WO2020162582A1 - Méthodes de traitement de cancers non induits par voie virale - Google Patents

Méthodes de traitement de cancers non induits par voie virale Download PDF

Info

Publication number
WO2020162582A1
WO2020162582A1 PCT/JP2020/004711 JP2020004711W WO2020162582A1 WO 2020162582 A1 WO2020162582 A1 WO 2020162582A1 JP 2020004711 W JP2020004711 W JP 2020004711W WO 2020162582 A1 WO2020162582 A1 WO 2020162582A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cationic peptide
pharmaceutically acceptable
virally
omiganan
Prior art date
Application number
PCT/JP2020/004711
Other languages
English (en)
Inventor
Lee FEISS Gary
Matthijs MOERLAND
Maria Gerarda JIRKA Silvana
Original Assignee
Maruho Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Maruho Co., Ltd. filed Critical Maruho Co., Ltd.
Publication of WO2020162582A1 publication Critical patent/WO2020162582A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1729Cationic antimicrobial peptides, e.g. defensins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • Cationic peptides (also referred to as cationic antimicrobial peptides) were identified as naturally occurring, small, positively charged peptides that were secreted by immune and epithelial cells that serve as a component of the host defense mechanism in response to bacterial infections. These peptides have therefore, been investigated as potential anti-infective agents. In addition to their anti-microbial activity, these peptides appear to have pleiotropic effects in innate immunity. For example, the synthetic cationic peptide, omiganan, has been demonstrated to have in vitro activity against a wide variety of bacteria, which is believed to be due to the disruption of the cytoplasmic membranes of microorganisms, resulting in depolarization and cell death.
  • Cancer is a disease characterized by uncontrolled cell division and growth within the body. Millions of people are diagnosed with cancer every year worldwide. Standard treatments such as chemotherapy and radiotherapy are often not curative and are accompanied by potential toxicity and numerous undesirable side effects. Therefore, there remains a need for an improved therapeutic approach to treat cancers.
  • the human immune system may recognize and destroy cancer cells but cancer cells can evade the host immune system.
  • Cancer immunotherapy makes use of the body’s own immune system to help fight cancer and while some progress has been made in the field of cancer immunotherapy, a need still exists for improved treatment options.
  • the present invention addresses these needs and provides cationic peptides that are effective as anti-cancer agents.
  • the present invention provides a method of treating a non-virally-induced cancer in a subject comprising administering to the subject a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof.
  • the invention provides the use of a cationic peptide or pharmaceutically acceptable salt thereof in the preparation of a medicament for treating a non-virally-induced cancer in a subject.
  • the present invention provides a cationic peptide or pharmaceutically acceptable salt thereof for use in treating a non-virally induced cancer in a subject.
  • the cationic peptide may be omiganan, LL-37, 11B32CN, 11B36CN, 11E3CN, 11F4CN, 11F5CN, 11F12CN, 11F17CN, 11F50CN, 11F56CN, 11F63CN, 11F64CN, 11F66CN, 11F67CN, 11F68CN, 11F93CN, 11G27CN, 11J02CN, 11J02ACN, 11J30CN, 11J36CN, 11J58CN, 11J67CN, 11J68CN, Nt-acryloyl-11B7CN, Nt-glucosyl-11J36CN, Nt-glucosyl-11J38CN or a pharmaceutically acceptable salt thereof.
  • the cationic peptide is omiganan or LL-37.
  • the cationic peptide is omiganan.
  • the cationic peptide is LL-37.
  • the non-virally-induced cancer is gastric cancer, sarcoma, lymphoma, leukemia, head and neck cancer, thymic cancer, epithelial cancer, salivary cancer, liver cancer, stomach cancer, thyroid cancer, lung cancer, ovarian cancer, breast cancer, prostate cancer, esophageal cancer, pancreatic cancer, glioma, leukemia, multiple myeloma, renal cell carcinoma, bladder cancer, choriocarcinoma, colon cancer, colorectal cancer, oral cancer, testicular cancer, bone cancer, skin cancer, or melanoma.
  • the non-virally-induced cancer is colon cancer (e.g., colon carcinoma).
  • the non-virally-induced cancer is melanoma.
  • Another aspect of the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a cationic peptide or pharmaceutically acceptable salt thereof as disclosed herein and a pharmaceutically acceptable carrier.
  • the cationic peptide may be provided in conjunction with a counter anion.
  • the counter anion may be any pharmaceutically acceptable counter anion.
  • the cationic peptide may be provided in the form of any pharmaceutically acceptable salt.
  • the cationic peptide is omiganan pentahydrochloride.
  • the cationic peptide or pharmaceutically acceptable salt thereof may be administered topically or parentally.
  • the parenteral administration includes, but is not limited to, subcutaneous, intravenous, intramuscular, intraarterial, intraabdominal, intraperitoneal, intraarticular, intraocular or retrobulbar, intraaural, intrathecal, intracavitary, intracelial, intraspinal, intrapulmonary or transpulmonary, intrasynovial, or intraurethral injection or infusion.
  • the topical administration includes but is not limited to transdermal or inhalational delivery.
  • the methods and uses disclosed herein further comprise administration to the subject of an additional anti-cancer agent or therapy.
  • the additional anti-cancer agent may be a checkpoint inhibitor.
  • the checkpoint inhibitor is an inhibitor of the PD-1 pathway.
  • the additional anti-cancer agent is a chemotherapeutic agent.
  • chemotherapeutic agents include, but are not limited to, aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, campothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gemcitabine, genistein, gose
  • the cationic peptide or pharmaceutically acceptable salt thereof and the additional anti-cancer agent are in the same composition. In some embodiments, the cationic peptide or pharmaceutically acceptable salt thereof and the additional anti-cancer agent are administered simultaneously to the subject with a non-virally-induced cancer. In some embodiments, the cationic peptide or pharmaceutically acceptable salt thereof and the additional anti-cancer agent are in different compositions. In some embodiments, the cationic peptide or pharmaceutically acceptable salt thereof and the additional anti-cancer agent are administered sequentially to the subject with a non-virally-induced cancer.
  • Figures 1A and 1B show tumor outgrowth after repeated intratumoral injections of omiganan or PBS in mice with subcutaneous TC-1 (Fig. 1A) or CT26 tumors (Fig. 1B). Tumor size was measured by caliper on the indicated days post-tumor inoculation.
  • Figures 2A-2D show the effect of omiganan on neutrophils in CT26 and TC-1 mouse tumor models.
  • Fig. 2A shows flow cytometric analysis after three intratumoral injections of omiganan or PBS in TC-1 (top panel) and CT26 (bottom panel) mice for total immune cells using the cell surface marker CD45 and neutrophils specifically using the Ly6G marker. The presence of neutrophils in the tumor, as a percentage of all immune cells present in the tumor is shown.
  • Fig. 2B shows the results of metal-based mass cytometry (CyTOF) of cells after omiganan treatment of TC-1 mice.
  • CyTOF metal-based mass cytometry
  • the heatmap shows clusters of cells representing different cell types as columns, and each row indicates the presence of a cell surface marker on that particular cluster on a color scale.
  • Ly6G+ clusters are indicated by an arrow.
  • Fig. 2C is a bar graph showing the average abundance of clusters of different cell types in the omiganan or PBS-treated groups. Fig. 2C confirms that neutrophils increase after omiganan treatment (red versus blue bars).
  • Fig. 2D describes the cell types in the different clusters, the percentages, and the presence or absence of specific markers.
  • Figures 3A and 3B show tumor outgrowth after repeated intratumoral injections of omiganan or PBS in TC-1 (Fig. 3A) and CT26 (Fig. 3B) mice in the presence or absence of an anti-neutrophil antibody, aLy6G.
  • Figures 4A-4C show the effect of omiganan on the percentages of immune cell subsets and cell surface markers on neutrophils in CT26 tumors treated with omiganan or a PBS control.
  • Fig. 4A is a FACS analysis of the numbers of T cells (divided into CD4+ and CD8+ T cells).
  • Fig. 4B shows the percentage of total myeloid immune cells (top panel) as well as the percentages of Ly6G+, Ly6C-low, and Ly6C-high cells.
  • Fig. 4A is a FACS analysis of the numbers of T cells (divided into CD4+ and CD8+ T cells).
  • Fig. 4B shows the percentage of total myeloid immune cells (top panel) as well as the percentages of Ly6G+, Ly6C-low, and Ly6C-high cells.
  • 4C shows the mean fluorescence intensity of the markers CD54 and PD-L1 on Ly6G+ cells (neutrophils) (top panel) as well as the percentages of CD54+PD-L1+ cells in the Ly6G+ subset (neutrophils) (bottom panel) in the omiganan- and PBS-treatment groups.
  • Figure 5 is a dot plot of CT26 neutrophils analyzed by flow cytometry after omiganan treatment, where the axes indicate the expression level of the markers PD-L1 and CD54. Each dot represents a single neutrophil. The population inside the black box indicates that these two markers are mostly co-expressed on the same cells.
  • compositions are described as having, including, or comprising, specific components, it is contemplated that compositions also may consist essentially of, or consist of, the recited components.
  • the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system.
  • non-virally-induced cancer refers to a cancer that is not caused by a virus.
  • a non-virally-induced cancer is not caused by a viral infection such as but not limited to HPV, EBV, hepatitis B virus (HBV), hepatitis C virus (HCV), Human T-lymphotropic virus 1 (HTLV 1), Kaposi’s sarcoma-associated herpesvirus (KSHV or HHV8), Merkel cell polyoma virus (MCV), or Human cytomegalovirus (CMV or HHV-5) infection.
  • a viral infection such as but not limited to HPV, EBV, hepatitis B virus (HBV), hepatitis C virus (HCV), Human T-lymphotropic virus 1 (HTLV 1), Kaposi’s sarcoma-associated herpesvirus (KSHV or HHV8), Merkel cell polyoma virus (MCV), or Human cytomegalovirus (CMV or HHV-5) infection.
  • omiganan includes omiganan and its pharmaceutically acceptable salts, hydrates, solvates, esters, prodrugs, analogs, or derivatives thereof.
  • the term “administration” of an agent to a subject includes any route of introducing or delivering the agent to a subject to perform its intended function.
