WO2005043155A1 - System and method for the treatment of cancer, including cancers of the central nervous system - Google Patents

System and method for the treatment of cancer, including cancers of the central nervous system Download PDF

Info

Publication number
WO2005043155A1
WO2005043155A1 PCT/US2004/034761 US2004034761W WO2005043155A1 WO 2005043155 A1 WO2005043155 A1 WO 2005043155A1 US 2004034761 W US2004034761 W US 2004034761W WO 2005043155 A1 WO2005043155 A1 WO 2005043155A1
Authority
WO
WIPO (PCT)
Prior art keywords
chemotherapy
vaccination
cancer
vaccine
tumor
Prior art date
Application number
PCT/US2004/034761
Other languages
French (fr)
Inventor
Christopher Wheeler
Asha Das
Keith Black
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 EP04795865.7A priority Critical patent/EP1676132B1/en
Priority to US10/575,438 priority patent/US7939090B2/en
Priority to JP2006536762A priority patent/JP5015601B2/en
Publication of WO2005043155A1 publication Critical patent/WO2005043155A1/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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61K39/46432Nervous system antigens
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal 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
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • 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
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/47Brain; Nervous system

Definitions

  • the invention relates to the treatment of cancer, and in particular embodiments to cancers of the central nervous system (“CNS”), such as those found in the brain.
  • CNS central nervous system
  • GBM ghoblastoma multiforme
  • GBM diagnosis carries with it an average survival between twelve and eighteen months (with 90-95% patients surviving less than two years), without' the possibility of spontaneous remission or effective treatment. See, e.g., Id; and W.JJ. Curran et al, "Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials," J. Natl Cancer Inst., 85:690 (1993). The consistently short survival and absence of spontaneous remission that makes GBM such a devastating disease also renders the evaluation of new therapies for this disease relatively rapid and unequivocal. Overall survival represents the standard by which therapies for GBM are evaluated, in part because tumor mass reduction (i.e., surgically) does not necessarily correlate with prolonged survival.
  • tumor mass reduction i.e., surgically
  • CTL induce death in their cellular targets, it is not unreasonable to expect that inefficient CTL killing might either incompletely trigger death pathways in targeted tumor cells, or select for CTL-resistant tumor variants.
  • vaccine-elicited tumor- responsive CTL might fundamentally alter tumors by "priming" their death machinery.
  • CTL could fundamentally alter tumor cell physiology and/or genetics. Both of these possibilities could, in theory, be exploited by additional therapeutic modalities. Therefore, the clinical insufficiency of cancer vaccines encourages the examination of synergy between vaccination and other therapies, particularly to the extent that such an examination might uncover a novel approach to cancer therapy.
  • the invention relates to a novel treatment for cancer, and particularly cancers of the CNS; for example, cancers of the brain, such as GBM.
  • the invention includes a dual therapeutic approach to the treatment of cancer with at least one vaccination of DC and at least one course or regimen of chemotherapy.
  • the two therapies may be administered concurrently with one another and/or with an initial vaccination preceding chemotherapy.
  • the dual therapeutic approach of the instant invention may further be implemented to beneficially influence the chemosensitivity of a mammal with cancer, including cancers of the CNS, by vaccinating the mammal with DC prior to and/or concurrently with administration of chemotherapy.
  • the DC used in connection with various embodiments of the invention may be autologous tumor antigen-presented DC or they may be "unprimed" DC. These cells can be prepared by a host of methodologies. Additionally, further therapeutic interventions may be implemented in connection with the inventive dual therapeutic approach, such as surgical resection of a tumor, radiation therapy and the like.
  • Fig. 1 depicts tumor progression, in accordance with an embodiment of the present invention.
  • Fig. 1 A illustrates tumor progression (recurrence) intervals monitored for each group of GBM patients. Progression times were monitored over intervals spanning vaccination or chemotherapy and subsequently thereafter.
  • Fig. IB illustrates time to tumor progression in vaccine, chemotherapy and vaccine + chemotherapy groups. Tumor progression was defined as the time from first diagnosis of brain tumor (de novo GBM in all cases) to the first new scan enhancement, if verified by subsequent scans or by histology, or time from diagnosis to death due to tumor progression. Mean times to tumor progression + standard error are shown for each group over specific intervals, as indicated.
  • Fig. 2 depicts overall survival in vaccine, chemotherapy and vaccine + chemotherapy groups, in accordance with an embodiment of the present invention.
  • Overall survival was defined as the time from first diagnosis of brain tumor (de novo GBM in all cases) to death due to tumor progression. Kaplan-Meyer survival plots with censored values in open circles are shown for each group.
  • Broken line chemotherapy group; solid thin line: vaccine group; solid bold line: vaccine + chemotherapy group.
  • Fig. 3 depicts tumor regression following post- vaccine chemotherapy, in accordance with an embodiment of the present invention.
  • Relative days after diagnosis are represented by numbers under individual MRI scans, with individual patients' scans in each row.
  • Patient A recurred 82 days after vaccine initiation; patient B recurred 147 days after vaccine initiation, was treated surgically, and recurred 227 additional days (374 days total) after vaccine initiation.
  • An additional patient suffering tumor recurrence 35 days after vaccine initiation and treated with subsequent chemotherapy experienced objective tumor regression, but a complete array of images was not available for this individual.
  • Fig. 4 illustrates that CD8+ T cell receptor excision circles ("TRECs") are strongly associated with chemotherapeutic responses following vaccination, in accordance with an embodiment of the present invention.
  • TRECs CD8+ T cell receptor excision circles
  • the increase in time to tumor progression in months (y-axis) was correlated in the same GBM patients with: (A) TRECs quantified within 50,000 purified CD8+ T cells from peripheral blood mononuclear cells (“PBMC”) collected at the time of surgery, or (B) patient age.
  • PBMC peripheral blood mononuclear cells
  • Fig. 5 depicts demographic and treatment parameters of GBM patient groups, in accordance with an embodiment of the present invention. Calculations of % 2-year survival and % 3-year survival illustrated therein excluded censored values.
  • Fig. 6 depicts vaccine trial composition and distinctions, in accordance with an embodiment of the present invention. CTL responsiveness illustrated therein was determined from five testable samples per trial.
  • Fig. 7 depicts chemotherapy use, in accordance with an embodiment of the present invention.
  • Temozolomide standard dose as illustrated therein, is 150-200 mg/m 2 qd x 5 days every 28 days.
  • Gliadel wafers are a timed-release encapsulation of l,3-bis(2-chloroethyl)- l-nitosourea ("BCNU"). What is meant by "CCNU” is l-(2-chloroethyl)-3-cyclohexyl-l- nitosourea.
  • BCNU l-(2-chloroethyl)-3-cyclohexyl-l- nitosourea.
  • “Alleviating" specific cancers and/or their pathology includes degrading a tumor, for example, breaking down the structural integrity or comiective tissue of a tumor, such that the tumor size is reduced when compared to the tumor size before treatment.
  • “Alleviating” metastasis of cancer includes reducing the rate at which the cancer spreads to other organs.
  • "Beneficial results” may include, but are in no way limited to, lessening or alleviating the severity of the disease condition, preventing the disease condition from worsening, curing the disease condition and prolonging a patient's life or life expectancy.
  • the disease conditions may relate to or may be modulated by the central nervous system.
  • 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, astrocytomas, ependymal tumors, GBM and primitive neuroectodermal tumors.
  • “Chemotherapy,” as used herein, refers to the use of chemicals, such as pharmaceuticals or drugs, in the treatment of a disease condition, such as cancer.
  • “Chemotherapeutic agents” denote particular chemicals, such as pharmaceuticals or drugs, which are used to effect chemotherapy.
  • “Conditions” and “disease conditions,” as used herein, may include, but are in no way limited to any form of cancer; by way of example, astrocytomas, ependymal tumors, glioma, GBM and primitive neuroectodermal tumors.
  • “Curing” cancer includes degrading a tumor such that a tumor cannot be detected after treatment.
  • the tumor may be reduced in size or become undetectable, for example, by atrophying from lack of blood supply or by being attacked or degraded by one or more components administered according to the invention.
  • “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.
  • "Pathology" of cancer includes all phenomena that compromise the well-being of the patient.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down the targeted pathologic disease condition or disorder even if the treatment is ultimately unsuccessful.
  • a therapeutic agent e.g., a chemotherapeutic agent
  • tumor may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutics, such as radiation therapy.
  • chemotherapeutic agent e.g., a chemotherapeutic agent
  • Tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • Vaccine refers to a preparation, typically in liquid or suspension form, of treated disease-producing microorganisms or their products, or of treated cells (e.g., dentritic cells harvested from the vaccine recipient), which are used to stimulate an immune response in the body so as to confer resistance to a disease condition or another beneficial result in the recipient.
  • treated cells e.g., dentritic cells harvested from the vaccine recipient
  • the present invention is based, in part, on the surprising results obtained by the inventors in conducting a retrospective examination of the impact of therapeutic vaccination on the efficacy of conventional GBM chemotherapy. Progression rates and overall survival was compared among twelve vaccine-treated, thirteen chemotherapy-treated, and thirteen vaccine plus chemotherapy-treated de novo GBM patients. The results suggested that chemotherapy synergizes with previous therapeutic vaccination to generate a uniquely effective treatment that slows GBM progression and significantly extends patient survival relative to individual therapies. This represents the first evidence that a vaccine-based therapeutic approach may benefit a majority of cancer patients, and represents a novel treatment strategy that may substantially prolong GBM survival across a wide age range and relative to standard radiation plus chemotherapy. Additional independent evidence implicated anti-tumor T cells as influencing GBM chemosensitivity.
  • a treatment for disease conditions includes at least one therapeutic vaccination with a DC-based cancer vaccine in connection with at least one course of chemotherapy.
  • This dual treatment may be administered to a mammal to alleviate, and potentially cure, a host of disease conditions; particularly cancer, and more particularly, cancers of the brain, such as GBM.
  • the cancer vaccine used in various embodiments of the instant invention may be selected from any dendritic cell (“DC")-based cancer vaccine, and can be administered by routine methods.
  • the DC-based cancer vaccine may be administered concurrently with a course of chemotherapy and/or with an initial vaccination preceding chemotherapy.
  • the DC may be autologous tumor antigen-presented DC that are "primed" ex vivo by conventional methods; for instance, the DC may be loaded with HLA-eluted peptides from cultured tumor cells or autologous tumor lysate.
  • the DC may be delivered in an "unprimed” state and essentially primed in vivo, as described in U.S. patent application serial No. 10/251,148, filed ⁇ September 20, 2002, the disclosure of which is incorporated by reference herein in its entirety.
  • Use of "unprimed" DC may be particularly advantageous in instances where a tumor is surgically inoperable, where surgery is otherwise undesirable, or where no portion of the tumor can be retrieved for priming DC ex vivo against the tumor.
