC), carboplatin (FIG. 10D), VP- 16 (FlG. 10A), and Taxol (FIG. 10B) as compared to autologous CD133 negative cells. CD133 positive cells isolated from No. 377 showed significant resistance to carboplatin at all concentrations tested (FIG. 1 IA) and to VP-16 at 200 μM (FIG. 1 IB). CDl 33 positive cells isolated from No. 1049 showed significant resistance to carboplatin at 200 μM compared to autologous CD 133 negative cells (FIG. 11 C). This example demonstrates increased drug resistance of CD 133 positive cancer stem cells as compared to autologous CD133 negative cells.
Example 8. Anti-Apoptotic Genes Are Upregulated in CD133 Positive Cancer Stem Cells Real-time PCR of FACs-sorted CD133 positive and CD133 negative cells was used to investigate the relative expression of multi-drug resistance genes and genes related to inhibiting cell apoptosis between these two populations. BCRPl has been demonstrated to play an important role in the drug resistance of normal stem cells and tumor stem cells (Zhou et al., Nat. Med., 7:1028-34, 2001; Hirschmann-Jax et al., Proc. Natl. Acad. Sci. USA, 101:14228-33, 2004). Higher expression of BCRPl (6.5 fold) was found in CD133 positive cells as compared to that of autologous CDl 33 negative cells. Furthermore, anti- apoptotic genes, such as FLIP, BCL-2, and BCL-XL, were found at significantly higher levels (294, 13.9 and 5.6 times higher) in CD133 positive cells than in CD133 negative cells. Also, inhibitor of apoptosis protein (IAPs) family members, XlAP, cIAPl, cIAP2, NAIP, and survivin were found at higher expression levels on CD133 positive cells 21.9, 39.04, 3.03, 12.1, 6.73, and 1.6 times higher, respectively, than in CD133 negative cells. It has been demonstrated that SIRTl deacetylates the DNA repair factor Ku70, causing it to sequester the pro-apoptotic factor, Bax, away from mitochondria, thereby inhibiting stress- induced apoptotic cell death (Cohen et al., Science, 305:390-392, 2004). SIRTl deacetylase mRNA expression was increased 4.92 times in CD 133 positive cells. The pro- apoptotic gene BAX was decreased 3 times in CDl 33 positive cells as compared to autologous CD 133 negative cells. Thus, gene expression differences were observed in CD133 positive cells as compared to CDl 33 negative cells.
Example 9. Recurrent Glioblastomas Express Higher Levels of CD 133
Malignant glioma is a highly recurrent tumor even after surgery, chemotherapy, radiation and immunotherapy. To address the potential role of CDl 33 positive tumor cells in glioblastoma recurrence, the CD133 expression upon first and second resection of tumor tissue from the same patient were compared. We obtained primary tumor tissue from five pathological confirmed grade IV astrocytoma (GBM) patients, and then re-operated following radiation, chemotherapy, and/or immunotherapy to obtain recurrent tumor tissue. RNA extraction and RT-PCR was performed for CDl 33 as described above. For all tested patients, CDl 33 expression was significantly higher in recurrent tumor tissue (relative expression ~2.5-19.1) than that in autologous primary tumor tissue (relative expression ~1)(FIG. 12). This example implicates a resistant CD133 positive tumor population in tumor recurrence.
Example 10. Immunization With Cancer Stem Cell Antigens Increased Survival A dendritic cell vaccine was generated using antigens from neurosphercs, daughter cells, and monolayer cells. Immature dendritic cells were generated from the bone marrow of 6-12-week-old Fisher F344 rats as previously described (Talmor et al., Eur. J. Immunol., 28:811-817, 1998). Briefly, bone marrow was harvested from the femoral and tibial marrow cavities and cultured in RPMI 1640 media supplemented with 10% fetal bovine serum (Gemini Biotechnologies, Calabasas, CA), 1% Penicillin/Streptomycin (Invitrogen, Carlsbad, CA), 50 ng/ml recombinant rat GM-CSF and 100 ng/ml recombinant rat IL-4 (R & D Systems, Minneapolis, MN). Cultures were fed every 2 days by removing 75% of the media and replacing it with fresh media containing cytokines (this washed away most of the lymphocytes and granulocytes). To determine the percentage of immature dendritic cells generated and cultured from the bone marrow of Fisher rats after 1 week of exposure to GM-CSF and IL-4, FACS analysis was run on cells imrnunostained with antibodies for CD86 (costimulatory marker B 7-1; DC marker), CD80 (costimulatory marker B7-2; DC marker), CD3 (T cell marker), or MHC II (DC marker). The dendritic cell populations obtained were positive for CD86, CD80, and MHC II, while negative for CD3.
Soluble peptides were generated for dendritic cell pulsing by cell lysis. 9L neurosphcres, daughter cells, and monolayer (adherent) cells were processed in the laboratory to produce a single cell suspension. The cells were then lyscd by 4 to 5 freeze cycles (on liquid nitrogen) and thaw cycles (room temperature). Lysis was monitored by light microscopy, and larger particles were removed by centrifugation (10 minutes at 60Ox g). The supematants were passed through a 0.2 μm filter, and protein concentration was determined by BioRad protein assay and aliquots frozen at -800C until use. To establish intracranial tumors, adult Fisher F344 rats were stereotactically inoculated in the right corpus striatum (from bregma in mm: anterior-posterior +1 mm; medial-lateral -3 mm; dorsal-ventral -5 mm) with 25,0009L-luciferase tumor cells as described above. For vaccination of the rats, freshly cultured immature dendritic cells were cocultured overnight for 24 hours with 80-100 μg of cell lysate one day prior to the vaccination at days 7, 14, and 21 post-opcratively. Vaccinations were given subcutaneously in the flanks on days 7, 14, and 21 with 50,000 DCs pulsed with antigens from either 9L neurospheres (NS), daughter cells (DtC), monolayer (adherent) cells (AC), or saline control. The animals were followed for survival and euthanized when terminal neurological signs developed, for example, inability to access food, water, seizure activity, weakness, and paralysis.
Tumor bearing rats injected with three successive vaccines (once per week) of dendritic cells pulsed with control, AC , DtC, or NS antigens, had median survival dates of 26.5, 32, 29 and 50 days, respectively (FIG. 13). Kaplan-Meier analysis showed that rats treated with 9L neurosphere lysate-pulsed DC had significantly longer survival time than each of the other groups (p = 0.0015).
Example 11. DCs Pulsed With Cancer Stem Cell Antigens Induced a Strong Cytotoxic T Cell Response Against 9L Tumor Cells
To determine whether the relative protective effect of NS-DC vaccination on survival is due to tumor-specific immunity, a cytotoxic T lymphocyte (CTL) assay was performed. Spleens were removed on day 28 post-intracranial 9L-lucifcrase tumor cells implantation from groups of rats treated with cither control or 9L peptide-pulsed dendritic cells. Splenocytes were isolated and re-stimulated in vitro, as described (Wunderlich et al., "Assays for T-cell function," In: Coligan et al., eds., Current Protocols in Immunology, New York, NY, John Wiley & Sons, Inc; 1997:3.11.1-3.11.20; Ehtesham et al., J. Immunother., 26:107-116, 2003) with either irradiated (10,000 rads) 9L adherent cells (AC), daughter cells (DtC) or neurospheres (NS). The effector: target (E:T) ratio was 6:1. Re-stimulated cells were cultured in RPMI-1640 with 10% fetal bovine scrum, 1% penicillin-streptomycin, and 1% HEPES in 6-wcll flat-bottom plates in a humidified 37 0C, 5% CCh incubator. Cells were incubated for 11 days with addition of IL-2 (300 units/mL) every 3 days. After 11 days of re-stimulation, the cells were re-exposed to their initial target (i.e., irradiated 9L AC, DtC, or NS). 24 hours later, culture media was harvested from the remaining cells and used for IFNγ protein quantification by ELISA (Ehtesham et al., J. Immunother., 26: 107-116, 2003). Re-stimulated splenocytes from rats treated with NS pulsed DCs showed significantly higher IFNγ release in response to exposure to tumor cell targets than re-stimulated splenocytes from rats treated with AC or DtC (FIG. 14), indicating that a higher cytotoxic T cell response was obtained in the NS-DC vaccinated rats. These data were also confirmed by PCR for IFNγ levels (data not shown). The higher IFNγ response in the NS-DC vaccinated group corresponds to the higher survival rate observed in the same group. These data show that a cancer stem cell antigen vaccine targets tumor cells more potently than vaccines based on tumor antigens from daughter cells, or cell populations that are not enriched for cancer stem cells.
Example 12. Intracranial T-CeIl Infiltration Is Associated With Prolonged Survival