  • subject refers to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovine, porcine, etc.), companion animals (e.g., canine, feline, etc.) and rodents (e.g., mice and rats).
  • mammals such as humans, primates, livestock animals (including bovine, porcine, etc.), companion animals (e.g., canine, feline, etc.) and rodents (e.g., mice and rats).
  • treating includes reversing, reducing, ameliorating, alleviating, or arresting the symptoms, clinical signs or underlying pathology of a condition in a manner to improve, or stabilize the subject’s condition.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired results include, but are not limited to, prevention, alleviation, amelioration, or slowing the progression of one or more symptoms or conditions associated with a condition, diminishment of extent of disease, stabilized state of disease, delay or slowing of disease progression, amelioration or palliation of disease state, and remission (partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • the term “therapeutically effective amount” refers to an amount of an agent (e.g., omiganan or a pharmaceutically acceptable salt thereof) or composition comprising an agent (e.g., omiganan or a pharmaceutically acceptable salt thereof) that when administered to a subject will have the intended therapeutic effect (e.g. treatment or reduction of a symptom or symptoms).
  • the full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses.
  • the particular “therapeutically effective amount” will depend upon e.g., the age, weight and medical condition of the subject, as well as on the method of administration and the therapeutic or combination of therapeutics selected for administration. Suitable amounts are readily determined by persons skilled in the art.
  • the present invention provides a method of treating a non-virally-induced cancer in a subject by administering to the subject a cationic peptide.
  • the cationic peptides described herein provide a novel therapeutic approach for the treatment of non-virally induced cancers.
  • Naturally occurring cationic peptides are small, positively charged peptides that serve as a component of the host defense mechanism.
  • LL-37 is an antimicrobial peptide belonging to the cathelicidin family that plays an important role as a first line of defense against bacteria and other pathogens by disintegrating (damaging and puncturing) the cell membranes of bacteria and other pathogens.
  • the synthetic cationic peptide omiganan has in vitro activity against a wide variety of bacteria, which is believed to be due to the disruption of the cytoplasmic membranes of microorganisms, resulting in depolarization and cell death. While omiganan and LL-37 have been demonstrated to elicit antimicrobial effects in the context of disrupting pathogen-related structures and signals, the present invention relates generally to the discovery that cationic peptides also modulate anti-cancer immune responses. Without being bound by theory, these peptides are believed to attract immune cells to the tumor environment and induce anti-cancer responses.
  • the present invention is based on the observation that cationic peptides are capable of decreasing tumor outgrowth in a mouse model of a non-virally-induced cancer. Further, it is demonstrated herein that cationic peptides are capable of inducing an anti-tumor lymphocyte and monocyte response. Further, the cationic peptides are capable of upregulating the PD-L1 marker on tumor neutrophils, suggesting the use of combination therapy with a checkpoint inhibitor (e.g., a PD-1 pathway inhibitor) to treat non-virally induced cancers. Accordingly, the present invention provides formulations and therapeutic uses of cationic peptides.
  • a checkpoint inhibitor e.g., a PD-1 pathway inhibitor
  • the cationic peptides described herein are able to decrease tumor outgrowth in a mouse model of a non-virally-induced cancer. Accordingly, the present invention provides a method of treating a non-virally-induced cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof.
  • the cationic peptide or pharmaceutically acceptable salt thereof treats a non-virally-induced cancer by decreasing tumor outgrowth.
  • the cationic peptide or pharmaceutically acceptable salt thereof treats a non-virally-induced cancer by inducing an anti-cancer immune response.
  • the anti-cancer immune response is a lymphocyte response.
  • the anti-cancer immune response includes a proliferation of lymphocytes.
  • the anti-cancer immune response includes recruitment of lymphocytes to the tumor site.
  • the lymphocytes are CD4 and/or CD8 T cells.
  • the lymphocytes are CD4 T cells.
  • the lymphocytes are CD8 T cells.
  • the lymphocytes are CD4 and CD8 T cells.
  • the anti-cancer immune response is driven monocytes.
  • the anti-cancer immune response includes a proliferation of monocytes. In some embodiments, the anti-cancer immune response includes recruitment of monocytes to the tumor site. In some embodiments, the anti-cancer immune response includes a proliferation of CD54+ immune cells. In some embodiments, the anti-cancer immune response includes recruitment of CD54+ immune cells to the tumor site.
  • the non-virally-induced cancer includes, but is not limited to, gastric cancer, sarcoma, lymphoma, leukemia, head and neck cancer, thymic cancer, epithelial cancer, salivary cancer, liver cancer, stomach cancer, thyroid cancer, lung cancer, ovarian cancer, breast cancer, prostate cancer, esophageal cancer, pancreatic cancer, glioma, leukemia, multiple myeloma, renal cell carcinoma, bladder cancer, choriocarcinoma, colon cancer, colorectal cancer, oral cancer, testicular cancer, bone cancer, skin cancer, and melanoma.
  • the non-virally induced cancer is colon cancer (e.g., colon carcinoma).
  • the non-virally induced cancer is melanoma.
  • the cationic peptide used in the methods described herein includes, but is not limited to omiganan, LL-37, 11B32CN, 11B36CN, 11E3CN, 11F4CN, 11F5CN, 11F12CN, 11F17CN, 11F50CN, 11F56CN, 11F63CN, 11F64CN, 11F66CN, 11F67CN, 11F68CN, 11F93CN, 11G27CN, 11J02CN, 11J02ACN, 11J30CN, 11J36CN, 11J58CN, 11J67CN, 11J68CN, Nt-acryloyl-11B7CN, Nt-glucosyl-11J36CN or Nt-glucosyl-11J38CN, or a pharmaceutically acceptable salt thereof.
  • the cationic peptide is omiganan or LL-37 or a pharmaceutically acceptable salt thereof. In some embodiments, the cationic peptide is omiganan or a pharmaceutically acceptable salt thereof. In some embodiments, the cationic peptide is LL-37 or a pharmaceutically acceptable salt thereof. In some embodiments, the cationic peptide is omiganan. In some embodiments, the cationic peptide is omiganan pentahydrochloride. In some embodiments, the cationic peptide is LL-37.
  • the present invention provides a cationic peptide or pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a non-virally-induced cancer. In some embodiments, the present invention provides a cationic peptide or pharmaceutically acceptable salt thereof for use in treating a non-virally-induced cancer.
  • the present invention provides a method for treating a non-virally-induced cancer in a subject by administering a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof as a monotherapy. In some embodiments, the present invention provides a method for treating a non-virally-induced cancer in a subject by administering a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof in combination with one or more additional anti-cancer agents or therapies.
  • cationic peptides or pharmaceutically acceptable salts thereof may be used in combination therapies for the treatment, prevention, or management of a non-virally-induced cancer.
  • the administration of a cationic peptide or pharmaceutically acceptable salt thereof in combination with one or more anti-cancer agents or therapies, either concomitantly or sequentially, may enhance the therapeutic effect of the anti-cancer agent or therapy or overcome cellular resistance to such anti-cancer agent or therapy.
  • the cationic peptide or pharmaceutically acceptable salt thereof may be administered, for example, prior to, concurrently with, or subsequent to the one or more additional cancer agents or therapies, or vice versa.
  • the cationic peptide or pharmaceutically acceptable salt thereof and the one or more additional cancer agents or therapies may be administered at different administration sites, or at the same administration site, by the same administration route, or by different administration routes.
  • the cationic peptides or pharmaceutically acceptable salts thereof may be administered to patients in combination with radiation, surgical treatment, cytotoxic chemotherapy, or immunotherapy. Concurrent or sequential administration of a cationic peptide or pharmaceutically acceptable salt thereof and one or more additional anti-cancer agents or therapies is expected to provide effective treatment of cancer. Such combination treatments may work synergistically and allow reduction of dosage of each of the individual treatments, thereby reducing the detrimental side effects exerted by each treatment at higher dosages.
  • the cationic peptide or pharmaceutically acceptable salt thereof and the one or more additional anti-cancer agents are used at doses below what is normally a therapeutically effective dose when these anti-cancer agents are used individually.
  • the additional anti-cancer agent and cationic peptide can be administered using a normally effective therapeutic dose for each agent.
  • the invention provides a method of treating subjects having malignancies that are refractory to treatment with other anti-cancer agents or therapies by administering to said subject a cationic peptide or pharmaceutically acceptable salt thereof as described herein.
  • Chemotherapeutic compounds that may be used for combinatory anti-tumor therapy include, but are not limited to any one or more of: aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, campothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone
  • chemotherapeutic anti-tumor compounds may be categorized by their mechanism of action into, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthr
  • the dosage and selection of the chemotherapeutic agent can be determined by a health care professional based on common knowledge in the art.
  • the cationic peptides or pharmaceutically acceptable salts thereof of the invention may be administered to patients in combination with a checkpoint inhibitor.
  • the checkpoint inhibitor may inhibit one or more immune checkpoints, such as but not limited to, programmed cell death protein 1 (PD-1), Cytotoxic T-lymphocyte antigen 4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte-activation gene 3 (LAG-3), or T-cell immunoreceptor with Ig and ITIM domains (TIGIT).
  • the checkpoint inhibitor may be an antagonistic antibody targeting an immune checkpoint or a member of its signaling pathway.