  • unprimed DC include those that do not rely upon the acquisition of rumor tissue as a protein source, and the subsequent culturing therewith.
  • DC are primed ex vivo. Priming in this manner typically involves culturing the DC with the tumor cells against which they will subsequently be utilized; thereby providing the DC access to the tumor proteins and allowing the DC to process the associated antigens in preparation for presentation of the digested antigens to T-cells upon administration to a patient.
  • DC may be delivered directly into a tumor bed or tumor region without first being primed ex vivo; the DC process the tumor antigens in vivo.
  • DC suitable for use in accordance with various embodiments of the present invention may be isolated or obtained from any tissue in which such cells are found, or may be otherwise cultured and provided.
  • antigen-presenting DC may be used in accordance with the present invention.
  • Such DC may 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 and may further be employed in conjunction with the present invention, and are included within the scope of DC used herein).
  • bone marrow may be harvested from a mammal and cultured in a medium that promotes the growth of DC.
  • GM-CSF, IL-4 and/or other cytokines, growth factors and supplements may be included in this medium.
  • clusters of DC may be harvested and/or subcultured and subsequently harvested for use in a cancer vaccine.
  • the DC-based cancer vaccine may be delivered to a recipient by any suitable delivery route, which may include, but is in no way limited to, injection, infusion, inoculation, direct surgical delivery, or any combination thereof.
  • the DC-based cancer vaccine may be administered to a mammal by direct inoculation via stereotactic surgery; a standard inoculation procedure known to those of skill in the art of neurosurgery. Moreover, the vaccine may be administered to a tumor itself, to a physiologic region in close proximity to the tumor or to a remote location within a mammal with respect to the target tumor or tumors. [0036]
  • the DC-based cancer vaccine of the present invention may include "primed" or
  • the pharmaceutical carrier is saline, although other carriers may be utilized depending upon the desired characteristics of the cancer vaccine.
  • a cancer vaccine differently in order to account for different delivery techniques for the vaccine, physiological differences among patients (e.g., sex, weight, age, etc.), or different types of tumors (e.g., brain, breast, lung, etc.), among other factors.
  • the DC-based cancer vaccine administered to a mammal in accordance with the present invention may be delivered in combination with any of a variety of additional substances and compounds; for example, any suitable carrier, vehicle, additive, excipient, pharmaceutical adjunct, or other suitable product.
  • the quantity of DC 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 may include, but are in no way limited to, the physical characteristics of the patient (e.g., age, weight, sex, etc.), the physical characteristics of the tumor (e.g., location, size, rate of growth, accessibility, etc.), and the extent to which other therapeutic methodologies (including chemotherapy, as well as beam radiation therapy) are being implemented in connection with an overall treatment regimen.
  • the physical characteristics of the patient e.g., age, weight, sex, etc.
  • the physical characteristics of the tumor e.g., location, size, rate of growth, accessibility, etc.
  • other therapeutic methodologies including chemotherapy, as well as beam radiation therapy
  • a mammal may be administered with from about 10 5 to about 10 7 DC in from about 0.05 mL to about 0.30 mL saline in a single administration, in one embodiment of the present invention. Additional administrations may be effected, depending upon the above- described and other factors, such as the severity of tumor pathology. In one embodiment of the present invention, from about one to about five administrations of about 10-40xl0 6 DC is performed at two-week intervals.
  • the chemotherapeutic agent used in connection with the present invention may be selected from any chemotherapeutic agent, as will be readily appreciated by one of skill in the art.
  • chemotherapeutic agents may include, but are in no way limited to, temozolomide, procarbazine, carboplatin, vincristine, BCNU, CCNU, thalidomide, irinotecan, isotretinoin (available from Hoffman-LaRoche, Inc.
  • chemotherapeutic agents may be administered to treat cancer in connection with various embodiments of the present invention, a wide array of combinations of chemotherapeutic agents may alternatively be administered in the treatment of cancer.
  • chemotherapeutic agents may be administered by any suitable delivery route, such as, without limitation, oral (PO), intravenous (IV), intrathecal (IT), intraarterial, intracavitary, intramuscular (IM), intralesional or topical.
  • PO oral
  • IV intravenous
  • IT intrathecal
  • I intraarterial
  • IM intramuscular
  • TM intralesional or topical.
  • kits for the treatment of cancer in a mammal.
  • the kit may be configured for cancers of the brain; for instance, for the treatment of GBM.
  • the kit is useful for practicing the inventive method of treating disease conditions.
  • the kit is an assemblage of materials or components, including at least one dose of a DC-based cancer vaccine and at least one dose of a chemotherapeutic agent. The exact nature of the components configured in the inventive kit depends on the particular DC-based cancer vaccine and chemotherapeutic regimens that are to be implemented.
  • the inventive kit may include components that can be used in connection with priming a quantity of DC ex vivo. It should be readily apparent that, in embodiments of the present invention wherein the DC- based cancer vaccine is to include only “unprimed” DC, components used solely for priming the DC would not be necessary in the kit.
  • Instructions for use may be included with the kit.
  • “Instructions for use” typically include a tangible expression describing the components of the kit and the treatment schedule, dosing and directions for administration of the DC-based cancer vaccine and chemotherapeutic agent or agents.
  • the instructions for use may describe the harvesting of DC from a mammal and/or a procedure to be implemented for priming the DC ex vivo.
  • the kit may also contain other useful components, such as diluents, buffers, pharmaceutically acceptable carriers, specimen containers, syringes, stents, catheters, pipetting or measuring tools, and the like.
  • the materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable manner that preserves their operability and utility.
  • 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).
  • packaging material refers to one or more physical structures used to house the contents of the kit.
  • 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 the field.
  • 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.
  • a package can be a glass vial used to contain suitable quantities of a composition containing a chemotherapeutic agent.
  • the packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components.
  • vaccinated (“vaccine” or “vaccine + chemotherapy”) patients received at least three vaccinations with autologous tumor antigen-pulsed DC, starting approximately fifteen weeks post-surgery and five weeks post-radiation therapy, respectively.
  • Patients receiving chemotherapy alone (“chemotherapy” patients) were treated (with surgery, radiation and chemotherapy) over the same time interval as vaccinated patients, as depicted in Fig. 7.
  • Serial MRI scans were performed every 2-3 months in all patients. Tumor progression and overall survival among vaccine, chemotherapy and vaccine + chemotherapy groups were determined and compared, as illustrated in Figs. 1 and 2.
  • Example 2 Effect of Vaccination on Chemotherapeutic Treatment of GBM Tumors
  • the inventors examined whether vaccination could alter GBM sensitivity to subsequent chemotherapy.
  • GBM patients receiving chemotherapy after vaccination enjoyed significantly prolonged tumor progression relative to those receiving vaccination or chemotherapy alone.
  • GBM patients receiving chemotherapy after vaccination exhibited significantly prolonged survival relative to those receiving either treatment individually.
  • Example 3 CD8+ TREC Correlation with Increased Tumor Progression Time
  • Thymic production of CD8+ T cells is accurately reflected by the concentration of TRECs in purified T cells. See, e.g., D.C. Douek et al, "Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution," The Lancet, 355:1875 (2000); D.C. Douek et al, “Changes in thymic function with age and during the treatment of HIV infection," Nature, 396:690 (1998); and B.D.
  • thymic production of CD8+ T cells accounts for age-dependent glioma prognosis and outcome and predominantly influences vaccine-induced anti-tumor responses in GBM patients.
  • Thymic CD8+ T cell production strongly influences tumor antigen recognition and age-dependent glioma mortality
  • J. Immunol, 171(9):4927 (2003) The inventors surmised that a direct influence of anti-tumor immunity on GBM chemosensitivity, itself an age- dependent phenomenon (See Fine et al. at 2585), would be reflected by a dominant relationship between CD8+ TRECs and chemotherapeutic responsiveness within the same GBM patients.
  • TREC content within purified CD8+ T cells dominantly correlated with the increase in tumor recurrence times following post- vaccine chemotherapy.
  • This relationship was not simply a function of an independent influence of age on chemosensitivity and thymic production of CD8+ T cells, because the strength and significance of this correlation surpassed that between increased recurrence times following post-vaccine chemotherapy and patient age (Fig. 4).
  • the close relationship between thymus products and glioma outcome is a direct result of CD 8+ T cell production and/or function. See Wheeler et al. at 4927.
  • CD8+ TRECs the dominant relationship between CD8+ TRECs and prolonged progression times following post- vaccine chemotherapy suggests that clinical responsiveness to chemotherapy is similarly impacted by production and/or function of newly emigrated CD8+ T cells. Because levels of such T cells were shown to predominantly mediate anti-tumor immune responsiveness following vaccination of GBM patients (Id.), this constitutes independent validation of the notion that anti- tumor immunity impacts GBM chemosensitivity.
  • Vaccinated patients were steroid-free during blood collection and vaccinations were administered as described in Yu at 842. Patients received three vaccines, two weeks apart, of 10-40x10 autologous DC loaded with either HLA7 eluted peptides from cultured tumor cells or 150 ⁇ g/ml autologous tumor freeze-thaw lysate, starting approximately fifteen weeks post- surgery. A fourth identical vaccination followed six weeks later only in phase II trial patients (12 of 25). Serial MRI scans were performed every 2 to 3 months for all patients.
  • Example 5 Cell Isolation and Lysis [0055] PBMC were prepared with Ficoll from patients' blood obtained at the time of surgery and/or from banked leukaphereses.
  • CD4+ and CD8+ T cells were purified from PBMC using MACS bead separation (obtained from Miltenyi Biotec; Auburn, CA). 10 7 CD4+ or CD8+ cells/ml were prepared for quantitative real-time PCR ("qPCR") by lysis in 100 ⁇ g/mL proteinase K (obtained fro Boehringer; Indianapolis, IN) 1 hr, 56°C, with inactivation at 95°C, lO min.
  • qPCR quantitative real-time PCR
  • TRECs were quantified in duplicate or triplicate by qPCR using the 5 'nuclease (TaqMan) method as described in D.C. Douek et al., "Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution," The Lancet, 355:1875 (2000), and detected on an iCycler system (obtained from BioRad; Hercules, CA). qPCR was performed on 5 ⁇ L cell lysate (from 50,000 cells) with the primers depicted in Table 1 (probe was obtained from MegaBases; Chicago, IL).
  • PCR reactions including 0.5 U Platinum Taq (obtained from Gibco; Grand Island, NY), 3.5 mM MgCl 2 , 0.2 mM dNTPs, 500 nM of each primer, 150 nM probe, were amplified at 95°C for 5 minutes, 95°C for 30 seconds, and 60°C for 1 minute for 45 cycles. Control ⁇ -actin reactions were performed to ensure nucleic acid content and negative samples were excluded from further analysis. TREC values were adjusted for T cell purity.
  • Example 5 Statistical analyses included 2-tailed Mann- Whitney log-rank tests for disease- free and overall survival, binomial distribution probability, and Pearson's correlation coefficients (r values) calculated with SAS and Excel software. Binomial distributions were determined for 2-year and 3-year survival frequencies between vaccine + chemotherapy and other (vaccine or chemotherapy) patient groups.

