US20130266666A1 - Combination Therapy with an Antitumor Alkaloid - Google Patents

Combination Therapy with an Antitumor Alkaloid Download PDF

Info

Publication number
US20130266666A1
US20130266666A1 US13/884,874 US201113884874A US2013266666A1 US 20130266666 A1 US20130266666 A1 US 20130266666A1 US 201113884874 A US201113884874 A US 201113884874A US 2013266666 A1 US2013266666 A1 US 2013266666A1
Authority
US
United States
Prior art keywords
combination
cancer
inhibitors
pharmaceutically acceptable
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/884,874
Other languages
English (en)
Inventor
Victoria Moneo Ocaña
Gema Santamaría Núñez
Luis Francisco García Fernández
Carlos Maria Galmarini
María José Guillén Navarro
Pablo Manuel Avilés Marín
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pharmamar SA
Original Assignee
Pharmamar SA
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 Pharmamar SA filed Critical Pharmamar SA
Assigned to PHARMA MAR, S.A. reassignment PHARMA MAR, S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AVILES MARIN, PABLO MANUEL, GALMARINI, CARLOS MARIA, GARCIA FERNANDEZ, LUIS FRANCISCO, GUILLEN NAVARRO, MARIA JOSE, MONEO OCANA, VICTORIA, SANTAMARIA NUNEZ, GEMA
Publication of US20130266666A1 publication Critical patent/US20130266666A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds 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
    • A61K31/7064Compounds 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 containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds 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 containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/15Depsipeptides; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the combination of PM01183 with other anticancer drugs, in particular other anticancer drugs selected from antitumor platinum coordination complexes, antimetabolites, mitotic inhibitors, anticancer antibiotics, topoisomerase I and/or II inhibitors, proteasome inhibitors, histone deacetylase inhibitors, nitrogen mustard alkylating agents, nitrosourea alkylating agents, nonclassical alkylating agents, estrogen antagonists, androgen antagonists, mTOR inhibitors, tyrosine kinase inhibitors, and other agents selected from aplidine, ET-743, PM02734, and PM00104 and the use of these combinations in the treatment of cancer.
  • other anticancer drugs selected from antitumor platinum coordination complexes, antimetabolites, mitotic inhibitors, anticancer antibiotics, topoisomerase I and/or II inhibitors, proteasome inhibitors, histone deacetylase inhibitors, nitrogen mustard alkylating agents, nitrosourea alky
  • Cancer develops when cells in a part of the body begin to grow out of control. Although there are many kinds of cancer, they all arise from out-of-control growth of abnormal cells. Cancer cells can invade nearby tissues and can spread through the bloodstream and lymphatic system to other parts of the body. There are several main types of cancer. Carcinoma is a malignant neoplasm, which is an uncontrolled and progressive abnormal growth, arising from epithelial cells. Epithelial cells cover internal and external surfaces of the body, including organs, lining of vessels, and other small cavities. Sarcoma is cancer arising from cells in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue. Leukemia is cancer that arises in blood-forming tissue such as the bone marrow, and causes large numbers of abnormal blood cells to be produced and enter the bloodstream. Lymphoma and multiple myeloma are cancers that arise from cells of the immune system.
  • cancer is invasive and tends to infiltrate the surrounding tissues and give rise to metastases. It can spread directly into surrounding tissues and also may be spread through the lymphatic and circulatory systems to other parts of the body.
  • Chemotherapy in its classic form, has been focused primarily on killing rapidly proliferating cancer cells by targeting general cellular metabolic processes, including DNA, RNA, and protein biosynthesis. Chemotherapy drugs are divided into several groups based on how they affect specific chemical substances within cancer cells, which cellular activities or processes the drug interferes with, and which specific phases of the cell cycle the drug affects.
  • DNA-alkylating drugs such as cyclophosphamide, ifosfamide, cisplatin, carboplatin, dacarbazine
  • antimetabolites (5-fluorouracil, capecitabine, 6-mercaptopurine, methotrexate, gemcitabine, cytarabine, fludarabine
  • mitotic inhibitors such as paclitaxel, docetaxel, vinblastine, vincristine
  • anticancer antibiotics such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone
  • topoisomerase I and/or II inhibitors such as topotecan, irinotecan, etoposide, teniposide
  • hormone therapy such as tamoxifen, flutamide
  • the ideal antitumor drug would kill cancer cells selectively, with a wide index relative to its toxicity towards non-cancer cells and it would also retain its efficacy against cancer cells, even after prolonged exposure to the drug.
  • none of the current chemotherapies with these agents posses an ideal profile. Most posses very narrow therapeutic indexes and, in addition, cancerous cells exposed to slightly sublethal concentrations of a chemotherapeutic agent may develop resistance to such an agent, and quite often cross-resistance to several other antitumor agents.
  • PM01183 also known as tryptamicidin, is a synthetic alkaloid which is currently in clinical trials for the treatment of cancer, and has the following chemical structure:
  • PM01183 has demonstrated a highly potent in vitro activity against solid and non-solid tumour cell lines as well as a significant in vivo activity in several xenografted human tumor cell lines in mice, such as those for breast, kidney and ovarian cancer.
  • PM01183 exerts its anticancer effects through the covalent modification of guanines in the DNA minor groove that eventually give rise to DNA double-strand break, S-phase arrest and apoptosis in cancer cells. Further information regarding this compound can be found in WO 03/01427; 100 th AACR Annual Meeting, Apr. 18-22, 2009, Denver, Colo., Abstract Nr. 2679 and Abstract Nr. 4525; and Leal J F M et al. Br. J. Pharmacol. 2010, 161, 1099-1110.
  • the problem to be solved by the present invention is to provide anticancer therapies that are useful in the treatment of cancer.
  • the present invention establishes that PM01183 potentiates the antitumor activity of other anticancer agents, in particular other anticancer drugs selected from antitumor platinum coordination complexes, antimetabolites, mitotic inhibitors, anticancer antibiotics, topoisomerase I and/or II inhibitors, proteasome inhibitors, histone deacetylase inhibitors, nitrogen mustard alkylating agents, nitrosourea alkylating agents, nonclassical alkylating agents, estrogen antagonists, androgen antagonists, mTOR inhibitors, tyrosine kinase inhibitors, and other agents selected from aplidine, ET-743, PM02734 and PM00104. Therefore PM01183 and said other anticancer agents can be successfully used in combination therapy for the treatment of cancer.
  • other anticancer drugs selected from antitumor platinum coordination complexes, antimetabolites, mitotic inhibitors, anticancer antibiotics, topoisomerase I and/or II inhibitors, proteasome inhibitors, histone deacety
  • this invention is directed to pharmaceutical compositions, kits, methods for the treatment of cancer using these combination therapies and uses of both drugs in the treatment of cancer and in the manufacture of medicaments for combination therapies.
  • the invention is directed to PM01183, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer comprising administering a therapeutically effective amount of PM01183, or a pharmaceutical acceptable salt thereof, in combination with a therapeutically effective amount of another anticancer drug.
  • the invention encompasses a method of treating cancer comprising administering to a patient in need of such treatment a therapeutically effective amount of PM01183, or a pharmaceutically acceptable salt thereof, and a therapeutically effective amount of another anticancer drug.
  • the invention encompasses a method of increasing or potentiating the therapeutic efficacy of an anticancer drug in the treatment of cancer, which comprises administering to a patient in need thereof a therapeutically effective amount of PM01183, or a pharmaceutically acceptable salt thereof, in conjunction with this other anticancer drug.
  • the invention encompasses the use of PM01183, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer by combination therapy employing PM01183, or a pharmaceutically acceptable salt thereof, with another anticancer drug.
  • the invention encompasses a pharmaceutical composition comprising PM01183, or a pharmaceutically acceptable salt thereof, and/or another anticancer drug, and a pharmaceutically acceptable carrier, to be used in combination therapy for the treatment of cancer.
  • the invention also encompasses a kit for use in the treatment of cancer which comprises a dosage form of PM01183, or a pharmaceutically acceptable salt thereof, and/or a dosage form of another anticancer drug, and instructions for the use of both drugs in combination.
  • the present invention is concerned with synergistic combinations of PM01183, or a pharmaceutically acceptable salt thereof, with another anticancer drug.
  • FIG. 1-20 In vitro activity data of PM01183 in combination with oxaliplatin, 5-fluorouracil, gemcitabine, paclitaxel, docetaxel, vincristine, daunorubicin, mitomycin C, actinomycin D, topotecan, etoposide, bortezomib, vorinostat, cyclophosphamide, carmustine, dacarbazine, temsirolimus, erlotinib, ET-743 and PM00104 respectively against A549 cells.
  • FIG. 21-41 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, cytarabine, gemcitabine, docetaxel, vincristine, vinorelbine, daunorubicin, mitomycin C, actinomycin D, topotecan, etoposide, vorinostat, cyclophosphamide, dacarbazine, temsirolimus, erlotinib, aplidine, ET-743, PM02734 and PM00104 respectively against A673 cells.
  • FIG. 42-56 In vitro activity data of PM01183 in combination with cisplatin, 5-fluorouracil, cytarabine, methotrexate, daunorubicin, doxorubicin, mitomycin C, topotecan, irinotecan, etoposide, dacarbazine, temsirolimus, ET-743, PM02734 and PM00104 respectively against SK-MEL-2 cells.
  • FIG. 57-80 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, docetaxel, paclitaxel, vinorelbine, daunorubicin, doxorubicin, mitomycin C, actinomycin D, topotecan, irinotecan, etoposide, bortezomib, vorinostat, flutamide, temsirolimus, erlotinib, ET-743, PM02734 and PM00104 respectively against PC-3 cells.
  • FIG. 81-98 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, cytarabine, gemcitabine, methotrexate, daunorubicin, doxorubicin, actinomycin D, topotecan, irinotecan, etoposide, bortezomib, vorinostat, temsirolimus, erlotinib, ET-743, PM02734 and PM00104 respectively against PANC-1 cells.
  • FIG. 99-123 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, paclitaxel, vincristine, vinorelbine, daunorubicin, doxorubicin, actinomycin D, topotecan, irinotecan, etoposide, bortezomib, vorinostat, cyclophosphamide, dacarbazine, temsirolimus, erlotinib, aplidine, ET-743, PM02734 and PM00104 respectively against HGC-27 cells.
  • FIG. 124-150 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, docetaxel, paclitaxel, vincristine, vinorelbine, daunorubicin, doxorubicin, actinomycin D, mitomycin C, topotecan, irinotecan, etoposide, vorinostat, cyclophosphamide, carmustine, dacarbazine, temsirolimus, erlotinib, aplidine, ET-743, PM02734 and PM00104 respectively against IGROV-1 cells.
  • FIG. 151-170 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, docetaxel, paclitaxel, vincristine, vinorelbine, daunorubicin, doxorubicin, topotecan, irinotecan, etoposide, bortezomib, cyclophosphamide, erlotinib, ET-743 and PM00104 respectively against HEP-G2 cells.
  • FIG. 171-197 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, docetaxel, paclitaxel, vincristine, vinorelbine, daunorubicin, doxorubicin, actinomycin D, mitomycin C, topotecan, irinotecan, etoposide, vorinostat, cyclophosphamide, carmustine, dacarbazine, tamoxifen, temsirolimus, erlotinib, ET-743, PM02734 and PM00104 respectively against MDA-MB-231 cells.
  • FIG. 198-219 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, cytarabine, gemcitabine, docetaxel, vinorelbine, daunorubicin, doxorubicin, actinomycin D, mitomycin C, topotecan, irinotecan, etoposide, bortezomib, vorinostat, cyclophosphamide, dacarbazine, temsirolimus, erlotinib, aplidine and PM02734 respectively against HT-29 cells.
  • FIG. 220-242 In vitro activity data of PM01183 in combination with cisplatin, 5-fluorouracil, cytarabine, gemcitabine, methotrexate, docetaxel, vincristine, vinorelbine, daunorubicin, doxorubicin, actinomycin D, mitomycin C, topotecan, irinotecan, etoposide, vorinostat, cyclophosphamide, dacarbazine, erlotinib, aplidine, ET-743, PM02734 and PM00104 respectively against RXF-393 cells.
  • FIG. 243-262 In vitro activity data of PM01183 in combination with cisplatin, oxaliplatin, 5-fluorouracil, gemcitabine, methotrexate, docetaxel, vincristine, daunorubicin, doxorubicin, topotecan, irinotecan, etoposide, bortezomib, vorinostat, dacarbazine, temsirolimus, erlotinib, aplidine, ET-743 and PM02734 respectively against U87-MG cells.
