WO2021097227A1 - Il-15 fusion protein enhanced adoptive cell therapeutics - Google Patents
Il-15 fusion protein enhanced adoptive cell therapeutics Download PDFInfo
- Publication number
- WO2021097227A1 WO2021097227A1 PCT/US2020/060440 US2020060440W WO2021097227A1 WO 2021097227 A1 WO2021097227 A1 WO 2021097227A1 US 2020060440 W US2020060440 W US 2020060440W WO 2021097227 A1 WO2021097227 A1 WO 2021097227A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- cell
- complex
- hspc
- cancer
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
- A61K38/1793—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2086—IL-13 to IL-16
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/15—Natural-killer [NK] cells; Natural-killer T [NKT] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0646—Natural killers cells [NK], NKT cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5443—IL-15
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/715—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
- C07K14/7155—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/30—Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
Definitions
- compositions include natural killer (NK) cell adoptive transfer combined with hypomethylating agents (HMAs) for the treatment of cancer.
- HMAs hypomethylating agents
- an IL-15 superagonist is included.
- Harnessing natural killer (NK) cells against cancer is an emerging therapeutic approach, which is increasingly being explored for both hematological malignancies and solid tumors.
- NK natural killer
- a method of preventing or treating cancer comprises administering to a patient in need thereof, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent, thereby treating cancer.
- the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL- 15R ⁇ complex.
- the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT- 803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy.
- the adoptive cell therapy comprises hematopoietic stem cell transplantation, donor leukocyte infusion, adoptive transfer of natural killer cells (NK), T cells, B cells, chimeric antigen receptor- T cells (CAR-T), chimeric antigen receptor natural killer cells (CAR-NK) or combinations thereof.
- the NK cell is an allogeneic progenitor-derived NK cell.
- the adoptive cell therapy comprises transfer of allogeneic, autologous, syngeneic, related, unrelated, HLA- matched, HLA-mismatched or haploidentical cells.
- at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof.
- the at least one chemotherapeutic agent is administered prior to the administration of the adoptive cell therapy.
- the at least one chemotherapeutic agent is administered concomitantly with the administration of the adoptive cell therapy.
- the at least one chemotherapeutic agent is administered after the administration of the adoptive cell therapy.
- the at least one chemotherapeutic agent comprises nucleoside analogs.
- the nucleoside analog is a hypomethylating agent.
- the hypomethylating agent comprises: 5-azacytidine, 5-aza-2'- deoxycytidine (5-AZA-CdR), zebularine, procainamide, procaine, hydralazine, epigallocathechin-3-gallate, RG108, MG98 or combinations thereof.
- the hypomethylating agent increases anti-tumor NK cell activity as compared to a non- hypomethylating agent treated control.
- the method of treating cancer further comprises administering one or more cytokines to the patient and/or culturing the cells prior to adoptive transfer with one or more cytokines.
- the method of treatment includes one or more chemotherapeutic agents specific for treatment of the type of cancer.
- the method of treating cancer further comprises administering to the patient, a therapeutically effective amount of an IL-15:IL-15R ⁇ complex.
- the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT-803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a method of treating cancer comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of a hypomethylating agent, thereby treating cancer.
- the NK cells are allogeneic cells. These cells can be generated or obtained from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC).
- the hypothemylating agent is 5-aza-2'-deoxycytidine (5-AZA-CdR).
- the method of treating cancer further comprises administering to the patient, a therapeutically effective amount of an IL-15:IL-15R ⁇ complex.
- the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT-803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a kit for treating cancer comprises an adoptive cell therapy, at least one chemotherapeutic agent and directions for the use of the kit for the treatment of a cancer.
- the adoptive cell therapy comprises hematopoietic stem cells, donor leukocytes, T cells, or natural killer (NK) cells.
- the NK cell is an allogeneic progenitor-derived NK cell.
- the NK cells are generated from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC).
- the chemotherapeutic agent is a hypomethylating agent.
- the kit includes an IL-15:IL-15R ⁇ complex, wherein the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT-803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a method of treating cancer comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of an IL-15:IL-15R ⁇ complex or IL-15, thereby treating cancer.
- thee NK cells are obtained from one or more sources comprising ascites, peritoneum, lymph, blood, plasma or combinations thereof.
- the NK cells are obtained from ascites fluids.
- the IL-15:IL-15R ⁇ complex is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof.
- the IL-15:IL-15R ⁇ complex is administered prior to the administration of the adoptive cell therapy. In certain embodiments, the IL-15:IL-15R ⁇ complex is administered concomitantly with the administration of the adoptive cell therapy. In certain embodiments, the IL-15:IL-15R ⁇ complex is administered after the administration of the adoptive cell therapy. In certain embodiments, the NK cells are optionally cultured with the IL-15:IL-15R ⁇ complex prior to the administration of the adoptive cell therapy. In certain embodiments, the method of treating cancer, further comprises administering one or more chemotherapeutic agents. [0017] Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs.
- analog is meant a molecule that is not identical, but has analogous functional or structural features.
- a polypeptide analog retains the biological activity of a corresponding naturally-occurring polypeptide, while having certain biochemical modifications that enhance the analog's function relative to a naturally occurring polypeptide. Such biochemical modifications could increase the analog's protease resistance, membrane permeability, or half-life, without altering, for example, ligand binding.
- An analog may include an unnatural amino acid.
- cancer as used herein is meant, a disease, condition, trait, genotype or phenotype characterized by unregulated cell growth or replication as is known in the art; including colorectal cancer, as well as, for example, leukemias, e.g., acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia, AIDS related cancers such as Kaposi's sarcoma; breast cancers; bone cancers such as Osteosarcoma, Chondrosarcomas, Ewing's sarcoma, Fibrosarcomas, Giant cell tumors, Adamantinomas, and Chordomas; Brain cancers such as Meningiomas, Glioblastomas, Lower-Grade Astrocytomas, Oligodendrocytomas, Pituitary Tumors, Schwannomas, and Metastatic brain cancers; cancers
- cancer cell is meant a cell exhibiting a neoplastic cellular phenotype, which may be characterized by one or more of, for example, abnormal cell growth, abnormal cellular proliferation, loss of density dependent growth inhibition, anchorage-independent growth potential, ability to promote tumor growth and/or development in an immunocompromised non-human animal model, and/or any appropriate indicator of cellular transformation.
- Cancer cell may be used interchangeably herein with “tumor cell”, “malignant cell” or “cancerous cell”, and encompasses cancer cells of a solid tumor, a semi- solid tumor, a primary tumor, a metastatic tumor, and the like.
- cancer therapy refers to a therapy useful in treating cancer.
- anti-cancer therapeutic agents include, but are not limited to, e.g., surgery, chemotherapeutic agents, immunotherapy, growth inhibitory agents, cytotoxic agents, agents used in radiation therapy, anti-angiogenesis agents, apoptotic agents, anti- tubulin agents, and other agents to treat cancer, such as anti-HER-2 antibodies (e.g., HERCEPTIN TM ), anti-CD20 antibodies, an epidermal growth factor receptor (EGFR) antagonist (e.g., a tyrosine kinase inhibitor), HER1/EGFR inhibitor (e.g., erlotinib (TARCEVA TM )), platelet derived growth factor inhibitors (e.g., GLEEVEC TM (Imatinib Mesylate)), a COX-2 inhibitor (e.g., celecoxib), interferons, cytokines, antagonists (e.g.,
- a disease is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. Examples of diseases include neoplasias and viral infections.
- an effective amount is meant an amount of a compound, alone or in a combination, required to ameliorate the symptoms of a disease relative to an untreated patient.
- the effective amount of active compound(s) used to practice the present invention for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an “effective” amount.
- the effective amount is administered to a patient that has been diagnosed with cancer.
- the effective amount can result in the prevention of the development, recurrence, or onset of cancer and one or more symptoms thereof, to enhance or improve the efficacy of another therapy, reduce the severity, the duration of cancer, ameliorate one or more symptoms of cancer, prevent the advancement of cancer, cause regression of cancer, and/or enhance or improve the therapeutic effect(s) of another therapy.
- Effective amount also refers to the amount of a therapy that is sufficient to result in the prevention of the development, recurrence, or onset of cancer and one or more symptoms thereof, to enhance or improve the prophylactic effect(s) of another therapy, reduce the severity, the duration of cancer, ameliorate one or more symptoms of cancer, prevent the advancement of cancer, cause regression of cancer, and/or enhance or improve the therapeutic effect(s) of another therapy.
- the amount of a therapy is effective to achieve one, two, three, or more results following the administration of one, two, three or more therapies: (1) a stabilization, reduction or elimination of the cancer stem cell population; (2) a stabilization, reduction or elimination in the cancer cell population; (3) a stabilization or reduction in the growth of a tumor or neoplasm; (4) an impairment in the formation of a tumor; (5) eradication, removal, or control of primary, regional and/or metastatic cancer; (6) a reduction in mortality; (7) an increase in disease-free, relapse-free, progression-free, and/or overall survival, duration, or rate; (8) an increase in the response rate, the durability of response, or number of patients who respond or are in remission; (9) a decrease in hospitalization rate, (10) a decrease in hospitalization lengths, (11) the size of the tumor is maintained and does not increase or increases by less than 10%, preferably less than 5%, preferably less than 4%, preferably less than 2%, (12) an increase
- the term “in combination” in the context of the administration of a therapy to a subject refers to the use of more than one therapy for therapeutic benefit.
- the term “in combination” in the context of the administration can also refer to the prophylactic use of a therapy to a subject when used with at least one additional therapy.
- the use of the term “in combination” does not restrict the order in which the therapies (e.g., a first and second therapy) are administered to a subject.
- a therapy can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject which had, has, or is susceptible to cancer.
- the therapies are administered to a subject in a sequence and within a time interval such that the therapies can act together.
- the therapies are administered to a subject in a sequence and within a time interval such that they provide an increased benefit than if they were administered otherwise.
- Any additional therapy can be administered in any order with the other additional therapy.
- fragment is meant a portion of a polypeptide or nucleic acid molecule. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acid molecule or polypeptide.
- a fragment may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 nucleotides or amino acids.
- the invention also comprises polypeptides and nucleic acid fragments, so long as they exhibit the desired biological activity of the full length polypeptides and nucleic acid, respectively.
- a nucleic acid fragment of almost any length is employed.
- illustrative polynucleotide segments with total lengths of about 10,000, about 5,000, about 3,000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length (including all intermediate lengths) are included in many implementations of this invention.
- polypeptide fragment of almost any length is employed.
- illustrative polypeptide segments with total lengths of about 10,000, about 5,000, about 3,000, about 2,000, about 1,000, about 5,000, about 1,000, about 500, about 200, about 100, or about 50 amino acids in length (including all intermediate lengths) are included in many implementations of this invention.
- isolated denotes a degree of separation from original source or surroundings.
- Purify denotes a degree of separation that is higher than isolation.
- a “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of this invention is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography. The term “purified” can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel.
- nucleotide or polypeptide that has been separated from the components that naturally accompany it.
- the nucleotides and polypeptides are substantially pure when they are at least 60%, 70%, 80%, 90%, 95%, or even 99%, by weight, free from the proteins and naturally-occurring organic molecules with they are naturally associated.
- marker is meant any protein or polynucleotide having an alteration in expression level or activity that is associated with a disease or disorder.
- Neoplastic condition refers to the disease state associated with the neoplasia.
- Colon cancer e.g., colorectal cancer
- lung cancer ovarian cancer are examples (non-limiting) of a neoplastic condition.
- Illustrative neoplasms for which the invention can be used include, but are not limited to leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease), Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, end
- the neoplasia is multiple myeloma, beta-cell lymphoma, urothelial/bladder carcinoma or melanoma.
- “obtaining” as in “obtaining an agent” includes synthesizing, purchasing, or otherwise acquiring the agent.
- “reduces” is meant a negative alteration of at least 5%, 10%, 25%, 50%, 75%, or 100%.
- “specifically binds” is meant a compound or antibody that recognizes and binds a polypeptide of the invention, but which does not substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the invention.
- subject is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, equine, canine, ovine, or feline.
- the subject is preferably a mammal in need of such treatment, e.g., a subject that has been diagnosed with B cell lymphoma or a predisposition thereto.
- the mammal is any mammal, e.g., a human, a primate, a mouse, a rat, a dog, a cat, a horse, as well as livestock or animals grown for food consumption, e.g., cattle, sheep, pigs, chickens, and goats.
- the mammal is a human.
- tumor means a mass of transformed cells that are characterized by neoplastic uncontrolled cell multiplication and at least in part, by containing angiogenic vasculature. The abnormal neoplastic cell growth is rapid and continues even after the stimuli that initiated the new growth has ceased.
- the term “tumor” is used broadly to include the tumor parenchymal cells as well as the supporting stroma, including the angiogenic blood vessels that infiltrate the tumor parenchymal cell mass.
- a tumor generally is a malignant tumor, i.e., a cancer having the ability to metastasize (i.e.
- a metastatic tumor a tumor also can be nonmalignant (i.e. non-metastatic tumor).
- Tumors are hallmarks of cancer, a neoplastic disease the natural course of which is fatal. Cancer cells exhibit the properties of invasion and metastasis and are highly anaplastic.
- a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50.
- the terms “treating” and “treatment” as used herein refer to the administration of an agent or formulation to a clinically symptomatic individual afflicted with an adverse condition, disorder, or disease, so as to affect a reduction in severity and/or frequency of symptoms, eliminate the symptoms and/or their underlying cause, and/or facilitate improvement or remediation of damage.
- Treatment of patients with neoplasia may include any of the following: Adjuvant therapy (also called adjunct therapy or adjunctive therapy) to destroy residual tumor cells that may be present after the known tumor is removed by the initial therapy (e.g.
- neoadjuvant therapy given prior to the surgical procedure to shrink the cancer
- induction therapy to cause a remission, typically for acute leukemia
- consolidation therapy also called intensification therapy
- maintenance therapy given in lower or less frequent doses to assist in prolonging a remission
- first line therapy also called standard therapy
- second (or 3rd, 4th, etc.) line therapy also called salvage therapy
- palliative therapy also called supportive therapy
- FIGS.1A-1F are a series of graphs and an experimental protocol showing that HMAs can sensitize AML cells to NK cell–mediated killing.
- FIG.1A Experimental design: THP-1 and KG1a cells were cultured in the presence of AZA or DAC at the indicated concentrations. Two days later, cells were harvested and used as targets for HSPC-NK cells with or without drug washout. The same numbers of viable AML cells were plated for each condition. The numbers of viable AML cells were determined by FCM after 1 to 3 days of coculture and were used for calculation of AML cell survival, NK- specific killing for each independent treatment, and overall effect of NK cells, as indicated.
- FIGS.1B-1D Effect of HMAs pretreatment and HSPC-NK cells on THP-1 cells at day+1. Data were obtained after drug washout and are depicted as the mean ⁇ standard error of the mean (SEM) of 3 independent experiments (1-way ANOVA; **P ⁇ .01).
- FIGS.1E-1F Effect of HMA pretreatment and HSPC-NK cells on KG1a cells. Data were obtained without drug washout and are depicted as the mean ⁇ SEM of 3 independent experiments. Data shown in FIG.1E were obtained at day+1 and in FIG.1F are depicted the relative numbers of viable AML cells quantified from day 0 to day+3.
- FIGS.2A-2E are a series of graphs and an experimental protocol showing that HSPC-NK cells in combination with HMAs potently combat primary AML cells in vitro.
- FIG.2A Experimental design: Primary AML cells obtained from 5 different patients at diagnosis were stained with carboxyfluorescein diacetate succinimidyl ester and cultured in the presence of AZA or DAC using the indicated concentrations (5 x 10 4 AML cells per well). One day after, HSPC-NK cells (2.5 x 10 5 cells per well) were added and the drugs were refreshed daily.
- FIG.2B Median HSPC-NK cell survival at day+3 (combined data obtained with 5 different primary AML samples). The number of NK cells quantified without HMAs was set at 100%.
- FIG.2C Median AML cell survival at day+3 (combined data obtained with 5 different primary AML samples). The number of AML cells quantified without HMAs and NK cells is set at 100%.
- Data depicted in FIGS.2B-2C represent combined data obtained with 5 different primary AML samples and were analyzed with 1-way ANOVA. *** ⁇ .001; ** ⁇ .01. n.s., not significant.
- FIGS.2D-2E The survival of AML cells from 2 different patients (pAML #4 and pAML #5) and corresponding effect of NK cells quantified at day+3 are depicted as the mean ⁇ SEM of data obtained with 4 different HSPC-NK cell donors. Data were analyzed with 2-way ANOVA. *** ⁇ .001; * ⁇ .05. “No NK” is indicated by open bars; “+NK” is indicated by solid bars (FIG.2D). n.s., not significant. [0050] FIGS.3A-3G are a series of graphs, dot plots and an experimental protocol showing that low-dose HMAs do not impair HSPC-NK cell viability, proliferation, and cytolytic functions.
- FIG.3A Experimental design: Carboxyfluorescein diacetate succinimidyl ester–labeled HSPC-NK cells were cultured under proliferative (high-dose IL-15 and IL-2) or steady-state (low-dose IL-15) conditions in the presence of AZA or DAC refreshed daily at the indicated concentrations. Cell proliferation, viability, and absolute numbers, as well as functionality and phenotype, were analyzed by FCM after 6 days of treatment.
- FIG.3B Percentages of proliferating HSPC-NK cells under steady-state (left panel) and proliferative (right panel) conditions. Combined data from 3 independent experiments (mean ⁇ SEM) are shown.
- FIG.3C Specific killing of K562 and THP-1 cells by HSPC-NK cells pretreated with HMAs. The same numbers of viable NK cells were plated in each experimental well after washout of the drug, and the killing of K562 and THP-1 cells was determined after overnight coculture using 1:1 E:T ratio. Data obtained with NK cells that were treated either under proliferative or steady-state conditions and performed with 6 different HSPC-NK cell donors are combined and depicted as mean ⁇ SEM.
- FIGS.3D-3E NK cell reactivity upon K562 stimulation and analyzed at the single- cell level by FCM.
- FIG.3F Expression level of the maturation markers NKG2A, CD16, and killer immunoglobulin-like receptor-positive (KIR) cells on HSPC- NK cells following culture upon proliferative conditions in the presence of DAC 0.1 pM, or without HMAs. Mean ⁇ standard deviation (SD) of 6 HSPC-NK cell donors is shown.
- SD standard deviation
- FIG.3G Representative dot plots of HSPC-NK cell IFN-y production capacity with respect to KIR expression following DAC 0.1 pM or no HMA treatment under proliferative conditions. Statistical analyses were performed with 1-way (FIG.3B-3D) and 2-way ANOVA (FIG.3E). NS, not stimulated. [0051] FIGS.4A-4D are a series of graphs showing that DAC, but not AZA, potentiates HSPC-NK cell anti-leukemic effect in vivo.
- FIGS.4B-4D THP-1–bearing mice were treated with HMAs with use of the same dosages as described in titration #2, and with a single infusion of HSPC-NK cells, applied at day 4. Survival of NK cells in vivo was supported by recombinant human IL-15, given subcutaneously every 2 to 3 days.
- FIG.4B Experimental design.
- FIG.4C Median tumor load at day 17.
- FIG.4D Fold increase in tumor load after 2 weeks of treatment with DAC alone or in combination with HSPC-NK cells (calculated as the ratio between day 17 and day 3 signals). Data were analyzed with an unpaired, 2-tailed Student t test. IF, intrafemoral.
- FIGS.5A-5C are a series of graphs and an experimental protocol showing treatment with HSPC-NK cell infusions and DAC improves control of AML in vivo.
- FIG. 5A Experimental design: THP-1–bearing mice received 1 or 2 cycles of DAC (1.25 mg/m 2 ), with or without HSPC-NK cells, which were infused on the first day from each cycle. Survival of HSPC-NK cells in vivo was supported by ALT-803 (an IL-15 superagonist complex), which was given subcutaneously every 3 to 4 days until day 35 (0.2 mg/kg per injection). Groups that were not treated with NK cells were also given ALT-803 as a control.
- FIG.5B Impact of DAC on tumor load progression.
- FIG.5C Impact of HSPC-NK cell infusions on tumor load progression in mice cotreated with 2 cycles of DAC. Data are shown as mean ⁇ SD and were analyzed with 2-way ANOVA. One mouse in each DAC X 2 and DAC+NK X 2 group died at day 17 and day 15, respectively, likely due to DAC-related toxicities (weight loss >20% after second treatment cycle). These mice were excluded from the complete data set shown in this figure.
- FIGS.6A and 6B show a graph and scatter plot demonstrating that DAC treatment upregulates NK-inducing ligands on THP-1 cells in vivo.
- FIGS.6A, 6B THP-1– bearing mice were treated with DAC (1.25 mg/m 2 ) for 5 consecutive days. One week after the start of treatment, mice were euthanized and bone marrow cells were isolated for ex vivo analysis of THP-1 cells. Data obtained from 5 individual mice per treatment group were pooled to reach enough events by FCM (500-900 single and viable THP-1 cells acquired per test). The relative expression level of NKG2D and DNAM-1 ligands, as well as death receptors, are depicted in FIG.6A, and overlay plots with mean fluorescence intensities are shown in FIG.6B. Iso, isotype control.
- FIGS.7A-7C are a series of graphs and plots showing that DAC enhances the anti-leukemic potential of HSPC-NK cells through modulation of their maturation, activation, cytolytic functions, and trafficking to the bone marrow.
- FIG.7A-7C Adult NSG mice were infused with HSPC-NK cells and treated with DAC (1.25 mg/m 2 ) for 5 consecutive days. Persistence of NK cells in vivo was supported by subcutaneous administration of IL-15 (1 mg/injection) every 2 to 3 days. Mice were euthanized 1 or 2 weeks after NK cell infusion for detailed ex vivo analysis.
- FIG.7A Phenotype of HSPC- NK cells analyzed 1 week after the start of DAC treatment. Analysis was performed on cells isolated from the spleen, including 5 mice per treatment group. The relative expression of various maturation and activation markers, as well as adhesion molecules and homing receptor (right panel) and representative dot plots (left panel), is shown.
- FIG.7B Gene expression profiling for the cytolytic machinery of NK cells, analyzed by quantitative reverse transcription polymerase chain reaction on cells isolated from livers, including 5 mice per treatment group. Data were normalized to human b-actin.
- FIG.7C Absolute numbers of HSPC-NK cells were determined in peripheral blood (absolute number per milliliter) and mouse bone marrow either 1 week (experiment #1) or 2 weeks (experiment #2) after the start of treatment. Two femurs per mouse were combined in experiment #1, whereas experiment #2 was performed in IF THP-1–bearing mice and absolute NK cell counts were determined in each femur, with (Tumor BM) or without (NBM) tumor. Data shown in panel A were analyzed with 2-way ANOVA and data from FIGS.7B and 7C with an unpaired, 2-tailed Student t test.
- BM bone marrow
- IF intrafemoral
- ND no drug
- PB peripheral blood.
- FIG.8 shows a series of graphs demonstrating that HMA have a direct and dose-dependent effect on AML cell viability and proliferation.
- THP-1 and KG-1a cells were stained with CFSE and cultured in the presence of Azacitidine (AZA) or Decitabine (DAC) using the indicated concentrations. Treatment was applied for 5 consecutive days and both HMA were refreshed daily. Cell number, viability and proliferation were analyzed by flow cytometry after 1, 3, and 5 days of culture. Cell viability was determined on forward/side scatter and exclusion of 7AAD, and the proliferation index was determined using the mean fluorescence intensity (MFI) of CFSE and calculated as the ratio between untreated (No HMA) and treated cells. Representative data from one experiment are shown (mean ⁇ SD).
- MFI mean fluorescence intensity
- FIG.9 shows a series of graphs demonstrating the effect of HMA on primary AML cell viability and proliferation.
- Patient-derived primary AML cells were stained with CFSE and cultured in the presence of Azacitidine (AZA) or Decitabine (DAC) using the indicated concentrations. Treatment was applied for 5 consecutive days and both HMA were refreshed daily. Cell number, viability and proliferation were analyzed by flow cytometry after 1, 3, and 5 days of culture. Cell viability was determined on forward/side scatter and exclusion of 7AAD, and the proliferation index was determined using the mean fluorescence intensity (MFI) of CFSE and calculated as the ratio between untreated (No HMA) and treated cells.
- MFI mean fluorescence intensity
- FIG.10 is a series of graphs demonstrating that HSPC-NK cells in combination with HMA potently combat primary AML cells in vitro.
- Primary AML cells obtained from 2 patients at diagnosis (pAML#4 and pAML#5) were cultured in the presence of HMA, with or without HSPC-NK cells which were added at day 0.
- the numbers of viable AML cells were determined by flow cytometry from day -1 (plating of AML cells) till day +3 of co-culture.
- FIG.11 is a series of graphs demonstrating that phenotypical analysis of HSPC-NK cells following exposure to HMA in vitro.
- FIGS.12A, 12B are graphs demonstrating the tolerability and anti-leukemic activity of Azacitidine and Decitabine in mice.
- Adult NSG mice were injected IF with THP- 1.LucGFP cells.
- FIG.12B Tumor load monitoring was performed weekly by bioluminescence imaging till day 28 (higher dosages) or 42 (for lower and tolerated dosages only). Dotted lines indicate upper detection limit for tumor load monitoring (signal saturation). Data are depicted as mean ⁇ SD and include 3-6 mice per group per time- point.
- FIGS.13A, 13B demonstrate the effect of HMA therapy on mouse CD45 + cells vs. HSPC-NK cells in vivo.
- Adult NSG mice were infused with 5x10 6 HSPC-NK cells and treated for 5 consecutive days with 12.5 mg/m 2 Azacitidine (AZA) or 1.25 mg/m 2 Decitabine (DAC). Survival of NK cells in vivo was supported by subcutaneous administration of IL-15 (1 ⁇ g/injection) every 2-3 days. Mice were sacrificed one week after cell infusion and percentages of HSPC-NK cells within total human and mouse CD45 + cells (FIG.13A) as well as absolute numbers (FIG.13B) were determined in peripheral blood and spleen.
- AZA Azacitidine
- DAC 1.25 mg/m 2 Decitabine
- FIGS.14A-14C are graphs demonstrating the head-to-head comparison of recombinant human IL-15 and ALT-803 to support survival and proliferation of HSPC-NK cells.
- HSPC-NK cells were stained with eFluor450 proliferation dye and cultured in basal medium only, or supplemented with equimolar concentrations of rhIL-15 vs. ALT-803.
- FIGS.15A-15D are graphs demonstrating the impact of Azacitidine on THP-1 cells and HSPC-NK cells in vivo.
- FIG.15C Gene expression profiling for the cytolytic machinery of NK cells, analyzed by RT-qPCR on cells isolated from liver. Data were normalized to human ⁇ -Actin.
- FIG.15D Absolute numbers of HSPC-NK cells determined in mouse bone marrow (2 femurs combined per mouse). Data were analyzed using two-way (FIG.15B) and one-way (FIGS 15C and 15D) ANOVA. *p ⁇ 0.05, ***p ⁇ 0.001, n.s. not significant. [0063]
- FIG.16 is a graph and histogram showing the impact of HMA on THP-1 cells and HSPC-NK cells in vitro. The phenotype of THP-1 cells was analyzed by flow cytometry following 3 days culture in the presence of azacytidine (AZA) or Decitabine (DAC) at the indicated concentrations.
- AZA azacytidine
- DAC Decitabine
- FIG.17 is a series of graphs showing that HSPC-NK cells are highly capable of killing ovarian cancer (OC) cell lines and SKOV-3 spheroids in vitro. At an E:T ratio of 1:1, 24% to 79% killing of OC cell line monolayers was observed after overnight co-culture. This increased to >90% using higher E:T ratios.
- FIG.18 is a series of graphs and images demonstrating that the HSPC-NK cells infiltrate and mediate efficient intra-tumoral killing in SKOV-3 spheroids. Progressive infiltration of the spheroids by HSPC-NK cells was demonstrated, peaking at 8 hours.
- FIG.19 is a schematic and a series of graphs demonstrating that infusion of HSPC-NK cells in NSG mice bearing SKOV-3 tumors decreases tumor progression and improves survival. A rapid increase in tumor growth in the no treatment group was shown.
- FIG.20 is a schematic and a graph demonstrating the anti-tumor effect of intraperitoneal (ip) HSPC-NK cell therapy in combination with ALT-803 or recombinant human IL-15 in NSG mice ip engrafted with SKOV-3 tumors using bioluminescence imaging (BLI). All mice received nanogam (IgGs).
- FIGS.21A-21H show the NK, NKT and T cell frequency in benign ascites and ascites from ovarian cancer patients.
- FIG.21A Fraction of CD45 + lymphocytes (white), CD45 + non-lymphocytes (grey) and CD45- cells (black) cell populations within peritoneal fluid of benign compared to malignant ovarian cancer patients, based on flow cytometric analysis of CD45 expression and forward/side scatter.
- FIG.21B Percentage of NK cells, T cells, NKT cells and other lymphocytes within benign and malignant ascites.
- FIG.21D The NK cell population is subdivided based on CD56 bright and CD56 dim cells.
- FIG.21E Overall survival curve of OC patients groups with low and high CD56 + NK cell frequencies in ascites.
- FIG.21F Overall survival curve of OC patients groups with low and high CD3 + T cell frequencies in ascites.
- FIG.22 shows the expression of activating receptors on CD45 + CD3-CD56+ NK cells. Percentage positive 2B4, NKG2D, NKp46, NKp30, DNAM-1 and NKG2A NK cells of CD56+ NK cells in benign and malignant peritoneal fluid.
- FIGS.23A-23D show results from a degranulation assay comparing NK cells in healthy donor (HD) peripheral blood mononuclear cells (PBMCs) with ascites mononuclear cells (MNCs). Percentage CD56 + NK cells positive for: FIG.23A. CD69, FIG. 23B. TRAIL, FIG.23C. CD107a, FIG.23D.
- FIGS.24A-24F show results from a degranulation assay comparing ascites CD56 + NK cells with and without monomeric IL-15 or ALT-803 stimulation.
- FIG.24A-24C Percentage CD56 + NK cells positive for CD107a after 4h co-culture with no stimulation (FIG.24A), K562 cells (FIG.24B) or SKOV-3 cells (FIG.24C).