  • antibody includes monoclonal and polyclonal antibodies and antibodies with polyepitopic specificity.
  • Antibody typically comprises any antibody known in the art (e.g., IgG, IgD, IgM, IgA, and IgE antibodies), such as naturally occurring antibodies, antibodies generated by immunization in a host organism, as well as chimeric antibodies, humanized antibodies, human antibodies, bispecific antibodies, etc.
  • the term “antibody” also includes derivates of antibodies such as antibody fragments, variants or adducts, that are capable of binding to the antigen. Antibody fragments may be selected from Fab, Fab’, F(ab’) 2 , Fd, Fv, and scFv fragments of full-length antibodies.
  • the checkpoint inhibitor may be an siRNA or antisense RNA directed against an immune checkpoint or a member of its signaling pathway.
  • the checkpoint inhibitor may also be a polypeptide or fragment thereof comprising an amino acid sequence capable of binding to an immune checkpoint or a member of its signaling pathway and inbiting signaling.
  • a checkpoint inhibitor may be a small molecule inhibitor capable of inhibiting immune checkpoint signaling, e.g., a small organic molecule.
  • the dosage and selection of the checkpoint inhibitor can be determined by a health care professional based on common knowledge in the art.
  • the cationic peptides or pharmaceutically acceptable salts thereof of the invention may be administered to patients in combination with an inhibitor of the PD-1 pathway.
  • PD-1 also known as CD279 is a cell surface membrane protein of the immunoglobulin superfamily, which is expressed in B cells and NK cells (Shinohara et al., 1995, Genomics 23:704-6; Blank et al., 2007, Cancer Immunol Immunother 56:739-45; Finger et al., 1997, Gene 197:177-87; Pardoll, 2012, Nature Reviews 12:252-264).
  • the PD-1 pathway plays a role in tumor immune evasion and blockade of the PD-1 pathway restores anti-tumor responses.
  • a PD-1 pathway inhibitor may be any compound directed against any member of the PD-1 pathway (e.g., PD-1, PD-L1, PD-L2, etc.) that is capable of antagonizing PD-1 pathway signaling, such as signaling mediated by the PD-1 receptor.
  • the inhibitor may be an antagonistic antibody targeting any member of the PD-1 pathway (e.g., PD-1, PD-L1, PD-L2, etc.).
  • the PD-1 pathway inhibitor may be siRNA or antisense RNA directed against a PD-1 pathway member (e.g., PD-1, PD-L1, PD-L2, etc.).
  • the PD-1 pathway inhibitor may also be a polypeptide or fragment thereof comprising an amino acid sequence capable of binding to PD-1 and preventing PD-1 signaling (e.g., a fusion protein of a fragment of PD-L1 or PD-L2 and the Fc part of an immunoglobulin or a soluble protein that competes with PD-1 for binding to PD-L1 or PD-L2.
  • a PD-1 pathway inhibitor may be a small molecule inhibitor capable of inhibiting PD-1 pathway signaling, e.g., a small organic molecule.
  • the PD-1 pathway inhibitor is an antibody.
  • An antibody may be selected from any antibody, e.g., any recombinantly produced or naturally occurring antibodies, directed against PD-1, PD-L1, or PD-L2.
  • the PD-1 pathway inhibitor is an anti-PD-1 antibody that is capable of inhibiting PD-1 signaling.
  • the antibody specifically binds the extracellular domain of PD-1.
  • the antibody binds at or near the binding site of PD-L1 or PD-L2 for PD-1, thus inhibiting the binding of the ligands to PD-1.
  • the anti-PD1 antibody includes, but is not limited to, any one or more of nivolumab , pembrolizumab, cemiplimab, or pidilizumab. Other suitable anti-PD-1 antibodies are known in the art.
  • the PD-1 pathway inhibitor is an anti-PD-L1 antibody.
  • the antibody binds at or near the binding site of PD-L1 for PD-1, thus inhibiting the binding of PDL-1 to PD-1.
  • the anti-PD-L1 antibody includes, but is not limited to, any one or more of atezolizumab, avelumab, durvalumab, or MDX-1105/BMS-936559. Other suitable anti-PD-L1 antibodies are known in the art.
  • the PD-1 pathway inhibitor is a fusion protein comprising the extracellular domain of PD-L1 or PD-L2 or a fragment thereof capable of binding to PD-1 and an Fc portion of an immunoglobulin.
  • the fusion protein is AMP-224 (extracellular domain of murine PD-L2/B7-DC fused to the unmodified Fc portion of murine IgG2a protein).
  • Other PD-1 pathway inhibitors are known in the art.
  • the dosage and selection of the PD-1 pathway inhibitor can be determined by a health care professional based on common knowledge in the art.
  • the cationic peptides or pharmaceutically acceptable salts thereof of the invention may be administered to patients in combination with a CTLA-4 inhibitor.
  • CTLA-4 also known as CD152 acts to inhibit T cell activation and is reported to inhibit helper T cell activity and enhance regulatory T cell immunosuppressive activity (Pardoll, 2012, Nature Reviews 12:252-264).
  • the CTLA-4 inhibitor may be an antagonistic antibody targeting CTLA-4 or a member of its signaling pathway.
  • Exemplary anti-CTLA4 antibodies include ipilimumab (Bristol-Myers Squibb) and tremelimumab (MedImmune).
  • the CTLA-4 inhibitor may be an siRNA or antisense RNA directed against CTLA-4 or a member of its signaling pathway.
  • the checkpoint inhibitor may also be a polypeptide or fragment thereof comprising an amino acid sequence capable of binding to CTLA-4 or a member of its signaling pathway and inbiting signaling.
  • a checkpoint inhibitor may be a small molecule inhibitor capable of inhibiting CTLA-4 signaling, e.g., a small organic molecule.
  • the additional anti-cancer therapy is a treatment that depletes neutrophils.
  • the treatment that depeletes neutrophils is an anti-neutrophil antibody (e.g., anti-Lys6G antibody).
  • the cationic peptides and the one or more additional agents may be administered either in the same formulation or in separate formulations, either concomitantly, sequentially, or on different schedules.
  • the one or more additional agents may be administered separately from the cationic peptide disclosed herein, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with a cationic peptide in a single composition.
  • the period between administration of the cationic peptide and the one or more additional agent may vary from minutes, hours, days or months from each. In general, each agent will be administered at a dose and/or on a time schedule determined for that particular agent.
  • composition(s) and dosing frequency(ies) of the combination therapy will depend on a variety of factors, including the route of administration, the condition being treated, any potential interactions between the active ingredients when combined into a single composition, any interactions between the active ingredients when they are administered to the subject, and various other factors known to those skilled in the art.
  • the present invention is directed generally to methods of treating a non-virally-induced cancer using cationic peptides or pharmaceutically acceptable salts thereof.
  • the cationic peptides useful in the methods and compositions described herein may be produced by a variety of methods (e.g., chemical or recombinant). Suitable cationic peptides include, but are not limited to, naturally occurring cationic peptides, which have been isolated, and derivatives or analogs thereof.
  • An “isolated peptide, polypeptide, or protein” is an amino acid sequence that is essentially free from contaminating cellular components, such as carbohydrate, lipid, nucleic acid (DNA or RNA), or other proteinaceous impurities associated with the polypeptide in nature. Preferably, the isolated polypeptide is sufficiently pure for therapeutic use at the desired dose.
  • the cationic peptide useful in the methods and compositions disclosed herein may be a recombinant peptide or a synthetic peptide, and is preferably a synthetic peptide.
  • Peptides may be synthesized by standard chemical methods, including synthesis by automated procedure.
  • Peptide analogues may be synthesized based on the standard solid-phase Fmoc protection strategy with HATU as the coupling agent.
  • the peptide is cleaved from the solid-phase resin with trifluoroacetic acid containing appropriate scavengers, which also deprotects side chain functional groups.
  • Peptide analogues may also be synthesized by liquid-phase synthesis. Crude peptide is further purified using preparative reversed-phase chromatography.
  • Peptides may be synthesized as a linear molecule or as branched molecules. Branched peptides typically contain a core peptide that provides a number of attachment points for additional peptides. Lysine is most commonly used for the core peptide because it has one carboxyl functional group and two (alpha and epsilon) amine functional groups. Other diamino acids can also be used. Preferably, either two or three levels of geometrically branched lysines are used; these cores form a tetrameric and octameric core structure, respectively.
  • the cationic peptides useful in the methods and compositions disclosed herein are peptides that typically exhibit a positive charge at a pH ranging from about 3 to about 10 (i.e., has an isoelectric point of at least about 9), and contain at least one basic amino acid (e.g., arginine, lysine, histidine).
  • the cationic peptide generally comprises an amino acid sequence having a molecular mass of about 0.5 kDa (i.e., approximately five amino acids in length) to about 10 kDa (i.e., approximately 100 amino acids in length), or a molecular mass of any integer, or fraction thereof (including a tenth and one hundredth of an integer), ranging from about 0.5 kDa to about 10 kDa.
  • the cationic peptide has a molecular mass ranging from about 0.5 kDa to about 5 kDa (i.e., approximately from about 5 amino acids to about 45 amino acids in length), from about 1 kDa to about 4 kDa (i.e., approximately from about 10 amino acids to about 35 amino acids in length), or from about 1 kDa to about 2 kDa (i.e., approximately from about 10 amino acids to about 18 amino acids in length).
  • the cationic peptide is part of a larger peptide or polypeptide sequence having, for example, a total of up to 100 amino acids, up to 50 amino acids, up to 35 amino acids, or up to 15 amino acids.
  • the methods of the invention contemplate a cationic peptide having an amino acid sequence of 5 to 100 amino acids, with the number of amino acids making up the peptide sequence comprising any integer in that range.
  • the cationic peptide may exhibit anti-cancer activity, and synergistic activity with other anti-cancer peptides or anti-cancer agents, or a combination thereof.
  • Exemplary peptides include, but are not limited to, cathelicidins, such as indolicidin and derivatives or analogues thereof from bovine neutrophils (Falla et al., J. Biol. Chem. 277:19298, 1996).