  • FIG. 263 Tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, paclitaxel and PM01183 plus paclitaxel.
  • FIG. 264 Tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, vinorelbine and PM01183 plus vinorelbine.
  • FIG. 265 Tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, doxorubicin and PM01183 plus doxorubicin.
  • FIG. 266 Tumor volume evaluation of HGC-27 tumors in mice treated with placebo, PM01183, cisplatin and PM01183 plus cisplatin.
  • FIG. 267 Tumor volume evaluation of HGC-27 tumors in mice treated with placebo, PM01183, 5-fluorouracil and PM01183 plus 5-fluorouracil.
  • FIG. 268 Tumor volume evaluation of SW1990 tumors in mice treated with placebo, PM01183, gemcitabine and PM01183 plus gemcitabine.
  • FIG. 269 Tumor volume evaluation of U87-MG tumors in mice treated with placebo, PM01183, temozolomide and PM01183 plus temozolomide.
  • FIG. 270 Tumor volume evaluation of H460 tumors in mice treated with placebo, PM01183, irinotecan and PM01183 plus irinotecan.
  • FIG. 271 Tumor volume evaluation of HT1080 tumors in mice treated with placebo, PM01183, dacarbazine and PM01183 plus dacarbazine.
  • FIG. 272 Tumor volume evaluation of HT-29 tumors in mice treated with placebo, PM01183, irinotecan and PM01183 plus irinotecan.
  • FIG. 273 Effects of the combination of PM01183 with methotrexate in JURKAT cell line.
  • FIG. 274 Effects of the combination of PM01183 with methotrexate in MOLT-4 cell line.
  • FIG. 275 Effects of the combination of PM01183 with daunorubicin in JURKAT cell line.
  • FIG. 276 Effects of the combination of PM01183 with aplidine in JURKAT cell line.
  • FIG. 277 Effects of the combination of PM01183 with aplidine in MOLT-4 cell line.
  • FIG. 278 Effects of the combination of PM01183 with ET-743 in JURKAT cell line.
  • FIG. 279 Effects of the combination of PM01183 with ET-743 in MOLT-4 cell line.
  • FIG. 280 Effects of the combination of PM01183 with PM00104 in JURKAT cell line.
  • FIG. 281 Effects of the combination of PM01183 with PM00104 in MOLT-4 cell line.
  • FIG. 282 Effects of the combination of PM01183 with PM02734 in JURKAT cell line.
  • FIG. 283 Effects of the combination of PM01183 with PM02734 in MOLT-4 cell line.
  • FIG. 284 Effects of the combination of PM01183 with cytarabine in RAMOS cell line.
  • FIG. 285 Effects of the combination of PM01183 with methotrexate in RAMOS cell line.
  • FIG. 286 Effects of the combination of PM01183 with methotrexate in U-937 cell line.
  • FIG. 287 Effects of the combination of PM01183 with gemcitabine in RAMOS cell line.
  • FIG. 288 Effects of the combination of PM01183 with gemcitabine in U-937 cell line.
  • FIG. 289 Effects of the combination of PM01183 with daunorubicin in RAMOS cell line.
  • FIG. 290 Effects of the combination of PM01183 with daunorubicin in U-937 cell line.
  • FIG. 291 Effects of the combination of PM01183 with ET-743 in RAMOS cell line.
  • FIG. 292 Effects of the combination of PM01183 with ET-743 in U-937 cell line.
  • FIG. 293 Effects of the combination of PM01183 with PM00104 in RAMOS cell line.
  • FIG. 294 Effects of the combination of PM01183 with PM00104 in U-937 cell line.
  • FIG. 295 Effects of the combination of PM01183 with PM02734 in RAMOS cell line.
  • FIG. 296 Effects of the combination of PM01183 with PM02734 in U-937 cell line.
  • the present invention is directed to provide an efficacious treatment of cancer based on the combination of PM01183, or a pharmaceutically acceptable salt thereof, with another anticancer drug.
  • cancer it is meant to include tumors, neoplasias, and any other malignant disease having as cause malignant tissue or cells.
  • treating means reversing, alleviating, or inhibiting the progress of the disease or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of treating as “treating” is defined immediately above.
  • PM01183 is a synthetic alkaloid, having the following structure:
  • PM01183 is intended here to cover any pharmaceutically acceptable salt, solvate, hydrate, prodrug, or any other compound which, upon administration to the patient is capable of providing (directly or indirectly) the compound as described herein.
  • the preparation of salts, solvates, hydrates, and prodrugs can be carried out by methods known in the art.
  • salts can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods.
  • such salts are, for example, prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent or in a mixture of the two.
  • nonaqueous media like ether, ethyl acetate, ethanol, isopropanol or acetonitrile are preferred.
  • acid addition salts include mineral acid addition salts such as, for example, hydrochloride, hydrobromide, hydroiodide, sulphate, nitrate, phosphate, and organic acid addition salts such as, for example, acetate, trifluoroacetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulphonate and p-toluenesulphonate.
  • mineral acid addition salts such as, for example, hydrochloride, hydrobromide, hydroiodide, sulphate, nitrate, phosphate
  • organic acid addition salts such as, for example, acetate, trifluoroacetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulphonate and p-toluenesulphonate.
  • alkali addition salts include inorganic salts such as, for example, sodium, potassium, calcium and ammonium salts, and organic alkali salts such as, for example, ethylenediamine, ethanolamine, N,N-dialkylenethanolamine, triethanolamine and basic aminoacids salts.
  • prodrug is used in its broadest sense and encompasses those derivatives that are converted in vivo to PM01183.
  • the prodrug can hydrolyze, oxidize, or otherwise react under biological conditions to provide PM01183.
  • prodrugs include, but are not limited to, derivatives and metabolites of PM01183 that include biohydrolyzable moeities such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • Prodrugs can typically be prepared using well-known methods, such as those described by Burger in “Medicinal Chemistry and Drug Discovery” 6th ed. (Donald J. Abraham ed., 2001, Wiley) and “Design and Applications of Prodrugs” (H. Bundgaard ed., 1985, Harwood Academic Publishers).
  • any drug referred to herein may be in amorphous form or crystalline form either as free compound or as solvates (e.g. hydrates) and it is intended that both forms are within the scope of the present invention.
  • Methods of solvation are generally known within the art.
  • PM01183 for use in accordance with the present invention may be prepared following the synthetic process such as the one disclosed in WO 03/014127, which is incorporated herein by reference.
  • compositions of PM01183, or of a pharmaceutically acceptable salt thereof, that can be used include solutions, suspensions, emulsions, lyophilised compositions, etc., with suitable excipients for intravenous administration.
  • PM01183 may be supplied and stored as a sterile lyophilized product, comprising PM01183 and excipients in a formulation adequate for therapeutic use.
  • pharmaceutical compositions of PM01183, or a pharmaceutically acceptable salt thereof see for example the formulations described in WO 2006/046079, which is incorporated herein by reference.
  • PM01183, or a pharmaceutically acceptable salt thereof, or pharmaceutical compositions comprising the compound is preferably by intravenous infusion.
  • Infusion times of up to 72 hours can be used, more preferably between 1 and 24 hours, with either about 1 hour or about 3 hours most preferred. Short infusion times which allow treatment to be carried out without an overnight stay in hospital are especially desirable. However, infusion may be around 24 hours or even longer if required.
  • the administration of PM01183 is performed in cycles.
  • an intravenous infusion of PM01183 is given to the patients the first week of each cycle and the patients are allowed to recover for the remainder of the cycle.
  • the preferred duration of each cycle is of either 3 or 4 weeks. Multiple cycles can be given as needed.
  • Administration of PM01183, or a pharmaceutically acceptable salt thereof, by intravenous infusion during about 1 hour once every 3 weeks is the most preferred administration schedule, although other protocols can be devised as variations.
  • another anticancer drug selected from antitumor platinum coordination complexes, antimetabolites, mitotic inhibitors, anticancer antibiotics, topoisomerase I and/or II inhibitors, proteasome inhibitors, histone deacetylase inhibitors, nitrogen mustard alkylating agents, nitrosourea alkylating agents, nonclassical alkylating
  • cancer types are those selected from lung cancer, sarcoma, malignant melanoma, bladder carcinoma, prostate cancer, pancreas carcinoma, thyroid cancer, gastric carcinoma, ovarian cancer, hepatoma (also known as liver cancer), breast cancer, colorectal cancer, kidney cancer, esophageal cancer, neuroblastoma, brain cancer, cervical cancer, anal cancer, testicular cancer, leukemia, multiple myeloma and lymphoma.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with an antitumor platinum coordination complex in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, brain cancer and lymphoma.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, brain cancer and lymphoma.
  • This chemotherapeutic group includes, but is not limited to cisplatin, oxaliplatin, carboplatin, triplatin tetranitrate (BBR3464), satraplatin, tetraplatin, ormiplatin, iproplatin, nedaplatin and lobaplatin.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with an antimetabolite in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, malignant melanoma, bladder carcinoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, esophageal cancer, brain cancer, anal cancer, leukaemia and lymphoma.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, bladder carcinoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, esophageal cancer, brain cancer, anal cancer, leukaemia and lymphoma.
  • This chemotherapeutic group includes, but is not limited to 5-fluorouracil, gemcitabine, cytarabine, capecitabine, decitabine, floxuridine, fludarabine, aminopterin, methotrexate, pemetrexed, raltitrexed, cladribine, clofarabine, mercaptopurine, pentostatin, and thioguanine.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancrea
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a mitotic inhibitor in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, prostate cancer, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, brain cancer, leukemia, and lymphoma.
  • a cancer selected from lung cancer, sarcoma, prostate cancer, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, brain cancer, leukemia, and lymphoma.
  • This chemotherapeutic group includes, but is not limited to paclitaxel, docetaxel, vinblastine, vincristine, vindesine, and vinorelbine.
  • the combination of PM01183, or a pharmaceutically acceptable salt thereof, with paclitaxel, docetaxel, vinblastine, vincristine, vindesine, and vinorelbine and even more preferred is the combination with paclitaxel, docetaxel, vincristine and vinorelbine in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, prostate cancer, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with an anticancer antibiotic in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, malignant melanoma, bladder carcinoma, prostate cancer, pancreas carcinoma, thyroid cancer, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, neuroblastoma, brain cancer, anal cancer, testicular cancer, leukemia, multiple myeloma and lymphoma.
  • This chemotherapeutic group includes, but is not limited to daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, pixantrone, valrubicin, mitomycin C, bleomycin, actinomycin A and mithramycin.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a topoisomerase I and/or II inhibitor in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, neuroblastoma, brain cancer, cervical cancer, testicular cancer, leukemia and lymphoma.
  • This chemotherapeutic group includes, but is not limited to topotecan, SN-38, irinotecan, camptothecin, rubitecan, etoposide, amsacrine and teniposide.
  • the combination of PM00104, or a pharmaceutically acceptable salt thereof, with topotecan, SN-38, irinotecan, camptothecin, rubitecan, etoposide, amsacrine and teniposide and even more preferred is the combination with topotecan, irinotecan and etoposide in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a proteosome inhibitor in the treatment of cancer, and more particularly in the treatment of lung cancer, prostate cancer, pancreas carcinoma, gastric carcinoma, hepatoma, colorectal cancer, brain cancer, multiple myeloma and lymphoma.
  • This chemotherapeutic group includes, but is not limited to bortezomib, disulfuram, epigallocatechin gallate, and salinosporamide A.
  • bortezomib particularly preferred is the combination of PM01183, or a pharmaceutically acceptable salt thereof, with bortezomib, disulfuram, epigallocatechin gallate, and salinosporamide A, and even more preferred is the combination with bortezomib in the treatment of cancer, and more particularly in the treatment of lung cancer, prostate cancer, pancreas carcinoma, gastric carcinoma, hepatoma, colorectal cancer and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a histone deacetylase inhibitor in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer, brain cancer and lymphoma.