Abstract

The invention relates to the treatment of cancer, and particularly to the treatment of cancers of the central nervous system, such as glioblastoma multiforme. A dual therapeutic approach is provided, including the administration of a dendritic cell-based cancer vaccine and a regimen of chemotherapy. The two therapies may be administered concurrently with one another and/or with an initial vaccination preceding chemotherapy. In various embodiments, the dendritic cell-based cancer vaccine includes either primed or unprimed dendritic cells; for instance, the dendritic cells may be autologous tumor antigen-presented dendritic cells. The dual therapeutic approach of the instant invention beneficially influences the chemosensitivity of a mammal with cancer.

Description

SYSTEMANDMETHODFORTHE TREATMENT OFCANCER,INCLUDING CANCERS OFTHECENTRALNERVOUS SYSTEM
BACKGROUND OFTHEINVENTION 1. Field of the Invention
[0001] The invention relates to the treatment of cancer, and in particular embodiments to cancers of the central nervous system ("CNS"), such as those found in the brain.
2. Description of Related Art [0002] Malignant brain tumors are among the gravest forms of cancer. The most common of these incurable tumors, ghoblastoma multiforme ("GBM"), is responsible for 50% of all intracranial gliomas and 25% of intracranial tumors in adults. See, e.g., L.M. DeAngelis, "Medical progress: Brain tumors," N. Engl J. Med., 344:114 (2001); and F.G. Davis et al, "Prevalence estimates for primary brain tumors in the United States by behavior and major histology groups," Neuro-oncol. 3:152 (2001). GBM diagnosis carries with it an average survival between twelve and eighteen months (with 90-95% patients surviving less than two years), without' the possibility of spontaneous remission or effective treatment. See, e.g., Id; and W.JJ. Curran et al, "Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials," J. Natl Cancer Inst., 85:690 (1993). The consistently short survival and absence of spontaneous remission that makes GBM such a devastating disease also renders the evaluation of new therapies for this disease relatively rapid and unequivocal. Overall survival represents the standard by which therapies for GBM are evaluated, in part because tumor mass reduction (i.e., surgically) does not necessarily correlate with prolonged survival. See, e.g., F.W. Kreth et al, "Surgical resection and radiation therapy versus biopsy and radiation therapy in the treatment of ghoblastoma multiforme," J. Neurosurg. , 78:762 (1993); M.R. Quigley et al, "Value of surgical intervention in the treatment of glioma," Stereotact. Fund. Neurosurg., 65:171 (1995); and SJ. Hentschel et al, "Current surgical management of ghoblastoma," Cancer J., 9:113 (2003). [0003] Unfortunately, conventional therapies are remarkably ineffective at improving GBM clinical outcome, despite their ability to confer significant benefits to patients with non- glioma tumors. See, e.g., Curran at 690; J.F. Reavey-Cantwell et al, "The prognostic value of tumor markers in patients with ghoblastoma multiforme: analysis of 32 patients and review of the literature," J. Neurooncol, 55:195 (2001); and R. Stupp et al, "Recent developments in the management of malignant glioma," J: Clin. Oncol, 1091-9118:779 (2001). Even the few treatments effective against GBM typically either exhibit small increases in survival that are evident only from large population studies, or primarily benefit certain (i.e., young) patient subpopulations. See, e.g., H.A. Fine et al, "Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults," Cancer, 71 :2585 (1993); and S. Diete et al, "Sex differences in length of survival with malignant astrocytoma, but not with ghoblastoma," J. Neurooncol, 53:47 (2001). Thus, there exists a need in the art for a novel therapy for GBM. [0004] Cancer vaccines represent one such therapy for GBM. See, e.g., R.P. Glick et al, "Intracerebral versus subcutaneous immunization with allogeneic fibroblasts genetically engineered to secrete interleukin-2 in the treatment of central nervous system glioma and melanoma," Neurosurg., 41:898 (1997); L.M. Liau et al, "Treatment of intracranial gliomas with bone marrow-derived dendritic cells pulsed with tumor antigens," J. Neurosurg., 90:1115 (1999); and J.S. Yu et al, "Vaccination of malignant glioma patients with peptide-pulsed DC elicits systemic cytotoxicity and intracranial T-cell infiltration," Cancer Res., 61 :842 (2001). The clinical efficacy of therapeutic vaccination for any human tumor, however, remains controversial because consistent tumor destruction or extended lifespan is not observed in most vaccinated cancer patients. See, e.g., S.A. Rosenberg et al, "hnmunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma," Nat. Med., 4:321 (1998); K.H. Lee et al, "Increased vaccine-specific T cell frequency after peptide based vaccination correlates with increased susceptibility to in vitro stimulation but does not lead to tumor regression," J. Immunol, 163:6292 (1999); and L. Fong et al, "Altered peptide ligand vaccination with Flt3 ligand expanded DC for tumor immunotherapy," Proc. Natl. Acad. Sci. USA, 98:8809 (2001). In contrast, current cancer vaccines do reliably elicit tumor-reactive cytotoxic T lymphocytes ("CTL") in most patients. See, e.g., Rosenberg at 321; Lee at 6292; and B. Bodey et al, "Failure of cancer vaccines: the significant limitations of this approach to immunotherapy," Anticancer Res., 20:2665 (2000). The reasons underlying the general clinical failure of cancer vaccines are unknown, but one possibility is that the kinetics of anti-tumor CTL killing in cancer patients may be too inefficient to keep pace with rapidly growing, mutating tumors in situ. Consistent with this notion, it was previously reported that therapeutic vaccination with autologous tumor antigen-pulsed DC is sufficient to enhance peripheral tumor-reactive CTL activity and CD8+ T cell infiltration into tumors in situ in GBM patients. See Yu et al at 842. Nevertheless, improvements in overall patient survival were not apparent in this initial study.
[0005] Because CTL induce death in their cellular targets, it is not unreasonable to expect that inefficient CTL killing might either incompletely trigger death pathways in targeted tumor cells, or select for CTL-resistant tumor variants. In the first case, vaccine-elicited tumor- responsive CTL might fundamentally alter tumors by "priming" their death machinery. In the second case, such CTL could fundamentally alter tumor cell physiology and/or genetics. Both of these possibilities could, in theory, be exploited by additional therapeutic modalities. Therefore, the clinical insufficiency of cancer vaccines encourages the examination of synergy between vaccination and other therapies, particularly to the extent that such an examination might uncover a novel approach to cancer therapy.
BRIEF SUMMARY OF THE INVENTION
[0006] The invention relates to a novel treatment for cancer, and particularly cancers of the CNS; for example, cancers of the brain, such as GBM. In one embodiment, the invention includes a dual therapeutic approach to the treatment of cancer with at least one vaccination of DC and at least one course or regimen of chemotherapy. The two therapies may be administered concurrently with one another and/or with an initial vaccination preceding chemotherapy. [0007] The dual therapeutic approach of the instant invention may further be implemented to beneficially influence the chemosensitivity of a mammal with cancer, including cancers of the CNS, by vaccinating the mammal with DC prior to and/or concurrently with administration of chemotherapy.
[0008] The DC used in connection with various embodiments of the invention may be autologous tumor antigen-presented DC or they may be "unprimed" DC. These cells can be prepared by a host of methodologies. Additionally, further therapeutic interventions may be implemented in connection with the inventive dual therapeutic approach, such as surgical resection of a tumor, radiation therapy and the like.
BRIEF DESCRIPTION OF THE DRAWINGS
[0009] Fig. 1 depicts tumor progression, in accordance with an embodiment of the present invention. Fig. 1 A illustrates tumor progression (recurrence) intervals monitored for each group of GBM patients. Progression times were monitored over intervals spanning vaccination or chemotherapy and subsequently thereafter. Fig. IB illustrates time to tumor progression in vaccine, chemotherapy and vaccine + chemotherapy groups. Tumor progression was defined as the time from first diagnosis of brain tumor (de novo GBM in all cases) to the first new scan enhancement, if verified by subsequent scans or by histology, or time from diagnosis to death due to tumor progression. Mean times to tumor progression + standard error are shown for each group over specific intervals, as indicated. Significance (p values) were derived from double-sided paired (initial recurrence after vaccine vs. subsequent recurrence after chemotherapy in vaccine + chemotherapy group) or unpaired double-sided T tests (all other comparisons). Initial recurrence times were identical among all three groups (P > 0.6). The small difference in subsequent recurrence times between vaccine and chemotherapy groups was not statistically significant (P = 0.07).
[0010] Fig. 2 depicts overall survival in vaccine, chemotherapy and vaccine + chemotherapy groups, in accordance with an embodiment of the present invention. Overall survival was defined as the time from first diagnosis of brain tumor (de novo GBM in all cases) to death due to tumor progression. Kaplan-Meyer survival plots with censored values in open circles are shown for each group. Broken line: chemotherapy group; solid thin line: vaccine group; solid bold line: vaccine + chemotherapy group. Survival of the vaccine group was identical to that of chemotherapy group (p = 0.7, log-rank). Survival of vaccine + chemotherapy group was significantly greater relative to both survival in the other two groups (p = 0.047, log- rank), and greater than survival in the chemotherapy group alone (p = 0.02, log-rank). Survival of the vaccine group tended to be lower but was not statistically different than that of the vaccine + chemotherapy group (p = 0.05, log-rank). [0011] Fig. 3 depicts tumor regression following post- vaccine chemotherapy, in accordance with an embodiment of the present invention. Relative days after diagnosis are represented by numbers under individual MRI scans, with individual patients' scans in each row. Patient A recurred 82 days after vaccine initiation; patient B recurred 147 days after vaccine initiation, was treated surgically, and recurred 227 additional days (374 days total) after vaccine initiation. An additional patient suffering tumor recurrence 35 days after vaccine initiation and treated with subsequent chemotherapy experienced objective tumor regression, but a complete array of images was not available for this individual. All scans except the pre-resection scan for patient B were performed post-contrast enhancement with gadolinium. Two of the three patients exhibiting objective tumor regression survived more than two years (730 days) post-diagnosis. [0012] Fig. 4 illustrates that CD8+ T cell receptor excision circles ("TRECs") are strongly associated with chemotherapeutic responses following vaccination, in accordance with an embodiment of the present invention. The increase in time to tumor progression in months (y-axis) was correlated in the same GBM patients with: (A) TRECs quantified within 50,000 purified CD8+ T cells from peripheral blood mononuclear cells ("PBMC") collected at the time of surgery, or (B) patient age. Data were derived from all vaccinated GBM patients for whom chemotherapeutic response and TREC results (n = 13) or age (n = 17) were available. A related parameter, time to tumor progression after chemotherapy divided by time to tumor progression after vaccination, also correlated significantly with CD8+ TRECs (r = 0.73; P < 0.01), but failed to do so with patient age (r = -0.4O; P > 0.05). [0013] Fig. 5 depicts demographic and treatment parameters of GBM patient groups, in accordance with an embodiment of the present invention. Calculations of % 2-year survival and % 3-year survival illustrated therein excluded censored values. [0014] Fig. 6 depicts vaccine trial composition and distinctions, in accordance with an embodiment of the present invention. CTL responsiveness illustrated therein was determined from five testable samples per trial.