A representative number of brains from DC-DtC and DC-NS vaccinated rats were carefully removed and post-fixed in 4% paraformaldehyde. Coronal sections of 20 μm were cut on a cryostat and blocked with normal horse serum for 1 hour. Slides were then incubated with anti-CD4 (clone OX-38 monoclonal antibody diluted in 1 :200 in PBS) for 2 hours at room temperature, followed by a 20 minute incubation at room temperature with the linking antibody (BioGenex biotinylated anti-mouse immunoglobulin). After washing in PBS, the labeled moiety (BioGenex Horse Radish Peroxidase-conjugated streptavidin) was added for 20 minutes at room temperature. DAB (3,3'-diaminobenzidine) wa? used as the chromogen. For analysis of CD8 expression, slides were incubated overnight with Anti-CD8 alpha Chain, clone OX-8 antibody (Chemicon).
Immunohistochemical assessment of brain sections from rats vaccinated with dendritic cells pulsed with NS vaccinated showed that there was a robust infiltration of CD4+ (FIG. 15B) and CD8+ lymphocytes that was not observed in the brain sections obtained from rats vaccinated with dendritic cells pulsed with daughter cell antigens (FIG. 15A). This infiltration of T cells correlates with increased survival of rats vaccinated with NS pulsed DCs. Example 13. Cancer Stem Cells Express MHC. Unlike Normal Stem Cells
Five glioblastoma multiforme (GBM) cancer stem cells (CSC) were generated by the methods described in Yuan et al. (Oncogene, 23:9392-9400, 2004). Briefly, tumor specimens were obtained within half an hour of surgical resection from five adult GBM patients, as approved by the Institutional Review Boards at the Cedars-Sinai Medical Center. Tumor tissue was washed, minced, and enzymatically dissociated (Reynolds et al., Science, 255:1707-10, 1992). Tumor cells were resuspended in DMEM/F12 medium containing 10% fetal bovine serum (FBS) as growth medium and plated at a density of 2x10δ live cells per 75 cm2 flask. The cells attached and grew as a monolayer in flasks. AU the five monolayer growing adult GBM cells were switched into a defined serum-free NSC medium (Reynolds et al., Science, 255:1707-10, 1992) containing 20 ng/ml of basic fibroblast growth factor (bFGF, Peprotech, Rocky Hill, NJ) and 20 ng/ml of epidermal growth factor (EGF, Peprotech). Normal human fetal neural stem cells (NSC) were cultured in the same defined scrum-free medium as for cancer stem cells. CSC and NSC cells were stained by FlTC-HLA-A1B1C antibody and isotype control antibody (BD Bioscience, San Diego, CA) and analyzed by flow cytometry. Representative CSC and NSC results were shown in the FIGs. 17A-17B and 18A-18B, respectively.
HLA expression was seen in 5 of 5 cancer stem cells from different patients. CSCs expressed high levels of HLA-A,B,C, however, NSC did not expression MHC class I (HLA-A,B,C) antigens on the surface. This unexpected result indicates that specific cancer stem cell antigens on cancer stem cells can be targeted using vaccines that generate T cells that recognize and kill cancer stem cell antigens in the context of MHC. Therefore cancer stem cells will be targeted, whereas normal stem cells will not.
Example 14. Isolation of CD133 T Cell Epitopes
A nucleic acid encoding a portion of CD 133 extracellular domain 1 (amino acid residues 116-270 of SEQ ID NO: 53) is used in cloning and expression on the surface of DC. DC are transfected with CD133-1 cDNA construct or empty vector (mock) for 48 hours. The successful transfection of DC-CD133 cells is identified either by anti-CD133 monoclonal antibody using flow cytometry or by EGFP cloned vector under fluorescent microscopy. Antigen presenting cells with CD133 receptor are stimulated by CD133 T cell epitopes. Overlapping peptides of 8-10 amino acids of residues 325-350 of SEQ ID NO: 53 are produced as MHC class I epitopes. Similarly, overlapping peptides of 13-20 amino acid of residues 325-350 arc produced as MHC class II epitopes. Stimulated APC using the peptide epitopes lead to enhanced production of CD8 T cells targeted to CDl 33 molecular on stem/progenitor cells in brain tumors. OTHER EMBODIMENTS
A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method of treating neural cancer in a patient, the method comprising: obtaining a population of dendritic cells; contacting the dendritic cells with a neural cancer stem cell antigen composition under conditions such that the dendritic cells present neural cancer stem cell antigens; and administering to a patient a composition comprising the dendritic cells.
2. The method of claim 1, wherein the dendritic cells are autologous.
3. The method of claim 1 , wherein the dendritic cells are allogeneic.
4. The method of claim 1, wherein the neural cancer stem cell antigen composition is obtained from a brain tumor.
5. The method of claim 4, wherein the brain tumor is a glioma.
6. The method of claim 1, wherein the neural cancer stem cell antigen composition comprises a lysate of neural cancer stem cells.
7. The method of claim 1, wherein the neural cancer stem cell antigen composition comprises an acid eluate of neural cancer stem cells.
8. The method of claim 6, wherein the neural cancer stem cells express CDl 33.
9. The method of claim 1, wherein the neural cancer stem cell antigen composition comprises one or more isolated peptides of CDl 33, CD90, CD44, CXCR4, Ncstin,
Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAPl, cIAP2, NAIP, or survivin.
10. The method of claim 9, wherein the one or more isolated peptides are synthetic.
11. A method of preparing a neural cancer vaccine, the method comprising: obtaining a population of dendritic cells; and contacting the dendritic cells with a neural cancer stem cell antigen composition under conditions such that the dendritic cells present neural cancer stem cell antigens, thus preparing a neural cancer vaccine.
12. A method of preparing a cell vaccine for treating a neural cancer, the method comprising: obtaining mononuclear cells from a subject; culturing the mononuclear cells in vitro under conditions in which mononuclear cells differentiate into antigen presenting cells; isolating neural cancer stem cells from the subject; obtaining a neural cancer stem cell antigen composition from the neural cancer stem cells; and culturing the antigen presenting cells in the presence of the neural cancer stem cell antigen composition, thus preparing a cell vaccine.
13. The method of claim 12, further comprising administering the cell vaccine to the subject.
14. A kit for preparing a cell vaccine for treating a brain cancer, the kit comprising: one or more isolated peptides of CD133, CD90, CD44, CXCR4, Nestin, Musashi- 1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRPl, MGMT, Bcl-2, FLIP, BCL-XL1 XIAP, cIAPl, CIAP2, NAIP, or survivin.
15. A composition consisting essentially of dendritic cells comprising one type or one or more different types of neural cancer stem cell antigens.
16. The composition of claim 15, wherein the neural cancer stem cell antigens include peptides of onc ormore of CD133, CD90, CD44, CXCR4, Nestin, Musashi-1 (Msil), maternal embryonic leucine zipper kinase (MELK), GLIl, PTCHl, Bmi-1, phosphoserine phosphatase (PSP), Snail, OCT4, BCRP 1 , MGMT, Bcl-2, FLIP, BCL-XL, XIAP, cIAP L , cIAP2, NAIP, or survivin.
17. A method of treating cancer in a patient, the method comprising: obtaining a population of dendritic cells; contacting the dendritic cells with a cancer stem cell antigen composition under conditions such that the dendritic cells present cancer stem cell antigens; and administering to a patient a composition comprising the dendritic cells.
18. A method of preparing a cancer vaccine, the method comprising: obtaining a population of dendritic cells; and contacting the dendritic cells with a cancer stem cell antigen composition under conditions such that the dendritic cells present cancer stem cell antigens, thus preparing a cancer vaccine.
19. A method of preparing a cell vaccine for treating a cancer, the method comprising: obtaining mononuclear cells from a subject; culturing the mononuclear cells in vitro under conditions in which mononuclear cells differentiate into antigen presenting cells; isolating cancer stem cells from the subject; obtaining a cancer stem cell antigen composition from the cancer stem cells; and culturing the antigen presenting cells in the presence of the cancer stem cell antigen composition, thus preparing a cell vaccine.
20. A method of treating cancer in a patient, the method comprising: administering to a patient a composition consisting essentially of dendritic cells that present cancer stem cell antigens.
PCT/US2007/079600 2006-09-26 2007-09-26 Cancer stem cell antigen vaccines and methods WO2008039874A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA2700573A CA2700573C (en) 2006-09-26 2007-09-26 Cancer stem cell antigen vaccines and methods
EP07843269.7A EP2084267B1 (en) 2006-09-26 2007-09-26 Cancer stem cell antigen vaccines and methods