  • the cationic peptides are indolicidins or analogs or derivatives thereof.
  • Natural indolicidins may be isolated from a variety of organisms, and, for example, the indolicidin isolated from bovine neutrophils is a 13 amino acid peptide, which is tryptophan-rich and amidated at the C-terminus (see Selsted et al., J. Biol. Chem. 267:4292, 1992).
  • an indolicidin or analog or derivative thereof comprises 5 to 45 amino acids, 7 to 35 amino acids, 8 to 25 amino acids, or 10 to 14 amino acids.
  • the cationic peptide used in the methods and compositions disclosed herein is a peptide of up to 35 amino acids, comprising one of the sequences in Table 1, infra.
  • the cationic peptide is LL-37. In some embodiments, the cationic peptide is LL-22, a 22 amino acid variant of LL-37. In some embodiments, the cationic peptide is an N-terminal 22 amino acid truncation of LL-37. In some embodiments, the cationic peptide is a C-terminal 22 amino acid truncation of LL-37. In some embodiments, the cationic peptide is a fragment of LL-37. In some embodiments, the fragment of LL-37 has residues 1-22 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 2-23 of SEQ ID NO: 1.
  • the fragment of LL-37 has residues 3-24 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 4-25 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 5-26 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 6-27 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 7-28 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 8-29 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 9-30 of SEQ ID NO: 1.
  • the fragment of LL-37 has residues 10-31 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 11-32 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 12-33 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 13-34 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 14-35 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 15-36 of SEQ ID NO: 1. In some embodiments, the fragment of LL-37 has residues 16-37 of SEQ ID NO: 1. LL-37 variants are described in van der Does et al.
  • the cationic peptide is omiganan.
  • the cationic peptide is 11B32CN.
  • the cationic peptide is 11B36CN.
  • the cationic peptide is 11E3CN.
  • the cationic peptide is 11F4CN.
  • the cationic peptide is 11F5CN.
  • the cationic peptide is 11F12CN.
  • the cationic peptide is 11F17CN.
  • the cationic peptide is 11F50CN.
  • the cationic peptide is 11F56CN. In other embodiments, the cationic peptide is 11F63CN. In some embodiments, the cationic peptide is 11F64CN. In other embodiments, the cationic peptide is 11F66CN. In some embodiments, the cationic peptide is 11B32CN. In other embodiments, the cationic peptide is 11F67CN. In some embodiments, the cationic peptide is 11F68CN. In other embodiments, the cationic peptide is 11F93CN. In some embodiments, the cationic peptide is 11G27CN. In other embodiments, the cationic peptide is 11J02CN.
  • the cationic peptide is 11J02ACN. In other embodiments, the cationic peptide is 11J30CN. In some embodiments, the cationic peptide is 11J36CN. In other embodiments, the cationic peptide is 11J58CN. In some embodiments, the cationic peptide is 11J67CN. In other embodiments, the cationic peptide is 11J68CN. In some embodiments, the cationic peptide is Nt-acryloyl-11B7CN. In other embodiments, the cationic peptide is Nt-glucosyl-11J36CN. In some embodiments, the cationic peptide is Nt-glucosyl-11J38CN. In some embodiments, the cationic peptide is a pharmaceutically acceptable salt of any of the foregoing.
  • the cationic peptide is provided in conjunction with a counter anion.
  • the counter anion may be any pharmaceutically acceptable counter anion.
  • the cationic peptide is omiganan pentahydrochloride.
  • the cationic peptide used in the methods and compositions described herein may be an analog or derivative thereof.
  • the terms “derivative” and “analog” when referring to a cationic peptide, polypeptide, or fusion protein refer to any cationic peptide, polypeptide, or fusion protein that retain essentially the same (at least 50%, 60% and preferably greater than 70, 80, or 90%) or enhanced biological function or activity as such cationic peptide, as noted above.
  • the biological function or activity of such analogs and derivatives can be determined using standard methods (e.g., anti-cancer), such as with the assays described herein.
  • an analog or derivative may be a proprotein that can be activated by cleavage to produce an active anti-cancer cationic peptide.
  • a cationic peptide analog or derivative thereof can be identified by the ability to specifically bind anti-cationic peptide antibodies.
  • the cationic peptide analog or derivative may have, for example, one or more deletion, insertion, or modification of any amino acid residue, including the N- or C- terminal amino acids.
  • the cationic peptide analog or derivative includes modified cationic peptides, such as, for example, peptides having an acetylated, acylated, acryloylated, alkylated, glycosylated (e.g., glucosylated), PEGylated, myristylated, and the like N-terminal amino acid modification; having an esterified, amidated, homoserinelhomoserine lactone, or caprolactam C-terminal amino acid modification; or having a polyalkylene glycol (e.g., polyethylene glycol) conjugated to any free amino group.
  • modified cationic peptides such as, for example, peptides having an acetylated, acylated, acryloylated, alkylated, glycos
  • a preferred modification of the C-terminal amino acid is amidation.
  • An analog or derivative may also be a cationic peptide fusion protein. Fusion proteins, or chimeras, include fusions of one or more cationic peptides, and fusions of cationic peptides with non-cationic peptides. Additionally, the peptide may be modified to form a polymer-modified peptide. The peptides may also be labeled, such as with a radioactive label, a fluorescent label, a mass spectrometry tag, biotin, and the like.
  • an analog or derivative includes a cationic peptide that has one or more conservative amino acid substitutions, as compared with the amino acid sequence of a cationic peptide of the present invention.
  • a “conservative amino acid substitution” is illustrated, for example, by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine, or a combination thereof.
  • an analog or derivative of a cationic peptide may include, for example, non-protein amino acids, such as precursors of normal amino acids (e.g., homoserine and diaminopimelate), intermediates in catabolic pathways (e.g., pipecolic acid and D-enantiomers of normal amino acids), and amino acid analogs (e.g., azetidine-2-carboxylic acid and canavanine).
  • non-protein amino acids such as precursors of normal amino acids (e.g., homoserine and diaminopimelate), intermediates in catabolic pathways (e.g., pipecolic acid and D-enantiomers of normal amino acids), and amino acid analogs (e.g., azetidine-2-carboxylic acid and canavanine).
  • the cationic peptides described herein may be used individually, or may be used in combination with one or more different cationic peptides, or with one or more conventional anti-cancer agents or therapies, as described herein.
  • nucleotide sequences encoding conservative amino acid analogs or derivatives can be obtained, for example, by oligonucleotide-directed mutagenesis, linker-scanning mutagenesis, mutagenesis using the polymerase chain reaction, and the like (see Ausubel, 1995, at page 8-10 through page 8-22; and McPherson (ed.), Directed Mutagenesis: A Practical Approach, IRL Press, 1991).
  • a DNA sequence encoding the desired cationic peptide is optimized for a particular host system, such as prokaryotic or eukaryotic cells.
  • a nucleotide sequence encoding a radish cationic peptide may include a codon that is commonly found in radish, but is rare for E. coli.
  • Peptides may be synthesized by recombinant techniques (see e.g., U.S. Patent No. 5,593,866) and a variety of host systems are suitable for production of the cationic peptides and analogues or derivatives thereof, including bacteria (e.g., E. coli), yeast (e.g., Saccharomyces cerevisiae), insect (e.g., Sf9), and mammalian cells (e.g., CHO, COS-7). Many expression vectors have been developed and are available for each of these hosts. Generally, vectors that are functional in bacteria are used in this invention. However, at times, it may be preferable to have vectors that are functional in other hosts.
  • bacteria e.g., E. coli
  • yeast e.g., Saccharomyces cerevisiae
  • insect e.g., Sf9
  • mammalian cells e.g., CHO, COS-7
  • a DNA sequence encoding a cationic peptide is introduced into an expression vector appropriate for the host.
  • the gene is cloned into a vector to create a fusion protein.
  • the fusion partner is chosen to contain an anionic region, such that a bacterial host is protected from the toxic effect of the peptide.
  • the fusion partner (carrier protein) of the invention may further function to transport the fusion peptide to inclusion bodies, the periplasm, the outer membrane, or the extracellular environment.
  • Carrier proteins suitable in the context of this invention specifically include, but are not limited to, glutathione-S-transferase (GST), protein A from Staphylococcus aureus, two synthetic IgG-binding domains (ZZ) of protein A, outer membrane protein F, ⁇ -galactosidase (lacZ), and various products of bacteriophage ⁇ and bacteriophage T7. From the teachings provided herein, it is apparent that other proteins may be used as carriers. Furthermore, the entire carrier protein need not be used, as long as the protective anionic region is present.
  • amino acids susceptible to chemical cleavage e.g., CNBr
  • enzymatic cleavage e.g., VB protease, trypsin
  • the fusion partner may be a normal intracellular protein that directs expression toward inclusion body formation. In such a case, following cleavage to release the final product, there is no requirement for renaturation of the peptide.
  • the DNA cassette, comprising fusion partner and peptide gene may be inserted into an expression vector, which can be a plasmid, virus or other vehicle known in the art.
  • the expression vector may be a plasmid that contains an inducible or constitutive promoter to facilitate the efficient transcription of the inserted DNA sequence in the host. Transformation of the host cell with the recombinant DNA may be carried out by Ca ++ -mediated techniques, by electroporation, or other methods well known to those skilled in the art. Briefly, a DNA fragment encoding a peptide is derived from an existing cDNA or genomic clone or synthesized. A convenient method is amplification of the gene from a single-stranded template. The template is generally the product of an automated oligonucleotide synthesis.