[0015] Fig. 7 depicts chemotherapy use, in accordance with an embodiment of the present invention. Temozolomide standard dose, as illustrated therein, is 150-200 mg/m2 qd x 5 days every 28 days. Gliadel wafers are a timed-release encapsulation of l,3-bis(2-chloroethyl)- l-nitosourea ("BCNU"). What is meant by "CCNU" is l-(2-chloroethyl)-3-cyclohexyl-l- nitosourea. DETAILED DESCRIPTION OF THE INVENTION
[0016] All references cited herein are incorporated by reference as if fully set forth.
[0017] Unless defined otherwise, 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. Singleton et al, Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, NY 1994); March, Advanced Organic Chemistry Reactions,
Mechanisms and Structure 4th ed., J. Wiley & Sons (New York, NY 1992); and Sambrook and Russel, Molecular Cloning: A Laboratory Manual 3rd ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, NY 2001) provide one skilled in the art with a general guide to many of the terms used in the present application. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below. [0018] "Alleviating" specific cancers and/or their pathology includes degrading a tumor, for example, breaking down the structural integrity or comiective tissue of a tumor, such that the tumor size is reduced when compared to the tumor size before treatment. "Alleviating" metastasis of cancer includes reducing the rate at which the cancer spreads to other organs. [0019] "Beneficial results" may include, but are in no way limited to, lessening or alleviating the severity of the disease condition, preventing the disease condition from worsening, curing the disease condition and prolonging a patient's life or life expectancy. The disease conditions may relate to or may be modulated by the central nervous system. [0020] "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, astrocytomas, ependymal tumors, GBM and primitive neuroectodermal tumors.
[0021] "Chemotherapy," as used herein, refers to the use of chemicals, such as pharmaceuticals or drugs, in the treatment of a disease condition, such as cancer. [0022] "Chemotherapeutic agents" denote particular chemicals, such as pharmaceuticals or drugs, which are used to effect chemotherapy. [0023] "Conditions" and "disease conditions," as used herein, may include, but are in no way limited to any form of cancer; by way of example, astrocytomas, ependymal tumors, glioma, GBM and primitive neuroectodermal tumors.
[0024] "Curing" cancer includes degrading a tumor such that a tumor cannot be detected after treatment. The tumor may be reduced in size or become undetectable, for example, by atrophying from lack of blood supply or by being attacked or degraded by one or more components administered according to the invention.
[0025] "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. [0026] "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. [0027] "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 the targeted pathologic disease condition or disorder even if the treatment is ultimately unsuccessful. Those in need of treatment include those already with the disease condition as well as those prone to have or develop the disease condition or those in whom the disease condition is to be prevented. In tumor (i.e., cancer) treatment, a therapeutic agent (e.g., a chemotherapeutic agent) may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutics, such as radiation therapy. [0028] "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. [0029] "Vaccine," as used herein (particularly with reference to "cancer vaccines"), refers to a preparation, typically in liquid or suspension form, of treated disease-producing microorganisms or their products, or of treated cells (e.g., dentritic cells harvested from the vaccine recipient), which are used to stimulate an immune response in the body so as to confer resistance to a disease condition or another beneficial result in the recipient.
[0030] The present invention is based, in part, on the surprising results obtained by the inventors in conducting a retrospective examination of the impact of therapeutic vaccination on the efficacy of conventional GBM chemotherapy. Progression rates and overall survival was compared among twelve vaccine-treated, thirteen chemotherapy-treated, and thirteen vaccine plus chemotherapy-treated de novo GBM patients. The results suggested that chemotherapy synergizes with previous therapeutic vaccination to generate a uniquely effective treatment that slows GBM progression and significantly extends patient survival relative to individual therapies. This represents the first evidence that a vaccine-based therapeutic approach may benefit a majority of cancer patients, and represents a novel treatment strategy that may substantially prolong GBM survival across a wide age range and relative to standard radiation plus chemotherapy. Additional independent evidence implicated anti-tumor T cells as influencing GBM chemosensitivity. [0031] In accordance with an embodiment of the instant invention, a treatment for disease conditions, such as cancer, includes at least one therapeutic vaccination with a DC-based cancer vaccine in connection with at least one course of chemotherapy. This dual treatment may be administered to a mammal to alleviate, and potentially cure, a host of disease conditions; particularly cancer, and more particularly, cancers of the brain, such as GBM. [0032] The cancer vaccine used in various embodiments of the instant invention may be selected from any dendritic cell ("DC")-based cancer vaccine, and can be administered by routine methods. The DC-based cancer vaccine may be administered concurrently with a course of chemotherapy and/or with an initial vaccination preceding chemotherapy. The DC may be autologous tumor antigen-presented DC that are "primed" ex vivo by conventional methods; for instance, the DC may be loaded with HLA-eluted peptides from cultured tumor cells or autologous tumor lysate. Alternatively, the DC may be delivered in an "unprimed" state and essentially primed in vivo, as described in U.S. patent application serial No. 10/251,148, filed { September 20, 2002, the disclosure of which is incorporated by reference herein in its entirety. Use of "unprimed" DC may be particularly advantageous in instances where a tumor is surgically inoperable, where surgery is otherwise undesirable, or where no portion of the tumor can be retrieved for priming DC ex vivo against the tumor. [0033] Briefly, "unprimed" DC include those that do not rely upon the acquisition of rumor tissue as a protein source, and the subsequent culturing therewith. In conventional methods, DC are primed ex vivo. Priming in this manner typically involves culturing the DC with the tumor cells against which they will subsequently be utilized; thereby providing the DC access to the tumor proteins and allowing the DC to process the associated antigens in preparation for presentation of the digested antigens to T-cells upon administration to a patient. However, in various embodiments of the present invention, DC may be delivered directly into a tumor bed or tumor region without first being primed ex vivo; the DC process the tumor antigens in vivo.
[0034] DC suitable for use in accordance with various embodiments of the present invention may be isolated or obtained from any tissue in which such cells are found, or may be otherwise cultured and provided. In particular, antigen-presenting DC may be used in accordance with the present invention. Such DC may 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 and may further be employed in conjunction with the present invention, and are included within the scope of DC used herein). For instance, in one embodiment of the present invention, bone marrow may be harvested from a mammal and cultured in a medium that promotes the growth of DC. GM-CSF, IL-4 and/or other cytokines, growth factors and supplements may be included in this medium. After a suitable amount of time, clusters of DC may be harvested and/or subcultured and subsequently harvested for use in a cancer vaccine. [0035] The DC-based cancer vaccine may be delivered to a recipient by any suitable delivery route, which may include, but is in no way limited to, injection, infusion, inoculation, direct surgical delivery, or any combination thereof. In one embodiment of the present invention, the DC-based cancer vaccine may be administered to a mammal by direct inoculation via stereotactic surgery; a standard inoculation procedure known to those of skill in the art of neurosurgery. Moreover, the vaccine may be administered to a tumor itself, to a physiologic region in close proximity to the tumor or to a remote location within a mammal with respect to the target tumor or tumors. [0036] The DC-based cancer vaccine of the present invention may include "primed" or
"unprimed" DC in a pharmaceutical carrier. Any conventional pharmaceutical carrier may be used in accordance with the present invention, and an appropriate carrier may be selected by one of skill in the art by routine techniques. In one embodiment of the present invention, the pharmaceutical carrier is saline, although other carriers may be utilized depending upon the desired characteristics of the cancer vaccine. For example, one may formulate a cancer vaccine differently in order to account for different delivery techniques for the vaccine, physiological differences among patients (e.g., sex, weight, age, etc.), or different types of tumors (e.g., brain, breast, lung, etc.), among other factors. The DC-based cancer vaccine administered to a mammal in accordance with the present invention may be delivered in combination with any of a variety of additional substances and compounds; for example, any suitable carrier, vehicle, additive, excipient, pharmaceutical adjunct, or other suitable product.