  • This chemotherapeutic group includes, but is not limited to romidepsin, panobinostat, vorinostat, mocetinostat, belinostat, entinostat, resminostat, PCI-24781, AR-42, CUDC-101, and valproic acid.
  • the combination of PM01183, or a pharmaceutically acceptable salt thereof, with romidepsin, panobinostat, vorinostat, mocetinostat, belinostat, entinostat, resminostat, PCI-24781, AR-42, CUDC-101, and valproic acid and even more preferred is the combination with vorinostat in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a nitrogen mustard alkylating agent in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, bladder carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, leukemia, multiple myeloma and lymphoma.
  • a nitrogen mustard alkylating agent in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, bladder carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, leukemia, multiple myeloma and lymphoma.
  • This chemotherapeutic group includes, but is not limited to melphalan, ifosfamide, chlorambucil, cyclophosphamide, mechlorethamine, uramustine, estramustine and bendamustine.
  • melphalan ifosfamide, chlorambucil, cyclophosphamide, mechlorethamine, uramustine, estramustine and bendamustine
  • cyclophosphamide in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer and kidney cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a nitrosourea alkylating agent in the treatment of cancer, and more particularly in the treatment of lung cancer, ovarian cancer, breast cancer, brain cancer, multiple myeloma and lymphoma.
  • This chemotherapeutic group includes, but is not limited to lomustine, semustine, carmustine, fotemustine and streptozotocin.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a nonclassical alkylating agent in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, malignant melanoma, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer, brain cancer, leukemia and lymphoma.
  • a chemotherapeutic group includes, but is not limited to procarbazine, dacarbazine, temozolomide and altretamine.
  • Particularly preferred is the combination of PM01183, or a pharmaceutically acceptable salt thereof, with procarbazine, dacarbazine, temozolomide and altretamine, and even more preferred is the combination with dacarbazine and tezolomide in the treatment of lung cancer, sarcoma, malignant melanoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with an estrogen antagonist in the treatment of cancer, and more particularly in the treatment of breast cancer.
  • This chemotherapeutic group includes, but is not limited to toremifene, fulvestrant, tamoxifen and nafoxidine.
  • Particularly preferred is the combination of PM01183, or a pharmaceutically acceptable salt thereof, with toremifene, fulvestrant, tamoxifen and nafoxidine, and even more preferred is the combination with tamoxifen in the treatment of breast cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with an androgen antagonist in the treatment of cancer, and more particularly in the treatment of prostate cancer.
  • This chemotherapeutic group includes, but is not limited to bicalutamide, flutamide, MDV3100 and nilutamide.
  • Particularly preferred is the combination of PM01183, or a pharmaceutically acceptable salt thereof, with bicalutamide, flutamide, MDV3100 and nilutamide, and even more preferred is the combination with flutamide in the treatment of prostate cancer.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a mTOR inhibitor in the treatment of cancer, and more particularly in the treatment of lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • This chemotherapeutic group includes, but is not limited to sirolimus, temsirolimus, everolimus, ridaforolimus, KU-0063794 and WYE-354.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with a tyrosine kinase inhibitor in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • a cancer selected from lung cancer, sarcoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer and brain cancer.
  • This chemotherapeutic group includes, but is not limited to erlotinib, sorafenib, axitinib, bosutinib, cediranib, crizotinib, dasatinib, gefitinib, imatinib, canertinib, lapatinib, lestaurtinib, neratinib, nilotinib, semaxanib, sunitinib, vatalanib and vandetanib.
  • a cancer selected from lung cancer, sarcoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with aplidine in the treatment of cancer, and more particularly in the treatment of a cancer selected from sarcoma, gastric carcinoma, ovarian cancer, colorectal cancer, kidney cancer, brain cancer and leukemia.
  • a cancer selected from sarcoma, gastric carcinoma, ovarian cancer, colorectal cancer, kidney cancer, brain cancer and leukemia.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with ET-743 (trabectedin) in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, kidney cancer, leukemia and lymphoma.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, kidney cancer, leukemia and lymphoma.
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with PM02734 in the treatment of cancer, and more particularly in the treatment of a cancer selected from sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer, brain cancer, leukemia and lymphoma.
  • a cancer selected from sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, breast cancer, colorectal cancer, kidney cancer, brain cancer, leukemia and lymphoma.
  • PM02734 ((4S)-MeHex-D-Val-L-Thr-L-Val-D-Val-D-Pro-L-Orn-D-allo-Ile-cyclo(D-allo-Thr-D-allo-Ile-D-Val-L-Phe-Z-Dhb-L-Val) is a synthetic depsipeptide related to the family of kahalalide compounds, which is currently in clinical trials for the treatment of cancer.
  • This compound is the subject of WO 2004/035613 and has the following structure:
  • the invention is directed to the combination of PM01183, or a pharmaceutically acceptable salt thereof, with PM00104 in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, kidney cancer, leukemia and lymphoma.
  • a cancer selected from lung cancer, sarcoma, malignant melanoma, prostate cancer, pancreas carcinoma, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, kidney cancer, leukemia and lymphoma.
  • PM00104 is a synthetic alkaloid related to jorumycin and renieramycins, and also to safracin and saframycin compounds, which is currently in clinical trials for the treatment of cancer, and has the following structure:
  • the invention includes any pharmaceutically acceptable salt of any drug referred to herein, which can be synthesized from the parent compound by conventional chemical methods as disclosed before.
  • the invention relates to synergistic combinations employing PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above.
  • An indication of synergism can be obtained by testing the combinations and analyzing the results, for example by the Chou-Talalay method or by any other suitable method, such as those provided in the Examples section.
  • the possible favorable outcomes for synergism include 1) increasing the efficacy of the therapeutic effect, 2) decreasing the dosage but increasing or maintaining the same efficacy to avoid toxicity, 3) minimizing or slowing down the development of drug resistance, and 4) providing selective synergism against target (or efficacy synergism) versus host (or toxicity antagonism). Accordingly, in a combination of two chemotherapeutic agents having synergism, the treatment regimen will be different of those in which the combination of the two drugs shows only an additive effect.
  • the invention in another embodiment, relates to a method of increasing or potentiating the therapeutic efficacy of an anticancer drug selected from the list of drugs given above in the treatment of cancer, which comprises administering to a patient in need thereof a therapeutically effective amount of PM01183, or a pharmaceutically acceptable salt thereof, in conjunction with this other anticancer drug.
  • An indication of increase or potentiation of the therapeutic efficacy can be obtained by testing the combinations and analyzing the results, for example the tumor growth inhibition. This tumor growth inhibition can be assessed by comparing the mean tumor volume of the treatment combining the two drugs (PM01183 and the other drug) with those of the other drug monotherapy treatment.
  • increase or potentiation of the therapeutic efficacy is determined when the response of the combination therapy is greater than the best response of the most active drug administered as single agent (monotherapy) on the same schedule and dose as used in the combination therapy.
  • This aspect of the invention is further illustrated in the Examples section, specifically in Examples 13-19.
  • the invention is directed to the use of PM01183, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer by combination therapy employing PM01183, or a pharmaceutically acceptable salt thereof, with another anticancer drug selected from the list of drugs given above.
  • the invention is directed to a method for the treatment of cancer comprising administering to a patient in need of such treatment a therapeutically effective amount of PM01183, or pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of another anticancer drug selected from the list of drugs given above.
  • the invention is directed to PM01183, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer comprising administering a therapeutically effective amount of PM01183, or a pharmaceutical acceptable salt thereof, in combination with a therapeutically effective amount of another anticancer drug selected from the list of drugs given above.
  • PM01183, or a pharmaceutically acceptable salt thereof, and the other anticancer drug may be provided in the same medicament or as separate medicaments for administration at the same time or at different times.
  • PM01183, or a pharmaceutically acceptable salt thereof, and the other anticancer drug are provided as separate medicaments for administration at different times.
  • either PM01183, or a pharmaceutically acceptable salt thereof, or the other anticancer drug may be administered first.
  • both drugs can be administered in the same day or at different days, and they can be administered using the same schedule or at different schedules during the treatment cycle.
  • the administration of both drugs can be done by using the same route of administration or different routes. For instance, both drugs can be administered by intravenous administration or, alternatively, one drug can be administered orally and the other one by intravenous administration.
  • compositions of the present invention may comprise all the components (drugs) in a single pharmaceutically acceptable formulation or, alternatively, the components may be formulated separately and administered in combination with one another.
  • Various pharmaceutically acceptable formulations well known to those of skill in the art can be used in the present invention.
  • selection of an appropriate formulation for use in the present invention can be performed by those skilled in the art by taking into account the route of administration and the solubility characteristics of the components of the composition.
  • both drugs in combination will vary according to the particular formulation, the mode of application, and the particular site, patient and tumour being treated. Other factors like age, body weight, sex, diet, time of administration, rate of excretion, condition of the patient, other drug combinations, reaction sensitivities and severity of the disease shall be taken into account. Administration can be carried out continuously or periodically within the maximum tolerated dose.
  • the combination of the invention may be used alone or in combination with one or more of a variety of anticancer agents or supportive care agents.
  • anticancer effects of the treatments of the present invention include, but are not limited to, inhibition of tumor growth, tumor growth delay, regression of tumor, shrinkage of tumor, increased time to regrowth of tumor on cessation of treatment, slowing of disease progression, and prevention of metastasis. It is expected that when a treatment of the present invention is administered to a patient, such as a human patient, in need of such treatment, said treatment will produce an effect, as measured by, for example, the extent of the anticancer effect, the response rate, the time to disease progression, or the survival rate.
  • the treatments of the invention are suited for human patients, especially those who are relapsing or refractory to previous chemotherapy. First line therapy is also envisaged.
  • the present invention is directed to a kit for use in the treatment of cancer, comprising a supply of PM01183, or a pharmaceutically acceptable salt thereof, in dosage units for at least one cycle, and printed instructions for the use of PM01183, or a pharmaceutically acceptable salt thereof, with another anticancer drug selected from the list of drugs given above in combination.