- FIGS.24D-24F Percentage CD56 + NK cells positive for CD107a after 4h co-culture with no stimulation.
- FIG.24A Percentage CD56 + NK cells positive for CD107a after 4h co-culture with no stimulation
- FIG.24B Percentage CD56 + NK cells positive for CD107a after 4h co-culture with no stimulation
- FIG.24B K562 cells
- FIGS.24C FIGS.24D-24F.
- AZA azacitidine
- DAC decitabine
- HSPC-NK cells CD341 hematopoietic stem and progenitor cells
- AML acute myeloid leukemia
- low-dose HMAs did not impair viability of HSPC-NK cells.
- low-dose DAC preserved HSPC-NK killing, proliferation, and interferon gamma production capacity, whereas AZA diminished their proliferation and reactivity.
- HMAs and HSPC-NK cells could potently work together to target AML cell lines and patient AML blasts.
- HSPC-NK cells showed sustained expression of most activating receptors, upregulated NKp44 expression, and remarkable killer cell immunoglobulin-like receptor acquisition.
- DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells.
- treatment resulted in increased numbers of HSPC- NK cells in the bone marrow compartment, providing evidence that DAC could positively modulate NK cell activity, trafficking, and tumor targeting.
- Nucleoside analogs have been used clinically for the treatment of viral infections and cancer. Most nucleoside analogs are classified as anti-metabolites. After they enter the cell, nucleoside analogs are successively phosphorylated to nucleoside 5'-mono- phosphates, di-phosphates, and tri-phosphates.
- the drug is a nucleoside drug.
- a “nucleoside” includes (e.g., consists of) a nucleobase (such as a purine or pyrimidine) bound covalently to a pentose monosaccharide, e.g., a 5-carbon sugar such as ribose.
- nucleoside analogue is meant a nucleoside in which either the nucleobase and/or the pentose monosaccharide are unnatural.
- a nucleoside drug includes a nucleoside analogue that causes a physiological change within the body of an individual.
- the nucleoside analogue is any one of an adenoside/deoxyadenosine analogue, cytidine/deoxycytidine, guanosine/deoxyguanosine analogue, thymidine/deoxythymidine analogue, or a deoxyuridine analogue.
- the nucleoside analogue may be any one of gemcitabine, cytarabine, troxacitabine, decitabine, cladribine, fludarabine, clofarabine, or 2'-C-cyano-2'-deoxy-1-3-D-arabino-pentofuranosylcytosine (CNDAC).
- CNDAC 2'-C-cyano-2'-deoxy-1-3-D-arabino-pentofuranosylcytosine
- the nucleoside analogue is decitabine (DAC).
- 5-Azacytidine (National Service Center designation NSC-102816; CAS Registry Number 320-67-2), also known as azacitidine, AZA, or 4-amino-1- ⁇ -D- ribofuranosyl-1,3,5-triazin-2(1H)-one, is currently marketed as the drug product VIDAZA TM .
- 5-Azacytidine is a nucleoside analog, more specifically a cytidine analog.5-Azacytidine is an antagonist of its related natural nucleoside, cytidine.5-Azacytidine and 5-aza-2'- deoxycytidine (also known as decitabine (DAC), an analog of deoxycytidine).
- DAC decitabine
- At least one chemotherapeutic agent comprises nucleoside analog.
- the nucleoside analog is a hypomethylating agent. Hypomethylating agents have been approved by the US Food and Drug Administration in the treatment of cancer.
- VIDAZA TM azacitidine for injection
- FAB French-American-British myelodysplastic syndromes subtypes: refractory anemia (RA) or refractory anemia with ringed sideroblasts (if accompanied by neutropenia or thrombocytopenia or requiring transfusions), refractory anemia with excess blasts (RAEB), refractory anemia with excess blasts in transformation (RAEB-T), and chronic myelomonocytic leukemia (CMMoL).
- FAB French-American-British
- RA refractory anemia
- RAEB refractory anemia with ringed sideroblasts
- RAEB refractory anemia with excess blasts
- RAEB-T refractory anemia with excess blasts in transformation
- CMMoL chronic myelomonocytic leukemia
- DACOGEN TM (decitabine for injection) is indicated for treatment of patients with myelodysplastic syndromes (MDS) including previously treated and untreated, de novo and secondary MDS of all French- American-British subtypes (refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation, and chronic myelomonocytic leukemia) and intermediate-1, Intermediate-2, and high-risk International Prognostic Scoring System groups.
- MDS myelodysplastic syndromes
- French- American-British subtypes refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation, and chronic myelomonocytic leukemia
- the hypomethylating agent comprises: 5-azacytidine, 5-aza-2'-deoxycytidine (5-AZA-CdR), zebularine, procainamide, procaine, hydralazine, epigallocathechin-3-gallate, RG108, MG98 or combinations thereof.
- the hypomethylating agent is 5-aza-2'-deoxycytidine ((decitabine (DAC)).
- DAC decitabine
- the at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof. In another embodiment, the at least one chemotherapeutic agent is administered prior to the administration of the adoptive cell therapy.
- the at least one chemotherapeutic agent is administered concomitantly with the administration of the adoptive cell therapy. In another embodiment, the at least one chemotherapeutic agent is administered after the administration of the adoptive cell therapy.
- the hypomethylating agent increases anti-tumor NK cell activity as compared to a non-hypomethylating agent treated control.
- the method of treating cancer further comprises administering one or more cytokines to the patient and/or culturing the cells prior to adoptive transfer with one or more cytokines.
- the methods of the invention may include administration of second therapeutic agent or treatment with a second therapy (e.g., a therapeutic agent or therapy that is standard in the art).
- chemotherapeutic agents include chemotherapeutic agents.
- a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
- chemotherapeutic agents include Erlotinib (TARCEVA TM , Genentech/OSI Pharm.), Bortezomib (VELCADE TM , Millennium Pharm.), Fulvestrant (FASLODEX TM , Astrazeneca), Sutent (SU11248, Pfizer), Letrozole (FEMARA TM , Novartis), Imatinib mesylate (GLEEVEC TM , Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin TM , Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE TM , Wyeth), Lapatinib (GSK572016, GlaxoSmithKline), Lonafarnib (SCH 6
- dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, anthramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN TM doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, 6-diazo-5-oxo-L-norleucine
- chemotherapeutic agent also included in this definition of “chemotherapeutic agent” are: (i) anti- hormonal agents that act to regulate or inhibit hormone action on tumors such as anti- estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX TM (tamoxifen)), raloxifene, droloxifene, 4- hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON TM (toremifene); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE TM (megestrol acetate), AROMASIN TM (exemestane), formestanie, fadrozole, RIVISOR TM (vorozole), FEMARA TM (
- Adoptive Cell Therapy As described in detail ion the examples section which follows, hypomethylating agent, DAC positively modulates the in vivo anti-leukemic potential of adoptively transferred HSPC-NK cells through AML cell sensitization, enhancement of NK cell maturation, and cytolytic functions, as well as improves on NK cell trafficking and accumulation at the tumor site. Allogeneic NK cells were generated from CD34 + hematopoietic stem and progenitor cells (HSPC-NK cells) for use in the adoptive therapy treatments.
- HSPC-NK cells CD34 + hematopoietic stem and progenitor cells
- Adoptive cell therapy (including allogeneic and autologous hematopoietic stem cell transplantation (HSCT) and recombinant cell (i.e., CAR T) therapies) is the treatment of choice for many malignant disorders (for reviews of HSCT and adoptive cell therapy approaches, see, Rager & Porter, Ther Adv Hematol (2011) 2(6) 409–428; Roddie & Peggs, Expert Opin. Biol. Ther. (2011) 11(4):473-487; Wang et al. Int. J. Cancer. (2015)136, 1751–1768; and Chang, Y.J. and X.J. Huang, Blood Rev, 2013.27(1): 55-62).
- Such adoptive cell therapies include, but are not limited to, allogeneic and autologous hematopoietic stem cell transplantation, donor leukocyte (or lymphocyte) infusion (DLI), adoptive transfer of tumor infiltrating lymphocytes, or adoptive transfer of T cells or NK cells (including recombinant cells, i.e., CAR T, CAR NK, gene-edited T cells or NK cells, see Hu et al. Acta Pharmacologica Sinica (2016) 39: 167–176, Irving et al. Front Immunol. (2017) 8: 267). Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from transferred cells is important for the elimination of residual tumor cells.
- the efficacy of ACT as a curative option for malignancies is influenced by a number of factors including the origin, composition and phenotype (lymphocyte subset, activation status) of the donor cells, the underlying disease, the pre-transplant conditioning regimen and post-transplant immune support (i.e., IL-2 therapy) and the graft-versus-tumor (GVT) effect mediated by donor cells within the graft. Additionally, these factors must be balanced against transplant-related mortality, typically arising from the conditioning regimen and/or excessive immune activity of donor cells within the host (i.e., graft-versus-host disease, cytokine release syndrome, etc.). [0085] Approaches utilizing adoptive NK cell therapy have become of significant interest.
- NK cell numbers recover very early after the transplant and the levels of NK cells correlate with a positive outcome (Rueff et al., 2014, Biol. Blood Marrow Transplant.20, 896–899).
- therapeutic strategies with autologous NK cell transfer have had limited success due to a number of factors, adoptive transfer of ex vivo-activated allogeneic (or haplo-identical) NK cells has emerged as a promising immunotherapeutic strategy for cancer (Guillerey et al.2016. Nature Immunol.17: 1025-1036). The activity of these cells is less likely to be suppressed by self-MHC molecules compared to autologous NK cells.
- NK cells or NK precursors i.e., stem cells
- pre-transplant conditioning and post-transplant immune support strategies use of NK cell lines or recombinant tumor-targeting NK cells
- combination therapies with other agents such as therapeutic Ab, immunomodulatory agents (lenalidomide), and anti-KIR and checkpoint Abs.
- these strategies could be complemented by the combination therapeutic approach of the invention, which has the capacity to augment NK cell proliferation and activation.
- DAC potentiated HSPC-NK cell anti-leukemic activity in vivo. Besides upregulation of NKG2D- and DNAM-1– activating ligands on AML cells, DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells. In addition, treatment resulted in increased numbers of HSPC-NK cells in the bone marrow compartment, providing evidence that DAC positively modulated NK cell activity, trafficking, and tumor targeting.
- Natural Killer Cells One of the major types of circulating mononuclear cells is that of the natural killer, or NK, cell (M.
- NK cells are now known as one of the components of the early, innate immune system. In addition to their cytotoxic capabilities, NK cells serve as regulators of the immune response by releasing a variety of cytokines. In addition, the generation of complex immune responses is facilitated by the direct interaction of NK cells with other cells via various surface molecules expressed on the NK cells.
- NK cells are derived from bone marrow precursors (O. Haller et al., Journal of Experimental Medicine 145:1411-1420, 1977).
- NK cells appear to be closely related to T cells, and the two cell types share many cell surface markers (M. Manoussaka et al., 1997). As noted above, these cell surface markers play a significant role in NK cell activity.
- murine NK cells express specific antigens on their surfaces, such as asialo GM1, NK1, and NK2 antigens (D. See et al., Scand. J. Immunol.46:217-224, 1997), and the administration of antibodies against these antigens results in depletion of NK cells in vivo (Id.).
- NK cells Similarly to cytotoxic T lymphocytes (CTL), NK cells exert a cytotoxic effect by lysing a variety of cell types (Srivastava, S., Lundqvist, A. & Childs, R. W. Natural killer cell immunotherapy for cancer: a new hope. Cytotherapy 10, 775–783; 2008). These include normal stem cells, infected cells, and transformed cells. The lysis of cells occurs through the action of cytoplasmic granules containing proteases, nucleases, and perforin. Cells that lack MHC class I are also susceptible to NK cell-mediated lysis (H. Reyburn et al., Immunol. Rev.155:119-125, 1997).
- NK cells exert cytotoxicity in a non-MHC restricted fashion (E. Ciccione et al., J. Exp. Med.172:47, 1990; A. Moretta et al., J. Exp. Med.172:1589, 1990; and E. Ciccione et al., J. Exp. Med.175:709).
- NK cells can also lyse cells by antibody-dependent cellular cytotoxicity.
- NK cells mediate some of their functions through the secretion of cytokines, such as interferon ⁇ (IFN- ⁇ ), granulocyte-macrophage colony- stimulating factors (GM-CSFs), tumor necrosis factor ⁇ (TNF- ⁇ ), macrophage colony- stimulating factor (M-CSF), interleukin-3 (IL-3), and IL-8.
- cytokines such as interferon ⁇ (IFN- ⁇ ), granulocyte-macrophage colony- stimulating factors (GM-CSFs), tumor necrosis factor ⁇ (TNF- ⁇ ), macrophage colony- stimulating factor (M-CSF), interleukin-3 (IL-3), and IL-8.
- IFN- ⁇ interferon ⁇
- GM-CSFs granulocyte-macrophage colony- stimulating factors
- TNF- ⁇ tumor necrosis factor ⁇
- M-CSF macrophage colony- stimulating factor
- IL-3 interleukin-3
- cytokines including IL-2, IL-12, TNF- ⁇ , and IL-1 can induce NK cells to produce cytokines.
- IFN- ⁇ and IL-2 are strong inducers of NK cell cytotoxic activity (G. Trinichieri et al., Journal of Experimental Medicine 160:1147-1169, 1984; G. Trinichieri and D. Santoli, Journal of Experimental Medicine 147: 1314-1333, 1977).
- the presence of IL-2 both stimulates and expands NK cells (K. Oshimi, International Journal of Hematology 63:279- 290, 1996).
- IL-12 has been shown to induce cytokine production from T and NK cells, and augment NK cell-mediated cytotoxicity (M.
- NK cells are involved in both the resistance to and control of cancer spread. Since the advent of the cancer immune surveillance concept, the adoptive transfer of immune cells, particularly T cells and natural killer (NK) cells, has emerged as a targeted method of harnessing the immune system against cancer (Kroemer, G., Senovilla, L., Galluzzi, L., Andre, F. & Zitvogel, L. Natural and therapy-induced immunosurveillance in breast cancer. Nat Med 21, 1128–1138, (2015)). NK cells have garnered immense attention as a promising immunotherapeutic agent for treating cancers.
- NK cells are critical to the body’s first line of defense against cancer due to their natural cytotoxicity against malignant cells (Srivastava, S., et al., Cytotherapy 10, 775–783; 2008).
- NK cells have been expanded from multiple sources, including peripheral blood and umbilical cord blood (CB) ((Denman, C. J. et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells.
- CB umbilical cord blood
- Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity.
- Ex vivo NK cell expansion methods have been developed using cytokines in combination with artificial antigen-presenting cells (aAPCs) as feeder cells ((Denman, C. J. et al. PLoS One 7, e30264, (2012); Berg, M. et al.
- a IL-15 receptor ⁇ /IgG1 Fc fusion protein (IL- 15N72D:IL-15R ⁇ Su/Fc) can be administered as part of the adoptive cell therapy and can include one or more chemotherapeutic agents.
- IL- 15N72D:IL-15R ⁇ Su/Fc can be administered as part of the adoptive cell therapy and can include one or more chemotherapeutic agents.
- ALT-803 comprises an IL-15 mutant with increased ability to bind IL-2R ⁇ and enhanced biological activity (U.S. Patent No.8,507, 222, incorporated herein by reference).
- This super-agonist mutant of IL-15 was described in a publication (Zu et al., 2009 J Immunol, 183: 3598-3607, incorporated herein by reference).
- This IL-15 super-agonist in combination with a soluble IL-15 ⁇ receptor fusion protein (IL- 15R ⁇ Su/Fc) results in a fusion protein complex with highly potent IL-15 activity in vitro and in vivo (Han et al., 2011, Cytokine, 56: 804-810; Xu, et al., 2013 Cancer Res.73:3075-86, Wong, et al., 2013, OncoImmunology 2:e26442).
- the IL-15 super agonist complex comprises an IL-15 mutant (IL-15N72D) bound to an IL-15 receptor ⁇ /IgG1 Fc fusion protein (IL-15N72D:IL-15R ⁇ Su/Fc) is referred to as “ALT-803.”
- IL-15N72D IL-15 receptor ⁇ /IgG1 Fc fusion protein
- Pharmacokinetic analysis indicated that the fusion protein complex has a half- life of 25 hours following i.v. administration in mice.
- ALT-803 exhibits impressive anti- tumor activity against aggressive solid and hematological tumor models in immunocompetent mice. It can be administered as a monotherapy using a twice weekly or weekly i.v. dose regimen or as combinatorial therapy with an antibody.
- the ALT-803 anti-tumor response is also durable.
- IL-15N72D associated with a dimeric IL- 15R ⁇ Su/Fc fusion protein
- SEQ ID NO: 1 IL-15N72D protein sequence (with leader peptide) METDTLLLWVLLLWVPGSTG- [Leader peptide] NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLESGDAS IHDTVENLIILANDSLSSNGNVTESGCKECEELEEKNIKEFLQSFVHIVQMFINTS [IL-15N72D] IL-15R ⁇ Su/Fc protein sequence (with leader peptide) MDRLTSSFLLLIVPAYVLS- [Leader peptide] ITCPPPMSVEHADIW
- the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL-15R ⁇ complex.
- the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT-803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy.
- the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL- 15R ⁇ complex.
- the IL-15/IL15R ⁇ complex is an IL-15N72D:IL-15R ⁇ Su/Fc complex (ALT- 803) comprising a dimeric IL-15R ⁇ Su/Fc and two IL-15N72D molecules.
- a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy.
- the NK cells are contacted with the ALT-803 fusion protein complex.
- NK cells The ex vivo incubation of NK cells with the fusion protein complex results in induction of CIML NK cell exhibiting elevated activation markers, increased cytotoxicity against tumor cells and enhanced production of IFN- ⁇ . Additionally, the fusion protein complex is capable of activating human NK cell lines. Moreover, methods are provided for augmenting immune responses and treating neoplasia and infection disease by direct administration of the fusion protein complex of the invention or administration of immune cells activated by the fusion protein complex of the invention. [0098] Immune Modulating Molecules [0099] In certain embodiments, one or more immune modulating compounds can be administered as part of the treatment plan.
- the immune-modulating molecules comprise, but are not limited to cytokines, lymphokines, NK cell stimulating factors, T cell co-stimulatory ligands, etc.
- An immune-modulating molecule positively and/or negatively influences the humoral and/or cellular immune system, particularly its cellular and/or non-cellular components, its functions, and/or its interactions with other physiological systems.
- the immune-modulating molecule may be selected from the group comprising cytokines, chemokines, macrophage migration inhibitory factor (MIF; as described, inter alia, in Bernhagen (1998), Mol Med 76(3-4); 151-61 or Metz (1997), Adv Immunol 66, 197-223), T- cell receptors or soluble MHC molecules.
- MIF macrophage migration inhibitory factor
- Immune cell activity that may be measured include, but is not limited to, (1) cell proliferation by measuring the DNA replication; (2) enhanced cytokine production, including specific measurements for cytokines, such as IFN- ⁇ , GM-CSF, or TNF- ⁇ ; (3) cell mediated target killing or lysis; (4) cell differentiation; (5) immunoglobulin production; (6) phenotypic changes; (7) production of chemotactic factors or chemotaxis, meaning the ability to respond to a chemotactin with chemotaxis; (8) immunosuppression, by inhibition of the activity of some other immune cell type; and, (9) apoptosis, which refers to fragmentation of activated immune cells under certain circumstances, as an indication of abnormal activation.
- Cytokines of the invention are defined by any factor produced by cells that affect other cells and are responsible for any of a number of multiple effects of cellular immunity.
- cytokines include but are not limited to the IL-2 family, interferon (IFN), IL-7, IL-10, IL-12, IL-15, IL-18, IL-1, IL-17, TGF and TNF cytokine families, and to IL-1 through IL-35, IFN- ⁇ , IFN- ⁇ , IFN ⁇ , TGF- ⁇ , TNF- ⁇ , and TNF ⁇ .
- Chemokines similar to cytokines, are defined as any chemical factor or molecule which when exposed to other cells are responsible for any of a number of multiple effects of cellular immunity.
- Suitable chemokines may include but are not limited to the CXC, CC, C, and CX3C chemokine families and to CCL-1 through CCL-28, CXC-1 through CXC-17, XCL-1, XCL-2, CX3CL1, MIP-1b, IL-8, MCP-1, and Rantes.
- Growth factors include any molecules which when exposed to a particular cell induce proliferation and/or differentiation of the affected cell. Growth factors include proteins and chemical molecules, some of which include: stem cell factors, GM-CSF, G-CSF, human growth factor and stem cell growth factor. Additional growth factors may also be suitable for uses described herein.
- NK cells also express a family of at least 11 serine proteases termed granzymes, which have four primary substrate specificities (Kam et al., Biochim. Biophys. Acta 1477:307, 2000).
- compositions comprising HSPC-NK cells and/or hypomethylating agent and/or second or third therapeutic agents such as for example, ALT-803, cytokines, chemotherapeutics, and the like, for use as a therapeutic.
- the pharmaceutical compositions are administered systemically, for example, formulated in a pharmaceutically-acceptable buffer such as physiological saline.
- Preferable routes of administration include, for example, instillation into the bladder, subcutaneous, intravenous, intraperitoneal, intramuscular, intratumoral or intradermal injections that provide continuous, sustained or effective levels of the composition in the patient.
- Treatment of human patients or other animals is carried out using a therapeutically effective amount of a therapeutic identified herein in a physiologically-acceptable carrier. Suitable carriers and their formulation are described, for example, in Remington's Pharmaceutical Sciences by E. W. Martin.
- the amount of the therapeutic agent to be administered varies depending upon the manner of administration, the age and body weight of the patient, and with the clinical symptoms of the neoplasia.
- compositions embodied herein are by any suitable means that results in a concentration of the therapeutic that, combined with other components, is effective in ameliorating, reducing, or stabilizing said neoplasia or infectious disease.
- compositions may be provided in a dosage form that is suitable for parenteral (e.g., subcutaneous, intravenous, intramuscular, intravesicular, intratumoral or intraperitoneal) administration route.
- parenteral e.g., subcutaneous, intravenous, intramuscular, intravesicular, intratumoral or intraperitoneal
- the pharmaceutical compositions are formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988- 1999, Marcel Dekker, New York).
- Human dosage amounts are initially determined by extrapolating from the amount of compound used in mice or non-human primates, as a skilled artisan recognizes it is routine in the art to modify the dosage for humans compared to animal models.
- the dosage may vary from between about 1 ⁇ g compound/kg body weight to about 5000 mg compound/kg body weight; or from about 5 mg/kg body weight to about 4,000 mg/kg body weight or from about 10 mg/kg body weight to about 3,000 mg/kg body weight; or from about 50 mg/kg body weight to about 2000 mg/kg body weight; or from about 100 mg/kg body weight to about 1000 mg/kg body weight; or from about 150 mg/kg body weight to about 500 mg/kg body weight.
- the dose is about 1, 5, 10, 25, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1,000, 1,050, 1,100, 1,150, 1,200, 1,250, 1,300, 1,350, 1,400, 1,450, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, 3,000, 3,500, 4,000, 4,500, or 5,000 mg/kg body weight.
- doses are in the range of about 5 mg compound/Kg body weight to about 20 mg compound/kg body weight.
- the doses are about 8, 10, 12, 14, 16 or 18 mg/kg body weight.
- the fusion protein complex is administered at 0.5 mg/kg-about 10 mg/kg (e.g., 0.5, 1, 3, 5, 10 mg/kg).
- this dosage amount may be adjusted upward or downward, as is routinely done in such treatment protocols, depending on the results of the initial clinical trials and the needs of a particular patient.
- Pharmaceutical compositions are formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the therapeutic in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, molecular complexes, nanoparticles, patches, and liposomes.
- compositions embodied herein are administered parenterally by injection, infusion or implantation (subcutaneous, intravenous, intramuscular, intratumoral, intravesicular, intraperitoneal) in dosage forms, formulations, or via suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants.
- suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants.
- Formulations can be found in Remington: The Science and Practice of Pharmacy, supra.
- Compositions comprising a hypomethylating agent or a cytokine or the fusion protein complex for parenteral use are provided in unit dosage forms (e.g., in single-dose ampoules).
- the composition is provided in vials containing several doses and in which a suitable preservative may be added (see below).
- the composition is in the form of a solution, a suspension, an emulsion, an infusion device, or a delivery device for implantation, or is presented as a dry powder to be reconstituted with water or another suitable vehicle before use.
- the composition includes suitable parenterally acceptable carriers and/or excipients.
- the active therapeutic agent(s) may be incorporated into microspheres, microcapsules, nanoparticles, liposomes for controlled release.
- the composition may include suspending, solubilizing, stabilizing, pH-adjusting agents, tonicity adjusting agents, and/or dispersing, agents.
- the pharmaceutical compositions may be in a form suitable for sterile injection.
- the suitable active therapeutic(s) are dissolved or suspended in a parenterally acceptable liquid vehicle.
- acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1,3-butanediol, Ringer's solution, and isotonic sodium chloride solution and dextrose solution.
- the aqueous formulation may also contain one or more preservatives (e.g., methyl, ethyl or n-propyl p-hydroxybenzoate).
- a dissolution enhancing or solubilizing agent can be added, or the solvent may include 10-60% w/w of propylene glycol.
- the present invention provides methods of treating neoplasia or infectious diseases or symptoms thereof which comprise administering a therapeutically effective amount of a pharmaceutical composition.
- one embodiment is a method of treating a subject suffering from or susceptible to a neoplasia or infectious disease or symptom thereof.
- the method includes the step of administering to the mammal a therapeutic amount of an amount of the therapeutic agents and HSPC-NK cells sufficient to treat the disease or disorder or symptom thereof, under conditions such that the disease or disorder is treated.
- the methods herein include administering to the subject (including a subject identified as in need of such treatment) an effective amount of a compound described herein, or a composition described herein to produce such effect. Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
- the therapeutic methods of the invention in general comprise administration of a therapeutically effective amount of the compounds herein, such as a compound of the formulae herein to a subject (e.g., animal, human) in need thereof, including a mammal, particularly a human.
- a subject e.g., animal, human
- Such treatment will be suitably administered to subjects, particularly humans, suffering from, having, susceptible to, or at risk for a neoplasia, infectious disease, disorder, or symptom thereof. Determination of those subjects "at risk” can be made by any objective or subjective determination by a diagnostic test or opinion of a subject or health care provider (e.g., genetic test, enzyme or protein marker, Marker (as defined herein), family history, and the like).
- the fusion protein complexes of the invention may be used in the treatment of any other disorders in which an increase in an immune response is desired.
- the invention also provides a method of monitoring treatment progress.
- the method includes the step of determining a level of diagnostic marker (Marker) (e.g., any target delineated herein modulated by a compound herein, a protein or indicator thereof, etc.) or diagnostic measurement (e.g., screen, assay) in a subject suffering from or susceptible to a disorder or symptoms thereof associated with neoplasia in which the subject has been administered a therapeutic amount of a compound herein sufficient to treat the disease or symptoms thereof.
- a level of diagnostic marker Marker
- diagnostic measurement e.g., screen, assay
- the level of Marker determined in the method can be compared to known levels of Marker in either healthy normal controls or in other afflicted patients to establish the subject’s disease status.
- a second level of Marker in the subject is determined at a time point later than the determination of the first level, and the two levels are compared to monitor the course of disease or the efficacy of the therapy.
- a pre- treatment level of Marker in the subject is determined prior to beginning treatment according to this invention; this pre-treatment level of Marker can then be compared to the level of Marker in the subject after the treatment commences, to determine the efficacy of the treatment.
- Kits or Pharmaceutical Systems Pharmaceutical compositions comprising the therapeutic components embodied herein, such as DACX, HSPC-NK cells, cytokines, fusion protein complex may be assembled into kits or pharmaceutical systems for use in ameliorating a neoplasia or infectious disease.
- Kits or pharmaceutical systems according to this aspect of the invention comprise a carrier means, such as a box, carton, tube, having in close confinement therein one or more container means, such as vials, tubes, ampoules, bottles and the like.
- the kits or pharmaceutical systems of the invention may also comprise associated instructions for using the fusion protein complex of the invention.
- the kit includes appropriate containers such as bags, bottles, tubes, to allow ex vivo treatment of immune cells using the fusion protein complex of the invention and/or administration of such cells to a patient.
- Kits may also include medical devices comprising the fusion protein complex of the invention.
- HSPC-NK cells cytokine-based culture systems for the generation of NK cell products from CD34 + hematopoietic stem and progenitor cells
- HMAs 2 hypomethylating agents
- NK-activating molecules such as NKG2D ligands
- their potential in upregulating NK-activating molecules, such as NKG2D ligands, on tumor cells through their epigenetic modulation and thereby sensitizing tumors to NK-cell-mediated killing has been reported in several studies and for different cancers, including AML. 14-17 Nonetheless, direct impact of HMAs on NK-cell functionalities has not been well established yet.
- conflicting data have been reported, describing either advantageous or a deleterious effect on NK cells. 15,18-22 Moreover, these data are mostly derived from in vitro studies, often performed at high drug concentrations that do not reflect plasma levels achieved in patients. Therefore, it remains unclear whether application of HMA therapy can augment NK cell–mediated anti-tumor responses in patients with AML.
- HSPC-NK cell generation NK cell products were generated from CD34 + HSPCs derived from umbilical cord blood obtained after normal full-term delivery and written informed consent (“Commissie Mensgebonden Onderzoek” CMO 2014/226).
- the culture protocol was adapted from Cany et al., 2 and Roeven et al., 23 and combined the use of the aryl hydrocarbon receptor antagonist StemRegenin-1 for the expansion of CD34 + HSPCs, together with interleukin-15 (IL-15) and IL-12 for the differentiation of NK cells.
- IL-15 interleukin-15
- IL-12 interleukin-15
- magnetic-activated cell sorting–isolated CD34 + cells (Miltenyi Biotec) were expanded for 9 to 10 days in the presence of stem cell factor, IL-7, Flt3L, and recombinant human thrombopoietin (all 25 ng/mL, ImmunoTools).
- IL-15 50 ng/mL, ImmunoTools
- stem cell factor (20 ng/mL
- IL-7 20 ng/mL
- IL-15 50 ng/mL
- IL-12 0.2 ng/mL, Miltenyi Biotech
- Medium was supplemented with 2 ⁇ M StemRegenin-1 (Cellagen Technology) from day 0 to day 21.