[0037] The quantity of DC 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 may include, but are in no way limited to, the physical characteristics of the patient (e.g., age, weight, sex, etc.), the physical characteristics of the tumor (e.g., location, size, rate of growth, accessibility, etc.), and the extent to which other therapeutic methodologies (including chemotherapy, as well as 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 may be administered with from about 105 to about 107 DC in from about 0.05 mL to about 0.30 mL saline in a single administration, in one embodiment of the present invention. Additional administrations may be effected, depending upon the above- described and other factors, such as the severity of tumor pathology. In one embodiment of the present invention, from about one to about five administrations of about 10-40xl06 DC is performed at two-week intervals.
[0038] The chemotherapeutic agent used in connection with the present invention may be selected from any chemotherapeutic agent, as will be readily appreciated by one of skill in the art. Examples of such chemotherapeutic agents may include, but are in no way limited to, temozolomide, procarbazine, carboplatin, vincristine, BCNU, CCNU, thalidomide, irinotecan, isotretinoin (available from Hoffman-LaRoche, Inc. under the tradename Accutane®), imatinib (available from Novartis Pharmaceuticals Corporation under the tradename Gleevec®), etoposide, cisplatin, daunorubicin, doxorubicin, methotrexate, mercaptopurine, fluorouracil, hydroxyurea, vinblastine and pacitaxel. It will also be readily appreciated by those of skill in the art that, while a single chemotherapeutic agent may be administered to treat cancer in connection with various embodiments of the present invention, a wide array of combinations of chemotherapeutic agents may alternatively be administered in the treatment of cancer. Moreover, chemotherapeutic agents may be administered by any suitable delivery route, such as, without limitation, oral (PO), intravenous (IV), intrathecal (IT), intraarterial, intracavitary, intramuscular (IM), intralesional or topical.
[0039] In a further embodiment of the present invention, a kit is provided for the treatment of cancer in a mammal. In one embodiment, the kit may be configured for cancers of the brain; for instance, for the treatment of GBM. The kit is useful for practicing the inventive method of treating disease conditions. The kit is an assemblage of materials or components, including at least one dose of a DC-based cancer vaccine and at least one dose of a chemotherapeutic agent. The exact nature of the components configured in the inventive kit depends on the particular DC-based cancer vaccine and chemotherapeutic regimens that are to be implemented. For instance, if the DC-based cancer vaccine used in accordance with a particular embodiment of the present invention is to include "primed" DC, then the inventive kit may include components that can be used in connection with priming a quantity of DC ex vivo. It should be readily apparent that, in embodiments of the present invention wherein the DC- based cancer vaccine is to include only "unprimed" DC, components used solely for priming the DC would not be necessary in the kit.
[0040] Instructions for use may be included with the kit. "Instructions for use" typically include a tangible expression describing the components of the kit and the treatment schedule, dosing and directions for administration of the DC-based cancer vaccine and chemotherapeutic agent or agents. In various embodiments, the instructions for use may describe the harvesting of DC from a mammal and/or a procedure to be implemented for priming the DC ex vivo. The kit may also contain other useful components, such as diluents, buffers, pharmaceutically acceptable carriers, specimen containers, syringes, stents, catheters, pipetting or measuring tools, and the like. The materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable manner that preserves 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. [0041] 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. 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 the field. 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 a composition containing a chemotherapeutic agent. The packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components. Example 1 Comparison Among Differentially Treated Patients [0042] Newly-diagnosed de novo GBM patients (GBM did not arise from malignant progression of initially lower- grade gliomas) were enrolled into one of three vaccine studies or were administered chemotherapy alone, after surgical tumor resection and standard radiation therapy. As indicated in Figs. 5 and 6, vaccinated ("vaccine" or "vaccine + chemotherapy") patients received at least three vaccinations with autologous tumor antigen-pulsed DC, starting approximately fifteen weeks post-surgery and five weeks post-radiation therapy, respectively. Patients receiving chemotherapy alone ("chemotherapy" patients) were treated (with surgery, radiation and chemotherapy) over the same time interval as vaccinated patients, as depicted in Fig. 7. Serial MRI scans were performed every 2-3 months in all patients. Tumor progression and overall survival among vaccine, chemotherapy and vaccine + chemotherapy groups were determined and compared, as illustrated in Figs. 1 and 2.
[0043] Treatment and demographic parameters, including the extent of non-survivors, surgery before or after vaccination (where appropriate), chemotherapy before vaccination (where appropriate), gender and age were not significantly different among the relevant groups, as shown in Fig. 5. In addition, Karnofsky perfonnance status ("KPS") was not significantly different between vaccine and vaccine + chemotherapy patient groups (Fig. 5). Inclusion criteria for vaccinated de novo GBM patients, the patient subgroup common among the three vaccine trials, were identical in the three vaccine trials (Fig. 6). Similarly, anti-tumor immune response rates were similar for the three vaccine trials (Fig. 6), suggesting that differences in antigen source and/or vaccine dosing among individual vaccine trials did not substantially impact their immunological efficacy. Moreover, vaccine, vaccine + chemotherapy and chemotherapy patients exhibited identical recurrence times following vaccination (Fig. 1), indicating a lack of inherent bias in tumor clinical behavior among all three groups after initial treatment. [0044] Only the extent of surgical resection differed significantly between the patient groups, with all vaccinated patients receiving image-complete resections and a portion of chemotherapy patients receiving partial resections. This particular bias was expected to produce longer survival selectively in vaccine relative to chemotherapy groups. Surprisingly, and quite to the contrary, mean times to progression and overall survival of chemotherapy patients was similar to those in previous reports (See, e.g., Stupp et al. at 779) and did not differ significantly from those of vaccinated patients (Figs. 1, 2 & 6). Thus, therapeutic vaccination by itself failed to significantly slow progression or prolong survival relative to conventional GBM chemotherapy (P = 0.7, log-rank) . These results suggested that anti-tumor immunity was either inefficiently induced or that GBM tumors were inherently resistant to vaccine-elicited immune destruction alone. Because efficient induction of anti-tumor CTL responses by this vaccine methodology was previously demonstrated (See Yu et al. at 842), the latter possibility appeared most likely.
Example 2 Effect of Vaccination on Chemotherapeutic Treatment of GBM Tumors [0045] To determine if vaccination was capable of eliciting more subtle affects on GBM tumors, the inventors examined whether vaccination could alter GBM sensitivity to subsequent chemotherapy. GBM patients receiving chemotherapy after vaccination enjoyed significantly prolonged tumor progression relative to those receiving vaccination or chemotherapy alone. Similarly, GBM patients receiving chemotherapy after vaccination exhibited significantly prolonged survival relative to those receiving either treatment individually. The possibility that an inadvertent selection bias resulted in inherently slower progressing tumors in patients receiving chemotherapy after vaccination is inconsistent with the statistically identical initial progression times (i.e., after initial vaccination or chemotherapy) among all three groups of patients ("vaccine", "chemotherapy" and "vaccine + chemotherapy"): tumors behaved the same, clinically, regardless of initial vaccination or chemotherapy, and slowed tumor progression was unique to the period of chemotherapy after vaccination, hi addition, the two groups of vaccinated patients were statistically identical in terms of all other treatment parameters, including number of craniotomies, radiation, SRS and chemotherapy prior to vaccination, and exhibited similar Karnofsky performance scores after vaccine therapy. [0046] Importantly, 2-, 3- and 4-year survival was also unique for patients receiving chemotherapy after vaccination. Whereas chemotherapy or vaccination alone resulted in 2-year survival within the established range for GBM (8%; Fig. 5), post-vaccination chemotherapy resulted in a substantial increase (42%; Fig. 5) in 2-year survivors (P < 0.05; binomial distribution). Similarly, no 3-year or 4-year survivors were evident after chemotherapy or vaccination alone, but such survivors persisted in post-vaccine chemotherapy patients (P < 0.01 for 3-year survivors; binomial distribution).