  • Amplification primers are derived from the 5' and 3' ends of the template and typically incorporate restriction sites chosen with regard to the cloning site of the vector. If necessary, translational initiation and termination codons can be engineered into the primer sequences.
  • the sequence encoding the protein may be codon optimized for expression in the particular host. Thus, for example, if the analogue fusion protein is expressed in bacteria, codons are optimized for bacterial usage. Codon optimization is accomplished by automated synthesis of the entire gene or gene region, ligation of multiple oligonucleotides, mutagenesis of the native sequence, or other techniques known to those in the art.
  • the vector is capable of replication in bacterial cells.
  • the vector may contain a bacterial origin of replication.
  • Preferred bacterial origins of replication include f1-ori and col E1 ori, especially the ori derived from pUC plasmids.
  • Low copy number vectors e.g., pPD100
  • the plasmids may also include at least one selectable marker that is functional in the host. A selectable marker gene confers a phenotype on the host that allows transformed cells to be identified and/or selectively grown.
  • Suitable selectable marker genes for bacterial hosts include the chloramphenicol resistance gene (Cm r ), ampicillin resistance gene (Amp r ), tetracycline resistance gene (Tc r ) kanamycin resistance gene (Kan r ), and others known in the art. To function in selection, some markers may require a complementary deficiency in the host.
  • the vector may also contain a gene coding for a repressor protein, which is capable of repressing the transcription of a promoter that contains a repressor binding site. Altering the physiological conditions of the cell can depress the promoter. For example, a molecule may be added that competitively binds the repressor, or the temperature of the growth media may be altered.
  • Repressor proteins include, but are not limited to the E. coli lacI repressor (responsive to induction by IPTG), the temperature sensitive ⁇ cl857 repressor, and the like.
  • the expression vector should contain a promoter sequence.
  • other regulatory sequences may also be included.
  • Such sequences include an enhancer, ribosome binding site, transcription termination signal sequence, secretion signal sequence, origin of replication, selectable marker, and the like.
  • the regulatory sequences are operably linked with one another to allow transcription and subsequent translation.
  • the plasmids used herein for expression include a promoter designed for expression of the proteins in bacteria. Suitable promoters, including both constitutive and inducible promoters, are widely available and are well known in the art. Commonly used promoters for expression in bacteria include promoters from T7, T3, T5, and SP6 phages, and the trp, lpp, and lac operons.
  • Hybrid promoters such as tac and trc
  • examples of plasmids for expression in bacteria include the pET expression vectors pET3a, pET 11a, pET 12a-c, and pET 15b (see U.S. Patent 4,952,496; available from Novagen, Madison, WI).
  • Low copy number vectors e.g., pPD1 00
  • pPD1 00 can be used for efficient overproduction of peptides deleterious to the E. coli host (Dersch et al., FEMS Microbial. Lett. 123: 19, 1994).
  • Bacterial hosts for the T7 expression vectors may contain chromosomal copies of DNA encoding T7 RNA polymerase operably linked to an inducible promoter (e.g., lacUV promoter; see, U.S. Patent No. 4,952,496), such as found in the E. coli strains HMS174(DE3)plysS, BL21(DE3)plysS, HMS174(DE3) and BL21 (DE3).
  • T7 RNA polymerase can also be present on plasmids compatible with the T7 expression vector.
  • the polymerase may be under control of a lambda promoter and repressor (e.g., pGP1-2; Tabor and Richardson, Proc. Natl. Acad. Sci. USA 82: 1074, 1985).
  • sequence of nucleotides encoding the peptide also encodes a secretion signal, such that the resulting peptide is synthesized as a precursor protein (i.e., proprotein), which is subsequently processed and secreted.
  • the resulting secreted peptide or fusion protein may be recovered from the periplasmic space or the fermentation medium. Sequences of secretion signals suitable for use are widely available and are well known (von Heijne, J. Mol. Biol. 184:99-105, 1985).
  • the peptide product is isolated by standard techniques, such as affinity, size exclusion, or ionic exchange chromatography, HPLC and the like.
  • An isolated peptide should show a major band by Coomassie blue stain of SDS-PAGE, which is at least 75%, 80%, 90%, or 95% of the purified peptide, polypeptide, or fusion protein.
  • a therapeutically effective amount of the cationic peptides and salts, analogs and derivatives thereof, as described herein, can be administered according to any route of administration, without limitation, known in the art (e.g., parenteral, oral, topical, sublingual, buccal, enteral, nasal, inhalation, intranasal, injection, bladder wash-out, vagina, rectal, suppository, etc.). It is within the skill in the art to determine the appropriate route of administration for a given subject.
  • the cationic peptide or salt, analog or derivative thereof is administered in combination with an additional anti-cancer agent or therapy.
  • cationic peptides and salts, analogs and derivatives thereof, as described herein, may be administered systemically.
  • Systemic administration means administration to a subject by a method that causes the compounds to be absorbed into the bloodstream.
  • the cationic peptides, and salts, analogs and derivatives thereof, as described herein, can be administered orally by any method known in the art.
  • oral administration can be by tablets, capsules, pills, troches, elixirs, suspensions, syrups, wafers, chewing gum and the like.
  • carriers which are commonly used include lactose and corn starch, and lubricating agents such as magnesium stearate are commonly added.
  • useful carriers include lactose and corn starch.
  • carriers and excipients for oral administration include milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, calcium stearate, talc, vegetable fats or oils, gums and glycols.
  • emulsifying and/or suspending agents are commonly added.
  • sweetening and/or flavoring agents may be added to the oral compositions.
  • cationic peptides and salts, analogs and derivatives thereof, as described herein may be administered enterally or parenterally.
  • Parenteral administration may be by any means know in the art such as but not limited to, subcutaneous, intravenous, intramuscular, intraarterial, intraabdominal, intraperitoneal, intraarticular, intraocular or retrobulbar, intraaural, intrathecal, intracavitary, intracelial, intraspinal, intrapulmonary or transpulmonary, intrasynovial, or intraurethral injection or infusion.
  • sterile solutions of the cationic peptide can be employed, and the pH of the solutions can be suitably adjusted and buffered.
  • the total concentration of the solute(s) can be controlled in order to render the preparation isotonic
  • the administration is epicutaneous, by inhalation, intranasal, an enema, eye drops, ear drops, or through a mucous membrane.
  • the administration may be administered in a local manner.
  • the administration may be intratumoral by, for example, but not limited to, injection.
  • the cationic peptides and salts, analogs and derivatives thereof, as described herein are formulated for topical application to a target site on a subject in need thereof (e.g., creams, ointments, skin patches, eye drops, ear drops, shampoos).
  • a target site on a subject in need thereof e.g., creams, ointments, skin patches, eye drops, ear drops, shampoos.
  • compositions of the present invention may beformulated so as to allow the cationic peptide contained therein to be bioavailable upon administration of the composition to a subject.
  • the level of peptide in serum and other tissues after administration can be monitored by various well-established techniques, such as chromatographic or antibody based (e.g., ELISA) assays.
  • compositions may be administered to a subject as a single dosage unit (e.g., a tablet, capsule, or gel). Alternatively, the compositions may be administered as a plurality of dosage units (e.g., in aerosol form).
  • the cationic peptide formulations may be sterilized and packaged in single-use, plastic laminated pouches or plastic tubes of dimensions selected to provide for routine, measured dispensing.
  • cationic peptides disclosed herein may be administered intermittently.
  • a cationic peptide may be administered once daily, 2 times a day, 3 times a day, 4 times a day, every other day, twice a week, once a week, bi-weekly, once a month, etc.
  • the treatment can last as long as it is needed, such as 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, 50, 51, or 52 weeks, or more.
  • the additional anti-cancer agent can be administered according to any route of administration, without limitation, known in the art (e.g., parenteral, oral, topical, sublingual, buccal, enteral, nasal, inhalation, intranasal, injection, bladder wash-out, vagina, rectal, suppository, etc.). It is within the skill in the art to determine the appropriate route of administration for a given subject.
  • route of administration without limitation, known in the art (e.g., parenteral, oral, topical, sublingual, buccal, enteral, nasal, inhalation, intranasal, injection, bladder wash-out, vagina, rectal, suppository, etc.). It is within the skill in the art to determine the appropriate route of administration for a given subject.
  • route of administration without limitation, known in the art (e.g., parenteral, oral, topical, sublingual, buccal, enteral, nasal, inhalation, intranasal, injection, bladder wash-out, vagina, rectal,
  • the present invention provides methods for treating a non-virally-induced cancer by administering to a subject a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof, as described herein.
  • the cationic peptide or pharmaceutically acceptable salt thereof is preferably part of a pharmaceutical composition when used in the methods of the present invention.
  • the pharmaceutical composition will include at least one of a pharmaceutically acceptable vehicle, carrier, diluent, or excipient, in addition to one or more cationic peptide and, optionally, other components.
  • Pharmaceutically acceptable vehicles, carriers, diluents, or excipients for therapeutic use are well known in the pharmaceutical art, and are described herein and, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A.R. Gennaro, ed., 18 th Edition, 1990) and in CRC Handbook of Food, Agent, and Cosmetic Excipients, CRC Press LLC (S.C. Smolinski, ed., 1992).