- HMAs AZA and DAC were purchased from Sigma-Aldrich.
- both drugs were dissolved in NaCl 0.9% at 0.1 to 1 mM, aliquoted them for single use, and stored them at -20°C. Drugs were used immediately after thawing, and treatment of the cell cultures was performed with limited light exposure.
- HMAs were used at concentrations similar to those achieved in plasma of HMA-treated patients, 24,25 with daily refreshments due to the short half-life of these agents. 26
- 2 to 4 mg of HMAs was kept on ice, protected from light, and dissolved with NaCl 0.9% just before the mice were injected. Treatments were applied daily, in line with current clinical practice.
- the preclinical xenograft model for AML was established by intrafemoral injection of the THP-1 cells, engineered to express the green fluorescent protein (GFP) and luciferase reporter genes (THP-1.LucGFP cells) for longitudinal tumor load monitoring by bioluminescence imaging, as previously described.
- GFP green fluorescent protein
- THP-1.LucGFP cells luciferase reporter genes
- HSPC-NK cells were resuspended in phosphate-buffered saline and injected intravenously through the tail vein. Survival of HSPC-NK cells in vivo was supported by subcutaneous administration of recombinant human IL-15 (1 ⁇ g per mouse; ImmunoTools), every 2 to 3 days after HSPC-NK cell infusion.
- ALT-803 (Altor BioScience) was administered subcutaneously at 0.2 mg/kg every 3 to 4 days.
- the experimental designs implemented for the treatment of mice with HMAs were based on current clinical practice (dosage, duration, and route of administration).
- the dosages applied in humans were translated for mouse studies (milligrams per kilogram) based on the study by Reagan-Shaw et al., 27 (see FIGS.12A, 12B for calculation).
- mice were given wet food to improve feeding and tolerability of the drugs. Mice were carefully monitored for weight and general conditions, and euthanized according to well-defined end points.
- Antibodies The following conjugated monoclonal antibodies were used for HSPC-NK cell phenotyping: CD56 (HCD56, Biolegend), NKG2A (Z199, Beckman Coulter), CD158a/h, b and e (clones HP-MA4, DX27, and DX9 respectively, used as a combined staining for KIRs), CD16 (3G8), NKG2D (1D11), NKp46 (9E2), NKp44 (P448), CD69 (FN50), TRAIL (RIK-2), FasL (NOK-1), CD11a (HI111), CXCR3 (G025H7, all Biolegend), and DNAM-1 (DX11; BD Bioscience).
- the panel for AML cell analysis included MIC-A/B (6D4, Biolegend), ULBP-1 (170818), ULBP-2 (165903), CD155 (300907), CD112 (610603, all R&D), TRAIL-R1 (DJR2-4), TRAIL-R2 (DJR2-1), and Fas (DX2, all Biolegend).
- IgG1 (MOPC21) and IgG2a (MOPC173, Biolegend) isotype controls were included for calculation of specific fluorescence intensities.
- HSPC-NK cells and THP-1.LucGFP cells were identified based on CD56 and GFP expression, respectively, in combination with human CD45 (J.33, Beckman Coulter) and mouse CD45 (30-F11, BD Biosciences) antibodies. Non-viable cells were excluded using SYTOX Blue (Life Technologies) or eFluor780 (eBiosciences) viability dye. Phenotypical analyses were performed using the Gallios flowcytometer and Kaluza analysis software (both Beckman Coulter).
- FCM Flow cytometry-based cellular assays: The K562, THP-1, and KG1a cell lines (ATCC) were cultured in Iscove's modified Dulbecco's medium (Invitrogen) supplemented with 10% fetal calf serum (Integro). The HLA-type of THP-1 and KG-1a cells is Bw6, C1/C1 and Bw4, C1/C2 respectively.
- Patient- derived primary AML cells were obtained from diagnostic leftover material in accordance with the Declaration of Helsinki and institutional guidelines and regulations (CMO 2013/064). All samples employed in this study were from bone marrow origin and contained >90% blasts based on flow cytometric determination of CD45, CD33 and CD34 expression.
- NK cell co-cultures with primary AML cells were supplemented with SCF and Flt3L (both 20 ng/ml), IL-3 (50 ng/ml, Immunotools), G-CSF (20 ng/ml; Amgen) and IL-15 (5 ng/ml). Cytotoxicity assays were performed as previously described 17 using CFSE-labeled target cells. In experiments where AML or NK cells were pre-treated with HMA, cells were harvested, counted, and equal numbers of viable cells were plated in all experimental conditions.
- the numbers of viable target cells were determined using the FC500 cytometer (Beckman Coulter) by gating on CFSE + cells, exclusion of the live/dead marker 7AAD (Sigma Aldrich) and back-gating on forward/side scatter. Target cells were plated alone as controls without killing. Proliferation of AML and NK cells was examined after labeling with CFSE, culture in the presence or absence of HMA, and subsequent FCM analysis.
- NK cell reactivity at the single cell level was determined following 4 hours stimulation with K562 cells in the presence of anti- CD107a (H4A3; BD Biosciences) and Brefeldin A (BD Biosciences) and subsequent intracellular staining for perforin (dG9, Biolegend) and IFN- ⁇ (B27; BD Biosciences).
- FCM analysis was performed with exclusion of dead cells using the Fixable Viability Dye eFluor780 (eBiosciences), gating on CD56 + Perforin + NK cells, and using unstimulated cells as control.
- Gene expression analysis Total RNA was isolated using PURELINK RNA mini-kit (Life Technologies) according to manufacturer’s instructions.
- cDNA was synthesized using M-MLV-reverse transcriptase (Invitrogen) in a standard reaction after which real-time PCR was performed using the following TAQMAN Gene expression assays (Life Technologies): IFNG (Hs00989291_m1), TNF (Hs01113624_g1), PRF1 (Hs00169473_m1), GZMB (Hs00188051_m1), TNFSF10 (Hs00921974_m1), and FASLG (Hs00181225_m1). Expression levels were normalized to ⁇ -Actin (ACTB, NM_001101.2) using the ⁇ Ct method. [0133] Statistical analysis: Statistical analyses was performed using GraphPad Prism 5 software.
- HMAs augment HSPC-NK efficacy against AML in vitro: To investigate the possibility of combining HSPC-NK cells with HMA therapy, the effect of AZA and DAC on 2 AML cell lines was first determined. THP-1 and KG1a cells were treated daily with HMAs by use of concentrations similar to those achieved in plasma of treated patients. 24,25 As expected, the number of viable AML cells decreased in a time- and dose-dependent manner compared with untreated cells (FIG.8).
- HMAs can sensitize AML cells to HSPC-NK cell– mediated killing.
- the combination treatment of HMAs and HSPC-NK cells was also studied across time for KG1a cells, without drug washout (FIGS. 1E-1F).
- the effects of HSPC-NK cells and HMAs were additive, resulting in potent reduction of AML cell numbers when combined together.
- the impact of HMAs was studied in HSPC-NK cell cocultures with patient-derived primary AML cells (Table 1; FIG. 2A).
- FIG. 9 a robust and dose-dependent effect of HMAs on primary AML cell viability and proliferation was confirmed (FIG. 9).
- HSPC-NK cell survival was not affected (FIG.
- HMAs differentially affect AML and HSPC-NK cells in vitro, and that HSPC-NK cells maintain potent anti-leukemic activity during HMA exposure. Most importantly, these findings demonstrate that HMAs can potentiate HSPC-NK cell killing activity, providing evidence that the combination of HPSC-NK cells with HMA therapy could result in additive to synergistic effects against AML. [0138] Exposure to low-dose HMA concentrations does not impair HSPC-NK cell cytolytic activity in vitro. Thereafter, the aim was to confirm that HMAs did not impair HSPC- NK cell functionalities.
- HSPC-NK cells were cultured either under proliferative (i.e., 20 ng/mL of IL-15 plus 1000 IU/mL of IL-2) or steady-state (i.e., 5 ng/mL of IL-15) conditions and treated with HMAs for 6 consecutive days (FIG.3A).
- HMA treatment did not affect HSPC-NK cell viability (data not shown); however, 1 ⁇ M AZA significantly decreased NK cell proliferation (FIG.3B).
- DAC had a minor impact, even at the highest concentration tested.
- Target-cell killing was only diminished after HSPC-NK pretreatment with the highest dosages of AZA and, to a lesser extent, with DAC (FIG. 3C). These observations were confirmed at the single-cell level with use of CD107a degranulation and intracellular interferon-gamma (IFN- ⁇ ) staining. As depicted in FIGS. 3D-3E, the proportion of CD107a + HSPC-NK cells on K562 stimulation was only negatively affected with 1- ⁇ M AZA but not with 0.1- ⁇ M DAC. The percentages of IFN- ⁇ + HSPC-NK cells remained similar. [0140] In addition, the phenotype of HSPC-NK cells cultured in the presence of HMAs was examined.
- KIR killer cell immunoglobulin-like receptor
- HSPC-NK cells Only the frequency of killer cell immunoglobulin-like receptor (KIR)–positive HSPC-NK cells increased, particularly after exposure to HMAs under proliferative conditions (FIG. 3F). NKG2A + KIR + HSPC-NK cells display higher IFN- ⁇ production capacity compared with NKG2A + KIR- cells. 2 Nonetheless, the increase in KIR expression after HMA treatment does not correlate with IFN- ⁇ responses (FIG. 3G). Expression of other maturation markers (NKG2A and CD16), as well as activating receptors, adhesion molecules, and death receptors, remained unchanged after exposure to HMAs in vitro (FIG.11).
- ALT-803 was used instead of rhIL-15 to support NK cell persistence in vivo.
- ALT- 803 is an IL-15 superagonist complex composed of an IL-15 mutant (N72D) bound to sushi domain of IL-15R ⁇ fused to IgG1 Fc. This complex has been shown to display higher stability and enhanced biological activity on NK cells in vivo as well as superior localization to the lymphoid organs when compared with rhIL-15. 28,29
- the effects of ALT-803 on HSPC-NK cells were validated in vitro (FIGS.14A, 14B).
- NK cell potentiation by DAC occurred in vivo
- the phenotype of THP-1 cells and HSPC-NK cells were analyzed after treatment of the mice with DAC.
- activating ligands and death receptors on THP-1 cells were examined by FCM (FIGS.6A, 6B).
- FCM FCM
- ULBP1 MIC-A/B
- CD112, TRAIL receptors, and Fas the expression levels of ULBP2 and CD155 were evidently increased with DAC, which supports the finding that HMAs can sensitize AML cells to HSPC- NK cell-mediated killing.
- HSPC-NK cells sustained expression of DNAM-1, NKp46, TRAIL, and CD69 was observed (FIG.
- DAC can potentiate HSPC-NK cell functionalities and anti-leukemic activity.
- combined HSPC-NK cell and DAC treatment resulted in improved control of THP-1 AML in NSG mice, whereas the combination with AZA did not yield an additive anti-leukemic effect.
- Multiple factors can explain the difference between both HMAs, including reduced NK cell proliferation and degranulation capacity upon AZA treatment, or a lower impact of AZA on NK cell cytolytic machinery, in vivo trafficking, and accumulation in the bone marrow compartment compared with DAC.
- HSPC-NK cell numbers were seen in the bone marrow of particularly DAC-treated mice. Shortly after infusion, NK cell homing to the bone marrow may be favored as a result of transient leukopenia occurring in HMA-treated mice. Moreover, at 2 weeks after NK cell transfer to tumor-bearing mice, HSPC-NK cell numbers were particularly increased within the tumor bed rather than in normal bone marrow, which provides evidence of a specific NK cell homing to the tumor site.
- HSPC-NK cells express and maintain high levels of the chemokine receptor CXCR3 in vivo and display a higher capacity of inflammatory cytokine production after DAC treatment.
- AML cells including THP-1
- THP-1 can secrete the inflammatory chemokines CXCL9, CXCL10, and CXCL11 upon exposure to IFN- ⁇ and TNF- ⁇ .
- HMAs can reactivate expression of endogenous retroviral elements, thereby eliciting type I and III IFN response, as well as induce expression of CXCL9 and CXCL10 by tumor cells.
- the DAC-induced IFN response may positively affect HSPC-NK cell activity and function as seen by the upregulation of immune mediators such as IFN- ⁇ , TNF- ⁇ , and perforin.
- DAC can modulate tumor environments and immune cell trafficking in vivo. Therefore, without wishing to be bound by theory, it was thought that, besides its direct anti-leukemic activity, DAC can potently maximize HSPC-NK cell responses presumably through epigenetic modulation. Besides sensitizing AML cells to NK cell–mediated killing, the data herein, also support that DAC can boost HSPC-NK cell cytolytic functions, enhance inflammatory responses, and upregulate expression of the activating receptor NKp44. This mechanism could result in a self-stimulatory loop, further promoting NK cell recruitment to the tumor site and sustained control of AML.
- DAC and HSPC-NK cell infusion with the IL-15 superagonist ALT-803 which has shown enhanced biological activity and a better half-life in vivo compared with rhIL-15, is a promising combinatorial approach to maximizing the anti-leukemic effect. This combination could further eradicate MRD and ultimately improve outcome of allogeneic stem cell transplantation. 33,34 In the model, a 5-day treatment with DAC was applied, whereas in patients it has been observed that 10-day treatment is more efficacious. 35-37 More investigations are being conducted regarding DAC-mediated changes in the tumor environment, inflammation, and chemokine and cytokine levels in diseased bone marrow.
- HSPC-NK cells On HSPC-NK cells, TIGIT is not expressed at the end of the culture process and is not upregulated in vivo regardless of DAC treatment. In contrast, CD96 is highly expressed on HSPC-NK cells. Screening of primary AML blasts with respect to risk group, cytogenetic abnormalities, or FAB classification could help identify best responders to combined DAC and HSPC-NK cell therapy. Finally, the studies herein, revealed that 2 cycles with combined DAC, HSPC-NK cells, and ALT-803 treatment is better, but the effect seems to decrease at 28 days. Therefore, further improvement with longer duration of DAC treatment of 10 days in each cycle and/or more treatment cycles is to be tested to maximize the combined effect of NK cells, DAC, and ALT-803.
- Umbilical cord blood CD34+ progenitor-derived NK cells efficiently kill ovarian cancer spheroids and in- traperitoneal tumors in NOD/SCI D/ IL2Rgnull mice. OncoImmunology. 2017; 6(8):e 1320630. 4. Dolstra H, Roeven MWH, Spanholtz J, et al. Successful transfer of umbilical cord blood CD34(+) hematopoietic stem and progenitor-derived NK cells in older acute myeloid leukemia patients. Clin Cancer Res.2017;23(15): 4107-4118. 5. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al.
- the aryl hydrocarbon receptor antagonist StemRegenin1 improves in vitro generation of highly functional natural killer cells from CD34 (+) hematopoietic stem and progenitor cells.
- Cutting edge CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J Immunol.2004;172(7):3994-3998. 39. Chan CJ, Martinet L, Gilfillan S, et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol.2014;15(5):431-438. 40. Blake SJ, Stannard K, Liu J, et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov.2016;6(4):446-459. 41. Hismenger E, Krueger TE, Lang JM.
- Example 2 A higher peritoneal NK cell frequency is correlated with better outcome in advanced ovarian cancer patients, and peritoneal NK cell functionality can be boosted by IL-15.
- Ovarian cancer OC
- OC has the highest mortality rate of female cancers. Because OC is generally asymptomatic until ascites or metastases beyond the ovaries have developed, patients are often diagnosed in advanced stage. Moreover, the presence and progression of ascites is associated with poor prognosis and poor quality of life. 1 Current therapy consists of debulking surgery combined with platinum/taxane chemotherapy, but the majority of patients develop a recurrence within 3 years.
- NK cells recognize stressed neoplastic cells through a balance of activating and inhibitory receptors. 18,19 Epidemiological research has shown that low NK cell activity is associated with increased cancer risk in humans.
- NK cells from ascites also have a low cytotoxic effect, but can be reinvigorated by IL-2 or IL-15 25 .
- IL-2 or IL-15 IL-15
- ALT-803 a fusion protein complex of IL-15 variant (N72D) bound to sushi domain of IL-15R ⁇ fused to IgG1 Fc, potently enhanced the function of ascites-derived NK cells and healthy donor peripheral blood NK cells exposed to ascites fluid. 26 Furthermore, many studies demonstrated that OC cells are susceptible to killing by cytokine-stimulated NK cells. 27-42 [0156] In this study, NK cell frequency, phenotype and functionality was characterized in ascites of advanced OC patients in relation to clinical outcome, and tested their responsiveness to IL-15 receptor mediated stimulation.
- Detection of cysts, endometriosis and adhesions at laparoscopy were exclusion criteria. These benign samples were processed and analyzed on the day of surgery. Medical records were retrospectively reviewed and relevant clinical and pathology data were extracted. Time of diagnosis was considered to be the date of the primary surgical procedure. Time from diagnosis to death was calculated for OS. PFS was calculated as time of last chemotherapy till diagnosis of histochemical or radiologic recurrence. Median survival was expressed in months. [0159] Flow cytometry.
- MNCs were stained with labeled antibodies, CD3 ECD (Biolegend), CD45 Krome Orange (R&D systems), CD56 PE-Cy5 (Biolegend), CD16 APC- Cy7 (Biologend), CD326 PerCPCy5.5 (Biolegend). Phenotypic analysis was performed using DNAM-1 FITC (Becton Dickinson), 2B4 FITC (Biolegend), NKG2A APC (Beckman Coulter), NKG2D APC (Biolegend), NKp30 PE (Biolegend) and NKp46 PE(Biolegend), isotype controls for IgG1 and IgG2a, (all Biolegend).
- MNCs or PBMCs were cultured overnight with 1 nM IL-15 (Immunotools), 1 nM ALT-803 (Altor Bioscience) or without cytokine support in IMDM with 10% FCS and 1% penicillin/streptomycin (p/s).
- 1 x 10 6 cells were co-cultured with 0.5 x 10 6 K562 cells, 0.5 x 10 6 SKOV-3 cells or without target cells for 4 hours in IMDM with 10% FCS and 1% p/s and anti-CD107a PE- Cy7 (Biolegend) in a 24-well plate. After 1h of co-culture, brefeldin A (BD) was added.
- BD brefeldin A
- the selected OC patient cohort was divided in two groups based on the median OS and PFS of 19 months and 6 months, respectively.
- the PFS and OS in the good survival group were 19.7 ⁇ 16.4 and 32.9 ⁇ 11.2 months, respectively.
- the PFS and OS in the poor survival group was only 3.2 ⁇ 2.3 and 10.3 ⁇ 4.4 months, respectively. Further characteristics of the two OC patient groups are shown in Table 1. Patients in the relatively good survival group were younger and were less often postmenopausal.
- NK, NKT and T cells were assessed by flow cytometry and compared those with peritoneal fluid of 10 patients with a benign gynecological disorder.
- OC ascites samples contained 38.8 ⁇ 24.8% lymphocytes, 40.5 ⁇ 24.7% CD45+ non-lymphocytes and 16.4 ⁇ 23.5% CD45- non- hematopoietic tumor cells, and the benign samples contained 58.7 ⁇ 40.4% lymphocytes and 36.5 ⁇ 34.1% non-lymphocytes within CD45+ leucocytes (Figure 21A).
- a significantly lower NK cell frequency was seen in OC patient ascites (mean 17.1 ⁇ 2.7%) compared to benign peritoneal fluid (48.1 ⁇ 6.8%, p ⁇ 0.0001; Figure 21B).
- lower CD3+ T cell and CD3+CD56+ NKT cell frequencies were observed within the lymphocyte population in OC patient ascites.
- Activating receptors on ascites-derived NK cells are lowly expressed in poor survival ovarian carcinoma patients.
- flow cytometry analysis was performed on the peritoneal NK cells of the selected patient cohort and benign ascites controls ( Figure 22). While 2B4 had equally high expression levels on both benign and malignant peritoneal fluid NK cells, NKG2D was low to undetectable on these NK cells.
- NKp30 was almost absent on NK cells in malignant samples (mean 3%), whereas it was significantly higher on NK cells from benign samples (mean 79%).
- NK cells recognize cancer cells through a balance of activating and inhibitory receptors. 18,19 Interestingly, epidemiological research has shown that low NK cell activity is associated with increased cancer risk in humans. 20 Moreover, NK cells have been identified to play a role in tumor surveillance due to enhanced surface expression of ligands for activating receptors by the DNA damage response.
- NK cell immunity there is limited data on the contribution of NK cell immunity on the clinical outcome of women with ovarian carcinoma.
- the percentage of CD56 + NK cells in ascites fluid is related to OS and PFS, and that ascites-derived NK cells have lower expression of activation markers than benign peritoneal fluid NK cells.
- ascites-resident NK cells have poor activity against SKOV-3 OC cells (similarly as PB-NK cells) they can be effectively boosted by IL-15 receptor mediated stimulation.
- OC patients were randomly selected from our ascites biobank. Overall, this is a relative poor prognosis group since these patients have large amounts of ascites. A remarkably high percentage of these patients did not undergo complete debulking surgery.
- NK cell anti-tumor activity Normally, for peripheral blood the best way to correlate clinical outcome parameters to NK cell anti-tumor activity would be to quantify absolute numbers. However, because volume and cellular density in ascites differs greatly between OC patients, it is believed that the frequency within lymphocyte population was the most objective way to compare cell populations. For future research, it would be interesting to investigate how the ascites NK cell frequency is compared to blood NK cell numbers, phenotype and function of the same OC patient. [0174] It has been previously reported that OC ascites NK cells have a lower expression of the activating markers NKp30, NKp46, NKG2D and DNAM-1 compared to healthy donor PB-NK cells.
- NKp30 is proposed as a prognostic biomarker based on its low expression on NK cells in poor prognosis patients.
- telomere frequency is correlated with better clinical outcome and addressed whether boosting with monomeric IL-15 or the IL-15 superagonist fusion complex, ALT803, could reinvigorate OC-activity.
- IL-15 or ALT-803 degranulation improved and IFN- ⁇ production increased, especially against SKOV-3 OC cells.
- ALT-803 is likely more potent in longer assays and in vivo because of its longer half-life (Han KP, et al. Cytokine.2011;56(3):804-810.
- Rhode PR et al.
- Combined IL-15 and IL-12 drives the generation of CD34-derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer.
- Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrow-residing human leukemia cells in NOD/SCID/IL2Rg(null) mice.
- the Aryl Hydrocarbon Receptor Antagonist StemRegenin1 Improves In Vitro Generation of Highly Functional Natural Killer Cells from CD34(+) Hematopoietic Stem and Progenitor Cells. Stem Cells Dev.2015;24(24):2886-2898. 31. Spanholtz J, Tordoir M, Eissens D, Preijers F, van der Meer A, Joosten I, Schaap N, de Witte TM, Dolstra H.
- Rosario M Liu B, Kong L, Collins LI, Schneider SE, Chen X, Han K, Jeng EK, Rhode PR, Leong JW, Schappe T, Jewell BA, Keppel CR, Shah K, Hess B, Romee R, Piwnica-Worms DR, Cashen AF, Bartlett NL, Wong HC, Fehniger TA.
- the IL-15-Based ALT-803 Complex Enhances FcgammaRIIIa-Triggered NK Cell Responses and In Vivo Clearance of B Cell Lymphomas. Clin Cancer Res. 2016;22(3):596-608. 43. Timmermans M, Sonke GS, Van de Vijver KK, van der Aa MA, Kruitwagen R.
- NKp30 expression is a prognostic immune biomarker for stratification of patients with intermediate-risk acute myeloid leukemia. Oncotarget.2017;8(30):49548-49563. 50.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmacology & Pharmacy (AREA)
- Genetics & Genomics (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Gastroenterology & Hepatology (AREA)
- General Engineering & Computer Science (AREA)
- Hematology (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Combination therapies which include natural killer (NK) cells, IL-15 super agonist and/or at least one chemotherapeutic agent enhance the anti-tumor activity of the NK cells. In certain embodiments, a method of preventing or treating cancer, comprises administering to a patient in need thereof, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent, thereby treating cancer.
Description
IL-15 FUSION PROTEIN ENHANCED ADOPTIVE CELL THERAPEUTICS CROSS-REFERENCE TO RELATED APPLICATION [0001] This application claims priority to U.S. provisional application no.62/935,464, filed on November 14, 2019, which is incorporated herein by reference in its entirety. SEQUENCE LISTING [0002] This application includes a Sequence Listing created on November 10, 2020 and submitted electronically as an ASCII file named 048277-535P01US_SL.ST25.txt that is 4,886 kilobytes and is incorporated herein in its entirety. FIELD OF THE INVENTION [0003] Embodiments of the invention are directed to combination therapies for the treatment of cancer or infectious diseases. In particular, compositions include natural killer (NK) cell adoptive transfer combined with hypomethylating agents (HMAs) for the treatment of cancer. As part of the therapy an IL-15 superagonist is included. BACKGROUND [0004] Harnessing natural killer (NK) cells against cancer is an emerging therapeutic approach, which is increasingly being explored for both hematological malignancies and solid tumors. Next to activation of the patient’s own NK cells by means of cytokine administration or redirection through treatment with tumor-targeting antibodies, current research also invests in ex vivo NK cell generation and expansion methods for adoptive cell therapy. SUMMARY [0005] In certain embodiments, a method of preventing or treating cancer, comprises administering to a patient in need thereof, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent, thereby treating cancer. [0006] In certain embodiments, the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL- 15Rα complex. The IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT-
803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. In certain embodiments, a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy. [0007] In certain embodiments, the adoptive cell therapy comprises hematopoietic stem cell transplantation, donor leukocyte infusion, adoptive transfer of natural killer cells (NK), T cells, B cells, chimeric antigen receptor- T cells (CAR-T), chimeric antigen receptor natural killer cells (CAR-NK) or combinations thereof. In certain embodiments, the NK cell is an allogeneic progenitor-derived NK cell. In certain embodiments, the adoptive cell therapy comprises transfer of allogeneic, autologous, syngeneic, related, unrelated, HLA- matched, HLA-mismatched or haploidentical cells. [0008] In certain embodiments, at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof. In another embodiment, the at least one chemotherapeutic agent is administered prior to the administration of the adoptive cell therapy. In another embodiment, the at least one chemotherapeutic agent is administered concomitantly with the administration of the adoptive cell therapy. In another embodiment, the at least one chemotherapeutic agent is administered after the administration of the adoptive cell therapy. [0009] In certain embodiments, the at least one chemotherapeutic agent comprises nucleoside analogs. In certain embodiments, the nucleoside analog is a hypomethylating agent. In certain embodiments, the hypomethylating agent comprises: 5-azacytidine, 5-aza-2'- deoxycytidine (5-AZA-CdR), zebularine, procainamide, procaine, hydralazine, epigallocathechin-3-gallate, RG108, MG98 or combinations thereof. In embodiments, the hypomethylating agent increases anti-tumor NK cell activity as compared to a non- hypomethylating agent treated control. In certain embodiments, the method of treating cancer further comprises administering one or more cytokines to the patient and/or culturing the cells prior to adoptive transfer with one or more cytokines. In certain embodiments, the method of treatment includes one or more chemotherapeutic agents specific for treatment of the type of cancer. [0010] In certain embodiments, the method of treating cancer further comprises administering to the patient, a therapeutically effective amount of an IL-15:IL-15Rα complex.
The IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. [0011] In certain embodiments, a method of treating cancer, comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of a hypomethylating agent, thereby treating cancer. In certain embodiments, the NK cells are allogeneic cells. These cells can be generated or obtained from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC). In certain embodiments, the hypothemylating agent is 5-aza-2'-deoxycytidine (5-AZA-CdR). In certain embodiments, the method of treating cancer further comprises administering to the patient, a therapeutically effective amount of an IL-15:IL-15Rα complex. The IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. [0012] In other embodiments, a kit for treating cancer comprises an adoptive cell therapy, at least one chemotherapeutic agent and directions for the use of the kit for the treatment of a cancer. In certain embodiments, the adoptive cell therapy comprises hematopoietic stem cells, donor leukocytes, T cells, or natural killer (NK) cells. In certain embodiments, the NK cell is an allogeneic progenitor-derived NK cell. In certain embodiments, the NK cells are generated from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC). In certain embodiments, the chemotherapeutic agent is a hypomethylating agent. [0013] In certain embodiments, the kit includes an IL-15:IL-15Rα complex, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. [0014] In certain embodiments, a method of treating cancer, comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of an IL-15:IL-15Rα complex or IL-15, thereby treating cancer. [0015] In certain embodiments, thee NK cells are obtained from one or more sources comprising ascites, peritoneum, lymph, blood, plasma or combinations thereof. In certain embodiments, the NK cells are obtained from ascites fluids.