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82695506P 2006-09-26 2006-09-26
US60/826,955 2006-09-26

Publications (2)

Publication Number Publication Date
WO2008039874A2 true WO2008039874A2 (en) 2008-04-03
WO2008039874A3 WO2008039874A3 (en) 2008-07-10

Family

ID=39230952

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/079600 WO2008039874A2 (en) 2006-09-26 2007-09-26 Cancer stem cell antigen vaccines and methods

Country Status (4)

Country Link
US (2) US8129184B2 (en)
EP (1) EP2084267B1 (en)
CA (1) CA2700573C (en)
WO (1) WO2008039874A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2109668A2 (en) * 2007-01-22 2009-10-21 Raven Biotechnologies Human cancer stem cells
WO2010011893A1 (en) * 2008-07-24 2010-01-28 University Of Central Florida Research Foundation, Inc. Therapy targeting cancer stem cells
WO2010124498A1 (en) * 2009-04-30 2010-11-04 Beijing Cellonis Biotechnology Co., Ltd A resistance-screened tumor stem cell, its antigen composition, an anti-tumor dendritic cell loading with said antigens, their preparation methods, uses and kits thereof as well as a dendritic cell vaccine
US7939090B2 (en) 2003-10-21 2011-05-10 Cedars-Sinai Medical Center System and method for the treatment of cancer, including cancers of the central nervous system
EP2328923A2 (en) * 2008-09-02 2011-06-08 Cedars-Sinai Medical Center Cd133 epitopes
US8097256B2 (en) 2006-09-28 2012-01-17 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
CN102692506A (en) * 2011-03-21 2012-09-26 复旦大学 Use of CD133 in preparation of tumor marker and kit of CD133
US8383768B2 (en) 2009-05-07 2013-02-26 Immunocellular Therapeutics, Ltd. CD133 epitopes
CN104812446A (en) * 2012-10-24 2015-07-29 密歇根大学董事会 Cancer stem cell vaccination and treatment
US20160060599A1 (en) * 2014-08-29 2016-03-03 China Medical University Hospital Dendritic cell tumor vaccine and method for preparing the same
EP2956164A4 (en) * 2013-02-14 2016-08-31 Immunocellular Therapeutics Ltd Ovarian cancer vaccines and vaccination methods
EP3120869A1 (en) * 2008-10-01 2017-01-25 Immatics Biotechnologies GmbH Novel immunotherapy against several tumors including neuronal and brain tumors
AU2016204710B2 (en) * 2008-10-01 2018-02-22 Immatics Biotechnologies Gmbh Novel immunotherapy against several tumors including neuronal and brain tumors
US10137182B2 (en) 2013-02-14 2018-11-27 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods
CN109620952A (en) * 2019-01-04 2019-04-16 北京中台恒基生物技术有限公司 A kind of tumor vaccine and preparation method thereof