  • the cationic peptide is provided in conjunction with a counter anion.
  • the counter anion may be any pharmaceutically acceptable counter anion.
  • the counter anion may include anionic groups such as carboxylates, phosphonates, sulphates and sulphonates.
  • the cationic peptide may be provided in the form of any pharmaceutically acceptable salt such as but not limited to trifluoroacetate, acetate, chloride and sulfate.
  • the cationic peptide is omiganan pentahydrochloride.
  • the cationic peptide composition may be provided in various forms, depending on the amount and number of different pharmaceutically acceptable vehicles, carriers, diluents, or excipients present.
  • the cationic peptide composition may be in the form of a solid, a semi-solid, a liquid, a lotion, a cream, an ointment, a cement, a paste, a gel, or an aerosol.
  • a pharmaceutically acceptable vehicle, carrier, diluent, or excipient typically includes the liquid or non-liquid basis of a composition comprising a cationic peptide of the invention.
  • suitable vehicles, carriers, or diluents include pyrogen-free water, isotonic saline or buffered aqueous solutions (e.g., phosphate, citrate, etc., buffered solutions).
  • isotonic saline or buffered aqueous solutions e.g., phosphate, citrate, etc., buffered solutions.
  • Sterile phosphate-buffered saline is one example of a pharmaceutically suitable excipient.
  • Other suitable excipients are well-known to those in the art.
  • the cationic peptide or pharmaceutically acceptable salt thereof can be formulated for intravenous administration, via, for example, bolus injection, slow infusion or continuous infusions.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • An injection buffer may be hypertonic, isotonic, or hypotonic with reference to a specific reference medium. Reference media include blood, lymph, cytosolic liquids, other bodily fluids, or common buffers or liquids known to a skilled person and used in “in vitro” methods.
  • Solvents useful in the present compositions are well known in the art and include without limitation water, glycerin, propylene glycol, mineral oil, isopropanol, ethanol, and methanol.
  • the solvent is glycerin, propylene glycol or mineral oil, at a concentration ranging from about 0.1% to about 20%, about 5% to about 15%, and about 9% to 11%.
  • the solvent is water or ethanol, preferably at a concentration up to about 99%, up to about 90%, and up to about 85%. Unless otherwise indicated, all percentages are on a w/w basis.
  • the solvent is at least one of water, glycerin, propylene glycol, mineral oil, isopropanol, ethanol, and methanol. In some embodiments, the solvent is glycerin or propylene glycol, mineral oil and ethanol. In other embodiments, the solvent is glycerin and ethanol. In yet other embodiments, the solvent is glycerin and water.
  • One embodiment is a composition
  • a composition comprising the cationic peptide, a viscosity-increasing agent, a solvent, wherein the solvent comprises at least one of water at a concentration up to 99%, glycerin at a concentration up to 20%, propylene glycol at a concentration up to 20%, ethanol at a concentration up to 99%, and methanol at a concentration up to 99%.
  • the cationic peptide compositions of the present invention include a viscosity-increasing agent, including without limitation carbomer homopolymer, dextran, polyvinylpyrrolidone, methylcellulose, carboxymethyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, and hydroxypropyl cellulose, and combinations thereof.
  • the viscosity-increasing agent is hydroxyethyl cellulose or hydroxypropyl methylcellulose, at a concentration ranging from about 0.5% to about 5%, from about 1% to about 3%, and from about 1.3% to about 1.7%.
  • the cationic peptide compositions have a first viscosity increasing agent, such as carbomer homopolymer, hydroxyethyl cellulose, hydroxypropyl methylcellulose, dextran, or polyvinylpyrrolidone, and a second viscosity-increasing agent such as carbomer homopolymer, hydroxyethyl cellulose, hydroxypropyl methyl cellulose, dextran, or polyvinylpyrrolidone.
  • a first viscosity increasing agent such as carbomer homopolymer, hydroxyethyl cellulose, hydroxypropyl methylcellulose, dextran, or polyvinylpyrrolidone
  • a second viscosity-increasing agent such as carbomer homopolymer, hydroxyethyl cellulose, hydroxypropyl methyl cellulose, dextran, or polyvinylpyrrolidone.
  • dextran and polyvinylpyrrolidone are preferably used at a concentration ranging from about 0.1% to about 5% and more preferably from about 0.5% to about 1%.
  • the first viscosity increasing agent is hydroxyethyl cellulose at a concentration up to 3% and the second viscosity-increasing agent is hydroxypropyl methylcellulose at a concentration up to 3%.
  • the amount of viscosity-increasing agent may be increased to shift the form of the composition from a liquid to a gel to a semi-solid form.
  • the amount of a viscosity-increasing agent used in a formulation may be varied depending on the intended use and location of administration of the peptide compositions provided herein.
  • a composition comprising the cationic peptide, a viscosity-increasing agent, and a solvent, may further comprise a buffering agent.
  • the buffering agent comprises a monocarboxylate or a dicarboxylate, and more specifically may be acetate, benzoate, fumarate, lactate, malonate, sorbate, succinate, or tartrate. In some embodiments, the buffering agent comprises benzoate. In some embodiments, the cationic peptide composition comprising the buffering agent has a pH ranging from about 3 to about 8, and more preferably from about 3.5 to 7. In some embodiment, the buffering agent is at a concentration ranging from about lmM to about 200mM, from about 2mM to about 20mM, and about 4mM to about 6mM.
  • cationic peptide composition may be stored at 2°C to 8°C.
  • the composition comprising a cationic peptide, a viscosity-increasing agent, and a solvent may further comprise a humectant, (e.g., sorbitol and the like), or a preservative, (e.g., benzoic acid, benzyl alcohol, phenoxyethanol, methylparaben, propylparaben, and the like).
  • a humectant e.g., sorbitol and the like
  • a preservative e.g., benzoic acid, benzyl alcohol, phenoxyethanol, methylparaben, propylparaben, and the like.
  • the cationic peptide may itself function as a preservative of the final therapeutic composition.
  • a preservative is optional in the gel formulations described herein because the gels may be sterilized by autoclaving and, furthermore, show the surprising quality of releasing (i.e., making bioavailable) the cationic peptide at a more optimal rate than other formulations, such as a cream.
  • particular embodiments may have in a single formulation a humectant, a preservative, and a buffering agent, or combinations thereof. Therefore, in some embodiments, the composition comprises a cationic peptide, a viscosity-increasing agent, a solvent, a humectant, and a buffering agent.
  • the composition comprises a cationic peptide, a viscosity-increasing agent, a buffering agent, and a solvent. In some embodiments, the composition comprises a cationic peptide, a buffering agent, and a solvent.
  • Each of the above formulations may be used to treat or prevent viral infection or to reduce the viral load in a subject or at a target site.
  • the cationic peptide formulation is in the form of a gel.
  • the pharmaceutically acceptable excipients suitable for use in the cationic peptide formulation compositions as described herein may include, for example, a viscosity-increasing agent, a buffering agent, a solvent, a humectant, a preservative, a chelating agent, an oleaginous compound, an emollient, an antioxidant, an adjuvant, and the like.
  • the function of each of these excipients is not mutually exclusive within the context of the present invention.
  • glycerin may be used as a solvent or as a humectant or as a viscosity-increasing agent.
  • the formulation is a composition comprising a cationic peptide, a viscosity-increasing agent, and a solvent.
  • the concentration of the cationic peptide used in the method of the invention is 0.01-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.01-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.01-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-100 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is 0.05-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.1-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.1-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.1-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.5-250 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is 0.5-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.5-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-25 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-10 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-5 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is 0.05-15 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-20 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-25 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-30 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is 0.05-40 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is 0.01, 0.05, 0.1, 0.5, 1, 10, 15, 20, 25, 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 400, or 500 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is about 0.01-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.01-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.01-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-100 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is about 0.05-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.1-250 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.1-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.1-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.5-250 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is about 0.5-100 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.5-50 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-25 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-10 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-5 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is about 0.05-15 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-20 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-25 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-30 ⁇ g/mL. In some embodiments, the concentration of the cationic peptide used in the method of the invention is about 0.05-40 ⁇ g/mL.
  • the concentration of the cationic peptide used in the method of the invention is about 0.01, 0.05, 0.1, 0.5, 1, 10, 15, 20, 25, 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 400, or 500 ⁇ g/mL.