[0047] Finally, objective (> 50%) regression of the entire tumor mass was observed in three of thirteen vaccine + chemotherapy patients and this occurred only after initiation of post- vaccine chemotherapy. A similar regression was also observed in a single grade III malignant glioma patient receiving chemotherapy after vaccination (data not shown). Such dramatic regression of de novo GBM was unique to this group and is unknown in the literature, although a single example of partial regression in a recurrent GBM following post-vaccine chemotherapy has recently been reported. See H. Okada et al, "Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of recurrent ghoblastoma: preliminary observations in a patient with a favorable response to therapy," J. Neuro-oncol, 64:13 (2003). In that report, however, rapid tumor recurrence, as judged by increased tumor on imaging studies, clearly occurred following vaccination, but was discounted to favor the notion that regression was related to vaccination itself. In this context, it is significant that tumor recurrence in all vaccinated patients in the current study was determined by increased tumor imaging on MRI scans. Moreover, 33% (4/12) of vaccine patients and 46% (6/13) of vaccine + chemotherapy patients were biopsied upon observation of post-vaccine increases in tumor imaging, with all exhibiting histologically verified recurrent tumor. This suggests that apparent increases in tumor imaging in our study were not due to vaccine-induced inflammatory responses and instead generally reflected bona fide tumor recurrence. This suggests that the specific therapeutic regimen of chemotherapy after vaccination, rather than vaccination alone, elicited the tumor regressions. In any case, this is the first demonstration of objective regression of entire tumor mass in any adoptive immunotherapy setting, as well as in the treatment of GBM generally. [0048] The inventors' studies support the notion that clinical outcome is significantly improved by the specific combination and sequence of vaccination plus chemotherapy in GBM patients. Stated more generally, it is believed that anti-tumor immunity directly impacts GBM chemosensitivity. Vaccinated patients receiving subsequent chemotherapy exhibited significantly delayed tumor progression and longer survival relative to those receiving vaccinations without subsequent chemotherapy or to those receiving chemotherapy alone. Improved clinical outcome appeared dependent on the specific combination and sequence of therapeutic vaccination followed by chemotherapy. These observations suggest a substantial therapeutic slowing of GBM progression and extension of overall survival for GBM patients. These clinical benefits appeared to markedly surpass those in previous vaccine studies as well as those in even the most hopeful analyses of GBM chemotherapy. See, e.g., Stupp et al at 779. Moreover, the more favorable clinical outcome conferred by post- vaccine chemotherapy did not appear to be confined to younger subgroups of patients. As such, this supports the notion that this specific treatment combination conferred clinical improvement to a majority of treated cancer patients, which is believed to be unique for a vaccine-based therapy.
Example 3 CD8+ TREC Correlation with Increased Tumor Progression Time [0049] Thymic production of CD8+ T cells is accurately reflected by the concentration of TRECs in purified T cells. See, e.g., D.C. Douek et al, "Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution," The Lancet, 355:1875 (2000); D.C. Douek et al, "Changes in thymic function with age and during the treatment of HIV infection," Nature, 396:690 (1998); and B.D. Jamieson et al, "Generation of functional thymocytes in the human adult," Immunity, 10:569 (1999). Moreover, thymic production of CD8+ T cells accounts for age-dependent glioma prognosis and outcome and predominantly influences vaccine-induced anti-tumor responses in GBM patients. See CJ. Wheeler et al, "Thymic CD8+ T cell production strongly influences tumor antigen recognition and age-dependent glioma mortality," J. Immunol, 171(9):4927 (2003). The inventors surmised that a direct influence of anti-tumor immunity on GBM chemosensitivity, itself an age- dependent phenomenon (See Fine et al. at 2585), would be reflected by a dominant relationship between CD8+ TRECs and chemotherapeutic responsiveness within the same GBM patients.
Accordingly, TREC content within purified CD8+ T cells dominantly correlated with the increase in tumor recurrence times following post- vaccine chemotherapy. This relationship was not simply a function of an independent influence of age on chemosensitivity and thymic production of CD8+ T cells, because the strength and significance of this correlation surpassed that between increased recurrence times following post-vaccine chemotherapy and patient age (Fig. 4). [0050] The close relationship between thymus products and glioma outcome is a direct result of CD 8+ T cell production and/or function. See Wheeler et al. at 4927. Thus, the dominant relationship between CD8+ TRECs and prolonged progression times following post- vaccine chemotherapy suggests that clinical responsiveness to chemotherapy is similarly impacted by production and/or function of newly emigrated CD8+ T cells. Because levels of such T cells were shown to predominantly mediate anti-tumor immune responsiveness following vaccination of GBM patients (Id.), this constitutes independent validation of the notion that anti- tumor immunity impacts GBM chemosensitivity.
[0051] Taken together, although not wishing to be bound by any particular theory, these findings suggest that GBM tumors are recognized and acted on in situ by cellular immune components. Overall, such activity may result in fundamental alteration of GBM tumors that renders them increasingly susceptible to DNA damaging chemotherapy, despite the inability of vaccination by itself to confer overt clinical benefits to patients.
[0052] Furthermore, both clinical outcome and chemotherapeutic responsiveness are known to be age-dependent processes in gliomas generally. See, e.g., W.J.J. Curran at 690; Fine at 2585; and K.L. Chandler et al, "Long-term survival in patients with ghoblastoma multiforme/'Newrøs'urg-., 32:716 (1993). Age-dependent glioma clinical outcome is critically impacted by the production of CD8+ T cells in the thymus in mice and a directly related parameter, TREC concentration within CD8+ T cells, accounts entirely for age-dependent prognosis in GBM patients. See Wheeler et al at 4927. This raises the possibility of a similar immune impact on additional age-dependent properties of GBM, such as responsiveness to chemotherapy, hi support of this, the inventors observed a robust correlation between CD8+ TRECs and GBM responsiveness to post-vaccine chemotherapy that surpassed that between age and responsiveness to chemotherapy. Thus, a particular cellular immune parameter appears to not only account for the strongest prognostic factor in GBM (i.e., age), but also appears to be closely linked to chemotherapeutic responsiveness of these tumors as well. Moreover, because age is the single most dominant factor influencing the outcome of most human tumors, establishing the generality of such an immune impact on distinct tumors could substantially broaden clinical expectations associated with immune-based cancer therapies. In this context, it will be additionally important to determine whether cellular immune processes similarly influence clinical outcome and chemotherapeutic efficacy in distinct human tumors.
Example 4 Patients and Clinical Parameters
[0053] All patients suffered from newly-diagnosed GBM (55 yrs average, 32-78 range) and provided informed consent to treatments and associated monitoring. Patients in the "vaccine group" underwent craniotomy (five patients underwent one craniotomy prior to receiving vaccine therapy, six underwent two craniotomies, and one patient underwent four craniotomies prior to receiving vaccine therapy). All of these patients received a course of radiation prior to vaccination. Four patients in this group also received chemotherapy and one patient received stereotactic radiosurgery ("SRS") prior to vaccination. After vaccination, five of these patients underwent another craniotomy, and three received additional SRS. None received chemotherapy following vaccination. All patients in the "chemotherapy group" underwent craniotomy, radiation and chemotherapy. Six of these patients underwent a second craniotomy, and five patients received additional SRS. Of note, the longest overall survivor in this group (991 days) suffered from post-operative intracranial abscess requiring multiple surgical procedures for drainage. Intracranial infections in malignant glioma patients are associated with prolonged survival and have been proposed to initiate an anti-tumor immune response. See A.PJ. Bowles et al, "Long-term remission of malignant brain tumors after intracranial infections: a report of four cases," Neurosurgery, 44:636 (1999). Patients in the "vaccine and chemotherapy group" underwent craniotomy (eight patients underwent one craniotomy and five underwent two craniotomies) prior to receiving the vaccine therapy. All of these patients received radiation therapy. Five patients received additional chemotherapy and three received SRS. Following vaccination, six of these patients underwent another craniotomy, and five received SRS. All patients received chemotherapy following vaccination at the time of tumor progression. Notably, a single patient in this group (surviving > 730 days and depicted in Fig. 3B) experienced a cutaneous ghoblastoma with single lymph node involvement prior to vaccination and at the site of irradiated tumor cell inoculation for DTH testing. These two tumors were removed surgically approximately one year prior to chemotherapy and did not recur.
[0054] Vaccinated patients were steroid-free during blood collection and vaccinations were administered as described in Yu at 842. Patients received three vaccines, two weeks apart, of 10-40x10 autologous DC loaded with either HLA7 eluted peptides from cultured tumor cells or 150 μg/ml autologous tumor freeze-thaw lysate, starting approximately fifteen weeks post- surgery. A fourth identical vaccination followed six weeks later only in phase II trial patients (12 of 25). Serial MRI scans were performed every 2 to 3 months for all patients. Example 5 Cell Isolation and Lysis [0055] PBMC were prepared with Ficoll from patients' blood obtained at the time of surgery and/or from banked leukaphereses. CD4+ and CD8+ T cells were purified from PBMC using MACS bead separation (obtained from Miltenyi Biotec; Auburn, CA). 107 CD4+ or CD8+ cells/ml were prepared for quantitative real-time PCR ("qPCR") by lysis in 100 μg/mL proteinase K (obtained fro Boehringer; Indianapolis, IN) 1 hr, 56°C, with inactivation at 95°C, lO min.
Example 6 • TREC Quantification [0056] TRECs were quantified in duplicate or triplicate by qPCR using the 5 'nuclease (TaqMan) method as described in D.C. Douek et al., "Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution," The Lancet, 355:1875 (2000), and detected on an iCycler system (obtained from BioRad; Hercules, CA). qPCR was performed on 5 μL cell lysate (from 50,000 cells) with the primers depicted in Table 1 (probe was obtained from MegaBases; Chicago, IL).
Table 1 : Primers Used in qPCR
Figure imgf000019_0001
[0057] PCR reactions including 0.5 U Platinum Taq (obtained from Gibco; Grand Island, NY), 3.5 mM MgCl2, 0.2 mM dNTPs, 500 nM of each primer, 150 nM probe, were amplified at 95°C for 5 minutes, 95°C for 30 seconds, and 60°C for 1 minute for 45 cycles. Control β-actin reactions were performed to ensure nucleic acid content and negative samples were excluded from further analysis. TREC values were adjusted for T cell purity. Example 5 Statistical analyses [0058] Statistical analyses included 2-tailed Mann- Whitney log-rank tests for disease- free and overall survival, binomial distribution probability, and Pearson's correlation coefficients (r values) calculated with SAS and Excel software. Binomial distributions were determined for 2-year and 3-year survival frequencies between vaccine + chemotherapy and other (vaccine or chemotherapy) patient groups.
[0059] While the description above refers to particular embodiments of the present invention, it will be understood that many modifications may be made without departing from the spirit thereof. The accompanying claims are intended to cover such modifications as would fall within the true scope and spirit of the present invention. The presently disclosed embodiments are therefore to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims, rather than the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.