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1670513B1 (en) * 2003-10-06 2013-01-23 Cedars-Sinai Medical Center Cox-2 inhibitors and dendritic cells for use in the treatment of cancer.
EP2084267B1 (en) 2006-09-26 2018-04-11 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
WO2008042407A1 (en) * 2006-10-03 2008-04-10 Ludwig Institute For Cancer Research Melanoma repopulating cells
ITMI20062100A1 (en) * 2006-10-31 2008-05-01 Fondazione I R C C S Istituto Neurologico METHOD FOR THE STIMULATION OF DENDRITIC CELLS AND CELL PRODUCT SO OBTAINED FOR AUTOLOGOUS IMMUNOTHERAPY OF HUMAN SOLID TUMORS
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
US20100285465A1 (en) * 2007-08-02 2010-11-11 Glaxosmithkline Biologicals S.A. Molecular Typing of Neisseria Strains by Determining the Presence of Genes Involved in Lipooligosaccharide (LOS) Biosynthesis
US20100048912A1 (en) 2008-03-14 2010-02-25 Angela Brodie Novel C-17-Heteroaryl Steroidal CYP17 Inhibitors/Antiandrogens, In Vitro Biological Activities, Pharmacokinetics and Antitumor Activity
WO2009129227A1 (en) 2008-04-17 2009-10-22 Pds Biotechnology Corporation Stimulation of an immune response by enantiomers of cationic lipids
US20110312916A1 (en) 2009-02-05 2011-12-22 Tokai Pharmaceuticals, Inc. Novel prodrugs of steroidal cyp17 inhibitors/antiandrogens
ES2825723T3 (en) 2009-07-13 2021-05-17 Biogencell Ltd Method of using lead cells for activation and differentiation of specific stem / progenitor cells
JP2011135864A (en) * 2009-12-30 2011-07-14 Korea Univ Research & Business Foundation Composition for retrodifferentiating somatic cell to embryonic stem cell-like cell, by using oct4 in combination with bmi1 or upstream regulator thereof, and method for generating embryonic stem cell-like cell using the same
US20130028915A1 (en) * 2011-07-27 2013-01-31 Baylor Research Institute Dendritic cell (dc)-vaccine therapy for pancreatic cancer
US8956870B2 (en) 2012-01-19 2015-02-17 Biogencell, Ltd. Method for using directing cells for specific stem/progenitor cell activation and differentiation
AU2013303012B2 (en) * 2012-08-15 2017-04-27 Neostem Oncology, Llc Rapid method production high purity cancer stem cells and population of high purity cancer stem cells
JP2015530413A (en) 2012-09-21 2015-10-15 ベデュ−アッド,フランク Improved vaccine compositions and methods of use
US8975290B2 (en) 2013-03-01 2015-03-10 Colorado State University Research Foundation Methods and compositions for enhancing an immune response, blocking monocyte migration, amplifying vaccine immunity and inhibiting tumor growth and metastasis
CA2903212A1 (en) * 2013-03-07 2014-09-12 Neostem Oncology, Llc Individualized high purity hepatocellular carcinoma stem cells, methods and use of the same
AU2014248713A1 (en) * 2013-03-12 2015-09-10 Neostem Oncology, Llc Individualized high-purity glioblastoma multiforme stem cells and methods for stimulating immune response
AU2014249346A1 (en) * 2013-03-12 2015-10-01 Caladrius Biosciences High purity ovarian cancer stem cells for active autologous immune therapy
AU2014248711A1 (en) * 2013-03-13 2015-09-10 Neostem Oncology, Llc Individualized high purity colon carcinoma stem cells, methods and use of the same
SG11201507093WA (en) 2013-03-14 2015-10-29 Univ Maryland Baltimore Office Of Technology Transfer Androgen receptor down-regulating agents and uses thereof
RU2016104643A (en) 2013-08-12 2017-09-19 Токай Фармасьютикалз, Инк. BIOMARKERS FOR THE TREATMENT OF NEOPLASTIC DISEASES WITH THE APPLICATION OF METHODS TREATED AT ANDROGEN
WO2015143074A1 (en) * 2014-03-20 2015-09-24 University Of Southern California Brain cancer immunotherapy
WO2016014613A1 (en) * 2014-07-22 2016-01-28 The Trustees Of The University Of Pennsylvania Compositions and methods for cancer immunotherapy
WO2016161309A1 (en) * 2015-04-01 2016-10-06 Colorado State University Research Foundation Optimized cancer stem cell vaccines
WO2017081686A1 (en) 2015-11-10 2017-05-18 B. G. Negev Technologies And Applications Ltd., At Ben-Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
WO2017083820A1 (en) 2015-11-13 2017-05-18 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
KR102549625B1 (en) 2017-03-03 2023-06-28 트레오스 바이오 리미티드 Personalized Immunogenic Peptide Identification Platform
MX2021002449A (en) 2018-09-04 2021-08-05 Treos Bio Ltd Peptide vaccines.
US20220313734A1 (en) * 2019-05-09 2022-10-06 Figene, Llc Fibroblast generated patient-specific vaccines

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US710903A (en) 1902-06-21 1902-10-07 Louis J Sticklin Lid-controlling device.
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5643741A (en) 1990-03-30 1997-07-01 Systemix, Inc. Identification and isolation of human hematopoietic stem cells
US5643786A (en) 1995-01-27 1997-07-01 The United States Of America As Represented By The Department Of Health And Human Services Method for isolating dendritic cells
US5650299A (en) 1993-10-06 1997-07-22 The University Of Florida Cells producing stem cell proliferation factor
US5824489A (en) 1992-07-27 1998-10-20 California Institute Of Technology In vitro method for obtaining an isolated population of mammalian neural crest stem cells
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
US5928947A (en) 1992-07-27 1999-07-27 California Institute Of Technology Mammalian multipotent neural stem cells
US6337184B1 (en) 1997-04-01 2002-01-08 Jeffrey B. Miller Molecular marker for muscle stem cells
US6645763B2 (en) 2001-10-12 2003-11-11 Naoya Kobayashi Immortalized bone marrow mesenchymal stem cell
US6800790B2 (en) 1998-07-24 2004-10-05 Carnegie Institution Of Washington Method for maintenance and propagation of germline stem cells using members of the TFG-β family of growth factors
US6875607B1 (en) 1998-11-09 2005-04-05 Es Cell International Pte Ltd Embryonic stem cells
WO2005037995A2 (en) 2003-10-06 2005-04-28 Cedars-Sinai Medical Center Use of cox-2 inhibitor to prevent t-cell anergy induced by dendritic cell therapy
WO2005043155A1 (en) 2003-10-21 2005-05-12 Cedars-Sinai Medical Center System and method for the treatment of cancer, including cancers of the central nervous system
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US7115267B2 (en) 1996-04-03 2006-10-03 Wellstat Therapeutics Corporation Stimulator of stem cell division
US7247480B2 (en) 1998-11-05 2007-07-24 Isis Innovation Limited Method for producing dendritic cells