[0016] In certain embodiments, the IL-15:IL-15Rα complex is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof. In certain embodiments, the IL-15:IL-15Rα complex is administered prior to the administration of the adoptive cell therapy. In certain embodiments, the IL-15:IL-15Rα complex is administered concomitantly with the administration of the adoptive cell therapy. In certain embodiments, the IL-15:IL-15Rα complex is administered after the administration of the adoptive cell therapy. In certain embodiments, the NK cells are optionally cultured with the IL-15:IL-15Rα complex prior to the administration of the adoptive cell therapy. In certain embodiments, the method of treating cancer, further comprises administering one or more chemotherapeutic agents. [0017] Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed.1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise. [0018] By “ameliorate” is meant decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease. [0019] By “analog” is meant a molecule that is not identical, but has analogous functional or structural features. For example, a polypeptide analog retains the biological activity of a corresponding naturally-occurring polypeptide, while having certain biochemical modifications that enhance the analog's function relative to a naturally occurring polypeptide. Such biochemical modifications could increase the analog's protease resistance, membrane permeability, or half-life, without altering, for example, ligand binding. An analog may include an unnatural amino acid. [0020] By “cancer” as used herein is meant, a disease, condition, trait, genotype or phenotype characterized by unregulated cell growth or replication as is known in the art; including colorectal cancer, as well as, for example, leukemias, e.g., acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia, AIDS related cancers such as Kaposi's sarcoma; breast cancers; bone cancers such as Osteosarcoma, Chondrosarcomas, Ewing's sarcoma,
Fibrosarcomas, Giant cell tumors, Adamantinomas, and Chordomas; Brain cancers such as Meningiomas, Glioblastomas, Lower-Grade Astrocytomas, Oligodendrocytomas, Pituitary Tumors, Schwannomas, and Metastatic brain cancers; cancers of the head and neck including various lymphomas such as mantle cell lymphoma, non-Hodgkins lymphoma, adenoma, squamous cell carcinoma, laryngeal carcinoma, gallbladder and bile duct cancers, cancers of the retina such as retinoblastoma, cancers of the esophagus, gastric cancers, multiple myeloma, ovarian cancer, uterine cancer, thyroid cancer, testicular cancer, endometrial cancer, melanoma, lung cancer, bladder cancer, prostate cancer, lung cancer (including non- small cell lung carcinoma), pancreatic cancer, sarcomas, Wilms' tumor, cervical cancer, head and neck cancer, skin cancers, nasopharyngeal carcinoma, liposarcoma, epithelial carcinoma, renal cell carcinoma, gallbladder adeno carcinoma, parotid adenocarcinoma, endometrial sarcoma, multidrug resistant cancers; and proliferative diseases and conditions, such as neovascularization associated with tumor angiogenesis, macular degeneration (e.g., wet/dry AMD), corneal neovascularization, diabetic retinopathy, neovascular glaucoma, myopic degeneration and other proliferative diseases and conditions such as restenosis and polycystic kidney disease, and other cancer or proliferative disease, condition, trait, genotype or phenotype that can respond to the modulation of its environment, alone or in combination with other therapies. [0021] By “cancer cell” is meant a cell exhibiting a neoplastic cellular phenotype, which may be characterized by one or more of, for example, abnormal cell growth, abnormal cellular proliferation, loss of density dependent growth inhibition, anchorage-independent growth potential, ability to promote tumor growth and/or development in an immunocompromised non-human animal model, and/or any appropriate indicator of cellular transformation. “Cancer cell” may be used interchangeably herein with “tumor cell”, “malignant cell” or “cancerous cell”, and encompasses cancer cells of a solid tumor, a semi- solid tumor, a primary tumor, a metastatic tumor, and the like. [0022] As used herein, the term “cancer therapy” refers to a therapy useful in treating cancer. Examples of anti-cancer therapeutic agents include, but are not limited to, e.g., surgery, chemotherapeutic agents, immunotherapy, growth inhibitory agents, cytotoxic agents, agents used in radiation therapy, anti-angiogenesis agents, apoptotic agents, anti- tubulin agents, and other agents to treat cancer, such as anti-HER-2 antibodies (e.g., HERCEPTINTM), anti-CD20 antibodies, an epidermal growth factor receptor (EGFR) antagonist (e.g., a tyrosine kinase inhibitor), HER1/EGFR inhibitor (e.g., erlotinib
(TARCEVATM)), platelet derived growth factor inhibitors (e.g., GLEEVECTM (Imatinib Mesylate)), a COX-2 inhibitor (e.g., celecoxib), interferons, cytokines, antagonists (e.g., neutralizing antibodies) that bind to one or more of the following targets ErbB2, ErbB3, ErbB4, PDGFR-beta, BlyS, APRIL, BCMA or VEGF receptor(s), TRAIL/Apo2, and other bioactive and organic chemical agents, etc. Combinations thereof are also contemplated for use with the methods described herein. [0023] By “disease” is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. Examples of diseases include neoplasias and viral infections. [0024] By the terms “effective amount” and “therapeutically effective amount” of a formulation or formulation component is meant a sufficient amount of the formulation or component, alone or in a combination, to provide the desired effect. For example, by “an effective amount” is meant an amount of a compound, alone or in a combination, required to ameliorate the symptoms of a disease relative to an untreated patient. The effective amount of active compound(s) used to practice the present invention for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an “effective” amount. [0025] In one embodiment, the effective amount is administered to a patient that has been diagnosed with cancer. The effective amount can result in the prevention of the development, recurrence, or onset of cancer and one or more symptoms thereof, to enhance or improve the efficacy of another therapy, reduce the severity, the duration of cancer, ameliorate one or more symptoms of cancer, prevent the advancement of cancer, cause regression of cancer, and/or enhance or improve the therapeutic effect(s) of another therapy. “Effective amount” also refers to the amount of a therapy that is sufficient to result in the prevention of the development, recurrence, or onset of cancer and one or more symptoms thereof, to enhance or improve the prophylactic effect(s) of another therapy, reduce the severity, the duration of cancer, ameliorate one or more symptoms of cancer, prevent the advancement of cancer, cause regression of cancer, and/or enhance or improve the therapeutic effect(s) of another therapy. In an embodiment of the invention, the amount of a therapy is effective to achieve one, two, three, or more results following the administration of one, two, three or more therapies: (1) a stabilization, reduction or elimination of the cancer
stem cell population; (2) a stabilization, reduction or elimination in the cancer cell population; (3) a stabilization or reduction in the growth of a tumor or neoplasm; (4) an impairment in the formation of a tumor; (5) eradication, removal, or control of primary, regional and/or metastatic cancer; (6) a reduction in mortality; (7) an increase in disease-free, relapse-free, progression-free, and/or overall survival, duration, or rate; (8) an increase in the response rate, the durability of response, or number of patients who respond or are in remission; (9) a decrease in hospitalization rate, (10) a decrease in hospitalization lengths, (11) the size of the tumor is maintained and does not increase or increases by less than 10%, preferably less than 5%, preferably less than 4%, preferably less than 2%, (12) an increase in the number of patients in remission, (13) an increase in the length or duration of remission, (14) a decrease in the recurrence rate of cancer, (15) an increase in the time to recurrence of cancer, and (16) an amelioration of cancer-related symptoms and/or quality of life. [0026] As used herein, the term “in combination” in the context of the administration of a therapy to a subject refers to the use of more than one therapy for therapeutic benefit. The term “in combination” in the context of the administration can also refer to the prophylactic use of a therapy to a subject when used with at least one additional therapy. The use of the term “in combination” does not restrict the order in which the therapies (e.g., a first and second therapy) are administered to a subject. A therapy can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject which had, has, or is susceptible to cancer. The therapies are administered to a subject in a sequence and within a time interval such that the therapies can act together. In a particular embodiment, the therapies are administered to a subject in a sequence and within a time interval such that they provide an increased benefit than if they were administered otherwise. Any additional therapy can be administered in any order with the other additional therapy. [0027] By “fragment” is meant a portion of a polypeptide or nucleic acid molecule. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acid molecule or polypeptide. For example, a fragment may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600,
700, 800, 900, or 1000 nucleotides or amino acids. However, the invention also comprises polypeptides and nucleic acid fragments, so long as they exhibit the desired biological activity of the full length polypeptides and nucleic acid, respectively. A nucleic acid fragment of almost any length is employed. For example, illustrative polynucleotide segments with total lengths of about 10,000, about 5,000, about 3,000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length (including all intermediate lengths) are included in many implementations of this invention. Similarly, a polypeptide fragment of almost any length is employed. For example, illustrative polypeptide segments with total lengths of about 10,000, about 5,000, about 3,000, about 2,000, about 1,000, about 5,000, about 1,000, about 500, about 200, about 100, or about 50 amino acids in length (including all intermediate lengths) are included in many implementations of this invention. [0028] The terms “isolated”, “purified”, or “biologically pure” refer to material that is free to varying degrees from components which normally accompany it as found in its native state. “Isolate” denotes a degree of separation from original source or surroundings. “Purify” denotes a degree of separation that is higher than isolation. [0029] A “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of this invention is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography. The term “purified” can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. For a protein that can be subjected to modifications, for example, phosphorylation or glycosylation, different modifications may give rise to different isolated proteins, which can be separately purified. [0030] Similarly, by “substantially pure” is meant a nucleotide or polypeptide that has been separated from the components that naturally accompany it. Typically, the nucleotides and polypeptides are substantially pure when they are at least 60%, 70%, 80%, 90%, 95%, or even 99%, by weight, free from the proteins and naturally-occurring organic molecules with they are naturally associated.
[0031] By “marker” is meant any protein or polynucleotide having an alteration in expression level or activity that is associated with a disease or disorder. [0032] By “neoplasia” is meant a disease or disorder characterized by excess proliferation or reduced apoptosis. Neoplastic conditions include, but are not limited to, cancers, sarcomas, tumors, leukemias, lymphomas, and the like. A neoplastic condition refers to the disease state associated with the neoplasia. Colon cancer (e.g., colorectal cancer), lung cancer and ovarian cancer are examples (non-limiting) of a neoplastic condition. Illustrative neoplasms for which the invention can be used include, but are not limited to leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease), Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, nile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, glioblastoma multiforme, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). In particular embodiments, the neoplasia is multiple myeloma, beta-cell lymphoma, urothelial/bladder carcinoma or melanoma. As used herein, “obtaining” as in “obtaining an agent” includes synthesizing, purchasing, or otherwise acquiring the agent. [0033] By “reduces” is meant a negative alteration of at least 5%, 10%, 25%, 50%, 75%, or 100%. [0034] By “specifically binds” is meant a compound or antibody that recognizes and binds a polypeptide of the invention, but which does not substantially recognize and bind
other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the invention. [0035] By “subject” is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, equine, canine, ovine, or feline. The subject is preferably a mammal in need of such treatment, e.g., a subject that has been diagnosed with B cell lymphoma or a predisposition thereto. The mammal is any mammal, e.g., a human, a primate, a mouse, a rat, a dog, a cat, a horse, as well as livestock or animals grown for food consumption, e.g., cattle, sheep, pigs, chickens, and goats. In a preferred embodiment, the mammal is a human. [0036] As used herein, the term “tumor” means a mass of transformed cells that are characterized by neoplastic uncontrolled cell multiplication and at least in part, by containing angiogenic vasculature. The abnormal neoplastic cell growth is rapid and continues even after the stimuli that initiated the new growth has ceased. The term “tumor” is used broadly to include the tumor parenchymal cells as well as the supporting stroma, including the angiogenic blood vessels that infiltrate the tumor parenchymal cell mass. Although a tumor generally is a malignant tumor, i.e., a cancer having the ability to metastasize (i.e. a metastatic tumor), a tumor also can be nonmalignant (i.e. non-metastatic tumor). Tumors are hallmarks of cancer, a neoplastic disease the natural course of which is fatal. Cancer cells exhibit the properties of invasion and metastasis and are highly anaplastic. [0037] Ranges provided herein are understood to be shorthand for all of the values within the range. For example, a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50. [0038] The terms “treating” and “treatment” as used herein refer to the administration of an agent or formulation to a clinically symptomatic individual afflicted with an adverse condition, disorder, or disease, so as to affect a reduction in severity and/or frequency of symptoms, eliminate the symptoms and/or their underlying cause, and/or facilitate improvement or remediation of damage. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated. Agents or formulations used in treatment may comprise cells or tissues.
[0039] Treatment of patients with neoplasia may include any of the following: Adjuvant therapy (also called adjunct therapy or adjunctive therapy) to destroy residual tumor cells that may be present after the known tumor is removed by the initial therapy (e.g. surgery), thereby preventing possible cancer reoccurrence; neoadjuvant therapy given prior to the surgical procedure to shrink the cancer; induction therapy to cause a remission, typically for acute leukemia; consolidation therapy (also called intensification therapy) given once a remission is achieved to sustain the remission; maintenance therapy given in lower or less frequent doses to assist in prolonging a remission; first line therapy (also called standard therapy); second (or 3rd, 4th, etc.) line therapy (also called salvage therapy) is given if a disease has not responded or reoccurred after first line therapy; and palliative therapy (also called supportive therapy) to address symptom management without expecting to significantly reduce the cancer. [0040] Unless specifically stated or obvious from context, as used herein, the term “or” is understood to be inclusive. Unless specifically stated or obvious from context, as used herein, the terms “a”, “an”, and “the” are understood to be singular or plural. [0041] Unless specifically stated or obvious from context, as used herein, the term “about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about. [0042] The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable or aspect herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof. [0043] Any compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein. [0044] The transitional term “comprising,” which is synonymous with “including,” “containing,” or “characterized by,” is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. By contrast, the transitional phrase “consisting of” excludes any element, step, or ingredient not specified in the claim. The transitional phrase “consisting essentially of” limits the scope of a claim to the specified
materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. [0045] Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All published foreign patents and patent applications cited herein are incorporated herein by reference. [0046] Genbank and NCBI submissions indicated by accession number cited herein are incorporated herein by reference. All other published references, documents, manuscripts and scientific literature cited herein are incorporated herein by reference. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. BRIEF DESCRIPTION OF THE DRAWINGS [0047] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee. [0048] FIGS.1A-1F are a series of graphs and an experimental protocol showing that HMAs can sensitize AML cells to NK cell–mediated killing. FIG.1A: Experimental design: THP-1 and KG1a cells were cultured in the presence of AZA or DAC at the indicated concentrations. Two days later, cells were harvested and used as targets for HSPC-NK cells with or without drug washout. The same numbers of viable AML cells were plated for each condition. The numbers of viable AML cells were determined by FCM after 1 to 3 days of coculture and were used for calculation of AML cell survival, NK- specific killing for each independent treatment, and overall effect of NK cells, as indicated. FIGS.1B-1D: Effect of HMAs pretreatment and HSPC-NK cells on THP-1 cells at day+1. Data were obtained after drug washout and are depicted as the mean ± standard error of the mean (SEM) of 3 independent experiments (1-way ANOVA; **P < .01). FIGS.1E-1F: Effect of HMA pretreatment and HSPC-NK cells on KG1a cells. Data were obtained without drug washout and are depicted as the mean ± SEM of 3 independent experiments.
Data shown in FIG.1E were obtained at day+1 and in FIG.1F are depicted the relative numbers of viable AML cells quantified from day 0 to day+3. “No NK” is indicated by open bars; “+NK” is indicated by solid bars (FIGS.1B, 1E). A, AZA; D, DAC. [0049] FIGS.2A-2E are a series of graphs and an experimental protocol showing that HSPC-NK cells in combination with HMAs potently combat primary AML cells in vitro. FIG.2A: Experimental design: Primary AML cells obtained from 5 different patients at diagnosis were stained with carboxyfluorescein diacetate succinimidyl ester and cultured in the presence of AZA or DAC using the indicated concentrations (5 x 104 AML cells per well). One day after, HSPC-NK cells (2.5 x 105 cells per well) were added and the drugs were refreshed daily. The numbers of viable AML cells were determined by FCM at day+3 of coculture. FIG.2B: Median HSPC-NK cell survival at day+3 (combined data obtained with 5 different primary AML samples). The number of NK cells quantified without HMAs was set at 100%. FIG.2C: Median AML cell survival at day+3 (combined data obtained with 5 different primary AML samples). The number of AML cells quantified without HMAs and NK cells is set at 100%. Data depicted in FIGS.2B-2C represent combined data obtained with 5 different primary AML samples and were analyzed with 1-way ANOVA. *** < .001; ** < .01. n.s., not significant. FIGS.2D-2E: The survival of AML cells from 2 different patients (pAML #4 and pAML #5) and corresponding effect of NK cells quantified at day+3 are depicted as the mean ± SEM of data obtained with 4 different HSPC-NK cell donors. Data were analyzed with 2-way ANOVA. *** < .001; * < .05. “No NK” is indicated by open bars; “+NK” is indicated by solid bars (FIG.2D). n.s., not significant. [0050] FIGS.3A-3G are a series of graphs, dot plots and an experimental protocol showing that low-dose HMAs do not impair HSPC-NK cell viability, proliferation, and cytolytic functions. FIG.3A: Experimental design: Carboxyfluorescein diacetate succinimidyl ester–labeled HSPC-NK cells were cultured under proliferative (high-dose IL-15 and IL-2) or steady-state (low-dose IL-15) conditions in the presence of AZA or DAC refreshed daily at the indicated concentrations. Cell proliferation, viability, and absolute numbers, as well as functionality and phenotype, were analyzed by FCM after 6 days of treatment. FIG.3B: Percentages of proliferating HSPC-NK cells under steady-state (left panel) and proliferative (right panel) conditions. Combined data from 3 independent experiments (mean ± SEM) are shown. FIG.3C: Specific killing of K562 and THP-1 cells by HSPC-NK cells pretreated with HMAs. The same numbers of viable NK cells were
plated in each experimental well after washout of the drug, and the killing of K562 and THP-1 cells was determined after overnight coculture using 1:1 E:T ratio. Data obtained with NK cells that were treated either under proliferative or steady-state conditions and performed with 6 different HSPC-NK cell donors are combined and depicted as mean ± SEM. FIGS.3D-3E: NK cell reactivity upon K562 stimulation and analyzed at the single- cell level by FCM. Combined data from 4 experiments using proliferative (n = 2) or steady-state (n = 2) conditions (FIG.3D), and representative dot plots of HSPC-NK cells cultured under proliferative conditions and treated with AZA 1 pM, DAC 0.1 pM, or without HMAs (FIG.3E) are shown. FIG.3F: Expression level of the maturation markers NKG2A, CD16, and killer immunoglobulin-like receptor-positive (KIR) cells on HSPC- NK cells following culture upon proliferative conditions in the presence of DAC 0.1 pM, or without HMAs. Mean ± standard deviation (SD) of 6 HSPC-NK cell donors is shown. FIG.3G: Representative dot plots of HSPC-NK cell IFN-y production capacity with respect to KIR expression following DAC 0.1 pM or no HMA treatment under proliferative conditions. Statistical analyses were performed with 1-way (FIG.3B-3D) and 2-way ANOVA (FIG.3E). NS, not stimulated. [0051] FIGS.4A-4D are a series of graphs showing that DAC, but not AZA, potentiates HSPC-NK cell anti-leukemic effect in vivo. FIG.4A: Adult NSG mice that were injected with luciferase-expressing THP-1 cells in their femur were treated with HMAs with use of dosages as indicated in the figure (in milligrams per millimeter squared) and monitored for tumor load progression every 3 to 4 days by bioluminescence imaging (BLI). Treatment was applied daily from days 4 to 9 in titration #1, and from days 2 to 8 and days 4 to 8 for AZA and DAC, respectively, in titration #2. AZA was injected subcutaneously and DAC intravenously based on current clinical practices. Data are depicted as mean 6 SD including 8 to 10 mice per group. Dotted lines indicate upper detection limit for tumor load monitoring (signal saturation). FIGS.4B-4D: THP-1–bearing mice were treated with HMAs with use of the same dosages as described in titration #2, and with a single infusion of HSPC-NK cells, applied at day 4. Survival of NK cells in vivo was supported by recombinant human IL-15, given subcutaneously every 2 to 3 days. FIG.4B: Experimental design. FIG.4C: Median tumor load at day 17. FIG.4D: Fold increase in tumor load after 2 weeks of treatment with DAC alone or in combination with HSPC-NK cells (calculated as the ratio between day 17 and day 3 signals). Data were analyzed with an unpaired, 2-tailed Student t test. IF, intrafemoral.
[0052] FIGS.5A-5C are a series of graphs and an experimental protocol showing treatment with HSPC-NK cell infusions and DAC improves control of AML in vivo. FIG. 5A: Experimental design: THP-1–bearing mice received 1 or 2 cycles of DAC (1.25 mg/m2), with or without HSPC-NK cells, which were infused on the first day from each cycle. Survival of HSPC-NK cells in vivo was supported by ALT-803 (an IL-15 superagonist complex), which was given subcutaneously every 3 to 4 days until day 35 (0.2 mg/kg per injection). Groups that were not treated with NK cells were also given ALT-803 as a control. FIG.5B: Impact of DAC on tumor load progression. Median tumor load from untreated mice and mice treated with 1 or 2 cycles of DAC is shown. FIG.5C: Impact of HSPC-NK cell infusions on tumor load progression in mice cotreated with 2 cycles of DAC. Data are shown as mean ± SD and were analyzed with 2-way ANOVA. One mouse in each DAC X 2 and DAC+NK X 2 group died at day 17 and day 15, respectively, likely due to DAC-related toxicities (weight loss >20% after second treatment cycle). These mice were excluded from the complete data set shown in this figure. [0053] FIGS.6A and 6B show a graph and scatter plot demonstrating that DAC treatment upregulates NK-inducing ligands on THP-1 cells in vivo. FIGS.6A, 6B: THP-1– bearing mice were treated with DAC (1.25 mg/m2) for 5 consecutive days. One week after the start of treatment, mice were euthanized and bone marrow cells were isolated for ex vivo analysis of THP-1 cells. Data obtained from 5 individual mice per treatment group were pooled to reach enough events by FCM (500-900 single and viable THP-1 cells acquired per test). The relative expression level of NKG2D and DNAM-1 ligands, as well as death receptors, are depicted in FIG.6A, and overlay plots with mean fluorescence intensities are shown in FIG.6B. Iso, isotype control. [0054] FIGS.7A-7C are a series of graphs and plots showing that DAC enhances the anti-leukemic potential of HSPC-NK cells through modulation of their maturation, activation, cytolytic functions, and trafficking to the bone marrow. FIG.7A-7C: Adult NSG mice were infused with HSPC-NK cells and treated with DAC (1.25 mg/m2) for 5 consecutive days. Persistence of NK cells in vivo was supported by subcutaneous administration of IL-15 (1 mg/injection) every 2 to 3 days. Mice were euthanized 1 or 2 weeks after NK cell infusion for detailed ex vivo analysis. FIG.7A: Phenotype of HSPC- NK cells analyzed 1 week after the start of DAC treatment. Analysis was performed on cells isolated from the spleen, including 5 mice per treatment group. The relative expression of various maturation and activation markers, as well as adhesion molecules and
homing receptor (right panel) and representative dot plots (left panel), is shown. FIG.7B: Gene expression profiling for the cytolytic machinery of NK cells, analyzed by quantitative reverse transcription polymerase chain reaction on cells isolated from livers, including 5 mice per treatment group. Data were normalized to human b-actin. FIG.7C: Absolute numbers of HSPC-NK cells were determined in peripheral blood (absolute number per milliliter) and mouse bone marrow either 1 week (experiment #1) or 2 weeks (experiment #2) after the start of treatment. Two femurs per mouse were combined in experiment #1, whereas experiment #2 was performed in IF THP-1–bearing mice and absolute NK cell counts were determined in each femur, with (Tumor BM) or without (NBM) tumor. Data shown in panel A were analyzed with 2-way ANOVA and data from FIGS.7B and 7C with an unpaired, 2-tailed Student t test. BM, bone marrow; IF, intrafemoral; ND, no drug; PB, peripheral blood. [0055] FIG.8 shows a series of graphs demonstrating that HMA have a direct and dose-dependent effect on AML cell viability and proliferation. THP-1 and KG-1a cells were stained with CFSE and cultured in the presence of Azacitidine (AZA) or Decitabine (DAC) using the indicated concentrations. Treatment was applied for 5 consecutive days and both HMA were refreshed daily. Cell number, viability and proliferation were analyzed by flow cytometry after 1, 3, and 5 days of culture. Cell viability was determined on forward/side scatter and exclusion of 7AAD, and the proliferation index was determined using the mean fluorescence intensity (MFI) of CFSE and calculated as the ratio between untreated (No HMA) and treated cells. Representative data from one experiment are shown (mean ± SD). [0056] FIG.9 shows a series of graphs demonstrating the effect of HMA on primary AML cell viability and proliferation. Patient-derived primary AML cells were stained with CFSE and cultured in the presence of Azacitidine (AZA) or Decitabine (DAC) using the indicated concentrations. Treatment was applied for 5 consecutive days and both HMA were refreshed daily. Cell number, viability and proliferation were analyzed by flow cytometry after 1, 3, and 5 days of culture. Cell viability was determined on forward/side scatter and exclusion of 7AAD, and the proliferation index was determined using the mean fluorescence intensity (MFI) of CFSE and calculated as the ratio between untreated (No HMA) and treated cells. Combined data obtained with 5 different AML samples and analyzed at day+5 are shown (mean ± sem). Data were analyzed using one-way ANOVA with comparison of each treatment condition to the control No HMA. *p<0.05, **p<0.01, ***p<0.001.
[0057] FIG.10 is a series of graphs demonstrating that HSPC-NK cells in combination with HMA potently combat primary AML cells in vitro. Primary AML cells obtained from 2 patients at diagnosis (pAML#4 and pAML#5) were cultured in the presence of HMA, with or without HSPC-NK cells which were added at day 0. The numbers of viable AML cells were determined by flow cytometry from day -1 (plating of AML cells) till day +3 of co-culture. All experimental conditions were tested in triplicates. The numbers of viable AML cells are depicted as the mean ± sem of data obtained with 4 different HSPC-NK cell donors. Data were analyzed using two-way ANOVA. ***p<0.001, *p<0.05, n.s. not significant. [0058] FIG.11 is a series of graphs demonstrating that phenotypical analysis of HSPC-NK cells following exposure to HMA in vitro. HSPC-NK cells cultured under steady state (5 ng/ml IL-15) or proliferative (20 ng/ml IL-15 and 103 U/ml IL-2) conditions and in the presence of Azacitidine (AZA) and Decitabine (DAC) were harvested at day+6 and analyzed by flow cytometry for their expression levels of maturation and activation markers, as well as adhesion molecules and homing receptor. Data obtained with one representative donor are shown. [0059] FIGS.12A, 12B are graphs demonstrating the tolerability and anti-leukemic activity of Azacitidine and Decitabine in mice. Adult NSG mice were injected IF with THP- 1.LucGFP cells. One week later, treatments with deescalating doses of HMA were initiated as indicated. Drugs were prepared and administered daily. Azacitidine (AZA) was injected subcutaneously and Decitabine (DAC) intravenously, based on current clinical practice. FIG. 12A: Weight follow-up (n=6 per group). Mice were monitored for weight and condition, and sacrificed when weight loss exceeded 20%. FIG.12B: Tumor load monitoring was performed weekly by bioluminescence imaging till day 28 (higher dosages) or 42 (for lower and tolerated dosages only). Dotted lines indicate upper detection limit for tumor load monitoring (signal saturation). Data are depicted as mean ± SD and include 3-6 mice per group per time- point. [0060] FIGS.13A, 13B demonstrate the effect of HMA therapy on mouse CD45+ cells vs. HSPC-NK cells in vivo. Adult NSG mice were infused with 5x106 HSPC-NK cells and treated for 5 consecutive days with 12.5 mg/m2 Azacitidine (AZA) or 1.25 mg/m2 Decitabine (DAC). Survival of NK cells in vivo was supported by subcutaneous administration of IL-15 (1 µg/injection) every 2-3 days. Mice were sacrificed one week after cell infusion and percentages of HSPC-NK cells within total human and mouse CD45+ cells
(FIG.13A) as well as absolute numbers (FIG.13B) were determined in peripheral blood and spleen. Data were analyzed using one-way ANOVA. **p<0.01, ***p<0.001, n.s. not significant. [0061] FIGS.14A-14C are graphs demonstrating the head-to-head comparison of recombinant human IL-15 and ALT-803 to support survival and proliferation of HSPC-NK cells. HSPC-NK cells were stained with eFluor450 proliferation dye and cultured in basal medium only, or supplemented with equimolar concentrations of rhIL-15 vs. ALT-803. Cytokines were refreshed after 3 days, and NK cell proliferation (FIG.14A), viability (using 7AAD live/dead marker, FIG.14B), and absolute numbers (FIG.14C) were determined at day+6 by flow cytometry. Combined data (mean ± sem) obtained with 3 HSPC-NK products are shown. Statistical analyses were performed using one-way ANOVA. *p<0.05, **p<0.01, ***p<0.001. [0062] FIGS.15A-15D are graphs demonstrating the impact of Azacitidine on THP-1 cells and HSPC-NK cells in vivo. Adult NSG mice were injected with THP-1 or HSPC-NK cells, treated with 12.5 mg/m2 Azacitidine (AZA) or 1.25 mg/m2 Decitabine (DAC), and sacrificed one week after the start of treatment for organ collection and detailed ex vivo analysis. FIGS.15A-15B: Phenotype of THP-1 cells (FIG.15A) and HSPC-NK cells (B, n=5) isolated from bone marrow and spleen respectively, and analyzed by flow cytometry. FIG.15C: Gene expression profiling for the cytolytic machinery of NK cells, analyzed by RT-qPCR on cells isolated from liver. Data were normalized to human β-Actin. FIG.15D: Absolute numbers of HSPC-NK cells determined in mouse bone marrow (2 femurs combined per mouse). Data were analyzed using two-way (FIG.15B) and one-way (FIGS 15C and 15D) ANOVA. *p<0.05, ***p<0.001, n.s. not significant. [0063] FIG.16 is a graph and histogram showing the impact of HMA on THP-1 cells and HSPC-NK cells in vitro. The phenotype of THP-1 cells was analyzed by flow cytometry following 3 days culture in the presence of azacytidine (AZA) or Decitabine (DAC) at the indicated concentrations. The mean fluorescence intensity (MFI) measured for several activating ligands and death receptors is shown in the upper panel. The MFI values were corrected by substraction of signals obtained with the relevant isotype control. The bottom panel illustrates the increase observed in ULBP-2 expression level. Histograms are gated on viable THP-1 cells and MFI are indicated into brackets.