  • the present invention is directed to a kit for use in the treatment of cancer, comprising a supply of PM01183, or a pharmaceutically acceptable salt thereof, in dosage units for at least one cycle, a supply of another anticancer drug selected from the list of drugs given above in dosage units for at least one cycle, and printed instructions for the use of both drugs in combination.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising PM01183, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient, for use in combination with another anticancer drug selected from the list of drugs given above in the treatment of cancer.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising PM01183, or a pharmaceutically acceptable salt thereof, another anticancer drug selected from the list of drugs given above, and a pharmaceutically acceptable carrier.
  • This pharmaceutical composition is preferable for use in the treatment of cancer.
  • the invention further provides for the use of PM01183, or a pharmaceutically acceptable salt thereof, in the preparation of a composition for use in combination with another anticancer drug selected from the list of drugs given above in the treatment of cancer.
  • the invention further provides for the use of PM01183, or a pharmaceutically acceptable salt thereof, for the treatment of cancer, in combination therapy with another anticancer drug selected from the list of drugs given above.
  • cancer cells are contacted, or otherwise treated, with a combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above.
  • the cancer cells are preferably human and include carcinoma cells, sarcoma cells, leukemia cells, lymphoma cells, and myeloma cells.
  • the cancer cells are cells of lung cancer, sarcoma, malignant melanoma, bladder carcinoma, prostate cancer, pancreas carcinoma, thyroid cancer, gastric carcinoma, ovarian cancer, hepatoma, breast cancer, colorectal cancer, kidney cancer, esophageal cancer, neuroblastoma, brain cancer, cervical cancer, anal cancer, testicular cancer, leukemia, multiple myeloma and lymphoma.
  • the combination provides a synergistic inhibitory effect against the cancer cells, particularly against the human cancer cells mentioned above.
  • the combination inhibits proliferation or survival of contacted cancer cells.
  • a lower level of proliferation or survival of the contacted cancer cells compared to the non-contacted cancer cells supports the combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above as being effective for treating a patient with cancer.
  • the invention provides for a method for inhibiting the growth of cancer cells comprising contacting said cancer cells with an effective amount of PM01183, or a pharmaceutically acceptable salt thereof, in combination with another anticancer drug selected from the list of drugs given above.
  • the invention provides for a method for inhibiting the growth of cancer cells comprising contacting said cancer cells with a synergistic combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above, wherein said combination provides improved inhibition against cancer cell growth as compared to (i) PM01183, or a pharmaceutically acceptable salt thereof, in the absence of the other anticancer drug, or (ii) the other anticancer drug in the absence of PM01183.
  • the invention provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a synergistic combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above for inhibiting the growth of cancer cells, wherein said combination provides improved inhibition against cancer cell growth as compared to (i) PM01183, or a pharmaceutically acceptable salt thereof, in the absence of the other anticancer drug, or (ii) the other anticancer drug in the absence of PM01183.
  • the combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above inhibits tumor growth or reduces the size of a tumor in vivo.
  • the combination inhibits in vivo growth and/or reduces the size of carcinoma, sarcoma, leukemia, lymphoma, and myeloma.
  • the combination inhibits in vivo tumor growth of lung, sarcoma, malignant melanoma, bladder, prostate, pancreas, thyroid, gastric, ovarian, hepatoma, breast, colorectal, kidney, esophageal, neuroblastoma, brain, cervical, anal, testicular, leukemia, multiple myeloma and lymphoma tumours.
  • these combinations inhibit tumor growth or reduce the size of human cancer xenografts, particularly human gastric, pancreas, sarcoma, lung, colorectal and ovary tumors xenografts, in animal models.
  • a reduced growth or reduced size of human cancer xenografts in animal models administered with these combinations further supports the combination of PM01183, or a pharmaceutically acceptable salt thereof, and another anticancer drug selected from the list of drugs given above as being effective for treating a patient with cancer.
  • the invention provides for a method for reducing the size of a tumor, comprising administering an effective amount of PM01183, or a pharmaceutically acceptable salt thereof, in combination with another anticancer drug selected from the list of drugs given above.
  • the invention provides for a method for inhibiting tumor growth, comprising administering an effective amount of PM01183, or a pharmaceutically acceptable salt thereof, in combination with another anticancer drug selected from the list of drugs given above.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of lung carcinoma.
  • oxaliplatin carmustine, cyclophosphamide, mytomicin C (stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.), 5-fluorouracil (5-FU), gemcitabine, paclitaxel, docetaxel, vincristine, daunorubicin, actinomycin D, topotecan, etoposide, bortezomib, vorinostat, dacarbazine, temsirolimus, erlotinib, ET-743 and PM00104 (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • A549 was the human lung carcinoma cell line selected for this assay.
  • A549 cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were used as starting concentrations for each compound (100% concentration).
  • IC 50 of PM01183 IC 50 of Agent 100% 0% 75% 25% 70% 30% 60% 40% 50% 50% 40% 60% 30% 70% 25% 75% 0% 100%
  • response values were plotted on a scatter plot with dose ratios given on the x-axis and % response values on the y-axis.
  • a horizontal line was drawn between the two endpoint response values (E.g. between the response values for 100% IC 50 PM01183 and 100% IC 50 standard chemotherapeutic agent).
  • points lying above or below this predicted line of additivity could be interpreted as representing antagonistic or synergistic drug interaction, respectively.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of sarcoma.
  • cisplatin oxaliplatin, cyclophosphamide, mytomicin C (stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.), gemcitabine, docetaxel, vincristine, vinorelbine, daunorubicin, cytarabine, actinomycin D, topotecan, etoposide, vorinostat, dacarbazine, temsirolimus, erlotinib, aplidine, PM02734, ET-743 and PM00104 (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • A673 was the human rhabdomyosarcoma cell line selected for this assay.
  • A673 cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the A673 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the A673 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 2.
  • A673 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of malignant melanoma.
  • cisplatin mytomicin C (stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.), 5-fluorouracil, doxorubicin, daunorubicin, cytarabine, topotecan, irinotecan, methotrexate, etoposide, dacarbazine, temsirolimus, PM02734, ET-743 and PM00104 (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • SK-MEL-2 was the human melanoma cell line selected for this assay.
  • SK-MEL-2 cells were maintained in Minimum Essential Medium Eagle (MEME) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • MEME Minimum Essential Medium Eagle
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the SK-MEL-2 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the SK ⁇ MEL-2 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 3.
  • SK-MEL-2 tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of prostate cancer.
  • PC-3 was the human prostate adenocarcinome cell line selected for this assay.
  • PC-3 cells were maintained in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • RPMI Roswell Park Memorial Institute medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the PC-3 tumor cell line.
  • the IC 50 values (72 hours drug exposure) of each individual agent for the PC-3 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 4.
  • PC-3 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in examples 1.
  • the combination of PM01183 with temsirolimus exhibited strong synergism ( FIG. 76 ).
  • j. The combination of PM01183 with erlotinib ( FIG. 77 ) showed synergism at almost all dose ratios.
  • k. The combination of PM01183 with ET-743 ( FIG. 78 ) showed synergism at almost all dose ratios.
  • l. The combination of PM01183 with PM02734 ( FIG. 79 ) showed synergism at the 75/25-70/30 and 30/70 dose ratios.
  • m. The combination of PM01183 with PM00104 exhibited strong synergism ( FIG. 80 ).
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of pancreatic carcinoma.
  • cisplatin oxaliplatin
  • gemcitabine daunorubicin
  • cytarabine doxorubicin
  • actinomycin D topotecan
  • irinotecan methotrexate
  • temsirolimus bortezomib
  • erlotinib PM02734, ET-743
  • PM00104 stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.
  • Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • PANC-1 was the human pancreatic carcinoma cell line selected for this assay.
  • PANC-1 cells were maintained in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • RPMI Roswell Park Memorial Institute medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the PANC-1 tumor cell line.
  • the IC 50 values (72 hours drug exposure) of each individual agent for the PANC-1 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 5.
  • PANC-1 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed example 1.
  • the combination of PM01183 with vorinostat ( FIG. 93 ) showed synergism at almost all dose ratios.
  • g. The combination of PM01183 with temsirolimus exhibited strong synergism ( FIG. 94 ).
  • h. The combination of PM01183 with erlotinib exhibited strong synergism ( FIG. 95 ).
  • i. The combination of PM01183 with ET-743 ( FIG. 96 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with PM02734 ( FIG. 97 ) showed synergism at almost all dose ratios.
  • k. The combination of PM01183 with PM00104 showed synergism ( FIG. 98 ) at the 75/25 and 50/50 dose ratios.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of gastric cancer.
  • HGC-27 was the human gastric carcinoma cell line selected for this assay. HGC-27 cells were maintained in Iscove's modified Dulbeco's medium (IDMD) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • IDMD Iscove's modified Dulbeco's medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the HGC-27 tumor cell line.
  • HGC-27 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed, as described before and the cytotoxic effect was measured by the MTT Assay, as disclosed in example 1.
  • the combination of PM01183 with paclitaxel exhibited strong synergism ( FIG. 105 ).
  • the combination of PM01183 with vincristine ( FIG. 106 ) and PM01183 with vinorelbine ( FIG. 107 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with daunorubicin ( FIG. 108 ) and PM01183 with actinomycin D ( FIG. 110 ) exhibited strong synergism.
  • the combination of PM01183 with doxorubicin ( FIG. 109 ) exhibited synergism at the 75/25-60/40 dose ratios.
  • the combination of PM01183 with topotecan exhibited strong synergism ( FIG. 111 ).
  • the combination of PM01183 with irinotecan ( FIG. 112 ) showed synergism at the 70/30-60/40 and 40/60 dose ratios, while the combination of PM01183 with etoposide ( FIG. 113 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with bortezomib exhibited strong synergism ( FIG. 114 ).
  • the combination of PM01183 with vorinostat ( FIG. 115 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with cyclophosphamide exhibited strong synergism ( FIG. 116 ).
  • the combination of PM01183 with dacarbazine exhibited strong synergism ( FIG. 117 ).
  • j. The combination of PM01183 with temsirolimus exhibited strong synergism ( FIG. 118 ).
  • k. The combination of PM01183 with erlotinib exhibited strong synergism ( FIG. 119 ).
  • l. The combination of PM01183 with aplidine showed strong synergism ( FIG. 120 ).
  • the combination of PM01183 with ET-743 FIG. 121 ) showed synergism at the 50/50 and 75/25 dose ratios.
  • n. The combination of PM01183 with PM02734 exhibited strong synergism ( FIG. 122 ).
  • o. The combination of PM01183 with PM00104 ( FIG. 123 ) showed synergism at almost all dose ratios.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of ovarian cancer.
  • IGROV-1 was the human ovarian adenocarcinoma cell line selected for this assay. IGROV-1 cells were maintained in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • RPMI Roswell Park Memorial Institute medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the IGROV-1 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the IGROV-1 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 7.
  • IGROV-1 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of hepatocellular cancer.
  • cisplatin oxaliplatin
  • cyclophosphamide stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.
  • 5-fluorouracil gemcitabine
  • paclitaxel docetaxel
  • vincristine vinorelbine
  • daunorubicin cytarabine
  • doxorubicin topotecan
  • irinotecan methotrexate
  • etoposide bortezomib
  • erlotinib ET-743
  • PM00104 stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.
  • Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • HepG2 was the human hepatocellular liver carcinoma cell line selected for this assay. HepG2 cells were maintained in Minimum Essential Medium Eagle (MEME) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • MEME Minimum Essential Medium Eagle
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the HepG2 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the HepG2 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 8.
  • HepG2 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the combination of PM01183 with irinotecan ( FIG. 164 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with bortezomib ( FIG. 166 ) showed synergism at the 75/25-60/40 dose ratios.
  • the combination of PM01183 with cyclophosphamide ( FIG. 167 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with erlotinib ( FIG. 168 ) exhibited strong synergism.
  • the combination of PM01183 with ET-743 ( FIG. 169 ) showed synergism at the 60/40-50/50 dose ratios.
  • the combination of PM01183 with PM00104 ( FIG. 170 ) exhibited strong synergism.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of breast cancer.
  • MDA-MB-231 was the human breast adenocarcinoma cell line selected for this assay.
  • MDA-MB-231 cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the MDA-MB-231 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the MDA-MB-231 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 9.
  • MDA-MB-231 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the combination of PM01183 with ET-743 exhibited strong synergism ( FIG. 195 ).
  • the combination of PM01183 with PM02734 ( FIG. 196 ) exhibited synergism at almost all dose ratios.
  • the combination of PM01183 with PM00104 ( FIG. 197 ) showed synergism at almost all dose ratios.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of colorectal cancer.
  • the following agents were evaluated in combination with PM01183: cisplatin, oxaliplatin, cyclophosphamide, mytomicin C (stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.), 5-fluorouracil, gemcitabine, docetaxel, vinorelbine, daunorubicin, dacarbazine, cytarabine, doxorubicin, actinomycin D, topotecan, irinotecan, etoposide, vorinostat, bortezomib, temsirolimus, erlotinib, PM02734 and aplidine (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • HT-29 was the human colon adenocarcinoma cell line selected for this assay.
  • HT-29 cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the HT-29 tumor cell line.
  • IC 50 values 72 hours drug exposure
  • HT-29 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the combination of PM01183 with temsirolimus exhibited strong synergism ( FIG. 216 ).
  • the combination of PM01183 with erlotinib showed synergism at almost all dose ratios ( FIG. 217 ).
  • the combination of PM01183 with aplidine ( FIG. 218 ) showed synergism at the 40/60-25/75 dose ratios.
  • the combination of PM01183 with PM02734 ( FIG. 219 ) showed synergism at almost all dose ratios.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of kidney cancer.
  • cisplatin cyclophosphamide
  • mytomicin C stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.
  • 5-fluorouracil gemcitabine, methotrexate, docetaxel, vincristine, vinorelbine, daunorubicin, dacarbazine, cytarabine, doxorubicin, actinomycin D, topotecan, irinotecan, etoposide, vorinostat, erlotinib, PM02734, ET-743, PM00104 and aplidine (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • RXF-393 was the human kidney carcinoma cell line selected for this assay.
  • RXF-393 cells were maintained in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • RPMI Roswell Park Memorial Institute medium
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the RXF-393 tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the RXF-393 tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 11.
  • RXF-393 human tumor cells were incubated with PM01183 in combination with each of the agents mentioned above in the same combination of unique IC 50 concentrations as those described in example 1.
  • Cell culture and cell plating were performed as described before and the cytotoxic effect was measured by the MTT Assay as disclosed in example 1.
  • the combination of PM01183 with daunorubicin ( FIG. 228 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with doxorubicin ( FIG. 229 ) showed synergism at the 75/25-60/40 dose ratios, while the combination of PM01183 with actinomycin D ( FIG. 230 ) showed synergism at the 75/25-70/30 and 30/70 dose ratios.
  • the combination of PM01183 with mitomycin C ( FIG. 231 ) exhibited strong synergism.
  • the combination of PM01183 with topotecan ( FIG. 232 ) exhibited strong synergism.
  • the combination of PM01183 with irinotecan ( FIG.
  • the combination of PM01183 with erlotinib exhibited strong synergism ( FIG. 238 ).
  • the combination of PM01183 with aplidine ( FIG. 239 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with ET-743 ( FIG. 240 ) showed synergism at almost all dose ratios.
  • the combination of PM01183 with PM02734 FIG. 241
  • the combination of PM01183 with PM00104 ( FIG. 242 ) exhibited strong synergism.
  • the objective of this study was to determine the ability of PM01183 to potentiate the antitumor activity of chemotherapeutic agents used in the treatment of glioblastoma.
  • cisplatin oxaliplatin (stock solutions of these compounds prepared in sterile double distilled water and stored at ⁇ 20° C.), 5-fluorouracil, gemcitabine, docetaxel, vincristine, daunorubicin, dacarbazine, doxorubicin, topotecan, irinotecan, methotrexate, etoposide, vorinostat, temsirolimus, bortezomib erlotinib, PM02734, ET-743 and aplidine (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • U87-MG was the human glioblastoma cell line selected for this assay.
  • U87-MG cells were maintained in Minimum Essential Medium Eagle (MEME) supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • MEME Minimum Essential Medium Eagle
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • IC 50 values were determined for each drug after 72 hours of drug exposure in the U87-MG tumor cell line.
  • IC 50 values (72 hours drug exposure) of each individual agent for the U87-MG tumor cell line were calculated by using the same methodology disclosed in example 1 and are shown in table 12.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of paclitaxel, vinorelbine and doxorubicin by using a xenograft model of human ovarian carcinoma.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was A2780 cell line, which was obtained from the European Collection of Cell Cultures (ECACC n o 93112519).
  • A2780 cells were grown at 37° C. with 5% CO 2 in RPMI-1640 medium. Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 1 ⁇ 10 7 A2780 cells (from in vitro passage 5 in PM01183 and doxorubicin and PM01183 and vinorelbine studies; and passage 9 in PM01183 and paclitaxel study), in 0.05 mL suspension of 50% Matrigel and 50% serum free medium, without antibiotics.
  • Treatment tolerability was assessed by monitoring body weight evolution, clinical signs as well as evidences of local damage in the injection site.
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Doxorubicin was provided in the form of a solid powder containing Doxorubicin HCl, which was reconstituted in 0.9% saline solution.
  • Vinorelbine was provided as a solution prepared by diluting the product with 0.9% saline solution.
  • Paclitaxel was provided in the form of a solution prepared by diluting the product with 5% glucose solution for injection to the target final concentration.
  • the combination index (CI) that quantitatively measures the degree of drug interactions, was obtained from the fractions affected by the treatment, Fa (defined as 1 ⁇ T/C) for each experimental group at the last measurement day (Day 10 for PM01183 and paclitaxel combination study, and PM01183 and doxorubicin study, and Day 9 for PM01183 and vinorelbine study) using the median-effect principle (Chou T. C. Pharmacol. Rev. 2006, 58, 621-681).
  • Table 13 reports the % T/C values obtained with PM01183 and paclitaxel both administered as single agents and in combination for each dose level
  • FIG. 263 shows the tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, paclitaxel, and the corresponding combinations for the groups dosed at the two highest ratios.
  • Table 14 reports the % T/C values obtained with PM01183 and vinorelbine both administered as single agents and in combination for each dose level
  • FIG. 264 shows the tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, vinorelbine, and the corresponding combinations for the groups dosed at the two highest ratios.
  • Table 15 reports the % T/C values obtained with PM01183 and doxorubicin both administered as single agents and in combination for each dose level, and FIG. 265 shows the tumor volume evaluation of A2780 tumors in mice treated with placebo, PM01183, doxorubicin, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (25 mg/kg paclitaxel+0.18 mg/kg PM01183) vs paclitaxel alone (25 mg/kg paclitaxel) were 28.8 vs 42.9 (day 5), 20.9 vs 34.0 (day 7), and 9.2 vs 19.8 (day 10)
  • the TC (%) values of the combination (18.75 mg/kg paclitaxel+0.135 mg/kg PM01183) vs paclitaxel alone (18.75 mg/kg paclitaxel) were 37.1 vs 43.2 (day 5), 36.0 vs 41.5 (day 7), and 22.6 vs 31.1 (day 10).
  • the combination of PM01183 and vinorelbine produced lower T/C values than the more active single agent in this experiment (vinorelbine at doses of 16 mg/kg and 12 mg/kg).
  • the TC (%) values of the combination (16 mg/kg vinorelbine+0.18 mg/kg PM01183) vs vinorelbine alone (16 mg/kg vinorelbine) were 10.9 vs 20.8 (day 5), 10.6 vs 24.5 (day 7), and 8.6 vs 20.0 (day 9)
  • the TC (%) values of the combination (12 mg/kg vinorelbine+0.135 mg/kg PM01183) vs vinorelbine alone (12 mg/kg vinorelbine) were 29.6 vs 39.1 (day 5), 31.2 vs 43 (day 7), and 26.8 vs 36.1 (day 9). Therefore, when PM01183 is combined with vinorelbine a potentiation of the antitumor activity is clearly observed.
  • the TC (%) values of the combination (8 mg/kg doxorubicin+0.18 mg/kg PM01183) vs doxorubicin alone (8 mg/kg doxorubicin) were 32.6 vs 49.8 (day 5), 30.3 vs 48.1 (day 7), and 21.1 vs 39.4 (day 10). Therefore, when PM01183 is combined with doxorubicin a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of cisplatin and 5-fluorouracil by using a xenograft model of human gastric carcinoma.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was HGC-27 cell line, which was obtained from the European Collection of Cell Cultures (ECACC n o 94042256).
  • HGC-27 cells were grown at 37° C. with 5% CO 2 in Iscove's modified Dulbeco's medium (IDMD). Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 HGC-27 cells (from in vitro passage 4 in PM01183 and cisplatin study, and passage 6 in PM01183 and 5-fluorouracil study), in 0.05 mL suspension of 50% Matrigel and 50% serum free medium, without antibiotics.
  • IDMD Iscove's modified Dulbeco's medium
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Cisplatin and 5-fluorouracil were provided as solutions prepared by diluting the product with 0.9% saline solution for injection to the target final concentration.
  • Table 16 reports the % T/C values obtained with PM01183 and cisplatin both administered as single agents and in combination for each dose level
  • FIG. 266 shows the tumor volume evaluation of HGC-27 tumors in mice treated with placebo, PM01183, cisplatin, and the corresponding combinations for the groups dosed at the two highest ratios.
  • Table 17 reports the % T/C values obtained with PM01183 and 5-fluorouracil both administered as single agents and in combination for each dose level
  • FIG. 267 shows the tumor volume evaluation of HGC-27 tumors in mice treated with placebo, PM01183, 5-fluorouracil, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (6 mg/kg cisplatin+0.18 mg/kg PM01183) vs PM01183 alone (0.18 mg/kg PM01183) were 12.9 vs 33.5 (day 10), 7.6 vs 24.3 (day 12), and 4.6 vs 24.3 (day 14), and the TC (%) values of the combination (4.5 mg/kg cisplatin+0.135 mg/kg PM01183) vs PM01183 alone (0.135 mg/kg PM01183) were 17.3 vs 39.3 (day 10), 12.1 vs 37.1 (day 12), and 9.8 vs 38.3 (day 14). Therefore, when PM01183 is combined with paclitaxel a potentiation of the antitumor activity is clearly observed.
  • the TC (%) values of the combination (50 mg/kg 5-fluorouracil+0.18 mg/kg PM01183) vs PM01183 alone (0.18 mg/kg PM01183) were 35.9 vs 43.3 (day 7), 31.5 vs 41.0 (day 9), 25.3 vs 33.0 (day 12), and 22.0 vs 29.2 (day 14). Therefore, when PM01183 is combined with 5-fluorouracil a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of gemcitabine by using a xenograft model of human pancreatic cancer.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was SW1990 cell line, which was obtained from the American Type Culture Collection (ATCC: CRL-2172TM).
  • SW1990 cells were grown at 37° C. with 5% CO 2 in RPMI-1640 medium. Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 SW1990 cells, from in vitro passage 12, in 0.05 mL suspension of 50% Matrigel and 50% serum free medium, without antibiotics.
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Gemcitabine was provided as a solution prepared by reconstituting the product with 0.9% saline solution for injection to a concentration of 40 mg/ml stock solution.