  • kits for the treatment of a non-virally-induced cancer comprises a therapeutic composition containing an effective amount of a cationic peptide or pharmaceutically acceptable salt thereof.
  • the therapeutic composition in a unit dosage form.
  • the kits comprise one or more additional anti-cancer agent as described herein.
  • the kit comprises a sterile container which contains the cationic peptide or salt thereof; such containers can be sachets, packets, boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art.
  • Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
  • the cationic peptide can be provided together with instructions for administering the composition to a subject having a non-virally-induced cancer.
  • the instructions will generally include information about the use of the composition for the treatment of cancer.
  • the instructions include at least one of the following: description of the therapeutic agent; dosage schedule and administration for treatment of cancer; precautions; warnings; indications; counter-indications; overdose information; adverse reactions; animal pharmacology; clinical studies; and/or references.
  • the instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
  • CT26 is a carcinogen-induced murine colon carcinoma cell line derived from BALB/c mice and TC-1 is an HPV16 E6/E7-expressing, Ras transformed cell line derived from primary lung epithelial cells of C57BL/6 mice.
  • mice are inoculated on day 0 with 100,000 TC-1 or CT26 tumor cells via subcutaneous injection resulting in established solid tumors on day 10.
  • the TC-1 mice are treated with three intratumoral injections of 30 nmole omiganan in 30 ⁇ L PBS, or empty PBS as a negative control on days 10, 12, and 14.
  • the CT26 mice are treated with six intratumoral injections 30 nmole omiganan in 30 ⁇ L PBS, or empty PBS as a negative control on days 10, 12, 15, 18, 20, and 22.
  • Tumor size is measured by caliper on days 10, 12, 14, and 16 in the TC-1 mice and on days 10, 12, 15, 18, and 20 in the CT26 mice.
  • a reduction in tumor outgrowth is observed for the omiganan-treated groups compared to PBS-treated mice (Figs. 1A and 1B).
  • Omiganan therefore inhibits growth of established subcutaneous mouse tumors in both models.
  • Fig. 2A depicts the presence of neutrophils in the TC-1 and CT26 tumors as a percentage of all immune cells present in the tumor, as analyzed by fluorescence-based flow cytometry (FACS).
  • Omiganan treatment increases the number of neutrophils in TC-1 tumors (Fig. 2A, top panel), but not in CT26 tumors (Fig. 2A, bottom panel).
  • the increase in neutrophils in the TC-1 tumors as observed by FACS is confirmed with a detailed analysis using metal-based mass cytometry (CyTOF) (Fig. 2B).
  • the heatmap shows clusters of cells representing different cell types as columns, and each row indicates the presence of a cell surface marker on that particular cluster on a color scale. Ly6G+ clusters (neutrophils) are indicated by an arrow.
  • the bar graph depicted in Fig. 2C confirms that these cells increase after omiganan treatment (red versus blue bars in clusters 1, 10, and 11).
  • Fig. 2D describes the cell type and percentage in each cluster as well as the presence or absence of specific markers.
  • omiganan induces different cell-based effects in virally- and non-virally-induced tumors.
  • mice are inoculated with a tumor (TC-1 or CT26) on day 0 followed by treatment with repeated intratumoral injections of omiganan or an equal volume of PBS (two groups) with or without the anti-neutrophil antibody aLy6G.
  • TC-1 mice omiganan or PBS is administered on days 9, 11, 13, 15 and 17.
  • One of the omiganan treatment groups and one of the PBS control groups are further treated with repeated injections aLy6G on days 9, 13, and 16.
  • Tumor growth is measured on days 9, 11, 13, 15, and 17.
  • CT26 mice omiganan or PBS is administered on days 8, 10, 12, 14, and 16.
  • One of the omiganan treatment groups and one of the PBS control groups are further treated with repeated injections aLy6G on days 8, 10, 12, 14, and 16. Tumor growth is also measured on these days.
  • Figs. 3A and 3B Tumor growth curves of the subcutaneous mouse tumors with or without omiganan treatment, combined with the aLy6G antibody that removes all neutrophils from the body are shown in in Figs. 3A and 3B.
  • omiganan efficacy is reduced in the absence of neutrophils, while removal of neutrophils from untreated tumors has no effect (Fig. 3A).
  • CT26 tumor growth is inhibited in the absence of neutrophils, and is further reduced upon omiganan treatment (Fig. 3B).
  • neutrophil depletion has an additive effect to omiganan treatment. This indicates opposite roles for neutrophils during omiganan treatment in the TC-1 and CT26 tumor models.
  • Omiganan-treated CT26 tumors are analyzed ex vivo by flow cytometry for the percentages of several immune cell subsets, and for cell surface markers on neutrophils that could potentially indicate functional changes in neutrophils.
  • T cells (divided into CD4+ and CD8+ T cells) are increased after omiganan treatment (Fig. 4A). Further, the total number of myeloid immune cells (Fig. 4B, top panel) is not changed, but omiganan causes a shift within myeloid cells from Ly6C-low to Ly6C-high cells (Fig. 4B, bottom panel). This suggests that omiganan induces a lymphocyte and monocyte-based anti-tumor inflammatory response in non-virally-induced cancers. Without wishing to be bound by theory, omiganan may be attracting T cells and monocytes to the tumor environment.
  • Fig. 4C shows an example of tumor neutrophils analyzed by flow cytometry, where the axes indicate the expression level of the markers PD-L1 and CD54. Each dot represents a single neutrophil. The population inside the black box indicates that these two markers are mostly co-expressed on the same cells.
  • CD54 is associated with effector functions of neutrophils, while PD-L1 is an inhibitory molecule. Without wishing to be bound by theory, this suggests that omiganan may exert synergistic effects in combination with an inhibitor of the PD-1 pathway.
  • a method of treating a non-virally-induced cancer in a subject comprising administering to the subject a therapeutically effective amount of a cationic peptide or pharmaceutically acceptable salt thereof. 2.
  • the cationic peptide is selected from the group consisting of: omiganan, LL-37, 11B32CN, 11B36CN, 11E3CN, 11F4CN, 11F5CN, 11F12CN, 11F17CN, 11F50CN, 11F56CN, 11F63CN, 11F64CN, 11F66CN, 11F67CN, 11F68CN, 11F93CN, 11G27CN, 11J02CN, 11J02ACN, 11J30CN, 11J36CN, 11J58CN, 11J67CN, 11J68CN, Nt-acryloyl-11B7CN, Nt-glucosyl-11J36CN, Nt-glucosyl-11J38CN, and pharmaceutically acceptable salts thereof.
  • the non-virally induced cancer is selected from the group consisting of gastric cancer, sarcoma, lymphoma, leukemia, head and neck cancer, thymic cancer, epithelial cancer, salivary cancer, liver cancer, stomach cancer, thyroid cancer, lung cancer, ovarian cancer, breast cancer, prostate cancer, esophageal cancer, pancreatic cancer, glioma, leukemia, multiple myeloma, renal cell carcinoma, bladder cancer, choriocarcinoma, colon cancer, colorectal cancer, oral cancer, testicular cancer, bone cancer, skin cancer, and melanoma.
  • parenteral administration is subcutaneous, intravenous, intramuscular, intraarterial, intraabdominal, intraperitoneal, intraarticular, intraocular or retrobulbar, intraaural, intrathecal, intracavitary, intracelial, intraspinal, intrapulmonary or transpulmonary, intrasynovial, or intraurethral injection or infusion.
  • the cationic peptide is selected from the group consisting of: omiganan, LL-37, 11B32CN, 11B36CN, 11E3CN, 11F4CN, 11F5CN, 11F12CN, 11F17CN, 11F50CN, 11F56CN, 11F63CN, 11F64CN, 11F66CN, 11F67CN, 11F68CN, 11F93CN, 11G27CN, 11J02CN, 11J02ACN, 11J30CN, 11J36CN, 11J58CN, 11J67CN, 11J68CN, Nt-acryloyl-11B7CN, Nt-glucosyl-11J36CN, Nt-glucosyl-11J38CN, and pharmaceutically acceptable salts thereof.
  • the non-virally induced cancer is selected from the group consisting of gastric cancer, sarcoma, lymphoma, leukemia, head and neck cancer, thymic cancer, epithelial cancer, salivary cancer, liver cancer, stomach cancer, thyroid cancer, lung cancer, ovarian cancer, breast cancer, prostate cancer, esophageal cancer, pancreatic cancer, glioma, leukemia, multiple myeloma, renal cell carcinoma, bladder cancer, choriocarcinoma, colon cancer, colorectal cancer, oral cancer, testicular cancer, bone cancer, skin cancer, and melanoma.
  • the medicament further comprises one or more additional anti-cancer agents or therapies.
  • the additional anti-cancer agent is a checkpoint inhibitor.
  • the checkpoint inhibitor is a PD-1 inhibitor.
  • the PD-1 inhibitor is an anti-PD-1 antibody.
  • the checkpoint inhibitor is a PD-L1 inhibitor. 22.
  • the use according to paragraph 21, wherein the PD-L1 inhibitor is an anti-PD-L1 antibody.
  • the medicament is for topical or parenteral administration. 24.
  • parenteral administration is subcutaneous, intravenous, intramuscular, intraarterial, intraabdominal, intraperitoneal, intraarticular, intraocular or retrobulbar, intraaural, intrathecal, intracavitary, intracelial, intraspinal, intrapulmonary or transpulmonary, intrasynovial, or intraurethral injection or infusion.
  • parenteral administration is subcutaneous, intravenous, intramuscular, intraarterial, intraabdominal, intraperitoneal, intraarticular, intraocular or retrobulbar, intraaural, intrathecal, intracavitary, intracelial, intraspinal, intrapulmonary or transpulmonary, intrasynovial, or intraurethral injection or infusion.
  • a cationic peptide or pharmaceutically acceptable salt thereof for use in treating a non-virally-induced cancer in a subject.
  • cationic peptide for use according to paragraph 25, wherein the cationic peptide is selected from the group consisting of: omiganan, LL-37, 11B32CN, 11B36CN, 11E3CN, 11F4CN, 11F5CN, 11F12CN, 11F17CN, 11F50CN, 11F56CN, 11F63CN, 11F64CN, 11F66CN, 11F67CN, 11F68CN, 11F93CN, 11G27CN, 11J02CN, 11J02ACN, 11J30CN, 11J36CN, 11J58CN, 11J67CN, 11J68CN, Nt-acryloyl-11B7CN, Nt-glucosyl-11J36CN, Nt-glucosyl-11J38CN, and pharmaceutically acceptable salts thereof.
  • the non-virally induced cancer is selected from the group consisting of gastric cancer, sarcoma, lymphoma, leukemia, head and neck cancer, thymic cancer, epithelial cancer, salivary cancer, liver cancer, stomach cancer, thyroid cancer, lung cancer, ovarian cancer, breast cancer, prostate cancer, esophageal cancer, pancreatic cancer, glioma, leukemia, multiple myeloma, renal cell carcinoma, bladder cancer, choriocarcinoma, colon cancer, colorectal cancer, oral cancer, testicular cancer, bone cancer, skin cancer, and melanoma.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Zoology (AREA)
  • Biochemistry (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)