[0064] FIG.17 is a series of graphs showing that HSPC-NK cells are highly capable of killing ovarian cancer (OC) cell lines and SKOV-3 spheroids in vitro. At an E:T ratio of 1:1, 24% to 79% killing of OC cell line monolayers was observed after overnight co-culture. This increased to >90% using higher E:T ratios. Next, we set up an OC spheroid culture system using SKOV-3 cells. After incubation with HSPC-NK cells for 24 hours, flow cytometry analysis of the co-cultures showed a dose response relationship between the amount of NK cells added and the percentage killing of OC spheroid cells, resulting in >90% target cell killing after co-culture with 2x106 HSPC-NK cells. [0065] FIG.18 is a series of graphs and images demonstrating that the HSPC-NK cells infiltrate and mediate efficient intra-tumoral killing in SKOV-3 spheroids. Progressive infiltration of the spheroids by HSPC-NK cells was demonstrated, peaking at 8 hours. At an E:T ratio with 6x105 HSPC-NK cells >50% of SKOV-3 target cells were killed by the infiltrated HSPC-NK cells. Migrating NK cells reached the core of the sphere after 5h of co- culture. Quantification by using the propidium iodide (PI) signal during co-culture showed that at 5h, the amount of PI-positive cells per nm2 is increasing 12 times, while in the untreated SKOV-3 spheres the amount only doubled. [0066] FIG.19 is a schematic and a series of graphs demonstrating that infusion of HSPC-NK cells in NSG mice bearing SKOV-3 tumors decreases tumor progression and improves survival. A rapid increase in tumor growth in the no treatment group was shown. In contrast, the tumor growth was effectively decreased in SKOV-3 bearing NOD/SCID/IL2Rgnull (NSG) mice that received 2 HSPC-NK cell injections either intravenously or intraperitoneally. Intraperitoneal NK cell infusion was superior in terms of limiting tumor size and dissemination in ovaries and the peritoneum compared to intravenous injection. [0067] FIG.20 is a schematic and a graph demonstrating the anti-tumor effect of intraperitoneal (ip) HSPC-NK cell therapy in combination with ALT-803 or recombinant human IL-15 in NSG mice ip engrafted with SKOV-3 tumors using bioluminescence imaging (BLI). All mice received nanogam (IgGs). Mice treated with HSPC-NK cells in combination with ALT-803 or IL-15 showed slower tumor growth compared to the control group (no HSPC-NK cells and no cytokine support). [0068] FIGS.21A-21H show the NK, NKT and T cell frequency in benign ascites and ascites from ovarian cancer patients. FIG.21A: Fraction of CD45+ lymphocytes (white),
CD45+ non-lymphocytes (grey) and CD45- cells (black) cell populations within peritoneal fluid of benign compared to malignant ovarian cancer patients, based on flow cytometric analysis of CD45 expression and forward/side scatter. FIG.21B: Percentage of NK cells, T cells, NKT cells and other lymphocytes within benign and malignant ascites. FIG.21C: Within the lymphocyte population the percentage of NK cells is depicted. The group of malignant ovarian carcinoma ascites patients is divided into good and poor survival based on the median survival of the analyzed patient cohort (n=20). FIG.21D: The NK cell population is subdivided based on CD56 bright and CD56 dim cells. FIG.21E: Overall survival curve of OC patients groups with low and high CD56+ NK cell frequencies in ascites. FIG.21F: Overall survival curve of OC patients groups with low and high CD3+ T cell frequencies in ascites. FIGS.21G-21H: Progression free survival curves for low and high CD56+ NK cell and CD3+ T cell frequencies in ascites. Error bars represent mean + SEM. When 2 groups were compared the Student T-test was used whereas a one-way ANOVA with Bonferroni correction was performed when comparing 3 groups. *** P=0.001 [0069] FIG.22 shows the expression of activating receptors on CD45+CD3-CD56+ NK cells. Percentage positive 2B4, NKG2D, NKp46, NKp30, DNAM-1 and NKG2A NK cells of CD56+ NK cells in benign and malignant peritoneal fluid. The group of malignant ovarian carcinoma ascites patients is divided into lower than median overall survival and higher than median overall survival. Error bars represent mean + SEM. One way ANOVA with Bonferroni correction was performed when comparing the groups. *= p<0.05, ***=p<0.001. [0070] FIGS.23A-23D show results from a degranulation assay comparing NK cells in healthy donor (HD) peripheral blood mononuclear cells (PBMCs) with ascites mononuclear cells (MNCs). Percentage CD56+ NK cells positive for: FIG.23A. CD69, FIG. 23B. TRAIL, FIG.23C. CD107a, FIG.23D. IFN-γ, after 4h stimulation with no target cells, K562 or SKOV-3 tumor cells. Error bars represent mean + SEM. Open symbols depict HD PBMCs, closed symbols depict ascites MNCs. One way ANOVA with Bonferroni correction was performed when comparing the groups. [0071] FIGS.24A-24F show results from a degranulation assay comparing ascites CD56+ NK cells with and without monomeric IL-15 or ALT-803 stimulation. FIG.24A-24C: Percentage CD56+ NK cells positive for CD107a after 4h co-culture with no stimulation (FIG.24A), K562 cells (FIG.24B) or SKOV-3 cells (FIG.24C). FIGS.24D-24F. Percentage CD56+ NK cells positive for IFN-γ after 4h co-culture with no stimulation (FIG.24D), K562
cells (FIG.24E) or SKOV-3 cells (FIG.24F). Error bars represent mean + SEM. One way ANOVA with Bonferroni correction was performed when comparing the groups. *=p<0.05, **=p<0.01, ***=p<0.001 DETAILED DESCRIPTION [0072] The invention is based in part, on the discovery that the combination of adoptive cell therapy and chemotherapeutic agents, results in an enhancement of the adoptive cell therapeutic response in the treatment of cancer. In the examples section which follows, the inventors report on a comparative study of azacitidine (AZA) and decitabine (DAC) in combination with allogeneic NK cells generated from CD341 hematopoietic stem and progenitor cells (HSPC-NK cells) in in vitro and in vivo acute myeloid leukemia (AML) models. In vitro, low-dose HMAs did not impair viability of HSPC-NK cells. Furthermore, low-dose DAC preserved HSPC-NK killing, proliferation, and interferon gamma production capacity, whereas AZA diminished their proliferation and reactivity. Importantly, it was shown that HMAs and HSPC-NK cells could potently work together to target AML cell lines and patient AML blasts. In vivo, both agents exerted a significant delay in AML progression in NOD/SCID/IL2Rgnull mice, but the persistence of adoptively transferred HSPC-NK cells was not affected. Infused NK cells showed sustained expression of most activating receptors, upregulated NKp44 expression, and remarkable killer cell immunoglobulin-like receptor acquisition. Besides upregulation of NKG2D- and DNAM-1– activating ligands on AML cells, DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells. In addition, treatment resulted in increased numbers of HSPC- NK cells in the bone marrow compartment, providing evidence that DAC could positively modulate NK cell activity, trafficking, and tumor targeting. [0073] Nucleoside Analogs [0074] Nucleoside analogs have been used clinically for the treatment of viral infections and cancer. Most nucleoside analogs are classified as anti-metabolites. After they enter the cell, nucleoside analogs are successively phosphorylated to nucleoside 5'-mono- phosphates, di-phosphates, and tri-phosphates. In certain aspects, the drug is a nucleoside drug. A “nucleoside” includes (e.g., consists of) a nucleobase (such as a purine or pyrimidine) bound covalently to a pentose monosaccharide, e.g., a 5-carbon sugar such as ribose. By “nucleoside analogue” is meant a nucleoside in which either the nucleobase and/or the pentose monosaccharide are unnatural. A nucleoside drug includes a nucleoside analogue
that causes a physiological change within the body of an individual. In certain aspects, the nucleoside analogue is any one of an adenoside/deoxyadenosine analogue, cytidine/deoxycytidine, guanosine/deoxyguanosine analogue, thymidine/deoxythymidine analogue, or a deoxyuridine analogue. In some embodiments, the nucleoside analogue may be any one of gemcitabine, cytarabine, troxacitabine, decitabine, cladribine, fludarabine, clofarabine, or 2'-C-cyano-2'-deoxy-1-3-D-arabino-pentofuranosylcytosine (CNDAC). According to one embodiment, the nucleoside analogue is decitabine (DAC). [0075] 5-Azacytidine (National Service Center designation NSC-102816; CAS Registry Number 320-67-2), also known as azacitidine, AZA, or 4-amino-1-β-D- ribofuranosyl-1,3,5-triazin-2(1H)-one, is currently marketed as the drug product VIDAZATM. 5-Azacytidine is a nucleoside analog, more specifically a cytidine analog.5-Azacytidine is an antagonist of its related natural nucleoside, cytidine.5-Azacytidine and 5-aza-2'- deoxycytidine (also known as decitabine (DAC), an analog of deoxycytidine). [0076] In certain embodiments, at least one chemotherapeutic agent comprises nucleoside analog. In certain embodiments, the nucleoside analog is a hypomethylating agent. Hypomethylating agents have been approved by the US Food and Drug Administration in the treatment of cancer. For example, VIDAZATM (azacitidine for injection) is indicated for treatment of patients with the following French-American-British (FAB) myelodysplastic syndromes subtypes: refractory anemia (RA) or refractory anemia with ringed sideroblasts (if accompanied by neutropenia or thrombocytopenia or requiring transfusions), refractory anemia with excess blasts (RAEB), refractory anemia with excess blasts in transformation (RAEB-T), and chronic myelomonocytic leukemia (CMMoL). DACOGENTM (decitabine for injection) is indicated for treatment of patients with myelodysplastic syndromes (MDS) including previously treated and untreated, de novo and secondary MDS of all French- American-British subtypes (refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation, and chronic myelomonocytic leukemia) and intermediate-1, Intermediate-2, and high-risk International Prognostic Scoring System groups. [0077] In certain embodiments, the hypomethylating agent comprises: 5-azacytidine, 5-aza-2'-deoxycytidine (5-AZA-CdR), zebularine, procainamide, procaine, hydralazine, epigallocathechin-3-gallate, RG108, MG98 or combinations thereof. In certain embodiments, the hypomethylating agent is 5-aza-2'-deoxycytidine ((decitabine (DAC)).
[0078] In certain embodiments, the at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof. In another embodiment, the at least one chemotherapeutic agent is administered prior to the administration of the adoptive cell therapy. In another embodiment, the at least one chemotherapeutic agent is administered concomitantly with the administration of the adoptive cell therapy. In another embodiment, the at least one chemotherapeutic agent is administered after the administration of the adoptive cell therapy. [0079] In embodiments, the hypomethylating agent increases anti-tumor NK cell activity as compared to a non-hypomethylating agent treated control. In certain embodiments, the method of treating cancer further comprises administering one or more cytokines to the patient and/or culturing the cells prior to adoptive transfer with one or more cytokines. [0080] The methods of the invention may include administration of second therapeutic agent or treatment with a second therapy (e.g., a therapeutic agent or therapy that is standard in the art). Exemplary therapeutic agents include chemotherapeutic agents. A “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include Erlotinib (TARCEVATM, Genentech/OSI Pharm.), Bortezomib (VELCADETM, Millennium Pharm.), Fulvestrant (FASLODEXTM, Astrazeneca), Sutent (SU11248, Pfizer), Letrozole (FEMARATM, Novartis), Imatinib mesylate (GLEEVECTM, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (EloxatinTM, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNETM, Wyeth), Lapatinib (GSK572016, GlaxoSmithKline), Lonafarnib (SCH 66336), Sorafenib (BAY43- 9006, Bayer Labs.), and Gefitinib (IRESSATM, Astrazeneca), AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as Thiotepa and CYTOXANTM cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozcicsin, carzcicsin and bizcicsin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride,
melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin γ1 and calicheamicin omega 1 (Angew Chem. Intl. Ed. Engl. (1994) 33:183- 186); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, anthramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCINTM doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, strcptonigrin, strcptozocin, tubcrcidin, ubenimcx, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacytidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSKTM polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2''- trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosinc; arabinoside (“Ara-C”); cyclophosphamidc; thiotcpa; taxoids, e.g., TAXOLTM paclitaxel (Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANETM Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Ill.), and TAXOTERETM doxetaxel (Rhone-Poulenc Rorer, Antony, France); chloranbucil; GEMZARTM gemcitabine; 6-thioguanine;
mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINETM vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. [0081] Also included in this definition of “chemotherapeutic agent” are: (i) anti- hormonal agents that act to regulate or inhibit hormone action on tumors such as anti- estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEXTM (tamoxifen)), raloxifene, droloxifene, 4- hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTONTM (toremifene); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASETM (megestrol acetate), AROMASINTM (exemestane), formestanie, fadrozole, RIVISORTM (vorozole), FEMARATM (letrozole), and ARIMIDEXTM (anastrozole); (iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); (iv) aromatase inhibitors; (v) protein kinase inhibitors; (vi) lipid kinase inhibitors; (vii) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras; (viii) ribozymes such as a VEGF expression inhibitor (e.g., ANGIOZYMETM (ribozyme)) and a HER2 expression inhibitor; (ix) vaccines such as gene therapy vaccines, for example, ALLOVECTINTM vaccine, LEUVECTINTM vaccine, and VAXIDTM vaccine; PROLEUKINTM rIL-2; LURTOTECANTM topoisomerase 1 inhibitor; ABARELIXTM rmRH; (x) anti-angiogenic agents such as bevacizumab (AVASTINTM, Genentech); and (xi) pharmaceutically acceptable salts, acids or derivatives of any of the above. [0082] Adoptive Cell Therapy [0083] As described in detail ion the examples section which follows, hypomethylating agent, DAC positively modulates the in vivo anti-leukemic potential of adoptively transferred HSPC-NK cells through AML cell sensitization, enhancement of NK cell maturation, and cytolytic functions, as well as improves on NK cell trafficking and accumulation at the tumor site. Allogeneic NK cells were generated from CD34+
hematopoietic stem and progenitor cells (HSPC-NK cells) for use in the adoptive therapy treatments. [0084] Adoptive cell therapy (ACT) (including allogeneic and autologous hematopoietic stem cell transplantation (HSCT) and recombinant cell (i.e., CAR T) therapies) is the treatment of choice for many malignant disorders (for reviews of HSCT and adoptive cell therapy approaches, see, Rager & Porter, Ther Adv Hematol (2011) 2(6) 409–428; Roddie & Peggs, Expert Opin. Biol. Ther. (2011) 11(4):473-487; Wang et al. Int. J. Cancer. (2015)136, 1751–1768; and Chang, Y.J. and X.J. Huang, Blood Rev, 2013.27(1): 55-62). Such adoptive cell therapies include, but are not limited to, allogeneic and autologous hematopoietic stem cell transplantation, donor leukocyte (or lymphocyte) infusion (DLI), adoptive transfer of tumor infiltrating lymphocytes, or adoptive transfer of T cells or NK cells (including recombinant cells, i.e., CAR T, CAR NK, gene-edited T cells or NK cells, see Hu et al. Acta Pharmacologica Sinica (2018) 39: 167–176, Irving et al. Front Immunol. (2017) 8: 267). Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from transferred cells is important for the elimination of residual tumor cells. The efficacy of ACT as a curative option for malignancies is influenced by a number of factors including the origin, composition and phenotype (lymphocyte subset, activation status) of the donor cells, the underlying disease, the pre-transplant conditioning regimen and post-transplant immune support (i.e., IL-2 therapy) and the graft-versus-tumor (GVT) effect mediated by donor cells within the graft. Additionally, these factors must be balanced against transplant-related mortality, typically arising from the conditioning regimen and/or excessive immune activity of donor cells within the host (i.e., graft-versus-host disease, cytokine release syndrome, etc.). [0085] Approaches utilizing adoptive NK cell therapy have become of significant interest. In patients receiving autologous HSCT, blood NK cell numbers recover very early after the transplant and the levels of NK cells correlate with a positive outcome (Rueff et al., 2014, Biol. Blood Marrow Transplant.20, 896–899). Although therapeutic strategies with autologous NK cell transfer have had limited success due to a number of factors, adoptive transfer of ex vivo-activated allogeneic (or haplo-identical) NK cells has emerged as a promising immunotherapeutic strategy for cancer (Guillerey et al.2016. Nature Immunol.17: 1025-1036). The activity of these cells is less likely to be suppressed by self-MHC molecules compared to autologous NK cells. A number of studies have shown that adoptive therapy with haploidentical NK cells to exploit alloreactivity against tumor cells is safe and can
mediate significant clinical activity in AML patients. Taking these findings further, recent studies have focused on optimizing ex vivo activation/expansion methods for NK cells or NK precursors (i.e., stem cells) and pre-transplant conditioning and post-transplant immune support strategies; use of NK cell lines or recombinant tumor-targeting NK cells; evaluation of combination therapies with other agents such as therapeutic Ab, immunomodulatory agents (lenalidomide), and anti-KIR and checkpoint Abs. In each case, these strategies could be complemented by the combination therapeutic approach of the invention, which has the capacity to augment NK cell proliferation and activation. As indicated herein, DAC potentiated HSPC-NK cell anti-leukemic activity in vivo. Besides upregulation of NKG2D- and DNAM-1– activating ligands on AML cells, DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells. In addition, treatment resulted in increased numbers of HSPC-NK cells in the bone marrow compartment, providing evidence that DAC positively modulated NK cell activity, trafficking, and tumor targeting. [0086] Natural Killer Cells: One of the major types of circulating mononuclear cells is that of the natural killer, or NK, cell (M. Manoussaka et al., Journal of Immunology 158:112-119, 1997). Originally defined based on their ability to kill certain tumors and virus- infected cells, NK cells are now known as one of the components of the early, innate immune system. In addition to their cytotoxic capabilities, NK cells serve as regulators of the immune response by releasing a variety of cytokines. In addition, the generation of complex immune responses is facilitated by the direct interaction of NK cells with other cells via various surface molecules expressed on the NK cells. [0087] NK cells are derived from bone marrow precursors (O. Haller et al., Journal of Experimental Medicine 145:1411-1420, 1977). NK cells appear to be closely related to T cells, and the two cell types share many cell surface markers (M. Manoussaka et al., 1997). As noted above, these cell surface markers play a significant role in NK cell activity. For example, murine NK cells express specific antigens on their surfaces, such as asialo GM1, NK1, and NK2 antigens (D. See et al., Scand. J. Immunol.46:217-224, 1997), and the administration of antibodies against these antigens results in depletion of NK cells in vivo (Id.). [0088] Similarly to cytotoxic T lymphocytes (CTL), NK cells exert a cytotoxic effect by lysing a variety of cell types (Srivastava, S., Lundqvist, A. & Childs, R. W. Natural killer cell immunotherapy for cancer: a new hope. Cytotherapy 10, 775–783; 2008). These include normal stem cells, infected cells, and transformed cells. The lysis of cells occurs
through the action of cytoplasmic granules containing proteases, nucleases, and perforin. Cells that lack MHC class I are also susceptible to NK cell-mediated lysis (H. Reyburn et al., Immunol. Rev.155:119-125, 1997). In addition, NK cells exert cytotoxicity in a non-MHC restricted fashion (E. Ciccione et al., J. Exp. Med.172:47, 1990; A. Moretta et al., J. Exp. Med.172:1589, 1990; and E. Ciccione et al., J. Exp. Med.175:709). NK cells can also lyse cells by antibody-dependent cellular cytotoxicity. [0089] As noted above, NK cells mediate some of their functions through the secretion of cytokines, such as interferon γ (IFN-γ), granulocyte-macrophage colony- stimulating factors (GM-CSFs), tumor necrosis factor α (TNF-α), macrophage colony- stimulating factor (M-CSF), interleukin-3 (IL-3), and IL-8. NK cell cytotoxic activity is regulated through a balance of activating and inhibitory receptors that enables fine-tuned control of cytotoxic activity, preventing cytotoxicity against healthy cells, while maintaining effective cytotoxic capacity against tumor cells. Indeed, multiple studies have demonstrated the safety of adoptive NK cell transfer and clinical anti-cancer effects, highlighting the potential for NK cells as an effective cancer immunotherapy ((Parkhurst, M. R., et al. Clin Cancer Res 17, 6287–6297 (2011); Ruggeri, L. et al. Science 295, 2097– 2100, (2002); Miller, J. S. et al. Blood 105, 3051–3057, (2005; Bachanova, V. et al. Blood 123, 3855–3863, (2014); Rubnitz, J. E. et al. J Clin Oncol 28, 955–959, (2010)). For example, cytokines including IL-2, IL-12, TNF-α, and IL-1 can induce NK cells to produce cytokines. IFN-α and IL-2 are strong inducers of NK cell cytotoxic activity (G. Trinichieri et al., Journal of Experimental Medicine 160:1147-1169, 1984; G. Trinichieri and D. Santoli, Journal of Experimental Medicine 147: 1314-1333, 1977). The presence of IL-2 both stimulates and expands NK cells (K. Oshimi, International Journal of Hematology 63:279- 290, 1996). IL-12 has been shown to induce cytokine production from T and NK cells, and augment NK cell-mediated cytotoxicity (M. Kobayashi et al., Journal of Experimental Medicine 170:827-846, 1989). [0090] NK cells are involved in both the resistance to and control of cancer spread. Since the advent of the cancer immune surveillance concept, the adoptive transfer of immune cells, particularly T cells and natural killer (NK) cells, has emerged as a targeted method of harnessing the immune system against cancer (Kroemer, G., Senovilla, L., Galluzzi, L., Andre, F. & Zitvogel, L. Natural and therapy-induced immunosurveillance in breast cancer. Nat Med 21, 1128–1138, (2015)). NK cells have garnered immense attention as a promising immunotherapeutic agent for treating cancers.
NK cells are critical to the body’s first line of defense against cancer due to their natural cytotoxicity against malignant cells (Srivastava, S., et al., Cytotherapy 10, 775–783; 2008). [0091] NK cells have been expanded from multiple sources, including peripheral blood and umbilical cord blood (CB) ((Denman, C. J. et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One 7, e30264, (2012); Knorr, D. A. et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med 2, 274-283, (2013); Shah, N. et al. Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity. PLoS One 8, e76781, (2013); Woll, P. S. et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood 113, 6094-6101, (2009)). Ex vivo NK cell expansion methods have been developed using cytokines in combination with artificial antigen-presenting cells (aAPCs) as feeder cells ((Denman, C. J. et al. PLoS One 7, e30264, (2012); Berg, M. et al. Cytotherapy 11, 341–355, (2009); Gong, W. et al. Tissue Antigens 76, 467–475, (2010); Zhang, H. et al., J Immunother 34, 187–195, (2011)). [0092] ALT-803 [0093] In certain embodiments, a IL-15 receptor α/IgG1 Fc fusion protein (IL- 15N72D:IL-15RαSu/Fc) can be administered as part of the adoptive cell therapy and can include one or more chemotherapeutic agents. ALT-803 comprises an IL-15 mutant with increased ability to bind IL-2Rβγ and enhanced biological activity (U.S. Patent No.8,507, 222, incorporated herein by reference). This super-agonist mutant of IL-15 was described in a publication (Zu et al., 2009 J Immunol, 183: 3598-3607, incorporated herein by reference). This IL-15 super-agonist in combination with a soluble IL-15α receptor fusion protein (IL- 15RαSu/Fc) results in a fusion protein complex with highly potent IL-15 activity in vitro and in vivo (Han et al., 2011, Cytokine, 56: 804-810; Xu, et al., 2013 Cancer Res.73:3075-86, Wong, et al., 2013, OncoImmunology 2:e26442). The IL-15 super agonist complex comprises an IL-15 mutant (IL-15N72D) bound to an IL-15 receptor α/IgG1 Fc fusion protein (IL-15N72D:IL-15RαSu/Fc) is referred to as “ALT-803.” [0094] Pharmacokinetic analysis indicated that the fusion protein complex has a half- life of 25 hours following i.v. administration in mice. ALT-803 exhibits impressive anti-
tumor activity against aggressive solid and hematological tumor models in immunocompetent mice. It can be administered as a monotherapy using a twice weekly or weekly i.v. dose regimen or as combinatorial therapy with an antibody. The ALT-803 anti-tumor response is also durable. Tumor-bearing mice that were cured after ALT-803 treatment were also highly resistant to re-challenge with the same tumor cells indicating that ALT-803 induces effective immunological memory responses against the re-introduced tumor cells. [0095] The sequence for ALT-803 (IL-15N72D associated with a dimeric IL- 15RαSu/Fc fusion protein) comprises SEQ ID NO: 1: IL-15N72D protein sequence (with leader peptide) METDTLLLWVLLLWVPGSTG- [Leader peptide] NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLESGDAS IHDTVENLIILANDSLSSNGNVTESGCKECEELEEKNIKEFLQSFVHIVQMFINTS [IL-15N72D] IL-15RαSu/Fc protein sequence (with leader peptide) MDRLTSSFLLLIVPAYVLS- [Leader peptide] ITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSSLTECVLNKATNVAHWT TPSLKCIR- [IL-15RαSu] EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDP EVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSN KALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWES NGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ KSLSLSPGK [IgG1 CH2-CH3 (Fc domain)]. [0096] Accordingly, in certain embodiments, in certain embodiments, the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL-15Rα complex. The IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. In certain embodiments, a pharmaceutical composition comprising a therapeutically effective
amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy. [0097] In certain embodiments, the method of treating cancer comprises administering to the patient, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of an IL-15:IL- 15Rα complex. The IL-15/IL15Rα complex is an IL-15N72D:IL-15RαSu/Fc complex (ALT- 803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules. In certain embodiments, a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent is also administered to the patient as part of a combination therapy. In certain embodiments, the NK cells are contacted with the ALT-803 fusion protein complex. The ex vivo incubation of NK cells with the fusion protein complex results in induction of CIML NK cell exhibiting elevated activation markers, increased cytotoxicity against tumor cells and enhanced production of IFN-γ. Additionally, the fusion protein complex is capable of activating human NK cell lines. Moreover, methods are provided for augmenting immune responses and treating neoplasia and infection disease by direct administration of the fusion protein complex of the invention or administration of immune cells activated by the fusion protein complex of the invention. [0098] Immune Modulating Molecules [0099] In certain embodiments, one or more immune modulating compounds can be administered as part of the treatment plan. The immune-modulating molecules comprise, but are not limited to cytokines, lymphokines, NK cell stimulating factors, T cell co-stimulatory ligands, etc. An immune-modulating molecule positively and/or negatively influences the humoral and/or cellular immune system, particularly its cellular and/or non-cellular components, its functions, and/or its interactions with other physiological systems. The immune-modulating molecule may be selected from the group comprising cytokines, chemokines, macrophage migration inhibitory factor (MIF; as described, inter alia, in Bernhagen (1998), Mol Med 76(3-4); 151-61 or Metz (1997), Adv Immunol 66, 197-223), T- cell receptors or soluble MHC molecules. Such immune-modulating effector molecules are well known in the art and are described, inter alia, in Paul, “Fundamental immunology”, Raven Press, New York (1989). In particular, known cytokines and chemokines are described in Meager, “The Molecular Biology of Cytokines” (1998), John Wiley & Sons, Ltd., Chichester, West Sussex, England; (Bacon (1998). Cytokine Growth Factor Rev
9(2):167-73; Oppenheim (1997). Clin Cancer Res 12, 2682-6; Taub, (1994) Ther. Immunol. 1(4), 229-46 or Michiel, (1992). Semin Cancer Biol 3(1), 3-15). [0100] Immune cell activity that may be measured include, but is not limited to, (1) cell proliferation by measuring the DNA replication; (2) enhanced cytokine production, including specific measurements for cytokines, such as IFN-γ, GM-CSF, or TNF-α; (3) cell mediated target killing or lysis; (4) cell differentiation; (5) immunoglobulin production; (6) phenotypic changes; (7) production of chemotactic factors or chemotaxis, meaning the ability to respond to a chemotactin with chemotaxis; (8) immunosuppression, by inhibition of the activity of some other immune cell type; and, (9) apoptosis, which refers to fragmentation of activated immune cells under certain circumstances, as an indication of abnormal activation. [0101] Cytokines of the invention are defined by any factor produced by cells that affect other cells and are responsible for any of a number of multiple effects of cellular immunity. Examples of cytokines include but are not limited to the IL-2 family, interferon (IFN), IL-7, IL-10, IL-12, IL-15, IL-18, IL-1, IL-17, TGF and TNF cytokine families, and to IL-1 through IL-35, IFN-α, IFN-β, IFNγ, TGF-β, TNF-α, and TNFβ. [0102] Chemokines, similar to cytokines, are defined as any chemical factor or molecule which when exposed to other cells are responsible for any of a number of multiple effects of cellular immunity. Suitable chemokines may include but are not limited to the CXC, CC, C, and CX3C chemokine families and to CCL-1 through CCL-28, CXC-1 through CXC-17, XCL-1, XCL-2, CX3CL1, MIP-1b, IL-8, MCP-1, and Rantes. [0103] Growth factors include any molecules which when exposed to a particular cell induce proliferation and/or differentiation of the affected cell. Growth factors include proteins and chemical molecules, some of which include: stem cell factors, GM-CSF, G-CSF, human growth factor and stem cell growth factor. Additional growth factors may also be suitable for uses described herein. [0104] Also of interest are enzymes present in the lytic package that NK cells, cytotoxic T lymphocytes or LAK cells deliver to their targets. Perforin, a pore-forming protein, and Fas ligand are major cytolytic molecules in these cells (Brandau et al., Clin. Cancer Res.6:3729, 2000; Cruz et al., Br. J. Cancer 81:881, 1999). CTLs also express a family of at least 11 serine proteases termed granzymes, which have four primary substrate specificities (Kam et al., Biochim. Biophys. Acta 1477:307, 2000). Low concentrations of
streptolysin O and pneumolysin facilitate granzyme B-dependent apoptosis (Browne et al., Mol. Cell Biol.19:8604, 1999). [0105] Pharmaceutical Therapeutics [0106] The invention provides pharmaceutical compositions comprising HSPC-NK cells and/or hypomethylating agent and/or second or third therapeutic agents such as for example, ALT-803, cytokines, chemotherapeutics, and the like, for use as a therapeutic. In one aspect, the pharmaceutical compositions are administered systemically, for example, formulated in a pharmaceutically-acceptable buffer such as physiological saline. Preferable routes of administration include, for example, instillation into the bladder, subcutaneous, intravenous, intraperitoneal, intramuscular, intratumoral or intradermal injections that provide continuous, sustained or effective levels of the composition in the patient. Treatment of human patients or other animals is carried out using a therapeutically effective amount of a therapeutic identified herein in a physiologically-acceptable carrier. Suitable carriers and their formulation are described, for example, in Remington's Pharmaceutical Sciences by E. W. Martin. The amount of the therapeutic agent to be administered varies depending upon the manner of administration, the age and body weight of the patient, and with the clinical symptoms of the neoplasia. Generally, amounts will be in the range of those used for other agents used in the treatment of other diseases associated with neoplasia or infectious diseases, although in certain instances lower amounts will be needed because of the increased specificity of the compound. A compound is administered at a dosage that enhances an immune response of a subject, or that reduces the proliferation, survival, or invasiveness of a neoplastic or, infected cell as determined by a method known to one skilled in the art. [0107] Formulation of Pharmaceutical Compositions [0108] The administration of compositions embodied herein, is by any suitable means that results in a concentration of the therapeutic that, combined with other components, is effective in ameliorating, reducing, or stabilizing said neoplasia or infectious disease. TThe composition may be provided in a dosage form that is suitable for parenteral (e.g., subcutaneous, intravenous, intramuscular, intravesicular, intratumoral or intraperitoneal) administration route. For example, the pharmaceutical compositions are formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000
and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988- 1999, Marcel Dekker, New York). [0109] Human dosage amounts are initially determined by extrapolating from the amount of compound used in mice or non-human primates, as a skilled artisan recognizes it is routine in the art to modify the dosage for humans compared to animal models. For example, the dosage may vary from between about 1 µg compound/kg body weight to about 5000 mg compound/kg body weight; or from about 5 mg/kg body weight to about 4,000 mg/kg body weight or from about 10 mg/kg body weight to about 3,000 mg/kg body weight; or from about 50 mg/kg body weight to about 2000 mg/kg body weight; or from about 100 mg/kg body weight to about 1000 mg/kg body weight; or from about 150 mg/kg body weight to about 500 mg/kg body weight. For example, the dose is about 1, 5, 10, 25, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1,000, 1,050, 1,100, 1,150, 1,200, 1,250, 1,300, 1,350, 1,400, 1,450, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, 3,000, 3,500, 4,000, 4,500, or 5,000 mg/kg body weight. Alternatively, doses are in the range of about 5 mg compound/Kg body weight to about 20 mg compound/kg body weight. In another example, the doses are about 8, 10, 12, 14, 16 or 18 mg/kg body weight. In embodiments whereby the ALT-803 is administered to a patient as part of the therapy, the fusion protein complex is administered at 0.5 mg/kg-about 10 mg/kg (e.g., 0.5, 1, 3, 5, 10 mg/kg). Of course, this dosage amount may be adjusted upward or downward, as is routinely done in such treatment protocols, depending on the results of the initial clinical trials and the needs of a particular patient. [0110] Pharmaceutical compositions are formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the therapeutic in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, molecular complexes, nanoparticles, patches, and liposomes. [0111] The pharmaceutical compositions embodied herein are administered parenterally by injection, infusion or implantation (subcutaneous, intravenous, intramuscular, intratumoral, intravesicular, intraperitoneal) in dosage forms, formulations, or via suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants. The formulation and preparation of such compositions are well known to those skilled in the art of pharmaceutical formulation. Formulations can be found in Remington: The Science and Practice of Pharmacy, supra.