Family Cites Families (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US4844893A (en) 1986-10-07 1989-07-04 Scripps Clinic And Research Foundation EX vivo effector cell activation for target cell killing
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
DK0474727T3 (en) * 1989-05-19 1998-01-12 Genentech Inc HER2 extracellular domain
US5541104A (en) 1991-05-23 1996-07-30 Ludwig Institute For Cancer Research Monoclonal antibodies which bind to tumor rejection antigen precursor mage-1
US5342774A (en) 1991-05-23 1994-08-30 Ludwig Institute For Cancer Research Nucleotide sequence encoding the tumor rejection antigen precursor, MAGE-1
US5925729A (en) 1991-05-23 1999-07-20 Ludwig Institute For Cancer Research Tumor rejection antigen precursors, tumor rejection antigens and uses thereof
US6037135A (en) * 1992-08-07 2000-03-14 Epimmune Inc. Methods for making HLA binding peptides and their uses
EP0633929B1 (en) * 1992-04-01 2004-03-03 The Rockefeller University METHOD FOR $i(IN VITRO) PROLIFERATION OF DENDRITIC CELL PRECURSORS AND THEIR USE TO PRODUCE IMMUNOGENS
US6077519A (en) * 1993-01-29 2000-06-20 University Of Pittsburgh Methods for isolation and use of T cell epitopes eluted from viable cells in vaccines for treating cancer patients
US5801005A (en) * 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US5869445A (en) * 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5419900A (en) 1993-05-19 1995-05-30 The United States Of America As Represented By The Department Of Of Health And Human Services Immunologic enhancement with intermittent interleukin-2 therapy
US5550214A (en) 1994-02-10 1996-08-27 Brigham And Women's Hospital Isolated antigenic oncogene peptide fragments and uses
EP0756604A1 (en) 1994-04-22 1997-02-05 THE UNITED STATES OF AMERICA, as represented by the Secretary of the Department of Health and Human Services Melanoma antigens
US5874560A (en) * 1994-04-22 1999-02-23 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
US6300090B1 (en) * 1994-07-29 2001-10-09 The Rockefeller University Methods of use of viral vectors to deliver antigen to dendritic cells
JPH10510988A (en) 1994-12-14 1998-10-27 ザ スクリップス リサーチ インスティテュート In vivo activation of tumor-specific cytotoxic T cells
US6010905A (en) * 1995-01-27 2000-01-04 The United States Of America As Represented By The Department Of Health & Human Services Method for inducing monocytes to exhibit the phenotype of activated myeloid dendritic cells
US5635363A (en) 1995-02-28 1997-06-03 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for the detection, quantitation and purification of antigen-specific T cells
US6514942B1 (en) * 1995-03-14 2003-02-04 The Board Of Regents, The University Of Texas System Methods and compositions for stimulating T-lymphocytes
US5788963A (en) * 1995-07-31 1998-08-04 Pacific Northwest Cancer Foundation Isolation and/or preservation of dendritic cells for prostate cancer immunotherapy
US5840839A (en) * 1996-02-09 1998-11-24 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Alternative open reading frame DNA of a normal gene and a novel human cancer antigen encoded therein
US5849589A (en) * 1996-03-11 1998-12-15 Duke University Culturing monocytes with IL-4, TNF-α and GM-CSF TO induce differentiation to dendric cells
US5843633A (en) * 1996-04-26 1998-12-01 Amcell Corporation Characterization of a human hematopoietic progenitor cell antigen
US6455678B1 (en) * 1996-04-26 2002-09-24 Amcell Corporation Human hematopoietic stem and progenitor cell antigen
US6458585B1 (en) * 1996-08-14 2002-10-01 Nexell Therapeutics Inc. Cytokine-free culture of dendritic cells
US7368531B2 (en) 1997-03-07 2008-05-06 Human Genome Sciences, Inc. Human secreted proteins
AU7499498A (en) * 1997-05-21 1998-12-11 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Methods and compositions for making dendritic cells from expanded populations ofmonocytes and for activating t cells
AU2306499A (en) 1997-12-18 1999-07-05 Human Genome Sciences, Inc. 110 human secreted proteins
US20020034819A1 (en) * 1998-02-23 2002-03-21 Alan K. Smith Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US20020192211A1 (en) 1998-03-17 2002-12-19 Hudziak Robert M. Method of treating tumor cells by inhibiting growth factor receptor function
US6248329B1 (en) 1998-06-01 2001-06-19 Ramaswamy Chandrashekar Parasitic helminth cuticlin nucleic acid molecules and uses thereof
US20020115213A1 (en) * 1998-08-14 2002-08-22 Northeastern Ohio Universities Of Medicine Engraftable neural progenitor & stem cells for brain tumor therapy
US7186409B2 (en) * 1998-08-14 2007-03-06 The Children's Medical Center Corporation Neural stem cells and use thereof for brain tumor therapy
CN1330557A (en) * 1998-09-15 2002-01-09 匹兹堡大学联邦系统高等教育 In situ injection of antigen-presenting cells with generatically enhanced cytokine expression cell factor
US7504490B1 (en) 1998-10-16 2009-03-17 Oscient Pharmaceuticals Corporation Nucleic acid and amino acid sequences relating to Apergillus fumigatus for diagnostics and therapeutics
AU1598500A (en) 1998-10-26 2000-05-15 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Hla-a2 and hla-dr specific peptide epitopes from the melanoma antigen trp2
EP1140182A2 (en) 1998-12-23 2001-10-10 G.D. Searle & Co. Use of a matrix metalloproteinase inhibitor together with an antineoplastic agent, optionally also together with radiation, in the treatment of neoplasia
WO2000040616A1 (en) 1999-01-08 2000-07-13 Panacos Pharmaceuticals, Inc. METHODS OF ELICITING BROADLY NEUTRALIZING ANTIBODIES TARGETING HIV-1 gp41
CA2371260A1 (en) 1999-05-03 2000-11-09 Karolinska Innovations Ab Materials and methods relating to neuronal development
US6566395B1 (en) * 1999-05-25 2003-05-20 Biomedicines, Inc. Methods of treating proliferative disorders
US7060284B1 (en) 1999-08-03 2006-06-13 The Ohio State University Polypeptides and polynucleotides for enhancing immune reactivity to HER-2 protein
US20070098776A1 (en) * 1999-12-13 2007-05-03 Fikes John D HLA class I A2 tumor associated antigen peptides and vaccine compositions
EP1242049A4 (en) 1999-12-13 2005-01-05 Epimmune Inc Hla class i a2 tumor associated antigen peptides and vaccine compositions
US20020197266A1 (en) 2000-02-08 2002-12-26 Waldemar Debinski Immunotherapy using interleukin 13 receptor subunit alpha 2
US20060003323A1 (en) 2000-03-01 2006-01-05 John Alsobrook Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2406664C (en) 2000-03-15 2011-05-10 Children's Medical Center Corporation Systemic delivery of neural stem cells to treat cancer
US20020076707A1 (en) 2000-06-30 2002-06-20 David Mack Novel methods of diagnosing cancer, compositions, and methods of screening for cancer modulators
US20020119121A1 (en) * 2000-09-15 2002-08-29 Vitiello Maria Antonella Compositions and methods for inducing specific cytolytic T cell responses
AU2002211502A1 (en) 2000-10-04 2002-04-15 Curagen Corporation Novel proteins and nucleic acids encoding same and antibodies directed against these proteins
US20030202963A1 (en) * 2000-10-12 2003-10-30 Cornell Research Foundation, Inc. Method of treating cancer
US20040121946A9 (en) 2000-12-11 2004-06-24 John Fikes Inducing cellular immune responses to her2/neu using peptide and nucleic acid compositions
US6632459B2 (en) 2000-12-11 2003-10-14 Nutricia N.V. Chlorogenic acid and an analog thereof for immune system stimulation
HUP0402656A3 (en) 2001-02-20 2012-03-28 Ortho Mcneil Pharm Inc A cell therapy method for the treatment of tumors
EP1236740B1 (en) 2001-02-28 2012-07-18 Bio Life Science Forschungs- und Entwicklungsges.m.b.H. Vaccine against HER-2/neu oncogene-associated cancers
CN1168740C (en) * 2001-04-04 2004-09-29 上海美恩生物技术有限公司 Cell factor gene modified antigen presentation cell/tumor cell conjugate, its preparatino and use
DE10132502A1 (en) * 2001-07-05 2003-01-23 Gsf Forschungszentrum Umwelt Attack on tumor cells with missing, low or abnormal MHC expression by combining non-MHC-restricted T cells / NK cells and MHC-restricted cells
JP2004535819A (en) 2001-07-25 2004-12-02 イ・デ・エム・イミュノ−デジネ・モレキュル New isolated dendritic cells, methods for preparing them, and their use in pharmaceutical compositions
GB0119346D0 (en) 2001-08-08 2001-10-03 Bioclones Proprietary Ltd Process for the maturation of dendritic cells
JP2004538331A (en) * 2001-08-17 2004-12-24 ロジャー ウィリアムス ホスピタル In situ immunization
KR101803702B1 (en) * 2001-09-06 2017-12-01 노쓰웨스트 바이오써라퓨틱스, 인크. Compositions and methods for priming monocytic dendritic cells and T cells for Th-1 response
US20040072246A1 (en) * 2001-09-11 2004-04-15 Roland Martin System and method for identifying t cell and other epitopes and the like
WO2003035004A2 (en) 2001-10-26 2003-05-01 Immuno-Rx, Inc. Immunotherapy for reversing immune suppression
CA2474778A1 (en) 2002-02-08 2003-08-14 Peter Carmeliet A novel target to inhibit angiogenesis
JP2005526842A (en) 2002-03-22 2005-09-08 ザ ペン ステート リサーチ ファウンデーション Cancer immunotherapy
US7413869B2 (en) * 2002-04-05 2008-08-19 Dendreon Corporation Method for determining potency of antigenic presenting cell based vaccines
US7204982B2 (en) * 2002-07-02 2007-04-17 The Regents Of The University Of California Compositions and methods for treatment and detection of multiple cancers
US20090110702A1 (en) 2002-07-12 2009-04-30 The Johns Hopkins University Mesothelin Vaccines and Model Systems and Control of Tumors
US20040057935A1 (en) * 2002-09-20 2004-03-25 Cedars-Sinai Medical Center Intratumoral delivery of dendritic cells
US20040197903A1 (en) * 2003-01-31 2004-10-07 Northwest Biotherapeutics, Inc. Method for induction of proliferation of natural killer cells by dendritic cells cultured with GM-CSF and IL-15
CA2517295C (en) * 2003-02-27 2016-01-12 Northwest Biotherapeutics, Inc. Generation of dendritic cells from monocytic dendritic precursor cells with gm-csf in the absence of additional cytokines
WO2005079581A1 (en) 2004-01-20 2005-09-01 Cedars-Sinai Medical Center Intratumoral delivery of dendritic cells
US7612162B2 (en) * 2004-09-21 2009-11-03 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Peptide analogs capable of enhancing stimulation of a glioma-specific CTL response
US20060204509A1 (en) * 2005-03-14 2006-09-14 Harty John T Accelerated CD8+ T-cell memory after dendritic cell vaccination
US7842466B1 (en) 2005-09-16 2010-11-30 Celera Corporation Colon disease targets and uses thereof
WO2007062138A2 (en) 2005-11-23 2007-05-31 Applera Corporation Methods and compositions for treating diseases targeting human prominin-1(cd133)
US20080107668A1 (en) * 2006-08-30 2008-05-08 Immunotope, Inc. Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
EP2084267B1 (en) 2006-09-26 2018-04-11 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
CA2700436C (en) 2006-09-28 2017-04-18 John S. Yu Cancer vaccines and vaccination methods
ITMI20062100A1 (en) 2006-10-31 2008-05-01 Fondazione I R C C S Istituto Neurologico METHOD FOR THE STIMULATION OF DENDRITIC CELLS AND CELL PRODUCT SO OBTAINED FOR AUTOLOGOUS IMMUNOTHERAPY OF HUMAN SOLID TUMORS
CA2668223A1 (en) 2006-10-31 2008-05-08 National Stem Cell Holding Inc. Methods for identifying, isolating, and utilizing endocrine progenitor cells from adult human pancreata
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
US8168586B1 (en) 2007-11-21 2012-05-01 Celera Corporation Cancer targets and uses thereof
ES2565779T3 (en) 2008-09-02 2016-04-06 Cedars-Sinai Medical Center CD133 epitopes
DK2427485T3 (en) 2009-05-07 2017-03-13 Immunocellular Therapeutics Ltd CD133 epitopes
ES2930809T3 (en) 2010-08-24 2022-12-22 Univ Pittsburgh Commonwealth Sys Higher Education Interleukin-13 alpha 2 receptor peptide-based brain cancer vaccines
KR102062407B1 (en) 2010-12-09 2020-01-03 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Use of chimeric antigen receptor-modified t cells to treat cancer
US9187534B2 (en) 2011-03-11 2015-11-17 Universite De Geneve Multi-epitopic vaccine
US20140234350A1 (en) 2013-02-14 2014-08-21 Cedars-Sinai Medical Center Ovarian cancer vaccines and vaccination methods
CA2898474A1 (en) 2013-02-14 2014-08-21 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US710903A (en) 1902-06-21 1902-10-07 Louis J Sticklin Lid-controlling device.
US5643741A (en) 1990-03-30 1997-07-01 Systemix, Inc. Identification and isolation of human hematopoietic stem cells
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5824489A (en) 1992-07-27 1998-10-20 California Institute Of Technology In vitro method for obtaining an isolated population of mammalian neural crest stem cells
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
US5928947A (en) 1992-07-27 1999-07-27 California Institute Of Technology Mammalian multipotent neural stem cells
US5981708A (en) 1993-10-06 1999-11-09 University Of Florida Stem cell proliferation factor
US5650299A (en) 1993-10-06 1997-07-22 The University Of Florida Cells producing stem cell proliferation factor
US5643786A (en) 1995-01-27 1997-07-01 The United States Of America As Represented By The Department Of Health And Human Services Method for isolating dendritic cells
US7115267B2 (en) 1996-04-03 2006-10-03 Wellstat Therapeutics Corporation Stimulator of stem cell division
US6337184B1 (en) 1997-04-01 2002-01-08 Jeffrey B. Miller Molecular marker for muscle stem cells
US6800790B2 (en) 1998-07-24 2004-10-05 Carnegie Institution Of Washington Method for maintenance and propagation of germline stem cells using members of the TFG-β family of growth factors
US7247480B2 (en) 1998-11-05 2007-07-24 Isis Innovation Limited Method for producing dendritic cells
US6875607B1 (en) 1998-11-09 2005-04-05 Es Cell International Pte Ltd Embryonic stem cells
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US7115360B2 (en) 2000-08-03 2006-10-03 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US6645763B2 (en) 2001-10-12 2003-11-11 Naoya Kobayashi Immortalized bone marrow mesenchymal stem cell
WO2005037995A2 (en) 2003-10-06 2005-04-28 Cedars-Sinai Medical Center Use of cox-2 inhibitor to prevent t-cell anergy induced by dendritic cell therapy
WO2005043155A1 (en) 2003-10-21 2005-05-12 Cedars-Sinai Medical Center System and method for the treatment of cancer, including cancers of the central nervous system
US20070020297A1 (en) 2003-10-21 2007-01-25 Cedars-Sinai Medcal Center System and method for the treatment of cancer, including cancers of the central nervous system