  • the gemcitabine stock solution was further diluted with 0.9% saline solution for injection to the target final concentration.
  • Table 18 reports the % T/C values obtained with PM01183 and gemcitabine both administered as single agents and in combination for each dose level
  • FIG. 268 shows the tumor volume evaluation of SW1990 tumors in mice treated with placebo, PM01183, gemcitabine, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (180 mg/kg gemcitabine+0.18 mg/kg PM01183) vs PM01183 alone (0.18 mg/kg PM01183) were 31.7 vs 44.2 (day 20), 28.0 vs 45.3 (day 22), 26.0 vs 44.8 (day 24), and 22.7 vs 38.9 (day 28). Therefore, when PM01183 is combined with gemcitabine a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of temozolomide by using a xenograft model of human brain tumor.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was U87-MG cell line, which was obtained from the American Type Culture Collection (ATCC HTB-14TM).
  • U87-MG cells were grown at 37° C. with 5% CO 2 in Minimum Essential Medium Eagle (MEME). Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 U87-MG cells, from in vitro passage 5, in 0.05 mL suspension of 50% Matrigel and 50% serum free medium, without antibiotics.
  • MEME Minimum Essential Medium Eagle
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Temozolomide was provided as a solution prepared by diluting the product in DMSO 10% in 0.9% saline solution for injection to the target final concentration.
  • PM01183 and temozolomide treatments were administered as follows: PM01183, intravenously once per week up to 3 consecutive weeks, on Days 0, 7 and 14, temozolomide orally, in a daily basis during 8 consecutive days (Days 0 to 7), and placebo was administered following the same schedule as those provided for PM01183 and temozolomide.
  • Dose level groups were administered either as single agents or in combination.
  • Table 19 reports the % T/C values obtained with PM01183 and temozolomide both administered as single agents and in combination for each dose level
  • FIG. 269 shows the tumor volume evaluation of U87-MG tumors in mice treated with placebo, PM01183, temozolomide, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (3 mg/kg temozolomide+0.18 mg/kg PM01183) vs temozolomide alone (3 mg/kg temozolomide) were 18.3 vs 22.9 (day 11), 16.6 vs 28.4 (day 14), and 17.4 vs 31.5 (day 16), and the TC (%) values of the combination (1.5 mg/kg temozolomide+0.135 mg/kg PM01183) vs temozolomide alone (1.5 mg/kg temozolomide) were 29.1 vs 59.3 (day 11), 29.0 vs 50.0 (day 14), and 30.9 vs 53.5 (day 16). Therefore, when PM01183 is combined with temozolomide a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of iriniotecan by using a xenograft model of human lung cancer.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was H460 cell line, which was obtained from the American Type Culture Collection of Cell Cultures (ATCC ref. HTB-177TM).
  • H460 cells were grown at 37° C. with 5% CO2 in Dulbecco's modified Eagle's medium (DMEM). Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 H460 cells, from in vitro passage 10, in 0.05 mL suspension of 50% Matrigel and 50% serum free medium, without antibiotics.
  • DMEM Dulbecco's modified Eagle's medium
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Irinotecan was provided in the form of a solution prepared by diluting the product with 5% glucose solution for injection to the target final concentration.
  • PM01183 and irinotecan treatments were intravenously administered as follows: PM01183 once per week up to 2 consecutive weeks, on Days 0 and 7, irinotecan was dosed every 4 days, on Days 0, 4 and 8, and placebo was administered following the same schedule as those provided for PM01183 and irinotecan.
  • Dose level groups were administered either as single agents or in combination.
  • Table 20 reports the % T/C values obtained with PM01183 and irinotecan both administered as single agents and in combination for each dose level
  • FIG. 270 shows the tumor volume evaluation of H460 tumors in mice treated with placebo, PM01183, irinotecan, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (50 mg/kg irinotecan+0.18 mg/kg PM01183) vs irinotecan alone (50 mg/kg irinotecan) were 19.4 vs 34.7 (day 5), 13.4 vs 27.5 (day 7), 10.9 vs 24.8 (day 9), and 9.0 vs 22.9 (day 12), and the TC (%) values of the combination (37.5 mg/kg irinotecan+0.135 mg/kg PM01183) vs irinotecan alone (37.5 mg/kg irinotecan) were 23.8 vs 44.0 (day 5), 18.4 vs 36.7 (day 7), 15.7 vs 35.6 (day 9), and 15.3 vs 37.0 (day 12). Therefore, when PM01183 is combined with irinotecan a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of temozolomide by using a xenograft model of human fibrosarcoma.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was HT1080 cell line, which was obtained from the American Type Culture Collection (ATCC CCL-121TM).
  • HT1080 cells were grown at 37° C. with 5% CO 2 in Minimum Essential Medium Eagle (MEME). Each animal was orthotopically implanted into gastroecnemius muscle by an intramuscular injection using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 HT1080 cells, from in vitro passage 9, suspended in serum free medium, without antibiotics.
  • MEME Minimum Essential Medium Eagle
  • Total diameter (tumor+leg) measurements were determined by using digital caliper (Fowler Sylvac, S235PAT). This total diameter and animal body weights were measured 2-3 times per week starting from the first day of treatment.
  • Treatment tolerability was assessed by monitoring body weight evolution, clinical signs as well as evidences of local damage in the injection site.
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • dacarbazine was provided in the form of a solution prepared by diluting the product with 5% glucose solution for injection to the target final concentration.
  • Table 21 reports the % T/C values obtained with PM01183 and dacarbazine both administered as single agents and in combination for each dose level, and FIG. 271 shows the total diameter (tumor+leg) evaluation of HT1080 tumors in mice treated with placebo, PM01183, dacarbazine, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination were 4.0 vs 26.7 (day 9), 10.4 vs 11.5 (day 11), ⁇ 4.2 vs 21.2 (day 14), and 1.0 vs 30.6 (day 16)
  • the TC (%) values of the combination 112.5 mg/kg dacarbazine+0.135 mg/kg PM01183) vs PM01183 alone (0.135 mg/kg PM01183) were ⁇ 8.0 vs 36.0 (day 9), ⁇ 17.7 vs 30.2 (day 11), ⁇ 6.8 vs 33.0 (day 14), and 7.3 vs 41.9 (day 16). Therefore, when PM01183 is combined with dacarbazine a potentiation of the antitumor activity is clearly observed.
  • the aim of these studies was to evaluate the ability of PM01183 to potentiate the antitumor activity of irinotecan by using a xenograft model of human colorectal carcinoma.
  • mice Female athymic nude mice (Harlan Laboratories Models, S.L. (Barcelona, Spain) were utilized for all experiments. Animals were housed in individually ventilated cages, up to ten per cage in a 12-hour light-dark cycle at 21-23° C. and 40-60% humidity. The mice were allowed free access to irradiated standard rodent diet and sterilized water. Animals were acclimated for at least 5 days prior to tumor implantation with a tumor cell suspension.
  • the tumor model used in these studies was HT-29 cell line, which was obtained from the American Type Culture Collection (ATCC ref. HTB-38TM).
  • HT-29 cells were grown at 37° C. with 5% CO 2 in Dulbecco's modified Eagle's medium (DMEM). Each animal was subcutaneously implanted on the right flank, using 26 G needle and a 1 cc syringe, with 5 ⁇ 10 6 HT-29 cells, from in vitro passage 10, in 0.05 mL of 0.9% Sodium Chloride for injection.
  • DMEM Dulbecco's modified Eagle's medium
  • Tumor measurements and treatment tolerability were performed and determined as disclosed in Example 13. Treatment tolerability was assessed by monitoring body weight evolution, clinical signs as well as evidences of local damage in the injection site.
  • PM01183 was provided in the form of vials of lyophilized PM01183 cake which was reconstituted with water for infusion to a concentration of 0.2 mg/mL.
  • the PM01183 stock solution was further diluted in 5% glucose solution for injection to the dosing formulation concentrations.
  • Irinotecan was provided in the form of a solution prepared by diluting the product with 5% glucose solution for injection to the target final concentration.
  • PM01183 and irinotecan treatments were intravenously administered as follows: PM01183 once per week up to 3 consecutive weeks, on Days 0, 7 and 14, irinotecan was dosed every 4 days, on Days 0, 4, 8, 12 and 16, and placebo was administered following the same schedule as those provided for PM01183 and irinotecan.
  • Dose level groups were administered either as single agents or in combination.
  • Table 22 reports the % T/C values obtained with PM01183 and irinotecan both administered as single agents and in combination for each dose level, and FIG. 272 shows the tumor volume evaluation of HT-29 tumors in mice treated with placebo, PM01183, irinotecan, and the corresponding combinations for the groups dosed at the two highest ratios.
  • the TC (%) values of the combination (50 mg/kg irinotecan+0.18 mg/kg PM01183) vs irinotecan alone (50 mg/kg irinotecan) were 30.4 vs 51.7 (day 14), 21.7 vs 41.4 (day 17), and 15.6 vs 33.3 (day 20)
  • the TC (%) values of the combination (37.5 mg/kg irinotecan+0.135 mg/kg PM01183) vs irinotecan alone (37.5 mg/kg irinotecan) were 40.1 vs 65.0 (day 14), 39.2 vs 58.4 (day 17), and 28.7 vs 49.4 (day 20). Therefore, when PM01183 is combined with irinotecan a potentiation of the antitumor activity is clearly observed.
  • JURKAT and MOLT-4 were the human leukemia cell lines selected for this assay, which were obtained from the American Type Culture Collection (ATCC). JURKAT and MOLT-4 cells were grown in phenol red-free RPMI medium supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • the screening was performed in two parts:
  • cells were seeded in 96 well microtiter plates at a density of 50000 cells per well in 150 ⁇ L of culture medium and incubated for 4-6 hours in drug-free medium before treatment with vehicle alone or test compounds for 72 hours.
  • cytotoxic effect was evaluated using a MTT reduction assay.
  • 50 ⁇ L of MTT solution (1 mg/mL) were added to the wells and incubated for 15-17 hours at 37° C. until formazan crystals were formed.
  • DMSO was added to dissolve the insoluble purple formazan product into a colored solution.
  • the absorbance of the wells was quantified by measuring the optical density at 540 nm. Results were expressed as percentage of control cell growth.
  • the EC50 values (half-maximal effective concentration) used for the combination studies were calculated using Prism v5.02 software (GraphPad). EC50 was expressed as molar concentration and represented the mean of at least three independent assays.
  • f a /f u (C/C m ) m (where C is the drug concentration, C m the median-effect concentration (i.e., IC50, ED50, or LD50, that inhibits the system under study by 50%), f a the cell fraction affected by the drug concentration C, f u the unaffected fraction, and m the sigmoidicity coefficient of the concentration-response curve), describes the relationship between the concentration and the effect of a drug on a given biological system.
  • C the drug concentration
  • C m the median-effect concentration (i.e., IC50, ED50, or LD50, that inhibits the system under study by 50%)
  • f a the cell fraction affected by the drug concentration C
  • f u the unaffected fraction
  • m the sigmoidicity coefficient of the concentration-response curve
  • CI combination index
  • (C x ) 1 is the concentration of drug 1 alone that inhibits an x percentage of a system
  • (C x ) 2 the concentration of drug 2 alone that inhibits the same x percentage of the system
  • (C 1 )+(C) 2 the concentrations of drug 1 and drug 2 that in combination also inhibits an X percentage of the system.
  • CI values were calculated by solving the equation for different values of f a (i.e., for different degrees of cell growth inhibition). CI values of ⁇ 1 indicate synergy, the value of 1 indicates additive effects, and values >1 indicate antagonism.
  • FIGS. 273-283 The effect of the tested drug combinations on cell proliferation is shown in FIGS. 273-283 :
  • the following agents were evaluated in combination with PM01183: gemcitabine, cytarabine, methotrexate, daunorubicin, ET-743, PM02734 and PM00104 (stock solutions of these compounds prepared in pure DMSO and stored at ⁇ 20° C.). Additional serial dilutions were prepared in serum-free culture medium to achieve a final 4 ⁇ concentration. Aliquots of 50 ⁇ L of each diluted compound were added per well.
  • RAMOS and U-937 were the human lymphoma cell lines selected for this assay, which were obtained from the American Type Culture Collection (ATCC). RAMOS and U-937 cells were grown in phenol red-free RPMI medium supplemented with 10% Fetal Bovine Serum (FBS), 2 mM L-glutamine and 100 units/mL of Penicillin-Streptomycin, at 37° C., 5% CO2 and 95% humidity.
  • FBS Fetal Bovine Serum
  • Penicillin-Streptomycin 100 units/mL of Penicillin-Streptomycin
  • the screening was performed in two parts, as previously described in example 20.
  • FIGS. 284-296 The effect of the tested drug combinations on cell proliferation is shown in FIGS. 284-296 :