[0112] Compositions comprising a hypomethylating agent or a cytokine or the fusion protein complex for parenteral use are provided in unit dosage forms (e.g., in single-dose ampoules). Alternatively, the composition is provided in vials containing several doses and in which a suitable preservative may be added (see below). The composition is in the form of a solution, a suspension, an emulsion, an infusion device, or a delivery device for implantation, or is presented as a dry powder to be reconstituted with water or another suitable vehicle before use. Apart from the active agent that reduces or ameliorates a neoplasia or infectious disease, the composition includes suitable parenterally acceptable carriers and/or excipients. The active therapeutic agent(s) may be incorporated into microspheres, microcapsules, nanoparticles, liposomes for controlled release. Furthermore, the composition may include suspending, solubilizing, stabilizing, pH-adjusting agents, tonicity adjusting agents, and/or dispersing, agents. [0113] As indicated above, the pharmaceutical compositions may be in a form suitable for sterile injection. To prepare such a composition, the suitable active therapeutic(s) are dissolved or suspended in a parenterally acceptable liquid vehicle. Among acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1,3-butanediol, Ringer's solution, and isotonic sodium chloride solution and dextrose solution. The aqueous formulation may also contain one or more preservatives (e.g., methyl, ethyl or n-propyl p-hydroxybenzoate). In cases where one of the compounds is only sparingly or slightly soluble in water, a dissolution enhancing or solubilizing agent can be added, or the solvent may include 10-60% w/w of propylene glycol. [0114] The present invention provides methods of treating neoplasia or infectious diseases or symptoms thereof which comprise administering a therapeutically effective amount of a pharmaceutical composition. Thus, one embodiment is a method of treating a subject suffering from or susceptible to a neoplasia or infectious disease or symptom thereof. The method includes the step of administering to the mammal a therapeutic amount of an amount of the therapeutic agents and HSPC-NK cells sufficient to treat the disease or disorder or symptom thereof, under conditions such that the disease or disorder is treated. [0115] The methods herein include administering to the subject (including a subject identified as in need of such treatment) an effective amount of a compound described herein, or a composition described herein to produce such effect. Identifying a subject in need of
such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method). [0116] The therapeutic methods of the invention (which include prophylactic treatment) in general comprise administration of a therapeutically effective amount of the compounds herein, such as a compound of the formulae herein to a subject (e.g., animal, human) in need thereof, including a mammal, particularly a human. Such treatment will be suitably administered to subjects, particularly humans, suffering from, having, susceptible to, or at risk for a neoplasia, infectious disease, disorder, or symptom thereof. Determination of those subjects "at risk" can be made by any objective or subjective determination by a diagnostic test or opinion of a subject or health care provider (e.g., genetic test, enzyme or protein marker, Marker (as defined herein), family history, and the like). The fusion protein complexes of the invention may be used in the treatment of any other disorders in which an increase in an immune response is desired. [0117] The invention also provides a method of monitoring treatment progress. The method includes the step of determining a level of diagnostic marker (Marker) (e.g., any target delineated herein modulated by a compound herein, a protein or indicator thereof, etc.) or diagnostic measurement (e.g., screen, assay) in a subject suffering from or susceptible to a disorder or symptoms thereof associated with neoplasia in which the subject has been administered a therapeutic amount of a compound herein sufficient to treat the disease or symptoms thereof. The level of Marker determined in the method can be compared to known levels of Marker in either healthy normal controls or in other afflicted patients to establish the subject’s disease status. In some cases, a second level of Marker in the subject is determined at a time point later than the determination of the first level, and the two levels are compared to monitor the course of disease or the efficacy of the therapy. In certain aspects, a pre- treatment level of Marker in the subject is determined prior to beginning treatment according to this invention; this pre-treatment level of Marker can then be compared to the level of Marker in the subject after the treatment commences, to determine the efficacy of the treatment. [0118] Kits or Pharmaceutical Systems [0119] Pharmaceutical compositions comprising the therapeutic components embodied herein, such as DACX, HSPC-NK cells, cytokines, fusion protein complex may be assembled into kits or pharmaceutical systems for use in ameliorating a neoplasia or
infectious disease. Kits or pharmaceutical systems according to this aspect of the invention comprise a carrier means, such as a box, carton, tube, having in close confinement therein one or more container means, such as vials, tubes, ampoules, bottles and the like. The kits or pharmaceutical systems of the invention may also comprise associated instructions for using the fusion protein complex of the invention. In one embodiment, the kit includes appropriate containers such as bags, bottles, tubes, to allow ex vivo treatment of immune cells using the fusion protein complex of the invention and/or administration of such cells to a patient. Kits may also include medical devices comprising the fusion protein complex of the invention. [0120] All citations to sequences, patents and publications in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference. EXAMPLES [0121] The following non-limiting Examples serve to illustrate selected embodiments of the invention. It will be appreciated that variations in proportions and alternatives in elements of the components shown will be apparent to those skilled in the art and are within the scope of embodiments of the present invention
[0123] Good manufacturing practice compliant, cytokine-based culture systems for the generation of NK cell products from CD34+ hematopoietic stem and progenitor cells (HSPC-NK cells) were developed.1-3 Using this platform, a first-in-human phase 1 study was conducted in older patients with acute myeloid leukemia (AML) who were ineligible for allogeneic stem cell transplantation.4 In this study, escalating dosages of ex vivo generated HSPC-NK cells were infused after lymphodepleting chemotherapy. Important to note that infusion of up to 30 million allogeneic HSPC-NK cells per kg body weight was feasible, well tolerated, and safe, without inducing graft-versus-host disease or nonhematological toxicities. Although this study included older patients with AML who were in morphological complete remission, 2 patients showed persistent minimal residual disease (MRD) before treatment. It is interesting to note that MRD decreased below the detection level by day 90 after infusion of HSPC-NK cells, emphasizing a possible therapeutic activity of adoptively transferred HSPC-NK cells. In a likewise manner, promising anti-tumor responses have been reported worldwide from patients with cancer
undergoing NK cell adoptive immunotherapy.5-10 However, responses in those reports were mostly transient and concern a minority of the patients. [0124] Azacitidine/5-azacytidine (Vidaza, AZA) and decitabine/5-aza-29- deoxycytidine (Dacogen, DAC) are 2 hypomethylating agents (HMAs) currently used for the treatment of AML and myelodysplastic syndromes.13 These HMAs are cytidine nucleoside analogues that incorporate into the DNA during cell replication and division. Although at higher dosages these agents exert direct toxicities toward myeloid cancer cells through DNA damage, at lower dosages they can modulate gene expression due to hypomethylating activity. Particularly, their potential in upregulating NK-activating molecules, such as NKG2D ligands, on tumor cells through their epigenetic modulation and thereby sensitizing tumors to NK-cell-mediated killing, has been reported in several studies and for different cancers, including AML.14-17 Nonetheless, direct impact of HMAs on NK-cell functionalities has not been well established yet. Conflicting data have been reported, describing either advantageous or a deleterious effect on NK cells.15,18-22 Moreover, these data are mostly derived from in vitro studies, often performed at high drug concentrations that do not reflect plasma levels achieved in patients. Therefore, it remains unclear whether application of HMA therapy can augment NK cell–mediated anti-tumor responses in patients with AML. [0125] In this report, the possibility to combine HSPC-NK cell therapy with HMAs was addressed. Through in vitro and in vivo studies, a head-to-head comparison of AZA and DAC was performed to evaluate their impact on HSPC-NK viability and functional-ities, as well as their capacity to potentiate HSPC-NK cell reactivity toward AML. Most importantly, it was demonstrated that DAC, but not AZA, positively modulates the in vivo anti-leukemic potential of adoptively transferred HSPC-NK cells through AML cell sensitization, enhancement of NK cell maturation, and cytolytic functions, as well as improves on NK cell trafficking and accumulation at the tumor site. Furthermore, the study reveals that HSPC-NK cells and DAC can potently work together to combat AML, providing a strong rationale to explore this combination strategy in AML treatment. [0126] Materials and Methods [0127] HSPC-NK cell generation: NK cell products were generated from CD34+ HSPCs derived from umbilical cord blood obtained after normal full-term delivery and
written informed consent (“Commissie Mensgebonden Onderzoek” CMO 2014/226). The culture protocol was adapted from Cany et al.,2 and Roeven et al.,23 and combined the use of the aryl hydrocarbon receptor antagonist StemRegenin-1 for the expansion of CD34+ HSPCs, together with interleukin-15 (IL-15) and IL-12 for the differentiation of NK cells. In brief, magnetic-activated cell sorting–isolated CD34+ cells (Miltenyi Biotec) were expanded for 9 to 10 days in the presence of stem cell factor, IL-7, Flt3L, and recombinant human thrombopoietin (all 25 ng/mL, ImmunoTools). Thereafter, recombinant human thrombopoietin was replaced by IL-15 (50 ng/mL, ImmunoTools) for 5 days. From days 14 to 15 onward, cell differentiation and expansion were performed in the presence of stem cell factor (20 ng/mL), IL-7 (20 ng/mL), IL-15 (50 ng/mL), and IL-12 (0.2 ng/mL, Miltenyi Biotech). Medium was supplemented with 2 μM StemRegenin-1 (Cellagen Technology) from day 0 to day 21. All media were prepared with basal CellGro DC medium (CellGenix) and supplemented with 10% or 2% pooled human serum (Sanquin Blood Bank Nijmegen) during the expansion and differentiation phases, respectively. The cell density and CD56 acquisition were checked twice per week by flow cytometry (FCM) and adjusted to 1.5 × 106 to 2.5 × 106 cells/mL by addition of a new medium. HSPC-NK cells were used at the end of the culture process with >80% CD561 cell purity, which was typically achieved within 5 to 6 weeks of culture. [0128] HMAs: AZA and DAC were purchased from Sigma-Aldrich. For in vitro studies, both drugs were dissolved in NaCl 0.9% at 0.1 to 1 mM, aliquoted them for single use, and stored them at -20°C. Drugs were used immediately after thawing, and treatment of the cell cultures was performed with limited light exposure. In vitro, HMAs were used at concentrations similar to those achieved in plasma of HMA-treated patients,24,25 with daily refreshments due to the short half-life of these agents.26 For in vivo studies, 2 to 4 mg of HMAs was kept on ice, protected from light, and dissolved with NaCl 0.9% just before the mice were injected. Treatments were applied daily, in line with current clinical practice. [0129] In vivo studies: All animal experiments were conducted in immune-deficient NOD/SCID/IL2Rgnull (NSG) mice originally purchased from Jackson Laboratories. Mice were housed and bred in the Radboud University Medical Center (Radboudumc) Central Animal Laboratory and were used in experiments at 6 to 12 weeks of age (20-30 g body weight). Studies were approved by the Animal Experimental Committee of Radboudumc and were conducted in accordance with institutional and national guidelines under the
university permit number 10300. The preclinical xenograft model for AML was established by intrafemoral injection of the THP-1 cells, engineered to express the green fluorescent protein (GFP) and luciferase reporter genes (THP-1.LucGFP cells) for longitudinal tumor load monitoring by bioluminescence imaging, as previously described.2 In adoptive transfer studies, HSPC-NK cells were resuspended in phosphate-buffered saline and injected intravenously through the tail vein. Survival of HSPC-NK cells in vivo was supported by subcutaneous administration of recombinant human IL-15 (1 μg per mouse; ImmunoTools), every 2 to 3 days after HSPC-NK cell infusion. Alternatively (for data shown in FIGS.5A-5C), ALT-803, (Altor BioScience), was administered subcutaneously at 0.2 mg/kg every 3 to 4 days. The experimental designs implemented for the treatment of mice with HMAs were based on current clinical practice (dosage, duration, and route of administration). The dosages applied in humans (milligrams per meter squared) were translated for mouse studies (milligrams per kilogram) based on the study by Reagan-Shaw et al.,27 (see FIGS.12A, 12B for calculation). During treatment with HMA, mice were given wet food to improve feeding and tolerability of the drugs. Mice were carefully monitored for weight and general conditions, and euthanized according to well-defined end points. Ex vivo FCM analysis and gene expression profiling were performed on cells isolated from the spleen, bone marrow, or liver with use of erythrocyte lysis solution or lympholyte-M (Cedarlane). [0130] Antibodies: The following conjugated monoclonal antibodies were used for HSPC-NK cell phenotyping: CD56 (HCD56, Biolegend), NKG2A (Z199, Beckman Coulter), CD158a/h, b and e (clones HP-MA4, DX27, and DX9 respectively, used as a combined staining for KIRs), CD16 (3G8), NKG2D (1D11), NKp46 (9E2), NKp44 (P448), CD69 (FN50), TRAIL (RIK-2), FasL (NOK-1), CD11a (HI111), CXCR3 (G025H7, all Biolegend), and DNAM-1 (DX11; BD Bioscience). The panel for AML cell analysis included MIC-A/B (6D4, Biolegend), ULBP-1 (170818), ULBP-2 (165903), CD155 (300907), CD112 (610603, all R&D), TRAIL-R1 (DJR2-4), TRAIL-R2 (DJR2-1), and Fas (DX2, all Biolegend). IgG1 (MOPC21) and IgG2a (MOPC173, Biolegend) isotype controls were included for calculation of specific fluorescence intensities. For ex vivo FCM analysis, HSPC-NK cells and THP-1.LucGFP cells were identified based on CD56 and GFP expression, respectively, in combination with human CD45 (J.33, Beckman Coulter) and mouse CD45 (30-F11, BD Biosciences) antibodies. Non-viable cells were excluded using
SYTOX Blue (Life Technologies) or eFluor780 (eBiosciences) viability dye. Phenotypical analyses were performed using the Gallios flowcytometer and Kaluza analysis software (both Beckman Coulter). [0131] Flow cytometry (FCM)-based cellular assays: The K562, THP-1, and KG1a cell lines (ATCC) were cultured in Iscove's modified Dulbecco's medium (Invitrogen) supplemented with 10% fetal calf serum (Integro). The HLA-type of THP-1 and KG-1a cells is Bw6, C1/C1 and Bw4, C1/C2 respectively. Patient- derived primary AML cells were obtained from diagnostic leftover material in accordance with the Declaration of Helsinki and institutional guidelines and regulations (CMO 2013/064). All samples employed in this study were from bone marrow origin and contained >90% blasts based on flow cytometric determination of CD45, CD33 and CD34 expression. The HLA-type of primary AML cells was not determined. NK cell co-cultures with primary AML cells were supplemented with SCF and Flt3L (both 20 ng/ml), IL-3 (50 ng/ml, Immunotools), G-CSF (20 ng/ml; Amgen) and IL-15 (5 ng/ml). Cytotoxicity assays were performed as previously described17 using CFSE-labeled target cells. In experiments where AML or NK cells were pre-treated with HMA, cells were harvested, counted, and equal numbers of viable cells were plated in all experimental conditions. After overnight co-culture, the numbers of viable target cells were determined using the FC500 cytometer (Beckman Coulter) by gating on CFSE+ cells, exclusion of the live/dead marker 7AAD (Sigma Aldrich) and back-gating on forward/side scatter. Target cells were plated alone as controls without killing. Proliferation of AML and NK cells was examined after labeling with CFSE, culture in the presence or absence of HMA, and subsequent FCM analysis. Analysis of NK cell reactivity at the single cell level was determined following 4 hours stimulation with K562 cells in the presence of anti- CD107a (H4A3; BD Biosciences) and Brefeldin A (BD Biosciences) and subsequent intracellular staining for perforin (dG9, Biolegend) and IFN-γ (B27; BD Biosciences). FCM analysis was performed with exclusion of dead cells using the Fixable Viability Dye eFluor780 (eBiosciences), gating on CD56+Perforin+ NK cells, and using unstimulated cells as control. [0132] Gene expression analysis: Total RNA was isolated using PURELINK RNA mini-kit (Life Technologies) according to manufacturer’s instructions. Next, cDNA was synthesized using M-MLV-reverse transcriptase (Invitrogen) in a standard
reaction after which real-time PCR was performed using the following TAQMAN Gene expression assays (Life Technologies): IFNG (Hs00989291_m1), TNF (Hs01113624_g1), PRF1 (Hs00169473_m1), GZMB (Hs00188051_m1), TNFSF10 (Hs00921974_m1), and FASLG (Hs00181225_m1). Expression levels were normalized to β-Actin (ACTB, NM_001101.2) using the ΔCt method. [0133] Statistical analysis: Statistical analyses was performed using GraphPad Prism 5 software. Student t-tests and 1- and 2-way analyses of variance (ANOVAs) were used when appropriate, as indicated in the figure legends. Differences were considered to be significant for P < 0.05. [0134] Results [0135] HMAs augment HSPC-NK efficacy against AML in vitro: To investigate the possibility of combining HSPC-NK cells with HMA therapy, the effect of AZA and DAC on 2 AML cell lines was first determined. THP-1 and KG1a cells were treated daily with HMAs by use of concentrations similar to those achieved in plasma of treated patients.24,25 As expected, the number of viable AML cells decreased in a time- and dose-dependent manner compared with untreated cells (FIG.8). AZA at 0.1 to 1 pM and DAC at 0.01 to 0.1 pM had a moderate impact on AML cell viability and proliferation, so these concen- trations were selected for subsequent studies. [0136] Next, it was tested whether pretreatment of AML cells with HMAs would influence their susceptibility to HSPC-NK cell–mediated killing. Therefore, THP-1 and KG1a cells were cultured in the presence or absence of HMAs for 2 days and were used as targets for HSPC-NK cells with or without washout of the drugs (FIG. 1A). At start of the coculture, equal numbers of viable AML cells were plated in each experimental condition. Analysis of AML cell survival showed that the effects of HMAs and HSPC-NK cells were at least additive. After coculture with HSPC-NK cells, the survival of THP-1 cells decreased from 60% without HMA pretreatment to 30% to 40% for pretreated cells (FIG.1B; P < .001 for all HMA groups vs no HMA pretreatment). Because AML cell counts were slightly diminished with the HMAs in the absence of NK cells (FIG.1B), these data translated into a significant increase in NK-specific killing for each treatment condition analyzed independently (FIG. 1C). It is remarkable that the effect of HSPC-NK cells, which was defined using the absolute number of AML cells actually killed by NK cells, was significantly increased when THP-1 cells were pretreated with the lower concentrations of HMAs (FIG.
1D). These data support that HMAs can sensitize AML cells to HSPC-NK cell– mediated killing. The combination treatment of HMAs and HSPC-NK cells was also studied across time for KG1a cells, without drug washout (FIGS. 1E-1F). Here, the effects of HSPC-NK cells and HMAs were additive, resulting in potent reduction of AML cell numbers when combined together. [0137] Based on these findings, the impact of HMAs was studied in HSPC-NK cell cocultures with patient-derived primary AML cells (Table 1; FIG. 2A). Here, a robust and dose-dependent effect of HMAs on primary AML cell viability and proliferation was confirmed (FIG. 9). In contrast, HSPC-NK cell survival was not affected (FIG. 2B). It is interesting to note that combined treatment demonstrated sustained killing of primary AML cells by HSPC-NK cells at all drug concentrations tested, and the combination with 1 pM AZA and 0.1 pM DAC showed superior AML-killing efficacy compared with NK cell treatment alone (FIG.2C). As seen with AML cell lines, HSPC-NK cells and HMAs potently worked together in reducing the survival of primary AML blasts (FIG. 10; Table 2). Although their effects were mostly additive (FIG.2D), the numbers of AML cells killed by NK cells eventually increased in the presence of HMAs, particularly with DAC (as illustrated for patient 4 [pAML#4], FIG. 2E). These data demonstrate that HMAs differentially affect AML and HSPC-NK cells in vitro, and that HSPC-NK cells maintain potent anti-leukemic activity during HMA exposure. Most importantly, these findings demonstrate that HMAs can potentiate HSPC-NK cell killing activity, providing evidence that the combination of HPSC-NK cells with HMA therapy could result in additive to synergistic effects against AML. [0138] Exposure to low-dose HMA concentrations does not impair HSPC-NK cell cytolytic activity in vitro. Thereafter, the aim was to confirm that HMAs did not impair HSPC- NK cell functionalities. To do this, HSPC-NK cells were cultured either under proliferative (i.e., 20 ng/mL of IL-15 plus 1000 IU/mL of IL-2) or steady-state (i.e., 5 ng/mL of IL-15) conditions and treated with HMAs for 6 consecutive days (FIG.3A). HMA treatment did not affect HSPC-NK cell viability (data not shown); however, 1 μM AZA significantly decreased NK cell proliferation (FIG.3B). DAC had a minor impact, even at the highest concentration tested. [0139] The cytolytic activity of HMA-treated HSPC-NK cells against MHC-Ineg K562 cells and MHC-Ipos THP-1 cells was analyzed. Target-cell killing was only diminished after HSPC-NK pretreatment with the highest dosages of AZA and, to a lesser extent, with
DAC (FIG. 3C). These observations were confirmed at the single-cell level with use of CD107a degranulation and intracellular interferon-gamma (IFN-γ) staining. As depicted in FIGS. 3D-3E, the proportion of CD107a+ HSPC-NK cells on K562 stimulation was only negatively affected with 1-μM AZA but not with 0.1-μM DAC. The percentages of IFN-γ+ HSPC-NK cells remained similar. [0140] In addition, the phenotype of HSPC-NK cells cultured in the presence of HMAs was examined. Only the frequency of killer cell immunoglobulin-like receptor (KIR)–positive HSPC-NK cells increased, particularly after exposure to HMAs under proliferative conditions (FIG. 3F). NKG2A+KIR+ HSPC-NK cells display higher IFN-γ production capacity compared with NKG2A+KIR- cells.2 Nonetheless, the increase in KIR expression after HMA treatment does not correlate with IFN-γ responses (FIG. 3G). Expression of other maturation markers (NKG2A and CD16), as well as activating receptors, adhesion molecules, and death receptors, remained unchanged after exposure to HMAs in vitro (FIG.11). These data confirm that AML and HSPC-NK cells are differently affected by HMAs, and that low drug concentration preserves HSPC-NK cell anti-leukemic activity. [0141] DAC, but not AZA, potentiates HSPC-NK cell anti-leukemic efficacy in vivo: To investigate combinatorial HMA and HSPC-NK cell therapy against AML in vivo, studies were conducted using the established intrafemoral THP-1 mouse model.2 First, the tolerability of HMA treatment in mice was tested by daily application of de-escalating dosages, which were defined based on current practice in patients (i.e., 75 mg/m2 for AZA and 20 mg/m2 for DAC). Here, we identified 12.5 mg/m2 of AZA X 7 injections and 5 mg/m2 of DAC × 5 injections as the highest tolerated dose in NSG mice based on body weight loss and reduced THP-1 cell progression (FIGS.12A, 12B). To obtain a moderate effect of HMAs alone in this model, 7.5 mg/m2 of AZA and 2.0 mg/m2 of DAC were subsequently tested, which is 10 times lower than current dosages applied in patients (FIG. 4A, HMA titration #1). These dosages were well tolerated and still reduced THP-1 growth. However, at these dosages, AZA exerted a limited anti-leukemic effect, whereas DAC strongly reduced tumor growth. Based on these observations, the treatment schedule was further refined to give the highest tolerated dose of AZA (12.5 mg/m2) for 7 consecutive days and lowered the dosage of DAC to 1.25 mg/m2 during 5 days (FIG. 4A, HMA titration #2). Finally, these dosages resulted in a similar and intermediate effect on THP-1 cells in vivo and were used to test the efficacy of combination therapy with HSPC-NK cells.
[0142] To assess the impact of HMAs on NK cells in vivo, infusions of HSPC-NK cells plus rhIL-15 were applied at the start of HMA treatment (FIG. 4B). Of note, treatment with only HSPC-NK cells was not included because such treatment was ineffective in this highly stringent THP-1 AML mouse model.2 Bioluminescence imaging revealed lower tumor load in mice treated with HSPC-NK cells in combination with DAC, compared with DAC monotherapy (FIG.4C). Although transient, this reduction in tumor progression was significant at 2 weeks after treatment (FIG. 4D). In contrast, no difference was observed in AZA-treated mice. This did not rely on a defective HSPC-NK cell persistence on AZA treatment, as the numbers of HSPC-NK cells present in the blood and spleen of treated mice were comparable to those seen in control animals (FIGS.13A, 13B). [0143] To validate these findings and further improve anti-leukemic potency, a new experiment was performed applying the same approach but with 2 treatment cycles (FIG.5A). In addition, ALT-803 was used instead of rhIL-15 to support NK cell persistence in vivo. ALT- 803 is an IL-15 superagonist complex composed of an IL-15 mutant (N72D) bound to sushi domain of IL-15Rα fused to IgG1 Fc. This complex has been shown to display higher stability and enhanced biological activity on NK cells in vivo as well as superior localization to the lymphoid organs when compared with rhIL-15.28,29 The effects of ALT-803 on HSPC-NK cells were validated in vitro (FIGS.14A, 14B). In this experimental model, treatment with 2 DAC cycles further slowed down THP-1 progression (FIG.5B). Notably, it was shown that 2 cycles of combined DAC, HSPC-NK cells, and ALT-803 treatment resulted in significantly reduced THP-1 progression for up to 4 weeks compared with DAC treatment only (FIG. 5C). These data demonstrate that DAC does not impair HSPC-NK cells in vivo, and that this combination strategy can boost the reactivity of adoptively transferred HSPC-NK cells against AML. [0144] DAC enhances HSPC-NK cell cytolytic functions and trafficking to the bone marrow in vivo. To understand how NK cell potentiation by DAC occurred in vivo, the phenotype of THP-1 cells and HSPC-NK cells were analyzed after treatment of the mice with DAC. First, activating ligands and death receptors on THP-1 cells were examined by FCM (FIGS.6A, 6B). Although only slight differences were seen for ULBP1, MIC-A/B, CD112, TRAIL receptors, and Fas, the expression levels of ULBP2 and CD155 were evidently increased with DAC, which supports the finding that HMAs can sensitize AML cells to HSPC- NK cell-mediated killing. On HSPC-NK cells, sustained expression of DNAM-1, NKp46, TRAIL, and CD69 was observed (FIG. 7A). The level of NKG2D was diminished but was
still present on NK cells, and expression of NKp44 was significantly increased. Similar phenotypical changes were observed with AZA (FIGS. 15A-15D). Furthermore, HSPC-NK cells maintained high expression of NKG2A following adoptive transfer into NSG mice, and acquired CD16 and KIR expression as compared with the cell product before infusion. This finding is in line with a previous report,2 but treatment with DAC further increased the frequency of KIR+ HPSC-NK cells. By quantitative reverse transcription polymerase chain reaction, enhanced gene expression of inflammatory cytokines, perforin, and TRAIL was also demonstrated after DAC treatment, which are important players in NK cell–mediated killing (FIG.7B). It is notable that no regulation, or less upregulation, of these inflammatory and cytolytic mediators was seen with AZA (FIGS.15A-15D). Finally, HPSC-NK cell counts were examined in the bone marrow compartment. In 3 independent experiments (data of 2 independent experiments shown in FIG.7C), increased numbers of HSPC-NK cells in mouse bone marrow were repeatedly found after DAC treatment, whereas the numbers of circulating NK cells were either similar (day+7) or lower (day+14) in peripheral blood compared with control mice. Altogether, these data strongly support that low-dose DAC can potentiate the anti-leukemic reactivity of HSPC-NK cells toward AML through modulation of gene expression and phenotype in AML and NK cells, and by influencing cell trafficking and tumor targeting in vivo. [0145] Table 1: Primary AML sample characteristics
[0146] Table 2: Effect of HSPC-NK cells and HMA on survival of primary AML cells
[0147] Discussion [0148] The capacity of AZA and DAC to enhance HSPC-NK cell reactivity against AML was investigated. This study is believed to be the first head-to-head comparison of AZA and DAC in promoting NK cell–mediated anti-leukemic reactivity in vivo. It was shown herein, that these HMAs exert direct effects on AML cell viability, proliferation, and phenotype that eventually result in a greater susceptibility to NK cell-mediated killing. Furthermore, increased expression of NKG2D and DNAM-1 ligands was also found on THP- 1 AML cells after treatment with HMAs. In contrast to AML cells, exposure to HMAs has a minimal impact on HSPC-NK cell viability and proliferation. Moreover, the data demonstrated, in vitro and in vivo, that DAC can potentiate HSPC-NK cell functionalities and anti-leukemic activity. Notable is that combined HSPC-NK cell and DAC treatment resulted in improved control of THP-1 AML in NSG mice, whereas the combination with AZA did not yield an additive anti-leukemic effect. Multiple factors can explain the difference between both HMAs, including reduced NK cell proliferation and degranulation capacity upon AZA treatment, or a lower impact of AZA on NK cell cytolytic machinery, in vivo trafficking, and accumulation in the bone marrow compartment compared with DAC. It is believed that this difference is not due to different dosing of the HMAs in the mouse studies, as these were carefully titrated AZA and DAC for exerting the same direct effect on THP-1 cells in vivo. [0149] It is interesting that increased anti-leukemic activity of NK cells could already be demonstrated in DAC-treated mice after a single infusion of HSPC-NK cells but was further
enhanced by the application of 2 treatment cycles. It is important to underline that this intrafemoral THP-1 AML model is very stringent: on the one hand, because of the fast and aggressive THP-1 cell progression in vivo,2 and on the other hand, because it requires effective NK homing to the tumor site after intravenous infusion.30 In this model, treatment with DAC did not eradicate THP-1 cells in vivo but allowed indolent disease progression, thereby favoring HSPC-NK cell responses to less bulky and residual disease. In addition, increased HSPC-NK cell numbers were seen in the bone marrow of particularly DAC-treated mice. Shortly after infusion, NK cell homing to the bone marrow may be favored as a result of transient leukopenia occurring in HMA-treated mice. Moreover, at 2 weeks after NK cell transfer to tumor-bearing mice, HSPC-NK cell numbers were particularly increased within the tumor bed rather than in normal bone marrow, which provides evidence of a specific NK cell homing to the tumor site. Therefore, it was shown that HSPC-NK cells express and maintain high levels of the chemokine receptor CXCR3 in vivo and display a higher capacity of inflammatory cytokine production after DAC treatment. These observations are in line with a recent publication by Wang et al.,31 which described a dual effect of DAC on inflammation and lymphocyte trafficking at the tumor site in a mouse ovarian cancer model. They showed that treatment with low-dose DAC increases the expression of chemokines that recruit NK cells and CD81 T cells as well as promotes their production of IFN-γ and tumor necrosis factor alpha (TNF-α). It is interesting to note that it was also observed herein, that AML cells, including THP-1, can secrete the inflammatory chemokines CXCL9, CXCL10, and CXCL11 upon exposure to IFN- γ and TNF-α. In a likewise manner, it was recently shown that HMAs can reactivate expression of endogenous retroviral elements, thereby eliciting type I and III IFN response, as well as induce expression of CXCL9 and CXCL10 by tumor cells.32 The DAC-induced IFN response may positively affect HSPC-NK cell activity and function as seen by the upregulation of immune mediators such as IFN-γ, TNF-α, and perforin. All of these findings bring evidence that DAC can modulate tumor environments and immune cell trafficking in vivo. Therefore, without wishing to be bound by theory, it was thought that, besides its direct anti-leukemic activity, DAC can potently maximize HSPC-NK cell responses presumably through epigenetic modulation. Besides sensitizing AML cells to NK cell–mediated killing, the data herein, also support that DAC can boost HSPC-NK cell cytolytic functions, enhance inflammatory responses, and upregulate expression of the activating receptor NKp44. This mechanism could result in a self-stimulatory loop, further promoting NK cell recruitment to the tumor site and sustained control of AML.