Claims

WHAT IS CLAIMED IS:
1. A method for treating a disease condition in a mammal, comprising: administering at least one vaccination of dendritic cells ("DC") to said mammal; and administering a regimen of chemotherapy to said mammal.
2. The method of claim 1, wherein said DC are primed ex vivo.
3. The method of claim 1 , wherein said DC are autologous tumor antigen-presented DC.
4. The method of claim 1, wherein said DC are unprimed.
5. The method of claim 1, wherein administering said at least one vaccination further comprises administering at least three vaccinations of DC.
6. The method of claim 1, wherein each of said at least one vaccination of DC comprises from about 105 to about 107 DC.
7. The method of claim 1, wherein each of said at least one vaccination of DC comprises about lOxlO6 to about 40x106 DC.
8. The method of claim 1, wherein said administering of said at least one vaccination of DC occurs prior to administering said regimen of chemotherapy to said mammal.
9. The method of claim 1 , wherein said regimen of chemotherapy includes the administration of" at least one chemotherapeutic agent selected from the group consisting of temozolomide, procarbazine, carboplatin, vincristine, BCNU, CCNU, thalidomide, irinotecan, isotretinoin, imatinib, etoposide, and combinations thereof.
10. The method of claim 1, wherein said disease condition is a cancer of the central nervous system.
11. The method of claim 1, wherein said disease condition is ghoblastoma multiforme.
12. A method of increasing chemosensitivity of a mammal, comprising: providing at least one vaccination of dendritic cells ("DC"); and administering said at least one vaccination of DC to said mammal.
13. The method of claim 12, wherein said DC are primed ex vivo.
14. The method of claim 12, wherein said DC are autologous tumor antigen-presented DC.
15. The method of claim 12, wherein said DC are unprimed.
16. The method of claim 12, wherein administering said at least one vaccination further comprises administering at least three vaccinations of DC.
17. The method of claim 12, wherein each of said at least one vaccination of DC comprises from about 105 to about 107 DC.
18. The method of claim 12, wherein each of said at least one vaccination of DC comprises about lOxlO6 to about 40xl06 DC.
19. The method of claim 12, whereby chemosensitivity of said mammal is increased with respect to chemotherapeutic treatment of a cancer of the central nervous system.
20. The method of claim 12, whereby chemosensitivity of said mammal is increased with respect to chemotherapeutic treatment of ghoblastoma multiforme.
21. A kit for use in the treatment of a disease condition, comprising: a quantity of dendritic cells ("DC"); a quantity of at least one chemotherapeutic agent; and instructions for use of said quantity of DC and said quantity of said at least one chemotherapeutic agent in the treatment of said disease condition.
22. The kit of claim 21, wherein said disease condition is a cancer of the central nervous system.
23. The kit of claim 21, wherein said disease condition is ghoblastoma multiforme.
24. The kit of claim 21, further including at least one component for use in priming a quantity of DC ex vivo.
PCT/US2004/034761 2003-10-21 2004-10-20 System and method for the treatment of cancer, including cancers of the central nervous system WO2005043155A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP04795865.7A EP1676132B1 (en) 2003-10-21 2004-10-20 Combination of chemotherapy and administration of glioma-antigen-pulsed dendritic cells in the treatment of glioma
US10/575,438 US7939090B2 (en) 2003-10-21 2004-10-20 System and method for the treatment of cancer, including cancers of the central nervous system
JP2006536762A JP5015601B2 (en) 2003-10-21 2004-10-20 Systems and methods for the treatment of cancer, including cancer of the central nervous system

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US51304003P 2003-10-21 2003-10-21
US60/513,040 2003-10-21

Publications (1)

Publication Number Publication Date
WO2005043155A1 true WO2005043155A1 (en) 2005-05-12

Family

ID=34549244

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/034761 WO2005043155A1 (en) 2003-10-21 2004-10-20 System and method for the treatment of cancer, including cancers of the central nervous system

Country Status (4)

Country Link
US (1) US7939090B2 (en)
EP (1) EP1676132B1 (en)
JP (2) JP5015601B2 (en)
WO (1) WO2005043155A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007034188A2 (en) * 2005-09-21 2007-03-29 Oxford Biomedica (Uk) Limited Chemo-immunotherapy method
WO2008039874A2 (en) 2006-09-26 2008-04-03 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
JP2009518287A (en) * 2005-11-02 2009-05-07 デューク ユニバーシティー Simultaneous chemotherapy and immunotherapy
US7705010B2 (en) 2005-02-22 2010-04-27 Cedars-Sinai Medical Center Use of minoxidil sulfate as an anti-tumor drug
US8097256B2 (en) 2006-09-28 2012-01-17 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US8383768B2 (en) 2009-05-07 2013-02-26 Immunocellular Therapeutics, Ltd. CD133 epitopes
US9068020B2 (en) 2008-09-02 2015-06-30 Cedars-Sinai Medical Center CD133 epitopes
US10137182B2 (en) 2013-02-14 2018-11-27 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPQ801700A0 (en) * 2000-06-07 2000-06-29 Peplin Research Pty Ltd Enzyme and viral activation
US20080199484A1 (en) * 2003-10-06 2008-08-21 Cedars-Sinai Medical Center Use Of Cox-2 Inhibitor to Prevent T-Cell Anergy Induced By Dendritic Cell Therapy
CA2589992C (en) * 2004-12-13 2014-04-22 Peplin Research Pty Ltd Treatment of solid cancers
KR20080077625A (en) * 2005-11-25 2008-08-25 페플린 리서치 피티이 리미티드 Methods for wound healing
EP2313506A1 (en) * 2008-07-11 2011-04-27 Medizinische Universität Innsbruck Antagonists of nr2f6 for augmenting the immune response
WO2010065876A2 (en) * 2008-12-06 2010-06-10 The Board Of Regents Of The University Of Texas System Methods and compositions related to th-1 dendritic cells
US9067005B2 (en) 2008-12-08 2015-06-30 Thoratec Corporation Centrifugal pump apparatus
US10085987B2 (en) 2012-01-27 2018-10-02 Thomas Jefferson University MCT protein inhibitor-related prognostic and therapeutic methods
EP2986290A4 (en) 2013-04-19 2017-01-04 Thomas Jefferson University Caveolin-1 related methods for treating glioblastoma with temozolomide
KR20200047660A (en) 2017-09-01 2020-05-07 다나-파버 캔서 인스티튜트 인크. Immunogenic peptides specific for BCMA and TACI antigens for the treatment of cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6566395B1 (en) * 1999-05-25 2003-05-20 Biomedicines, Inc. Methods of treating proliferative disorders