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Biomedical Technology (AREA)
  • Obesity (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US13/884,874 2010-11-12 2011-11-11 Combination Therapy with an Antitumor Alkaloid Abandoned US20130266666A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10382300.1 2010-11-12
EP10382300 2010-11-12
PCT/EP2011/069976 WO2012062920A1 (en) 2010-11-12 2011-11-11 Combination therapy with an antitumor alkaloid

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/069976 A-371-Of-International WO2012062920A1 (en) 2010-11-12 2011-11-11 Combination therapy with an antitumor alkaloid

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US15/711,478 Continuation US20180008602A1 (en) 2010-11-12 2017-09-21 Combination therapy with an antitumor alkaloid
US15/824,506 Continuation US20180078550A1 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid
US15/824,551 Continuation US11590129B2 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid

Publications (1)

Publication Number Publication Date
US20130266666A1 true US20130266666A1 (en) 2013-10-10

Family

ID=44936282

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/884,874 Abandoned US20130266666A1 (en) 2010-11-12 2011-11-11 Combination Therapy with an Antitumor Alkaloid
US15/711,478 Abandoned US20180008602A1 (en) 2010-11-12 2017-09-21 Combination therapy with an antitumor alkaloid
US15/824,551 Active US11590129B2 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid
US15/824,506 Abandoned US20180078550A1 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid
US18/095,877 Pending US20230404999A1 (en) 2010-11-12 2023-01-11 Combination therapy with an antitumor alkaloid

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/711,478 Abandoned US20180008602A1 (en) 2010-11-12 2017-09-21 Combination therapy with an antitumor alkaloid
US15/824,551 Active US11590129B2 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid
US15/824,506 Abandoned US20180078550A1 (en) 2010-11-12 2017-11-28 Combination therapy with an antitumor alkaloid
US18/095,877 Pending US20230404999A1 (en) 2010-11-12 2023-01-11 Combination therapy with an antitumor alkaloid

Country Status (24)

Country Link
US (5) US20130266666A1 (pt)
EP (6) EP2786753B1 (pt)
JP (4) JP6382516B2 (pt)
KR (9) KR101716804B1 (pt)
CN (3) CN106860870B (pt)
AU (4) AU2011328004C1 (pt)
BR (4) BR122017028568B1 (pt)
CA (4) CA2817420C (pt)
CY (4) CY1117466T1 (pt)
DK (4) DK2637663T3 (pt)
ES (4) ES2790414T3 (pt)
HK (5) HK1202421A1 (pt)
HR (4) HRP20160361T1 (pt)
HU (4) HUE042801T2 (pt)
LT (3) LT2786753T (pt)
ME (3) ME03408B (pt)
PL (4) PL2786754T3 (pt)
PT (3) PT2786756T (pt)
RS (4) RS60236B1 (pt)
RU (4) RU2743643C2 (pt)
SI (4) SI2786753T1 (pt)
SM (1) SMT201600112B (pt)
TR (2) TR201903859T4 (pt)
WO (1) WO2012062920A1 (pt)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120310183A1 (en) * 2009-12-02 2012-12-06 Nimble Epitech, Llc Pharmaceutical composition containing a hypomethylating agent and a histone deacetylase inhibitor
US10538535B2 (en) 2017-04-27 2020-01-21 Pharma Mar, S.A. Antitumoral compounds
US20220235422A1 (en) * 2015-01-30 2022-07-28 GLAX Health LLC Compositions and methods for monitoring, diagnosis, prognosis, detection, and treatment of cancer
US11491154B2 (en) 2013-04-08 2022-11-08 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI2786753T1 (sl) * 2010-11-12 2019-05-31 Pharma Mar S.A. Kombinacijska terapija s protitumorskim antibiotikom
KR101588949B1 (ko) * 2014-03-13 2016-01-29 아주대학교산학협력단 플루나리진을 유효성분으로 함유하는 뇌암에 대한 항암 활성 보조제
KR20210049403A (ko) * 2019-10-25 2021-05-06 연세대학교 산학협력단 암의 예방 또는 치료용 조성물
CA3158733A1 (en) * 2019-11-21 2021-05-27 Pilar Calvo Methods of treating small cell lung cancer with lurbinectedin formulations
MA56827B2 (fr) 2019-11-21 2023-09-27 Pharma Mar Sa Procédés de traitement du cancer du poumon à petites cellules avec des formulations de lurbinectédine
WO2021228414A1 (en) * 2020-05-14 2021-11-18 Pharma Mar, S.A. Methods of treating small cell lung cancer with lurbinectedin formulations
CN111298122B (zh) * 2019-12-04 2021-12-21 哈尔滨商业大学 用于治疗小细胞肺癌的药物组合物及其应用
CN113491771A (zh) * 2020-03-18 2021-10-12 苏州大学 小分子化合物在制备glut5摄取转运果糖的抑制剂中的应用
WO2021215952A1 (ru) 2020-04-24 2021-10-28 Общество С Ограниченной Ответственностью "Индженик" Способ получения частиц бактериофагов семейства levivirus
CN114470216A (zh) * 2020-10-23 2022-05-13 和记黄埔医药(上海)有限公司 多受体酪氨酸激酶抑制剂与化疗剂的药物组合及其使用方法
CN112870194B (zh) * 2021-01-06 2022-06-21 广州医科大学附属肿瘤医院 治疗肝癌的组合物及其应用
CN115246846A (zh) * 2021-11-19 2022-10-28 江苏慧聚药业股份有限公司 卢比替定新晶型及其制备
WO2023165603A1 (en) * 2022-03-03 2023-09-07 Zai Lab (Shanghai) Co., Ltd. Dna-pk inhibitor and combination use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040108086A1 (en) * 2000-11-06 2004-06-10 Naoto Takahashi Effective antitumor treatments
WO2005049031A1 (en) * 2003-11-13 2005-06-02 Pharma Mar, S.A.U. Combination