Non-Patent Citations (84)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2000, WILLIAMS & WILKINS PA
ADVANI ET AL., CANCER RES., vol. 59, 1999, pages 2055 - 58
AL-HAJJ ET AL., PROC. NATL. ACAD. SCI. USA, vol. 100, 2003, pages 3983 - 88
ANDERSEN ET AL., J. IMMUNOL., vol. 175, 2005, pages 2709 - 14
ANDREEFF ET AL., LEUKEMIA, vol. 13, 1999, pages 1881 - 92
BACHINSKY ET AL., CANCER IMMUN., vol. 5, 2005, pages 6
BENDA ET AL., J. NEUROSURG., vol. 34, 1971, pages 310 - 323
BJERKVIG ET AL., NAT. REV. CANCER, vol. 5, 2005, pages 899 - 904
BOISE ET AL., CELL, vol. 74, 1993, pages 597 - 608
BONNET; DICK, NAT. MED., vol. 3, 1997, pages 730 - 737
CAI ET AL., CANCER RES., vol. 65, 2005, pages 3319 - 27
CAMPOS ET AL., BLOOD, vol. 81, 1993, pages 3091 - 3096
COHEN ET AL., SCIENCE, vol. 305, 2004, pages 390 - 392
DAHLSTRAND ET AL., J. CELL SCI., vol. 103, 1992, pages 589 - 597
DEVERAUX ET AL., NATURE, vol. 388, 1997, pages 300 - 304
DOYLE ET AL., PROC. NAT. ACAD. SCI. USA, vol. 95, 1998
EHTESHAM ET AL., J. IMMUNOTHER., vol. 26, 2003, pages 107 - 116
FREUDENTHAL; STEINMAN, PROC. NAT. ACAD. SCI. USA, vol. 57, 1990, pages 7698 - 7702
GESCHWIND ET AL., NEURON, vol. 29, 2001, pages 325 - 339
GOOD ET AL., GENOMICS, vol. 52, 1998, pages 382 - 384
GRAY ET AL., CANCER RES., vol. 65, 2005, pages 9751 - 61
GROSZER ET AL., SCIENCE, vol. 294, 2001, pages 2186 - 89
HEMMATI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 100, 2003, pages 15178 - 83
HIRSCHMANN-JAX ET AL., PROC. NATL. ACAD. SCI. USA, vol. 101, 2004, pages 14228 - 33
IRMLER ET AL., NATURE, vol. 388, 1997, pages 190 - 195
KABOS ET AL., EXP. NEUROL., vol. 178, 2002, pages 288 - 293
KASPER ET AL., EUR. J. CANCER, vol. 42, 2006, pages 437 - 445
KATOH ET AL., CANCER BIOL. THER., vol. 4, 2005, pages 1050 - 54
KINZLER ET AL., NATURE, vol. 332, 1988, pages 371 - 374
KINZLER ET AL., SCIENCE, vol. 236, 1987, pages 70 - 73
L1U GENTAO ET AL.: "Chemoresistance of stem-like cells isolated from glioblastoma", PROCEEDINGS OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH ANNUAL MEETING, vol. 47, 2006, pages 75
LEE ET AL., NAT. NEUROSCI., vol. 8, 2005, pages 723 - 729
LESSARD ET AL., NATURE, vol. 423, 2003, pages 255 - 260
LI ET AL., NATURE, vol. 396, 1998, pages 580 - 584
LISTON ET AL., NATURE, vol. 379, 1996, pages 349 - 353
MARHABA ET AL., J. MOL. HISTOL., vol. 35, 2004, pages 211 - 31
MARIC ET AL., J. NEUROSCI., vol. 23, 2003, pages 240 - 251
MARKOWICZ; ENGLEMAN, J. CLIN. INVEST., vol. 85, 1990, pages 955 - 961
MEHTA-DAMANI ET AL., J. IMMUNOL., vol. 153, 1994, pages 996 - 1003
MOLOFSKY ET AL., NATURE, vol. 425, 2003, pages 962 - 967
MULLER ET AL., NATURE, vol. 410, 2001, pages 50 - 56
NAGAO ET AL., GENOMICS, vol. 85, 2005, pages 462 - 71
NICHOLS ET AL., CELL, vol. 95, 1998, pages 379 - 391
NOTON ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 1286 - 95
O'BRIEN ET AL., NATURE, vol. 445, 2007, pages 106 - 110
O'DOHERTY ET AL., J. EXP. MED., vol. 178, 1993, pages 1067 - 78
OKANO ET AL., EXP. CELL RES., vol. 306, 2005, pages 349 - 356
PARK ET AL., NATURE, vol. 423, 2003, pages 302 - 305
PARKER ET AL., J. IMMUNOL., vol. 152, 1994, pages 163
POTTEN ET AL., DEVELOPMENT, vol. 110, 1990, pages 1001 - 1020
RABIK ET AL., CANCER TREAT. REV., vol. 32, 2006, pages 261 - 276
RAMMENSEE ET AL., IMMUNOGENET., vol. 50, 1999, pages 213 - 219
REHEMTULLA ET AL., NEOPLASIA, vol. 2, 2000, pages 491 - 495
REKER ET AL., CANCER BIOL. THER., vol. 3, 2004, pages 173 - 179
REYA ET AL., NATURE, vol. 414, 2001, pages 105 - 111
REYNOLDS ET AL., J. NEUROSCI., vol. 12, 1992, pages 4565 - 74
REYNOLDS ET AL., SCIENCE, vol. 255, 1992, pages 1707 - 10
ROTHE ET AL., CELL, vol. 83, 1995, pages 1243 - 1252
ROY ET AL., CELL, vol. 80, 1995, pages 167 - 178
SANAI ET AL., N. ENGL. J. MED., vol. 353, 2005, pages 811 - 822
SCHMIDEK ET AL., J. NEUROSURG., vol. 34, 1971, pages 335 - 340
SCHMITZ ET AL., CANCER RES., vol. 60, 2000, pages 4845 - 4849
SEABERG ET AL., TRENDS NEUROSCI., vol. 26, 2003, pages 125 - 131
See also references of EP2084267A4
SIDDALL ET AL., PROC. NAT. ACAD. SCI. USA, vol. 103, 2006, pages 84 - 8407
SIEGEL ET AL., BR. J. HAEMATOL., vol. 122, 2003, pages 911 - 914
SINGH ET AL., CANCER RES., vol. 63, 2003, pages 5821 - 28
SINGH ET AL., NATURE, vol. 432, 2004, pages 396 - 401
TALMOR ET AL., EUR. J. IMMUNOL., vol. 28, 1998, pages 811 - 817
TAUBERT ET AL., ONCOGENE, vol. 24, 2005, pages 5258 - 61
THOMAS ET AL., J. IMMUNOL., vol. 151, 1993, pages 6840 - 6852
TUNICI ET AL., MOL. CANCER, vol. 3, 2004, pages 25
TUNICI PATRIZIA ET AL.: "Brain tumor stem cells: new targets for clinical treatments?", NEUROSURGICAL FOCUS, vol. 20, no. 4, 2006, pages E27
VAN LOHUIZEN ET AL., NATURE, vol. 353, 1991, pages 353 - 355
VEGA ET AL., GENES DEV., vol. 18, 2004, pages 1131 - 1143
WEISSMAN ET AL., ANNU. REV. CELL. DEV. BIOL., vol. 17, 2001, pages 387 - 403
WHEATLEY ET AL., INT. REV. CYTOL., vol. 247, 2005, pages 35 - 88
WUNDERLICH ET AL.: "Current Protocols in Immunology, New York", 1997, JOHN WILEY & SONS, INC, article "Assays for T-cell function", pages: 3.11.1 - 3.11.20
YIN ET AL., BLOOD, vol. 90, 1997, pages 5002 - 12
YOUNG; STEINMAN, J. EXP. MED., vol. 171, 1990, pages 1315 - 32
YU ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 20711 - 16
YUAN ET AL., ONCOGENE, vol. 23, 2004, pages 9392 - 9400
ZHOU ET AL., NAT. MED., vol. 7, 2001, pages 1028 - 34
ZOELLER, CANCER IMMUNOL. IMMUNOTHER., vol. 53, 2004, pages 567 - 79