Abstract

La présente invention concerne un procédé de traitement de cancer non induits par voie virale chez un sujet.
PCT/JP2020/004711 2019-02-08 2020-02-07 Méthodes de traitement de cancers non induits par voie virale WO2020162582A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962803094P 2019-02-08 2019-02-08
US62/803,094 2019-02-08

Publications (1)

Publication Number Publication Date
WO2020162582A1 true WO2020162582A1 (fr) 2020-08-13

Family

ID=71948320

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2020/004711 WO2020162582A1 (fr) 2019-02-08 2020-02-07 Méthodes de traitement de cancers non induits par voie virale

Country Status (1)

Country Link
WO (1) WO2020162582A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999065506A2 (fr) * 1998-06-12 1999-12-23 Micrologix Biotech Inc. Traitement du cancer avec des peptides cationiques
US20030171281A1 (en) * 2001-08-21 2003-09-11 Micrologix Biotech Inc. Antimicrobial cationic peptides and formulations thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999065506A2 (fr) * 1998-06-12 1999-12-23 Micrologix Biotech Inc. Traitement du cancer avec des peptides cationiques
US20030171281A1 (en) * 2001-08-21 2003-09-11 Micrologix Biotech Inc. Antimicrobial cationic peptides and formulations thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHAE Y.K. ET AL.: "Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC)", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 6, no. 1, 2018, pages 1 - 27, XP021256513, DOI: 10.1186/s40425-018-0349-3 *
HARADA K. ET AL.: "Recent advances in the management of gastric adenocarcinoma patients", F1000RESEARCH, vol. 7, 1365, 2018, pages 1 - 12, XP055723484, DOI: 10.12688/f1000research.15133.1 *
MADER J.S. ET AL.: "The human host defense peptide LL-37 induces apoptosis in a calpain- and apoptosis-inducing factor-dependent manner involving Bax activity", MOLECULAR CANCER RESEARCH, vol. 7, no. 5, 2009, pages 689 - 702, XP055723484, DOI: 10.1158/1541-7786.MCR-08-0274 *
WU W.K.K. ET AL.: "The host defense peptide LL-37 activates the tumor-suppressing bone morphogenetic protein signaling via inhibition of proteasome in gastric cancer cells", JOURNAL OF CELLULAR PHYSIOLOGY, vol. 223, no. 1, 2010, pages 178 - 186, XP055723482 *

Similar Documents

Publication Publication Date Title
JP6714751B2 (ja) 単鎖trail受容体アゴニストタンパク質
KR102110127B1 (ko) 세포 내재화를 유도하기 위한 cd20-결합 면역독소 및 이의 사용 방법
EP3082857B1 (fr) Thérapie de combinaison avec un anticorps anti-ang2 et un agoniste de cd40
AU2018206015A1 (en) Anti-human 4-1 BB antibodies and use thereof
KR102645411B1 (ko) Her2, nkg2d 및 cd16에 결합하는 다중-특이적 결합 단백질 및 사용 방법
KR20170035945A (ko) 인간 cd40을 활성화시키는 항체 및 인간 pd-l1에 대한 항체의 병용 요법
JP2018515072A (ja) 単鎖cd40受容体アゴニストタンパク質
KR20150090919A (ko) 결합제를 사용한 면역요법
RU2725954C1 (ru) Пептид, обладающий высокой аффинностью к белку pd-l1, и его применение
EP3534965A1 (fr) Améliorations de la thérapie de blocage de cd47 par des inhibiteurs de hdac
EP3504239B1 (fr) Dosage intermittent d'un anticorps anti-csf-1r en combinaison avec un agent d'activation de macrophages
CA2849746A1 (fr) Anticorps anti-icam-1 destines au traitement de troubles lies a un myelome multiple
JP2023184773A (ja) 癌治療のためのtrpv6阻害剤および併用療法
US20120087916A1 (en) Anti-icam-1 antibody, uses and methods
EP2654769B1 (fr) Peptide antitumoral dérivé d'une région de détermination de la complémentarité d'un anticorps monoclonal humanisé anti-transporteur napi2b
CA3002741A1 (fr) Proteines agonistes du recepteur gitr a chaine unique
EP2222692B1 (fr) Peptide synthétique de fixation de cd154 chez l'homme et utilisations associées
WO2020162582A1 (fr) Méthodes de traitement de cancers non induits par voie virale
EP3333184B1 (fr) Compositions et procédés pour inhiber des interactions cd279
CA3002600A1 (fr) Proteines agonistes du recepteur ox40 a chaine unique
CA3218832A1 (fr) Polytherapie utilisant un anticorps anti-cd300c
WO2023089083A1 (fr) Fractions de liaison multispécifique comprenant des domaines de liaison de pd-1 et de tgf-brii
CN117192118A (zh) 一种抗cldn 18.2和cd47的双特异性抗体治疗疾病的用途
CN112512547A (zh) 嗜中性粒细胞吞噬能力增强剂

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: 20752386

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20752386

Country of ref document: EP

Kind code of ref document: A1