[0150] Altogether, the findings herein, provide a strong rationale to investigate HMA therapy in patients not only prior to adoptive NK cell immunotherapy, but also concomitantly with HSPC-NK cell infusion to exploit superior efficacy of these 2 treatment regimens. Potential clinical applications are broad. For AML, current remission induction and consolidation regimens could be complemented with adoptive transfer of allogeneic HSPC- NK cells combined with low or increasing doses of DAC as an adjuvant consolidation or a bridge-to-transplant strategy. Furthermore, combining DAC and HSPC-NK cell infusion with the IL-15 superagonist ALT-803, which has shown enhanced biological activity and a better half-life in vivo compared with rhIL-15, is a promising combinatorial approach to maximizing the anti-leukemic effect. This combination could further eradicate MRD and ultimately improve outcome of allogeneic stem cell transplantation.33,34 In the model, a 5-day treatment with DAC was applied, whereas in patients it has been observed that 10-day treatment is more efficacious.35-37 More investigations are being conducted regarding DAC-mediated changes in the tumor environment, inflammation, and chemokine and cytokine levels in diseased bone marrow. Moreover, this study highlights a possible synergy when combining HSPC-NK cells with DAC as demonstrated by the capacity to potentiate HSPC-NK cell reactivity toward AML. This action was seen in coculture experiments, whereas exposure to HMAs had a minor influence on the phenotype of THP-1 cells (FIG.16) or primary AML blasts in vitro, so other factors might be implicated in modulating NK cell reactivity. One explanation could be changes in inhibitory pathways. Higher CD155 expression was observed on THP-1 cells after treatment with DAC, but this molecule also binds to CD96 and TIGIT, which compete with DNAM-1 on NK cells. On HSPC-NK cells, TIGIT is not expressed at the end of the culture process and is not upregulated in vivo regardless of DAC treatment. In contrast, CD96 is highly expressed on HSPC-NK cells. Screening of primary AML blasts with respect to risk group, cytogenetic abnormalities, or FAB classification could help identify best responders to combined DAC and HSPC-NK cell therapy. Finally, the studies herein, revealed that 2 cycles with combined DAC, HSPC-NK cells, and ALT-803 treatment is better, but the effect seems to decrease at 28 days. Therefore, further improvement with longer duration of DAC treatment of 10 days in each cycle and/or more treatment cycles is to be tested to maximize the combined effect of NK cells, DAC, and ALT-803. Opportunities also could be considered in the post- transplant setting, where low-dose maintenance therapy with HMAs has been shown to be feasible.13 In this situation, using HSPC-NK cells generated from the donor stem cell graft would be beneficial to allow longer persistence of adoptively transferred NK cells.23 Furthermore, the influence of DAC on inflammation and immune-cell trafficking to the tumor
site, epigenetic modulation leading to tumor-associated antigen upregulation, and further stimulation of NK and dendritic cell cross-talk could also boost anti-leukemic T-cell responses.41-43 Therefore, this study, which shows that HSPC-NK cells and DAC can potently work together to eradicate AML, provides a strong rationale to explore this combination strategy in the treatment of AML. [0151] References 1. Spanholtz J, Preijers F, Tordoir M, et al. Clinical-grade generation of active NK cells from cord blood hematopoietic progenitor cells for immunotherapy using a closed- system culture process. PLoS One.2011;6(6): e20740. 2. Cany J, van der Waart AB, Spanholtz J, et al. Combined IL-15 and IL-12 drives the generation of CD34(+)-derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer. OncoImmunology.2015;4(7):e1017701. 3. Hoogstad-van Evert JS, Cany J, van den Brand D, et al. Umbilical cord blood CD34+ progenitor-derived NK cells efficiently kill ovarian cancer spheroids and in- traperitoneal tumors in NOD/SCI D/ IL2Rgnull mice. OncoImmunology. 2017; 6(8):e 1320630. 4. Dolstra H, Roeven MWH, Spanholtz J, et al. Successful transfer of umbilical cord blood CD34(+) hematopoietic stem and progenitor-derived NK cells in older acute myeloid leukemia patients. Clin Cancer Res.2017;23(15): 4107-4118. 5. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105(8): 3051-3057. 6. Arai S, Meagher R, Swearingen M, et al. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial. Cytotherapy.2008; 10(6):625-632. 7. Iliopoulou EG, Kountourakis P, Karamouzis MV, et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother.2010;59(12): 1781-1789. 8. Curti A, Ruggeri L, D’Addio A, et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood.2011;118(12): 3273-3279. 9. Szmania S, Lapteva N, Garg T, et al. Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients. J Immunother.2015;38(1):24-36. 10. Pérez-Martínez A, Fernández L, Valentín J, et al. A phase I/II trial of interleukin- 15– stimulated natural killer cell infusion after haploidentical stem cell transplantation for pediatric refractory solid tumors. Cytotherapy.2015;17(11):1594-1603. 11. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell.2015;28(6): 690- 714.
12. Gotwals P, Cameron S, Cipolletta D, et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer.2017;17(5):286-301. 13. Cruijsen M, Lübbert M, Wijermans P, Huls G. Clinical results of hypomethylating agents in AML treatment. J Clin Med. 2014; 4(1):1-17. 14. Rohner A, Langenkamp U, Siegler U, Kalberer CP, Wodnar-Filipowicz A. Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk Res.2007;31(10): 1393-1402. 15. Schmiedel BJ, Arélin V, Gruenebach F, Krusch M, Schmidt SM, Salih HR. Azacytidine impairs NK cell reactivity while decitabine augments NK cell responsiveness toward stimulation. Int J Cancer.2011;128(12): 2911-2922. 16. Zhu Z, Lu X, Jiang L, et al. STAT3 signaling pathway is involved in decitabine induced biological phenotype regulation of acute myeloid leukemia cells. Am J Transl Res. 2015; 7(10):1896-1907. 17. Baragano Raneros A, Martín-Palanco V, Fernandez AF, et al. Methylation of NKG2D ligands contributes to immune system evasion in acute myeloid leukemia. Genes Immun. 2015;16(1):71-82. 18. Gao XN, Lin J, Wang LL, Yu L. Demethylating treatment suppresses natural killer cell cytolytic activity. Mol Immunol.2009;46(10): 2064-2070. 19. Triozzi PL, Aldrich W, Achberger S, Ponnazhagan S, Alcazar O, Saunthararajah Y. Differential effects of low-dose decitabine on immune effector and suppressor responses in melanoma-bearing mice. Cancer Immunol Immunother.2012;61(9):1441-1450. 20. Kopp LM, Ray A, Denman CJ, et al. Decitabine has a biphasic effect on natural killer cell viability, phenotype, and function under proliferative conditions. Mol Immunol.2013; 54(3-4):296-301. 21. Gang AO, Frøsig TM, Brimnes MK, et al.5-Azacytidine treatment sensitizes tumor cells to T-cell mediated cytotoxicity and modulates NK cells in patients with myeloid malignancies. Blood Cancer J.2014;4(3):e197. 22. Raneros AB, Puras AM, Rodriguez RM, et al. Increasing TIMP3 expression by hypo-methylating agents diminishes soluble MICA, MICB and ULBP2 shedding in acute myeloid leukemia, facilitating NK cell-mediated immune recognition. Oncotarget. 2017;8(19): 31959-31976. 23. Roeven MW, Thordardottir S, Kohela A, et al. The aryl hydrocarbon receptor antagonist StemRegenin1 improves in vitro generation of highly functional natural killer cells from CD34 (+) hematopoietic stem and progenitor cells. Stem Cells Dev. 2015;24(24):2886- 2898. 24. Uchida T, Ogawa Y, Kobayashi Y, et al. Phase I and II study of azacitidine in Japanese patients with myelodysplastic syndromes. Cancer Sci.2011;102(9):1680-1686. 25. Oki Y, Kondo Y, Yamamoto K, et al. Phase I/II study of decitabine in patients with myelodysplastic syndrome: a multi-center study in Japan. Cancer Sci.2012;103(10):1839- 1847. 26. Liu Z, Marcucci G, Byrd JC, Grever M, Xiao J, Chan KK. Characterization of decomposition products and preclinical and low dose clinical pharmacokinetics of decitabine (5-aza-29-deoxycytidine) by a new liquid chromatography/tandem mass
spectrometry quantification method. Rapid Commun Mass Spectrom. 2006;20(7): 1117-1126. 27. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J.2008;22(3):659-661. 28. Rhode PR, Egan JO, Xu W, et al. Comparison of the superagonist complex, ALT- 803, to IL15 as cancer immunotherapeutics in animal models. Cancer Immunol Res. 2016;4(1): 49-60. 29. Rosario M, Liu B, Kong L, et al. The IL-15-based ALT-803 complex enhances FcgRIIIa-triggered NK cell responses and in vivo clearance of B cell lymphomas. Clin Cancer Res.2016;22(3):596-608. 30. Cany J, van der Waart AB, Tordoir M, et al. Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrow-residing human leukemia cells in NOD/SCID/IL2Rg(null) mice. PLoS One.2013;8(6):e64384. 31. Wang L, Amoozgar Z, Huang J, et al. Decitabine enhances lymphocyte migration and function and synergizes with CTLA-4 blockade in a murine ovarian cancer model. Cancer Immunol Res.2015;3(9):1030-1041. 32. Wolff F, Leisch M, Greil R, Risch A, Pleyer L. The double-edged sword of (re)expression of genes by hypomethylating agents: from viral mimicry to exploitation as priming agents for targeted immune checkpoint modulation. Cell Commun Signal. 2017;15(1):13. 33. Araki D, Wood BL, Othus M, et al. Allogeneic hematopoietic cell transplantation for acute myeloid leukemia: time to move toward a minimal residual disease-based definition of complete remission? J Clin Oncol.2016;34(4): 329-336. 34. Ivey A, Hills RK, Simpson MA, et al; UK National Cancer Research Institute AML Working Group. Assessment of minimal residual disease in standard-risk AML. N Engl J Med.2016;374(5):422-433. 35. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci USA.2010;107(16):7473-7478. 36. Ritchie EK, Feldman EJ, Christos PJ, et al. Decitabine in patients with newly diagnosed and relapsed acute myeloid leukemia. Leuk Lymphoma.2013;54(9):2003-2007. 37. Cruijsen M, Hobo W, van der Velden WJFM, et al. Addition of 10-day decitabine to fludarabine/total body irradiation conditioning is feasible and induces tumor-associated antigen-specific T cell responses. Biol Blood Marrow Transplant.2016;22(6): 1000-1008. 38. Fuchs A, Cella M, Giurisato E, Shaw AS, Colonna M. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J Immunol.2004;172(7):3994-3998. 39. Chan CJ, Martinet L, Gilfillan S, et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol.2014;15(5):431-438. 40. Blake SJ, Stannard K, Liu J, et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov.2016;6(4):446-459. 41. Héninger E, Krueger TE, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Front Immunol.2015;6:29.
42. Thordardottir S, Hangalapura BN, Hutten T, et al. The aryl hydrocarbon receptor antagonist StemRegenin 1 promotes human plasmacytoid and myeloid dendritic cell development from CD341 hematopoietic progenitor cells. Stem Cells Dev.2014;23(9):955- 967. 43. Cany J, Dolstra H, Shah N. Umbilical cord blood-derived cellular products for cancer immunotherapy. Cytotherapy.2015;17(6): 739-748. [0152] Example 2: A higher peritoneal NK cell frequency is correlated with better outcome in advanced ovarian cancer patients, and peritoneal NK cell functionality can be boosted by IL-15. [0153] Ovarian cancer (OC) has the highest mortality rate of female cancers. Because OC is generally asymptomatic until ascites or metastases beyond the ovaries have developed, patients are often diagnosed in advanced stage. Moreover, the presence and progression of ascites is associated with poor prognosis and poor quality of life.1 Current therapy consists of debulking surgery combined with platinum/taxane chemotherapy, but the majority of patients develop a recurrence within 3 years. Especially, for women with advanced stage disease the prognosis is dismal, and despite therapeutic advances the 5-year survival is only 28%.2 [0154] Since many studies demonstrated that OC is an immunogenic disease, development of immunotherapeutic interventions could be an attractive strategy to improve clinical outcome.3 In this regard it is essential to study immune cell function in the OC microenvironment in order to develop effective immunotherapeutic approaches. Particularly, ascites is an attractive source in OC patients because tumor cells and immune cells are both present. Furthermore, ascites contains a variety of immunosuppressive cellular and soluble components that influence the function of tumor-targeting lymphocytes.4-9 Nevertheless, several studies showed that presence of tumor-infiltrating lymphocytes positively correlated with survival in cancer patients.10-17 While the importance of CD8+ T cell infiltration has been clearly demonstrated, the role of infiltrating innate natural killer (NK) cells remains unclear. However, it was reported that CD103+ tumor-infiltrating NK cells often co-infiltrate with CD8+CD103+ T cells, yet the contribution of NK cells to improving outcome is difficult to assess.12 Therefore, more research is required to decipher the role of NK cell immunity in the OC microenvironment. [0155] NK cells recognize stressed neoplastic cells through a balance of activating and inhibitory receptors.18,19 Epidemiological research has shown that low NK cell activity is associated with increased cancer risk in humans.20 For OC patients, decreased functionality of
ascites-derived NK cells has been observed21, which could be partially attributed to the low expression of various activating NK cell receptors including CD16, DNAM-1 and NKp30.9,22,23 Similarly, ascites-derived T cells are rather inactive, though proliferation and functionality can be partially restored by cytokine stimulation.24 NK cells from ascites also have a low cytotoxic effect, but can be reinvigorated by IL-2 or IL-1525. In this regard, Felices et al. recently reported that the, ALT-803, a fusion protein complex of IL-15 variant (N72D) bound to sushi domain of IL-15Rα fused to IgG1 Fc, potently enhanced the function of ascites-derived NK cells and healthy donor peripheral blood NK cells exposed to ascites fluid.26 Furthermore, many studies demonstrated that OC cells are susceptible to killing by cytokine-stimulated NK cells.27-42 [0156] In this study, NK cell frequency, phenotype and functionality was characterized in ascites of advanced OC patients in relation to clinical outcome, and tested their responsiveness to IL-15 receptor mediated stimulation. A higher CD56+ NK cell frequency was observed in ascites was associated with a better progression free survival (PFS; p=0.01) and overall survival (OS; p=0.002) in OC patients. Furthermore, it was demonstrated that the cytolytic function of ascites-derived NK cells can be efficiently reinvigorated with either monomeric IL-15 or the IL-15 superagonist fusion complex, ALT-803. These findings indicate that boosting NK cell expansion and functionality by immunotherapeutic strategies could improve survival in OC patients. [0157] Material and Methods [0158] Ascites fluid samples were prospectively collected at diagnosis or first surgery of patients with stage IIIc or IV high-grade serous papillary OC between January 2009 and January 2013 at the Radboud University Medical Center (RUMC). Study approval was given by the Regional Committee for Medical Research Ethics (CMO 2013-516) and performed according to the Code for Proper Secondary Use of Human Tissue (Dutch Federation of Biomedical Scientific Societies). Ascites was filtered using a 100µm filter, washed and MNCs were isolated by Ficoll-Hypaque density gradient centrifugation. Subsequently, obtained cells were cryopreserved and stored in liquid nitrogen until use. From this biobank ascitic cell samples were randomly selected from 20 OC patients. For the benign control group, samples were collected at benign gynecological surgeries. Indications for diagnostic laparoscopy were abdominal pain, in the absence of pathological finding. Detection of cysts, endometriosis and adhesions at laparoscopy were exclusion criteria. These benign samples were processed and analyzed on the day of surgery. Medical records were retrospectively reviewed and relevant clinical and pathology data were extracted. Time of diagnosis was considered to be the date of
the primary surgical procedure. Time from diagnosis to death was calculated for OS. PFS was calculated as time of last chemotherapy till diagnosis of histochemical or radiologic recurrence. Median survival was expressed in months. [0159] Flow cytometry. MNCs were stained with labeled antibodies, CD3 ECD (Biolegend), CD45 Krome Orange (R&D systems), CD56 PE-Cy5 (Biolegend), CD16 APC- Cy7 (Biologend), CD326 PerCPCy5.5 (Biolegend). Phenotypic analysis was performed using DNAM-1 FITC (Becton Dickinson), 2B4 FITC (Biolegend), NKG2A APC (Beckman Coulter), NKG2D APC (Biolegend), NKp30 PE (Biolegend) and NKp46 PE(Biolegend), isotype controls for IgG1 and IgG2a, (all Biolegend). Dead cells were stained with 1:1000 diluted sytox blue (Life Technologies; Invitrogen). Flow cytometry analysis was performed on a Gallios flow cytometer from Beckman Coulter. Analysis was done in Kaluza 1.5 (Beckman Coulter). [0160] K562 and SKOV-3 cell lines. OC cell line SKOV-3 was cultured in Roswell Park Memorial Institute medium (RPMI 1640; Gibco) medium supplemented with 10% Fetal Calf Serum (FCS; Integro). The chronic myeloid leukemia cell line K562 was cultured in Iscove's Modified Dulbecco’s Medium (IMDM) with 10% FCS. [0161] Functional assay (CD107a and IFN-γ). After thawing ascites MNCs or PBMCs were cultured overnight with 1 nM IL-15 (Immunotools), 1 nM ALT-803 (Altor Bioscience) or without cytokine support in IMDM with 10% FCS and 1% penicillin/streptomycin (p/s). Subsequently, 1 x 106 cells were co-cultured with 0.5 x 106 K562 cells, 0.5 x 106 SKOV-3 cells or without target cells for 4 hours in IMDM with 10% FCS and 1% p/s and anti-CD107a PE- Cy7 (Biolegend) in a 24-well plate. After 1h of co-culture, brefeldin A (BD) was added. Finally, cells in each well were gently resuspended and stained with labeled antibodies, CD56 BV510 (Biolegend), CD45 AF700 (Invitrogen), CD3 ECD (Beckman Coulter), CD69 BV421 (Biolegend) or isotype IgG1BV421 (BD biosciences), and TRAIL APC or isotype IgG1 APC (both Biolegend). Dead cells were stained with 1:1000 in PBS diluted eFluor780. Next, cells were fixed, permeabilized and stained with anti-IFN-γ or isotype IgG1 FITC (BD biosciences) and anti-perforin or isotype IgG2b PE (Biolegend). Flow cytometry acquisition was performed on a Gallios flow cytometer from Beckman Coulter. Analysis was done in Kaluza 1.5 (Beckman Coulter). [0162] Statistical analysis. Statistical analysis was performed in Graphpad Prism software package version 5.03. Flow cytometry data was expressed as percentage positive cells.
Data were analyzed using two-way ANOVA for group comparison or one-way ANOVA with Bonferroni post-hoc correction if more than two groups were compared. Unpaired T-tests were performed for comparison of two single groups, as indicated. Differences were considered significant when the p value was <0.05. Survival curves were analyzed by Log rank (Mantel- Cox) test. [0163] Results [0164] Patient cohort characteristics. The mean age of the selected OC patient cohort (n=20) was 64 ± 8.8 years and 48 ± 8.1 years for the benign gynecological disorder control group (n=10). The selected OC patient cohort was divided in two groups based on the median OS and PFS of 19 months and 6 months, respectively. The PFS and OS in the good survival group were 19.7 ± 16.4 and 32.9 ± 11.2 months, respectively. Whereas the PFS and OS in the poor survival group was only 3.2 ± 2.3 and 10.3 ± 4.4 months, respectively. Further characteristics of the two OC patient groups are shown in Table 1. Patients in the relatively good survival group were younger and were less often postmenopausal. In both groups, half of the OC patients were treated with primary surgery, and half with neo-adjuvant chemotherapy. CA-125 levels were higher in the good survival group. [0165] A higher peritoneal NK cell frequency is associated with better survival. First, the frequencies of NK, NKT and T cells in cryopreserved ascites samples of the selected OC patients were assessed by flow cytometry and compared those with peritoneal fluid of 10 patients with a benign gynecological disorder. OC ascites samples contained 38.8 ± 24.8% lymphocytes, 40.5 ± 24.7% CD45+ non-lymphocytes and 16.4 ± 23.5% CD45- non- hematopoietic tumor cells, and the benign samples contained 58.7 ± 40.4% lymphocytes and 36.5 ± 34.1% non-lymphocytes within CD45+ leucocytes (Figure 21A). A significantly lower NK cell frequency was seen in OC patient ascites (mean 17.1 ± 2.7%) compared to benign peritoneal fluid (48.1 ± 6.8%, p<0.0001; Figure 21B). Furthermore, lower CD3+ T cell and CD3+CD56+ NKT cell frequencies were observed within the lymphocyte population in OC patient ascites. The group of non T-, non-NKT, non-NK cells in the lymphocyte gate, presumably B cells, is more prominent in the malignant samples (Figure 21B). Interestingly, the group of OC patients with poor survival had only 14.5 ± 3.6% NK cells versus 23.6 ± 4.0% in the patients with good survival (Figure 21C). [0166] In addition, a significant shift in the NKdim/bright ratio was observed among patients in comparison to peripheral blood (PB) of benign donors (Figure 21D). Generally, in
healthy donor blood around 90% cytotoxic NKdim and 10% regulatory NKbright cells are present. In contrast, in the benign samples 32.4 ± 3.7% NKdim cells and 67.5 ± 3.7% NKbright cells was found, respectively. In OC patient ascites, the ratio appears to be more in favor of the cytotoxic NKdim population with 54.7 ± 4.0% NKdim and 45.4 ± 4.0% NKbright cells, compared to the benign peritoneal fluids (Figure 21D). Next, the OC patients were divided in two groups based on the median ascites NK cell frequency into high (mean 31.4 ± 9.4%) and low (mean 8.9 ± 4.6%) frequency groups. In line with division of the OC patients by poor versus good survival, we observed that both PFS (p=0.01, hazard ratio=4.7) and OS (p=0.002, hazard ratio=5.7) are significantly better in OC patients with a high NK cell frequency versus patients with a low NK cell frequency in ascites (Figure 21E). Notably, this relationship was not observed for CD3+ T cell percentages in ascites (Figure 21E-21H). Notably, in the high NK group three patients are still alive after a follow-up of ≥50 months. Altogether, these data indicate that the frequency of NK cells in ascites fluid of OC patients is positively correlated with clinical outcome. [0167] Activating receptors on ascites-derived NK cells are lowly expressed in poor survival ovarian carcinoma patients. Next to the frequency of NK cells in ascites, it was studied whether better survival in OC patients could be also related to differential expression of NK cell activating receptors. Hereto, flow cytometry analysis was performed on the peritoneal NK cells of the selected patient cohort and benign ascites controls (Figure 22). While 2B4 had equally high expression levels on both benign and malignant peritoneal fluid NK cells, NKG2D was low to undetectable on these NK cells. Remarkably, NKp30 was almost absent on NK cells in malignant samples (mean 3%), whereas it was significantly higher on NK cells from benign samples (mean 79%). Moreover, NKp46 and DNAM-1 were significantly lower expressed on NK cells in malignant samples, especially in patients with a poor OS. These data indicate that activation receptors on the surface of ascites NK cells are significantly lower in OC patients with poor survival. [0168] Ascites-derived NK cells possess equal cytotoxic function as PB-NK cells from healthy donors. Next, the functional activity of peritoneal fluid NK cells of OC patients was addressed in comparison to peripheral blood NK cells from healthy donors. Here, MNCs from ascites of OC patients or peripheral blood of healthy controls were cultured overnight in the presence of IL-15, where after total cells were challenged for 4 hours with either K562, SKOV- 3 or no tumor cell line and subsequently analyzed by flow cytometry. Expression of the activation markers CD69 and TRAIL, the degranulation of NK cells using CD107a, and
intracellular IFN-γ positivity was examined. Both ascites and control NK cells showed high CD69 expression, while TRAIL levels decreased upon tumor challenge (Figure 23A and 23B). CD107a and IFN-γ expression levels of ascites-derived NK cells in response to K562 stimulation varied between different OC patients (Figure 23C and 23D). However, no significant differences were observed as compared to healthy donor NK cells. Notably, for both OC ascites and healthy donor NK cells the response against SKOV-3 ovarian cancer cells was poor. Together, these data demonstrate that OC ascites-derived NK cells have equivalent degranulation and IFN-γ secretion capacity as healthy donor PB-NK cells, yet responsiveness against SKOV-3 OC cells is limited. [0169] Functionality of ascites NK cells can be effectively improved by IL-15 or ALT- 803 stimulation. To improve peritoneal NK cell reactivity against OC, it was investigated whether ascites-derived NK cells could be boosted with monomeric recombinant human IL-15 or the human IL-15 superagonist fusion complex, ALT-803. Interestingly, IL-15 receptor- mediated stimulation already enhanced CD107a expression and IFN-γ secretion capacity of the NK cells in the absence of tumor challenge (Figure 24A and 24D). Most importantly, ascites- derived NK responsiveness against K562 and especially SKOV-3 tumor cells could be potently augmented by IL-15 or ALT-803 stimulation (p<0.001 for CD107a and p<0.01 for IFN-γ; Figures 24A-24F). These data demonstrate that the function activation of NK cells in ascites can be efficiently rescued with IL-15 or ALT-803.