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7939090B2 (en) 2003-10-21 2011-05-10 Cedars-Sinai Medical Center System and method for the treatment of cancer, including cancers of the central nervous system
US10226519B2 (en) 2006-09-28 2019-03-12 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US8871211B2 (en) 2006-09-28 2014-10-28 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US8097256B2 (en) 2006-09-28 2012-01-17 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US8309354B2 (en) 2007-01-22 2012-11-13 Macrogenics West, Inc. Human cancer stem cells
EP2109668A4 (en) * 2007-01-22 2011-10-05 Macrogenics West Inc Human cancer stem cells
EP2109668A2 (en) * 2007-01-22 2009-10-21 Raven Biotechnologies Human cancer stem cells
WO2010011893A1 (en) * 2008-07-24 2010-01-28 University Of Central Florida Research Foundation, Inc. Therapy targeting cancer stem cells
JP2011529080A (en) * 2008-07-24 2011-12-01 ユニバーシティ オブ セントラル フロリダ リサーチ ファウンデーション,インコーポレイテッド Therapies targeting cancer stem cells
EP2328923A4 (en) * 2008-09-02 2012-05-30 Cedars Sinai Medical Center Cd133 epitopes
US9382308B2 (en) 2008-09-02 2016-07-05 Cedars-Sinai Medical Center CD133 epitopes
EP2328923A2 (en) * 2008-09-02 2011-06-08 Cedars-Sinai Medical Center Cd133 epitopes
AU2016204710B2 (en) * 2008-10-01 2018-02-22 Immatics Biotechnologies Gmbh Novel immunotherapy against several tumors including neuronal and brain tumors
EP3120869A1 (en) * 2008-10-01 2017-01-25 Immatics Biotechnologies GmbH Novel immunotherapy against several tumors including neuronal and brain tumors
WO2010124498A1 (en) * 2009-04-30 2010-11-04 Beijing Cellonis Biotechnology Co., Ltd A resistance-screened tumor stem cell, its antigen composition, an anti-tumor dendritic cell loading with said antigens, their preparation methods, uses and kits thereof as well as a dendritic cell vaccine
US8604167B2 (en) 2009-05-07 2013-12-10 Immunocellular Therapeutics, Ltd. CD133 epitopes
US8383768B2 (en) 2009-05-07 2013-02-26 Immunocellular Therapeutics, Ltd. CD133 epitopes
CN102692506A (en) * 2011-03-21 2012-09-26 复旦大学 Use of CD133 in preparation of tumor marker and kit of CD133
EP2911748A4 (en) * 2012-10-24 2016-06-15 Univ Michigan Cancer stem cell vaccination and treatment
CN104812446A (en) * 2012-10-24 2015-07-29 密歇根大学董事会 Cancer stem cell vaccination and treatment
US10173074B2 (en) 2012-10-24 2019-01-08 The Regents Of The University Of Michigan Cancer stem cell vaccination and treatment
EP2956164A4 (en) * 2013-02-14 2016-08-31 Immunocellular Therapeutics Ltd Ovarian cancer vaccines and vaccination methods
US10137182B2 (en) 2013-02-14 2018-11-27 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods
US11096996B2 (en) 2013-02-14 2021-08-24 Precision Lifesciences Group Llc Cancer vaccines and vaccination methods
US20160060599A1 (en) * 2014-08-29 2016-03-03 China Medical University Hospital Dendritic cell tumor vaccine and method for preparing the same
CN109620952A (en) * 2019-01-04 2019-04-16 北京中台恒基生物技术有限公司 A kind of tumor vaccine and preparation method thereof