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
US6037135A (en) 1992-08-07 2000-03-14 Epimmune Inc. Methods for making HLA binding peptides and their uses
ATE260971T1 (en) 1992-04-01 2004-03-15 Univ Rockefeller METHOD FOR THE IN VITRO CULTIVATION OF DENDRITIC PRECURSOR CELLS AND THEIR USE FOR IMMUNOGENIC PRODUCTION
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
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
EP1630229B1 (en) 1994-04-22 2013-04-03 THE UNITED STATES OF AMERICA, as represented by the Secretary of the Department of Health and Human Services Melanoma antigens
WO1996018409A1 (en) 1994-12-14 1996-06-20 The Scripps Research Institute 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
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
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
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
US6458585B1 (en) 1996-08-14 2002-10-01 Nexell Therapeutics Inc. Cytokine-free culture of dendritic cells
EP0983345A1 (en) 1997-05-21 2000-03-08 THE UNITED STATES OF AMERICA, as represented by the Secretary of the Department of Health and Human Services Methods and compositions for making dendritic cells from expanded populations of monocytes and for activating t cells
US6565831B1 (en) * 1999-02-24 2003-05-20 Oncolytics Biotech Inc. Methods for preventing reovirus recognition for the treatment of cellular proliferative disorders
US20020192211A1 (en) 1998-03-17 2002-12-19 Hudziak Robert M. Method of treating tumor cells by inhibiting growth factor receptor function
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
WO2000024778A1 (en) 1998-10-26 2000-05-04 The Government Of The United States Of America Represented By The Secretary, Department Of Health And Human Services Hla-a2 and hla-dr specific peptide epitopes from the melanoma antigen trp2
GB9824306D0 (en) 1998-11-05 1998-12-30 Isis Innovation Method for producing dendritic dells
EP1140179A2 (en) 1998-12-23 2001-10-10 G.D. SEARLE &amp; CO. Use of a cyclooxygenase 2 inhibitor and an integrin antagonist as a combination therapy in the treatment of neoplasia
WO2001008636A2 (en) 1999-08-03 2001-02-08 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
WO2001041741A1 (en) 1999-12-13 2001-06-14 Epimmune Inc. Hla class i a2 tumor associated antigen peptides and vaccine compositions
EP1257289A1 (en) 2000-02-08 2002-11-20 The Penn State Research Foundation Immunotherapy using interleukin 13 receptor subunit alpha 2
DK1326961T3 (en) * 2000-09-15 2008-01-07 Ortho Mcneil Pharm Inc Compositions and Methods for Inducing Specific Cytolytic T Cell Reactions
US20030202963A1 (en) 2000-10-12 2003-10-30 Cornell Research Foundation, Inc. Method of treating cancer
US6632459B2 (en) 2000-12-11 2003-10-14 Nutricia N.V. Chlorogenic acid and an analog thereof for immune system stimulation
ES2394293T3 (en) 2001-02-28 2013-01-30 Bio Life Science Forschungs- Und Entwicklungsges.M.B.H. Vaccine against cancers that are associated with the HER-2 / neu oncogene
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
EP1409648A1 (en) * 2001-07-25 2004-04-21 I.D.M. Immuno-Designed Molecules New isolated dendritic cells, a process for preparing the same and their use in pharmaceutical compositions
GB0119346D0 (en) 2001-08-08 2001-10-03 Bioclones Proprietary Ltd Process for the maturation of dendritic cells
WO2003015705A2 (en) 2001-08-17 2003-02-27 Roger Williams Hospital In situ immunization
IL160720A0 (en) 2001-09-06 2004-08-31 Northwest Biotherapeutics Inc Composition 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
JP2005510491A (en) 2001-10-26 2005-04-21 イミュノ−アールエックス, インコーポレイテッド Immunotherapy to reverse immunosuppression
US7413869B2 (en) 2002-04-05 2008-08-19 Dendreon Corporation Method for determining potency of antigenic presenting cell based vaccines
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
DE602004019057D1 (en) 2003-02-27 2009-03-05 Northwest Biotherapeutics Inc GENERATION OF DENDRITIC CELLS FROM MONOCYTIC DENDRITIC CELL PRESERVATIVES WITH GM-CSF WITHOUT THE PRESENCE OF ADDITIONAL CYTOKINS
US20080199484A1 (en) 2003-10-06 2008-08-21 Cedars-Sinai Medical Center Use Of Cox-2 Inhibitor to Prevent T-Cell Anergy Induced By Dendritic Cell Therapy
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
WO2006099448A2 (en) 2005-03-14 2006-09-21 University Of Iowa Research Foundation Accelerated cd8+ t-cell memory after dendritic cell vaccination
US20080107668A1 (en) 2006-08-30 2008-05-08 Immunotope, Inc. Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
CA2700573C (en) 2006-09-26 2016-11-22 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
CA2700579A1 (en) 2006-09-28 2008-04-03 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6566395B1 (en) * 1999-05-25 2003-05-20 Biomedicines, Inc. Methods of treating proliferative disorders

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MARCH: "Advanced Organic Chemistry Reactions, Mechanisms and Structure", 1992, J. WILEY & SONS
SAMBROOK; RUSSEL: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
See also references of EP1676132A4
SINGLETON ET AL.: "Dictionary of Microbiology and Molecular Biology", 1994, J. WILEY & SONS

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7705010B2 (en) 2005-02-22 2010-04-27 Cedars-Sinai Medical Center Use of minoxidil sulfate as an anti-tumor drug
WO2007034188A3 (en) * 2005-09-21 2007-06-14 Oxford Biomedica Ltd Chemo-immunotherapy method
WO2007034188A2 (en) * 2005-09-21 2007-03-29 Oxford Biomedica (Uk) Limited Chemo-immunotherapy method
US8133681B2 (en) 2005-09-21 2012-03-13 Oxford Biomedica (Uk) Limited Chemo-immunotherapy method
US9399662B2 (en) 2005-11-02 2016-07-26 Duke University Concurrent chemotherapy and immunotherapy
JP2009518287A (en) * 2005-11-02 2009-05-07 デューク ユニバーシティー Simultaneous chemotherapy and immunotherapy
JP2013126994A (en) * 2005-11-02 2013-06-27 Duke Univ Concurrent chemotherapy and immunotherapy
US8129184B2 (en) 2006-09-26 2012-03-06 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
EP2084267A4 (en) * 2006-09-26 2011-03-02 Cedars Sinai Medical Center Cancer stem cell antigen vaccines and methods
EP2084267A2 (en) * 2006-09-26 2009-08-05 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
US9023338B2 (en) 2006-09-26 2015-05-05 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
WO2008039874A2 (en) 2006-09-26 2008-04-03 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
US8097256B2 (en) 2006-09-28 2012-01-17 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US10226519B2 (en) 2006-09-28 2019-03-12 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US9433667B2 (en) 2006-09-28 2016-09-06 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
US9095538B2 (en) 2006-09-28 2015-08-04 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
US9068020B2 (en) 2008-09-02 2015-06-30 Cedars-Sinai Medical Center CD133 epitopes
US9382308B2 (en) 2008-09-02 2016-07-05 Cedars-Sinai Medical Center CD133 epitopes
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
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

Also Published As

Publication number Publication date
US7939090B2 (en) 2011-05-10
EP1676132A4 (en) 2007-05-09
JP2010132684A (en) 2010-06-17
JP5015601B2 (en) 2012-08-29
US20070020297A1 (en) 2007-01-25
EP1676132B1 (en) 2014-01-22
EP1676132A1 (en) 2006-07-05
JP2007509156A (en) 2007-04-12

Similar Documents

Publication Publication Date Title
EP1676132B1 (en) Combination of chemotherapy and administration of glioma-antigen-pulsed dendritic cells in the treatment of glioma
Prins et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy
US11779555B2 (en) Combination of immunotherapy with local chemotherapy for the treatment of malignancies
US20110313229A1 (en) Therapy Targeting Cancer Stem Cells
Wood et al. A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma
EP3592841B1 (en) Compositions for treating cancers using antisense
TW201130975A (en) Furin-knockdown and GM-CSF-augmented (FANG) cancer vaccine
WO2018229132A1 (en) Tinostamustine for use in the treatment of t-cell prolymphocytic leukaemia
JP6764790B2 (en) Highly soluble aquaporin-4 extracellular loop peptide immunization for the treatment of neuromyelitis optica
Sun et al. Clinical research on dendritic cell vaccines to prevent postoperative recurrence and metastasis of liver cancer
US20210220458A1 (en) Development of dual whole cell-based vaccine against pancreatic cancer
CN104812446B (en) Cancer stem cell vaccination and treatment
RU2749610C2 (en) Methods related to activated dendritic cells compositions and to immunotherapeutic treatment of individuals with advanced cancer
Zheng et al. Effects of Shikonin on the functions of myeloid dendritic cells in a mouse model of severe aplastic anemia
CN114306614B (en) Biological response immune gel and preparation method and application thereof
US20230136088A1 (en) miRNA-193a for Promoting Immunogenic Cell Death
CN116694568A (en) Medium formulation for activating whole anti-tumor immune system and method for preparing agonist activated whole immune effector cells
JP7275131B2 (en) Methods of mobilizing bone marrow-infiltrating lymphocytes and uses thereof
Demaria et al. Cross-talk of breast cancer cells with the immune system
WO2018160699A1 (en) Biomarkers for diagnosis, prediction and/or prognosis of pancreatic cancer and uses thereof
US20220378890A1 (en) Immunogenic egfr peptide compositions and their use in the treatment of cancer
JP2010508364A (en) Dendritic cell tumor injection therapy and related vaccines
JP2022527473A (en) Methods for treating cancer with antisense
CN107073079A (en) Including the method for the treating cancer for giving PPAR gamma agonists
Pilones et al. Mechanisms of Invariant Natural Killer T Cell-Mediated Immunoregulation in Cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004795865

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007020297

Country of ref document: US

Ref document number: 10575438

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2006536762

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 2004795865

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10575438

Country of ref document: US