[0170] Discussion [0171] NK cells recognize cancer cells through a balance of activating and inhibitory receptors.18,19 Interestingly, epidemiological research has shown that low NK cell activity is associated with increased cancer risk in humans.20 Moreover, NK cells have been identified to play a role in tumor surveillance due to enhanced surface expression of ligands for activating receptors by the DNA damage response. However, there is limited data on the contribution of NK cell immunity on the clinical outcome of women with ovarian carcinoma. In the present study, it was found that the percentage of CD56+ NK cells in ascites fluid is related to OS and PFS, and that ascites-derived NK cells have lower expression of activation markers than benign peritoneal fluid NK cells. Although, ascites-resident NK cells have poor activity against SKOV-3 OC cells (similarly as PB-NK cells) they can be effectively boosted by IL-15 receptor mediated stimulation. [0172] OC patients were randomly selected from our ascites biobank. Overall, this is a relative poor prognosis group since these patients have large amounts of ascites. A remarkably high percentage of these patients did not undergo complete debulking surgery. However, since rates of complete debulking were comparable in both the poor and good OS groups, it was thought that the impact of incomplete debulking on the NK cell correlative results was limited. Notably, the OS of the patients is comparable to a large national cohort.43 Although there were age differences between the benign control and the OC groups, it is known that NK cell percentages in peripheral blood are very stable and do not change with age. Thus, age-related differences were not expected to have an impact on these findings. [0173] Notably, a higher NK cell frequency was found in peritoneal fluid of benign control subjects than in ascites of OC patients. Although never studied in relationship to cancer, peritoneal fluid of healthy individuals is measured in endometriosis research. Interestingly, also in endometriosis a significantly lower percentage of NK cells in peritoneal fluid is seen.44 In OC the only relation between clinical outcome and NK cells in ascites was described by Dong et al, and they related CD16 positive cells to poorer outcome of ovarian cancer patients. Unfortunately, CD56 expression was not evaluated in their report13. The study herein is the first report to show a relationship between frequency of NK cells in OC ascites and clinical outcome parameters. While the importance of CD8+ T cell infiltration within OC tumors has been clearly demonstrated, and also CD3+ T cells in ascites have been found to correlate with better outcome45,46, a relation between CD3+ T cell frequency and survival was not observed in this cohort. Normally, for peripheral blood the best way to correlate clinical outcome
parameters to NK cell anti-tumor activity would be to quantify absolute numbers. However, because volume and cellular density in ascites differs greatly between OC patients, it is believed that the frequency within lymphocyte population was the most objective way to compare cell populations. For future research, it would be interesting to investigate how the ascites NK cell frequency is compared to blood NK cell numbers, phenotype and function of the same OC patient. [0174] It has been previously reported that OC ascites NK cells have a lower expression of the activating markers NKp30, NKp46, NKG2D and DNAM-1 compared to healthy donor PB-NK cells.9,48 Here, the same significantly lower expression of NKp30, NKp46 and DNAM- 1 was observed on OC ascites-derived NK cells compared to NK cells from benign peritoneal fluid. However, the relationship between NKG2D and prognosis is not seen in our dataset. Interestingly, even a significant lower NK cell expression of NKp30 in poor prognosis ascites samples versus good prognosis was observed, suggesting that NKp30 expression changes in poor prognosis OC microenvironment. The same is seen in AML where NKp30 is proposed as a prognostic biomarker based on its low expression on NK cells in poor prognosis patients.49 Before markers on NK cells in ovarian cancer ascites can be used as a prognostic biomarker, validation in a larger cohort is required. [0175] In the functional studies, ascites-derived NK cells showed equal degranulation and cytokine secretion potential as healthy donor PB-NK cells. In contrast, other reports demonstrated that ascites-derived NK cells were dysfunctional, with decreased CD16 expression and low cytotoxic capacity.7,51,52 Here, it was shown that ascites-derived NK cells were highly capable of recognizing K562 tumor cells, but not SKOV-3 OC cells. However, as was shown here, high peritoneal NK cell frequency is correlated with better clinical outcome and addressed whether boosting with monomeric IL-15 or the IL-15 superagonist fusion complex, ALT803, could reinvigorate OC-activity. In this regard, it was demonstrated that after stimulation with IL-15 or ALT-803 degranulation improved and IFN-γ production increased, especially against SKOV-3 OC cells. Although the ex vivo studies did not show any difference between IL-15 and ALT-803, ALT-803 is likely more potent in longer assays and in vivo because of its longer half-life (Han KP, et al. Cytokine.2011;56(3):804-810. Rhode PR, et al. Cancer Immunol Res.2016 Jan;4(1):49-60). [0176] Concluding, this report shows a significant association between survival and frequency of NK cells in peritoneal fluid of OC patients. Moreover, it was demonstrated that peritoneal NK cell reactivity against OC tumor cells can be efficiently boosted by IL-15
receptor-mediated stimulation. The relationship between availability of NK cells in the abdominal cavity and the potentiating effect of IL-15 indicates that intraperitoneal NK cell adoptive transfer combined with IL-15 administration could be an interesting new therapy for OC patients to improve outcome. Currently, a phase 1 clinical trial testing intraperitoneal ALT- 803 therapy in OC patients is enrolling patients in the US (NCT0354909) and a phase 1 clinical trial on intraperitoneal NK cell therapy is open in the Netherlands (NCT03539406). By showing that a higher NK cell frequency is related to better outcome in OC patients and NK cell functionality can be boosted by IL-15 receptor stimulation, a part of NK cell immunity in OC is further deciphered to exploit NK cell based immunotherapy in these poor-prognosis patients. [0177] References 1. Ahmed N, Stenvers KL. Getting to know ovarian cancer ascites: opportunities for targeted therapy-based translational research. Front Oncol.2013;3:256. 2. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin. 2017;67(1):7-30. 3. Uppendahl LD, Dahl CM, Miller JS, Felices M, Geller MA. Natural Killer Cell- Based Immunotherapy in Gynecologic Malignancy: A Review. Front Immunol.2017;8:1825. 4. Fumita Y, Tanaka F, Saji F, Nakamuro K. Immunosuppressive factors in ascites fluids from ovarian cancer patients. Am J Reprod Immunol.1984;6(4):175-178. 5. Giuntoli RL, 2nd, Webb TJ, Zoso A, Rogers O, Diaz-Montes TP, Bristow RE, Oelke M. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res.2009;29(8):2875-2884. 6. Onsrud M, Bosnes V, Grahm I. cis-Platinum as adjunctive to surgery in early stage ovarian carcinoma: effects on lymphoid cell subpopulations. Gynecol Oncol.1986;23(3):323- 328. 7. Lukesova S, Vroblova V, Tosner J, Kopecky J, Sedlakova I, Cermakova E, Vokurkova D, Kopecky O. Comparative study of various subpopulations of cytotoxic cells in blood and ascites from patients with ovarian carcinoma. Contemp Oncol (Pozn). 2015;19(4):290-299. 8. Santin AD, Hermonat PL, Ravaggi A, Bellone S, Roman JJ, Smith CV, Pecorelli S, Radominska-Pandya A, Cannon MJ, Parham GP. Phenotypic and functional analysis of tumor-infiltrating lymphocytes compared with tumor-associated lymphocytes from ascitic fluid and peripheral blood lymphocytes in patients with advanced ovarian cancer. Gynecol Obstet Invest.2001;51(4):254-261. 9. Belisle JA, Gubbels JA, Raphael CA, Migneault M, Rancourt C, Connor JP, Patankar MS. Peritoneal natural killer cells from epithelial ovarian cancer patients show an altered phenotype and bind to the tumour marker MUC16 (CA125). Immunology.2007;122(3):418- 429. 10. Coca S, Perez-Piqueras J, Martinez D, Colmenarejo A, Saez MA, Vallejo C, Martos JA, Moreno M. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer.1997;79(12):2320-2328.
11. Gooden M, Lampen M, Jordanova ES, Leffers N, Trimbos JB, van der Burg SH, Nijman H, van Hall T. HLA-E expression by gynecological cancers restrains tumor- infiltrating CD8(+) T lymphocytes. Proc Natl Acad Sci U S A.2011;108(26):10656-10661. 12. Webb JR, Milne K, Watson P, Deleeuw RJ, Nelson BH. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer. Clin Cancer Res.2014;20(2):434-444. 13. Dong HP, Elstrand MB, Holth A, Silins I, Berner A, Trope CG, Davidson B, Risberg B. NK- and B-cell infiltration correlates with worse outcome in metastatic ovarian carcinoma. Am J Clin Pathol.2006;125(3):451-458. 14. Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Che X, Iwashige H, Aridome K, Hokita S, Aikou T. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer.2000;88(3):577-583. 15. Tian W, Wang L, Yuan L, Duan W, Zhao W, Wang S, Zhang Q. A prognostic risk model for patients with triple negative breast cancer based on stromal natural killer cells, tumor-associated macrophages and growth-arrest specific protein 6. Cancer Sci. 2016;107(7):882-889. 16. Turkseven MR, Oygur T. Evaluation of natural killer cell defense in oral squamous cell carcinoma. Oral Oncol.2010;46(5):e34-37. 17. Sconocchia G, Eppenberger S, Spagnoli GC, Tornillo L, Droeser R, Caratelli S, Ferrelli F, Coppola A, Arriga R, Lauro D, Iezzi G, Terracciano L, Ferrone S. NK cells and T cells cooperate during the clinical course of colorectal cancer. Oncoimmunology. 2014;3(8):e952197. 18. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol.2008;9(5):503-510. 19. Andre P, Castriconi R, Espeli M, Anfossi N, Juarez T, Hue S, Conway H, Romagne F, Dondero A, Nanni M, Caillat-Zucman S, Raulet DH, Bottino C, Vivier E, Moretta A, Paul P. Comparative analysis of human NK cell activation induced by NKG2D and natural cytotoxicity receptors. Eur J Immunol.2004;34(4):961-971. 20. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet.2000;356(9244):1795-1799. 21. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer.2016;16(1):7-19. 22. Pesce S, Tabellini G, Cantoni C, Patrizi O, Coltrini D, Rampinelli F, Matta J, Vivier E, Moretta A, Parolini S, Marcenaro E. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology. 2015;4(4):e1001224. 23. Berek JS, Bast RC, Jr., Lichtenstein A, Hacker NF, Spina CA, Lagasse LD, Knapp RC, Zighelboim J. Lymphocyte cytotoxicity in the peritoneal cavity and blood of patients with ovarian cancer. Obstet Gynecol.1984;64(5):708-714. 24. Tran E, Nielsen JS, Wick DA, Ng AV, Johnson LD, Nesslinger NJ, McMurtrie E, Webb JR, Nelson BH. Polyfunctional T-cell responses are disrupted by the ovarian cancer ascites environment and only partially restored by clinically relevant cytokines. PLoS One. 2010;5(12):e15625. 25. da Silva RF, Yoshida A, Cardozo DM, Jales RM, Paust S, Derchain S, Guimaraes F. Natural Killer Cells Response to IL-2 Stimulation Is Distinct between Ascites with the Presence or Absence of Malignant Cells in Ovarian Cancer Patients. Int J Mol Sci. 2017;18(5).
26. Felices M, Chu S, Kodal B, Bendzick L, Ryan C, Lenvik AJ, Boylan KLM, Wong HC, Skubitz APN, Miller JS, Geller MA. IL-15 super-agonist (ALT-803) enhances natural killer (NK) cell function against ovarian cancer. Gynecol Oncol.2017;145(3):453-461. 27. Kloss S, Chambron N, Gardlowski T, Weil S, Koch J, Esser R, Pogge von Strandmann E, Morgan MA, Arseniev L, Seitz O, Kohl U. Cetuximab Reconstitutes Pro- Inflammatory Cytokine Secretions and Tumor-Infiltrating Capabilities of sMICA-Inhibited NK Cells in HNSCC Tumor Spheroids. Front Immunol.2015;6:543. 28. Cany J, van der Waart AB, Spanholtz J, Tordoir M, Jansen JH, van der Voort R, Schaap NM, Dolstra H. Combined IL-15 and IL-12 drives the generation of CD34-derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer. Oncoimmunology.2015;4(7):e1017701. 29. Cany J, van der Waart AB, Tordoir M, Franssen GM, Hangalapura BN, de Vries J, Boerman O, Schaap N, van der Voort R, Spanholtz J, Dolstra H. Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrow-residing human leukemia cells in NOD/SCID/IL2Rg(null) mice. PLoS One.2013;8(6):e64384. 30. Roeven MW, Thordardottir S, Kohela A, Maas F, Preijers F, Jansen JH, Blijlevens NM, Cany J, Schaap N, Dolstra H. The Aryl Hydrocarbon Receptor Antagonist StemRegenin1 Improves In Vitro Generation of Highly Functional Natural Killer Cells from CD34(+) Hematopoietic Stem and Progenitor Cells. Stem Cells Dev.2015;24(24):2886-2898. 31. Spanholtz J, Tordoir M, Eissens D, Preijers F, van der Meer A, Joosten I, Schaap N, de Witte TM, Dolstra H. High log-scale expansion of functional human natural killer cells from umbilical cord blood CD34-positive cells for adoptive cancer immunotherapy. PLoS One.2010;5(2):e9221. 32. Geller MA, Knorr DA, Hermanson DA, Pribyl L, Bendzick L, McCullar V, Miller JS, Kaufman DS. Intraperitoneal delivery of human natural killer cells for treatment of ovarian cancer in a mouse xenograft model. Cytotherapy.2013;15(10):1297-1306. 33. Geller MA, Miller JS. Use of allogeneic NK cells for cancer immunotherapy. Immunotherapy.2011;3(12):1445-1459. 34. Hermanson DL, Bendzick L, Pribyl L, McCullar V, Vogel RI, Miller JS, Geller MA, Kaufman DS. Induced Pluripotent Stem Cell-Derived Natural Killer Cells for Treatment of Ovarian Cancer. Stem Cells.2016;34(1):93-101. 35. Bachanova V, Cooley S, Defor TE, Verneris MR, Zhang B, McKenna DH, Curtsinger J, Panoskaltsis-Mortari A, Lewis D, Hippen K, McGlave P, Weisdorf DJ, Blazar BR, Miller JS. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood.2014;123(25):3855-3863. 36. Giannattasio A, Weil S, Kloess S, Ansari N, Stelzer EH, Cerwenka A, Steinle A, Koehl U, Koch J. Cytotoxicity and infiltration of human NK cells in in vivo-like tumor spheroids. BMC Cancer.2015;15:351. 37. Bhat R, Watzl C. Serial killing of tumor cells by human natural killer cells-- enhancement by therapeutic antibodies. PLoS One.2007;2(3):e326. 38. Ames E, Canter RJ, Grossenbacher SK, Mac S, Chen M, Smith RC, Hagino T, Perez- Cunningham J, Sckisel GD, Urayama S, Monjazeb AM, Fragoso RC, Sayers TJ, Murphy WJ. NK Cells Preferentially Target Tumor Cells with a Cancer Stem Cell Phenotype. J Immunol. 2015;195(8):4010-4019. 39. Kozlowska AK, Kaur K, Topchyan P, Jewett A. Novel strategies to target cancer stem cells by NK cells; studies in humanized mice. Front Biosci (Landmark Ed). 2017;22:370-384.
40. Koh J, Lee SB, Park H, Lee HJ, Cho NH, Kim J. Susceptibility of CD24(+) ovarian cancer cells to anti-cancer drugs and natural killer cells. Biochem Biophys Res Commun. 2012;427(2):373-378. 41. Choi PJ, Mitchison TJ. Imaging burst kinetics and spatial coordination during serial killing by single natural killer cells. Proc Natl Acad Sci U S A.2013;110(16):6488-6493. 42. Rosario M, Liu B, Kong L, Collins LI, Schneider SE, Chen X, Han K, Jeng EK, Rhode PR, Leong JW, Schappe T, Jewell BA, Keppel CR, Shah K, Hess B, Romee R, Piwnica-Worms DR, Cashen AF, Bartlett NL, Wong HC, Fehniger TA. The IL-15-Based ALT-803 Complex Enhances FcgammaRIIIa-Triggered NK Cell Responses and In Vivo Clearance of B Cell Lymphomas. Clin Cancer Res. 2016;22(3):596-608. 43. Timmermans M, Sonke GS, Van de Vijver KK, van der Aa MA, Kruitwagen R. No improvement in long-term survival for epithelial ovarian cancer patients: A population-based study between 1989 and 2014 in the Netherlands. Eur J Cancer.2017;88:31-37. 44. Kang YJ, Jeung IC, Park A, Park YJ, Jung H, Kim TD, Lee HG, Choi I, Yoon SR. An increased level of IL-6 suppresses NK cell activity in peritoneal fluid of patients with endometriosis via regulation of SHP-2 expression. Hum Reprod.2014;29(10):2176-2189. 45. Lieber S, Reinartz S, Raifer H, Finkernagel F, Dreyer T, Bronger H, Jansen JM, Wagner U, Worzfeld T, Muller R, Huber M. Prognosis of ovarian cancer is associated with effector memory CD8(+) T cell accumulation in ascites, CXCL9 levels and activation- triggered signal transduction in T cells. Oncoimmunology.2018;7(5):e1424672. 46. Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S, Muller R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front Oncol.2017;7:24. 47. Funamizu A, Fukui A, Kamoi M, Fuchinoue K, Yokota M, Fukuhara R, Mizunuma H. Expression of natural cytotoxicity receptors on peritoneal fluid natural killer cell and cytokine production by peritoneal fluid natural killer cell in women with endometriosis. Am J Reprod Immunol.2014;71(4):359-367. 48. Nham T, Poznanski SM, Fan IY, Shenouda MM, Chew MV, Lee AJ, Vahedi F, Karimi Y, Butcher M, Lee DA, Hirte H, Ashkar AA. Ex vivo-expanded NK cells from blood and ascites of ovarian cancer patients are cytotoxic against autologous primary ovarian cancer cells. Cancer Immunol Immunother.2018;67(4):575-587. 49. Chretien AS, Fauriat C, Orlanducci F, Rey J, Borg GB, Gautherot E, Granjeaud S, Demerle C, Hamel JF, Cerwenka A, von Strandmann EP, Ifrah N, Lacombe C, Cornillet- Lefebvre P, Delaunay J, Toubert A, Arnoulet C, Vey N, Olive D. NKp30 expression is a prognostic immune biomarker for stratification of patients with intermediate-risk acute myeloid leukemia. Oncotarget.2017;8(30):49548-49563. 50. Pesce S, Greppi M, Tabellini G, Rampinelli F, Parolini S, Olive D, Moretta L, Moretta A, Marcenaro E. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J Allergy Clin Immunol.2017;139(1):335-346 e333. 51. Lai P, Rabinowich H, Crowley-Nowick PA, Bell MC, Mantovani G, Whiteside TL. Alterations in expression and function of signal-transducing proteins in tumor-associated T and natural killer cells in patients with ovarian carcinoma. Clin Cancer Res.1996;2(1):161- 173. 52. Ioannides CG, Platsoucas CD, Rashed S, Wharton JT, Edwards CL, Freedman RS. Tumor cytolysis by lymphocytes infiltrating ovarian malignant ascites. Cancer Res. 1991;51(16):4257-4265. Equivalents
[0178] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
Claims
What is claimed: 1. A method of preventing or treating cancer, comprising administering to a patient in need thereof, an effective amount of an adoptive cell therapy and a pharmaceutical composition comprising a therapeutically effective amount of at least one chemotherapeutic agent, thereby treating cancer.
2. The method of claim 1, wherein the adoptive cell therapy comprises hematopoietic stem cell transplantation, donor leukocyte infusion, adoptive transfer of natural killer cells (NK), T cells, B cells, chimeric antigen receptor- T cells (CAR-T), chimeric antigen receptor natural killer cells (CAR-NK) or combinations thereof.
3. The method of claim 2, wherein the adoptive cell therapy comprises transfer of allogeneic, autologous, syngeneic, related, unrelated, HLA-matched, HLA-mismatched or haploidentical cells.
4. The method of claim 2, wherein the NK cell is an allogeneic progenitor-derived NK cell.
5. The method of claim 1 wherein the at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof.
6. The method of claim 5, wherein the at least one chemotherapeutic agent is administered prior to the administration of the adoptive cell therapy.
7. The method of claim 5, wherein the at least one chemotherapeutic agent is administered concomitantly with the administration of the adoptive cell therapy.
8. The method of claim 5, wherein the at least one chemotherapeutic agent is administered after the administration of the adoptive cell therapy.
9. The method of claim 1, wherein the at least one chemotherapeutic agent comprises nucleoside analogs.
10. The method of claim 9, wherein the nucleoside analog is a hypomethylating agent.
11. The method of claim 9, wherein the hypomethylating agent comprises: 5- azacytidine, 5-aza-2'-deoxycytidine (5-AZA-CdR), zebularine, procainamide, procaine, hydralazine, epigallocathechin-3-gallate, RG108, MG98 or combinations thereof.
12. The method of claim 1, further comprising administering to the patient, a therapeutically effective amount of an IL-15:IL-15Rα complex.
13. The method of claim 12, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL- 15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules.
14. The method of claim 10, wherein the hypomethylating agent increases anti-tumor NK cell activity as compared to a non-hypomethylating agent treated control.
15. The method of claim 10, optionally comprising administering one or more cytokines.
16. A method of treating cancer, comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of a hypomethylating agent, thereby treating cancer.
17. The method of claim 16, wherein the NK cells are allogeneic cells.
18. The method of claim 16, wherein the NK cells are generated from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC).
19. The method of claim 16, wherein the hypomethylating agent is 5-aza-2'- deoxycytidine (5-AZA-CdR).
20. The method of claim 16, wherein the at least one chemotherapeutic agent is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof.
21. The method of claim 16, further comprising administering to the patient, a therapeutically effective amount of an IL-15:IL-15Rα complex.
22. The method of claim 21, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL- 15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules.
23. A kit for treating cancer, the kit comprising an adoptive cell therapy, at least one chemotherapeutic agent and directions for the use of the kit for the treatment of a cancer.
24. The kit of claim 23, wherein said adoptive cell therapy comprises hematopoietic stem cells, donor leukocytes, T cells, or natural killer (NK) cells.
25. The kit of claim 24, wherein the adoptive cell therapy comprises adoptively transferred allogeneic NK cells.
26. The kit of claim 25, wherein the NK cells are generated from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC).
27. The kit of claim 23, wherein the chemotherapeutic agent is a hypomethylating agent.
28. The kit of claim 23, further comprising an IL-15:IL-15Rα complex.
29. The kit of claim 28, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL- 15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules.
30. A method of treating cancer, comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of an IL-15:IL-15Rα complex, thereby treating cancer.
31. The method of claim 30, wherein the NK cells are allogeneic cells.
32. The method of claim 31, wherein the NK cells are generated from hematopoietic progenitor cell antigen CD34 positive hematopoietic stem and progenitor cells (HSPC).
33. The method of claim 30, further comprising administering one or more chemotherapeutic agents.
34. The method of claim 33, wherein the chemotherapeutic agent is a hypomethylating agent is 5-aza-2'-deoxycytidine (5-AZA-CdR).
35. The method of claim 34, wherein the hypomethylating agent is 5-aza-2'- deoxycytidine (5-AZA-CdR).
36. The method of claim 30, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL- 15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules.
37. A method of treating cancer, comprising administering to a patient in need thereof, an effective amount of adoptively transferred natural killer (NK) cells and, a composition comprising a therapeutically effective amount of an IL-15:IL-15Rα complex or IL-15, thereby treating cancer.
38. The method of claim 37, wherein the NK cells are obtained from one or more sources comprising ascites, peritoneum, lymph, blood, plasma or combinations thereof.
39. The method of claim 38, wherein the NK cells are obtained from ascites fluids.
40. The method of claim 37, wherein the IL-15/IL15Rα complex is an IL-15N72D:IL- 15RαSu/Fc complex (ALT-803) comprising a dimeric IL-15RαSu/Fc and two IL-15N72D molecules.
41. The method of claim 37, wherein the IL-15:IL-15Rα complex is administered prior to, simultaneously with, sequentially to the adoptive cell therapy, or any combination thereof.
42. The method of claim 41, wherein the IL-15:IL-15Rα complex is administered prior to the administration of the adoptive cell therapy.
43. The method of claim 41, wherein the IL-15:IL-15Rα complex is administered concomitantly with the administration of the adoptive cell therapy.
44. The method of claim 41, wherein the IL-15:IL-15Rα complex is administered after the administration of the adoptive cell therapy.
45. The method of claim 41, wherein the NK cells are optionally cultured with the IL- 15:IL-15Rα complex prior to the administration of the adoptive cell therapy.
46. The method of claim 37, further comprising administering one or more chemotherapeutic agents.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US201962935464P | 2019-11-14 | 2019-11-14 | |
| US62/935,464 | 2019-11-14 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2021097227A1 true WO2021097227A1 (en) | 2021-05-20 |
Family
ID=75912332
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2020/060440 Ceased WO2021097227A1 (en) | 2019-11-14 | 2020-11-13 | Il-15 fusion protein enhanced adoptive cell therapeutics |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2021097227A1 (en) |
Cited By (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN113599395A (en) * | 2021-09-22 | 2021-11-05 | 北京创世客生物技术有限公司 | Pharmaceutical composition for treating cancer containing NK cells |
| CN115778951A (en) * | 2022-12-20 | 2023-03-14 | 南京邮电大学 | Application of amoxicillin in enhancing anti-tumor activity of natural killer cells |
| CN118389428A (en) * | 2024-06-25 | 2024-07-26 | 深圳市中佳生物医疗科技有限公司 | Natural killer cell culture medium and application thereof |
| EP4450614A1 (en) * | 2023-04-19 | 2024-10-23 | Johann-Wolfgang-Goethe-Universität Frankfurt am Main | Treatment of leukemia by combining a hypomethylating agent in combination with engineered car-nk cells |
| WO2025114696A1 (en) * | 2023-11-30 | 2025-06-05 | Cambridge Enterprise Limited | Il-15 in combination with chemotherapy for treating cancer |
| WO2025151706A1 (en) * | 2024-01-12 | 2025-07-17 | Board Of Regents, The University Of Texas System | Epigenetic reagents and utilization of car nk cells |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20170071965A1 (en) * | 2003-02-19 | 2017-03-16 | Case Western Reserve University | Antimetabolite agent combinations in the treatment of cancer |
| US20170088597A1 (en) * | 2015-09-25 | 2017-03-30 | Altor Bioscience Corporation | Interleukin-15 superagonist significantly enhances graft-versus-tumor activity |
| US20190046623A1 (en) * | 2017-03-06 | 2019-02-14 | Altor Bioscience Corporation | Il-15-based fusions to il-12 and il-18 |
-
2020
- 2020-11-13 WO PCT/US2020/060440 patent/WO2021097227A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20170071965A1 (en) * | 2003-02-19 | 2017-03-16 | Case Western Reserve University | Antimetabolite agent combinations in the treatment of cancer |
| US20170088597A1 (en) * | 2015-09-25 | 2017-03-30 | Altor Bioscience Corporation | Interleukin-15 superagonist significantly enhances graft-versus-tumor activity |
| US20190046623A1 (en) * | 2017-03-06 | 2019-02-14 | Altor Bioscience Corporation | Il-15-based fusions to il-12 and il-18 |
Non-Patent Citations (2)
| Title |
|---|
| HOOGSTAD-VAN EVERT JANNEKE S, MAAS RALPH J, VAN DER MEER JOLIEN, CANY JEANNETTE, VAN DER STEEN SOPHIEKE, JANSEN JOOP H, MILLER JEF: "Peritoneal NK cells are responsive to IL-15 and percentages are correlated with outcome in advanced ovarian cancer patients", ONCOTARGET, vol. 9, 5 October 2018 (2018-10-05), pages 34810 - 34820, XP055823780 * |
| LUEVANO ET AL.: "Generation of natural killer cells from hematopoietic stem cells in vitro for immunotherapy", CELLULAR & MOLECULAR IMMUNOLOGY, vol. 9, 18 June 2012 (2012-06-18), pages 310 - 320, XP055561634, DOI: 10.1038/cmi.2012.17 * |
Cited By (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN113599395A (en) * | 2021-09-22 | 2021-11-05 | 北京创世客生物技术有限公司 | Pharmaceutical composition for treating cancer containing NK cells |
| CN113599395B (en) * | 2021-09-22 | 2022-08-12 | 郑州源创吉因实业有限公司 | Pharmaceutical composition for treating cancer comprising NK cells |
| CN115778951A (en) * | 2022-12-20 | 2023-03-14 | 南京邮电大学 | Application of amoxicillin in enhancing anti-tumor activity of natural killer cells |
| CN115778951B (en) * | 2022-12-20 | 2024-04-09 | 南京邮电大学 | Use of amoxicillin in enhancing anti-tumor activity of natural killer cells |
| EP4450614A1 (en) * | 2023-04-19 | 2024-10-23 | Johann-Wolfgang-Goethe-Universität Frankfurt am Main | Treatment of leukemia by combining a hypomethylating agent in combination with engineered car-nk cells |
| WO2025114696A1 (en) * | 2023-11-30 | 2025-06-05 | Cambridge Enterprise Limited | Il-15 in combination with chemotherapy for treating cancer |
| WO2025151706A1 (en) * | 2024-01-12 | 2025-07-17 | Board Of Regents, The University Of Texas System | Epigenetic reagents and utilization of car nk cells |
| CN118389428A (en) * | 2024-06-25 | 2024-07-26 | 深圳市中佳生物医疗科技有限公司 | Natural killer cell culture medium and application thereof |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US11717539B2 (en) | Combination immune therapy and cytokine control therapy for cancer treatment | |
| WO2021097227A1 (en) | Il-15 fusion protein enhanced adoptive cell therapeutics | |
| US12274714B2 (en) | Early apoptotic cells for use treating sepsis | |
| US11497767B2 (en) | Combination immune therapy and cytokine control therapy for cancer treatment | |
| US20190247431A1 (en) | Activated cd26-high immune cells and cd26-negative immune cells and uses thereof | |
| CN108697737A (en) | The composition used in immunotherapy | |
| AU2018282127B2 (en) | Therapeutic apoptotic cells for cancer therapy | |
| US20210008110A1 (en) | Activated lymphocytic cells and methods of using the same to treat cancer and infectious conditions | |
| US20250161412A1 (en) | Combination therapies for multiple myeloma | |
| Meehan et al. | Adoptive cellular therapy using cells enriched for NKG2D+ CD3+ CD8+ T cells after autologous transplantation for myeloma | |
| US12427184B2 (en) | Treatment of immunosuppressed subjects | |
| EP4031655A2 (en) | Combination cancer therapy and cytokine control therapy for cancer treatment | |
| CN114729314A (en) | Combination cancer therapy and cytokine control therapy for cancer treatment | |
| Kudo-Saito et al. | Targeting myeloid villains in the treatment with immune checkpoint inhibitors in gastrointestinal cancer | |
| KR20200035999A (en) | Treatment and suppression of leukemia using NK-92 cells | |
| WO2019213462A1 (en) | Inositol-based immunotherapies | |
| Cany et al. | Decitabine enhances targeting of AML cells by CD34+ stem and progenitor-derived NK cells in NOD/SCID/IL2Rgnull mice | |
| WO2020105034A1 (en) | Early apoptotic cells for use treating sepsis | |
| Langenkamp | Immunorecognition of leukemic stem cells by NK cells: the role of HDAC inhibitors in NKG2D ligand-mediated anti-tumor responses in acute myeloid leukemia |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20888714 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 32PN | Ep: public notification in the ep bulletin as address of the adressee cannot be established |
Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 19.08.2022) |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 20888714 Country of ref document: EP Kind code of ref document: A1 |