Also Published As

Publication number Publication date
US9023338B2 (en) 2015-05-05
US8129184B2 (en) 2012-03-06
CA2700573C (en) 2016-11-22
WO2008039874A3 (en) 2008-07-10
EP2084267A2 (en) 2009-08-05
US20120189664A1 (en) 2012-07-26
CA2700573A1 (en) 2008-04-03
EP2084267B1 (en) 2018-04-11
US20080206286A1 (en) 2008-08-28
EP2084267A4 (en) 2011-03-02

Similar Documents

Publication Publication Date Title
US9023338B2 (en) Cancer stem cell antigen vaccines and methods
JP5816627B2 (en) Method for the proliferation of antigen-specific T cells
AU2002307243A1 (en) Antigen-presenting cell populations and their use as reagents for enhancing or reducing immune tolerance
US20220000930A1 (en) Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same
JP2016510756A (en) Individual high-purity hepatocellular carcinoma stem cells, preparation method thereof and use thereof
US9687537B2 (en) Dendritic cell vaccines for asparaginyl-β-hydroxylase expressing tumors
WO2014165103A1 (en) Individualized high-purity glioblastoma multiforme stem cells and methods for stimulating immune response
Takeda et al. Immature dendritic cell/tumor cell fusions induce potent antitumour immunity
Pellegatta et al. Dendritic cell vaccines for cancer stem cells
US9149515B2 (en) Interferon-alpha-producing bone marrow dendritic cells
RU2395572C1 (en) Human melanoma cell line ig which secretes recombinant granulocytic-macrophagal colony-stimulating factor
A Chistiakov et al. Approaches to improve efficiency of dendritic cell-based therapy of high grade gliomas
EP1178822B1 (en) Whole cell cancer vaccine comprising co-cultured malignant and non-malignant cells
US10022402B2 (en) Allogenic mesendritic vector for ovarian cancer
CN117402218B (en) Individualized dendritic cell vaccine for Survivin positive tumor and preparation method thereof
RU2395571C1 (en) Human melanoma cell line 26g which secretes recombinant granulocytic-macrophagal colony-stimulating factor
RU2395574C1 (en) Human melanoma cell line ilg which secretes recombinant granulocytic-macrophagal colony-stimulating factor
CN116999461A (en) HLA-DR + CD8 + Application of Treg in preparation of kit or medicine
CN115927200A (en) Dendritic cell vaccine targeting receptor HER2
US20090136458A1 (en) Renal Carcinoma Cell Line and Uses thereof
RU2395570C1 (en) Human melanoma cell line pg which secretes recombinant granulocytic-macrophagal colony-stimulating factor
AU2008200315B2 (en) Antigen-presenting cell populations and their use as reagents for enhancing or reducing immune tolerance
RU2395573C1 (en) Human melanoma cell line 31g which secretes recombinant granulocytic-macrophagal colony-stimulating factor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07843269

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007843269

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2700573

Country of ref document: CA