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59225189A (ja) 1983-06-03 1984-12-18 Shionogi & Co Ltd キノナミン誘導体およびその製造法
JPS6084288A (ja) 1983-10-13 1985-05-13 Shionogi & Co Ltd シアノキノナミンアセテ−ト類およびその製造法
US5256663A (en) 1986-06-09 1993-10-26 The Board Of Trustees Of The University Of Illinois Compositions comprising ecteinascidins and a method of treating herpes simplex virus infections therewith
EP0309477B1 (en) 1986-06-09 1991-11-06 The Board Of Trustees Of The University Of Illinois Ecteinascidins 729, 743, 745, 759a, 759b and 770
US5089273A (en) 1986-06-09 1992-02-18 Board Of Trustees Of The University Of Illinois Ecteinascidins 729, 743, 745, 759A, 759B and 770
US5149804A (en) 1990-11-30 1992-09-22 The Board Of Trustees Of The University Of Illinois Ecteinascidins 736 and 722
US5478932A (en) 1993-12-02 1995-12-26 The Board Of Trustees Of The University Of Illinois Ecteinascidins
US5721362A (en) 1996-09-18 1998-02-24 President And Fellows Of Harvard College Process for producing ecteinascidin compounds
US5985876A (en) 1997-04-15 1999-11-16 Univ Illinois Nucleophile substituted ecteinascidins and N-oxide ecteinascidins
EP1067933B1 (en) 1998-04-06 2007-08-15 The Board Of Trustees Of The University Of Illinois Semi-synthetic ecteinascidins
IL139607A0 (en) 1998-05-11 2002-02-10 Pharma Mar Sa Metabolites of ecteinascidin 743
US6124292A (en) 1998-09-30 2000-09-26 President And Fellows Of Harvard College Synthetic analogs of ecteinascidin-743
US7311924B2 (en) * 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
MY130271A (en) 1999-05-14 2007-06-29 Pharma Mar Sa Hemisynthetic method and new compounds
EP1254140A4 (en) 2000-01-19 2003-03-12 Univ Columbia SAFRAMYCIN-ECTEINASCIDIN SERIES COMPOUNDS, USES AND SYNTHESIS
IL152094A (en) 2000-04-12 2011-10-31 Pharma Mar Sa History of actinacidin and pharmaceutical preparations containing them as anti-cancer substances
MXPA02011319A (es) 2000-05-15 2003-06-06 Pharma Mar Sa Analogos antitumorales de ecteinascidina 743.
SE0102232L (sv) * 2001-06-25 2003-02-06 Anoto Ab Förfarande och anordning i ett digitalt kommunikationssystem
GB0119243D0 (en) 2001-08-07 2001-10-03 Pharma Mar Sa Antitumoral analogs of ET-743
AU2003250483A1 (en) 2002-08-19 2004-03-11 Pfizer Products Inc. Combination therapy for hyperproliferative diseases
ES2357644T3 (es) 2002-10-18 2011-04-28 Pharma Mar, S.A.U. Compuesto 4-metilhexanoico kahalalido f.
FR2856688B1 (fr) * 2003-06-25 2008-05-30 Sod Conseils Rech Applic PRODUIT COMPRENANT AU MOINS UN INHIBITEUR DE PHOSPHATASE CDc25 EN ASSOCIATION AVEC AU MOINS UN AUTRE AGENT ANTI-CANCEREUX
US7393277B2 (en) * 2003-08-25 2008-07-01 Igt Horseshoe payline system and games using that system
GB0326486D0 (en) * 2003-11-14 2003-12-17 Pharma Mar Sau Combination treatment
CN1897949A (zh) * 2003-11-14 2007-01-17 马尔药品公司 包含使用et-743和紫杉醇来治疗癌症的联合疗法
MXPA06013902A (es) * 2004-05-28 2007-01-26 Pfizer Prod Inc Procedimiento para tratar el crecimiento de celulas anormal.
DK1658848T3 (da) 2004-10-29 2007-11-26 Pharma Mar Sa Formuleringer omfattende ecteinascidin og et disaccharid
CA2589521C (en) 2004-12-15 2014-04-01 Novartis Ag Combinations of 7-t-butoxyiminomethylcamptothecin with one or more chemotherapeutic agents for treating cancers
TW200744603A (en) * 2005-08-22 2007-12-16 Chugai Pharmaceutical Co Ltd Novel anticancer concomitant drug
EP2029155B1 (en) * 2006-02-28 2016-04-13 Pharma Mar S.A. Improved treatment of multiple myeloma
KR20110025178A (ko) * 2008-05-16 2011-03-09 파르마 마르 에스.에이. Pm00104 및 다른 항종양제를 이용한 복합 치료법
SI2786753T1 (sl) * 2010-11-12 2019-05-31 Pharma Mar S.A. Kombinacijska terapija s protitumorskim antibiotikom

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040108086A1 (en) * 2000-11-06 2004-06-10 Naoto Takahashi Effective antitumor treatments
WO2005049031A1 (en) * 2003-11-13 2005-06-02 Pharma Mar, S.A.U. Combination

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Leal et al. British Journal of Pharmacology, Published online on July 2, 2010, 161, pages 1099-1110. *
Messersmith et al. Cancer Chemother. Pharmacol., Volume 63, 2008, pages 181-188. *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120310183A1 (en) * 2009-12-02 2012-12-06 Nimble Epitech, Llc Pharmaceutical composition containing a hypomethylating agent and a histone deacetylase inhibitor
US10434090B2 (en) * 2009-12-02 2019-10-08 Nimble Epitech, Llc Pharmaceutical composition containing a hypomethylating agent and a histone deacetylase inhibitor
US11491154B2 (en) 2013-04-08 2022-11-08 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds
US20220235422A1 (en) * 2015-01-30 2022-07-28 GLAX Health LLC Compositions and methods for monitoring, diagnosis, prognosis, detection, and treatment of cancer
US20220243282A1 (en) * 2015-01-30 2022-08-04 GLAX Health LLC. Compositions and Methods for Monitoring, Diagnosis, Detection and Treatment of Cancer
US10538535B2 (en) 2017-04-27 2020-01-21 Pharma Mar, S.A. Antitumoral compounds
US11332480B2 (en) 2017-04-27 2022-05-17 Pharma Mar, S.A. Antitumoral compounds
US11339180B2 (en) 2017-04-27 2022-05-24 Pharma Mar, S.A. Antitumoral compounds
US11713325B2 (en) 2017-04-27 2023-08-01 Pharma Mar, S.A. Antitumoral compounds

Also Published As

Publication number Publication date
RU2018102080A3 (pt) 2021-04-12
CA2995025C (en) 2020-04-28
KR102452022B1 (ko) 2022-10-06
WO2012062920A1 (en) 2012-05-18
AU2011328004C1 (en) 2017-01-19
EP2786754A3 (en) 2014-10-29
ME03501B (me) 2020-04-20
AU2016200191A1 (en) 2016-02-04
AU2017276157A1 (en) 2018-01-04
AU2016200191B2 (en) 2018-01-18
RS58608B1 (sr) 2019-05-31
HK1188572A1 (zh) 2014-05-09
EP2786755A3 (en) 2014-10-29
EP3666275A3 (en) 2020-09-09
HK1202421A1 (en) 2015-10-02
DK2786754T3 (en) 2019-04-15
PL2637663T3 (pl) 2016-08-31
KR102301175B1 (ko) 2021-09-10
ES2790414T3 (es) 2020-10-27
US20230404999A1 (en) 2023-12-21
RU2018102078A (ru) 2019-07-19
KR101716804B1 (ko) 2017-03-15
AU2011328004B2 (en) 2016-07-14
EP2786755A2 (en) 2014-10-08
KR20210046853A (ko) 2021-04-28
EP2786756B1 (en) 2020-03-11
EP2786753A2 (en) 2014-10-08
KR20130140071A (ko) 2013-12-23
SI2786756T1 (sl) 2020-07-31
RU2605335C2 (ru) 2016-12-20
HRP20190554T1 (hr) 2019-05-03
EP2786753B1 (en) 2019-01-09
ME02395B (me) 2016-09-20
TR201903859T4 (tr) 2019-04-22
KR20170124641A (ko) 2017-11-10
RU2757373C2 (ru) 2021-10-14
ME03408B (me) 2020-01-20
SMT201600112B (it) 2016-07-01
CA2995018C (en) 2021-04-20
HUE049389T2 (hu) 2020-09-28
EP2786756A2 (en) 2014-10-08
HUE027516T2 (en) 2016-10-28
CY1121501T1 (el) 2020-05-29
EP2786754B1 (en) 2019-01-09
PT2786756T (pt) 2020-05-07
RS60236B1 (sr) 2020-06-30
CA2817420C (en) 2019-09-24
EP2786754A2 (en) 2014-10-08
HK1202419A1 (en) 2015-10-02
HK1202422A1 (en) 2015-10-02
CN105664165A (zh) 2016-06-15
AU2017276155A1 (en) 2018-01-04
RU2016144668A3 (pt) 2020-03-25
ES2719052T3 (es) 2019-07-08
SI2637663T1 (sl) 2016-05-31
JP6382516B2 (ja) 2018-08-29
DK2786756T3 (en) 2020-04-27
CN106860870A (zh) 2017-06-20
KR20170057472A (ko) 2017-05-24
JP2018100303A (ja) 2018-06-28
JP2016138133A (ja) 2016-08-04
RU2018102080A (ru) 2019-07-19
US20180078550A1 (en) 2018-03-22
BR122017028568B1 (pt) 2022-03-03
EP2637663A1 (en) 2013-09-18
CN103282037B (zh) 2017-04-05
BR112013011480B1 (pt) 2022-03-22
CY1122904T1 (el) 2021-10-29
RS54648B1 (en) 2016-08-31
SI2786754T1 (sl) 2019-05-31
JP6723815B2 (ja) 2020-07-15
JP6724055B2 (ja) 2020-07-15
LT2786756T (lt) 2020-05-11
LT2786753T (lt) 2019-04-10
RU2767664C2 (ru) 2022-03-18
HRP20160361T1 (hr) 2016-05-06
CY1117466T1 (el) 2017-04-26
AU2017276157B2 (en) 2019-03-07
HRP20200696T1 (hr) 2020-07-24
JP6724056B2 (ja) 2020-07-15
HK1202423A1 (en) 2015-10-02
CA2995018A1 (en) 2012-05-18
PT2786753T (pt) 2019-04-24
CN103282037A (zh) 2013-09-04
JP2013542241A (ja) 2013-11-21
HUE042802T2 (hu) 2019-07-29
KR102247982B1 (ko) 2021-05-04
PL2786756T3 (pl) 2020-07-13
BR112013011480A2 (pt) 2016-08-09
HUE042801T2 (hu) 2019-07-29
AU2017276155B2 (en) 2019-03-07
KR20170058454A (ko) 2017-05-26
BR122017028570B1 (pt) 2022-03-03
PT2786754T (pt) 2019-04-24
EP2637663B1 (en) 2016-02-24
AU2011328004A1 (en) 2013-05-30
CA2995033A1 (en) 2012-05-18
CA2995025A1 (en) 2012-05-18
CY1121504T1 (el) 2020-05-29
EP3666275A2 (en) 2020-06-17
ES2719091T3 (es) 2019-07-08
CN106860870B (zh) 2020-03-31
RS58609B1 (sr) 2019-05-31
HRP20190565T1 (hr) 2019-05-17
BR112013011480A8 (pt) 2018-01-02
TR201904459T4 (tr) 2019-05-21
PL2786753T3 (pl) 2019-09-30
KR20160088956A (ko) 2016-07-26
KR20190130078A (ko) 2019-11-20
DK2637663T3 (en) 2016-04-18
EP2786756A3 (en) 2014-10-29
SI2786753T1 (sl) 2019-05-31
PL2786754T3 (pl) 2019-07-31
KR20170096224A (ko) 2017-08-23
RU2016144668A (ru) 2018-12-18
KR20170096065A (ko) 2017-08-23
LT2786754T (lt) 2019-04-10
RU2013126630A (ru) 2014-12-20
US11590129B2 (en) 2023-02-28
US20180078551A1 (en) 2018-03-22
ES2569180T3 (es) 2016-05-09
BR122017028566B1 (pt) 2022-03-03
DK2786753T3 (en) 2019-04-15
JP2018100302A (ja) 2018-06-28
CA2995033C (en) 2020-04-28
RU2018102078A3 (pt) 2021-04-20
RU2743643C2 (ru) 2021-02-20
EP2786753A3 (en) 2014-10-29
CA2817420A1 (en) 2012-05-18
US20180008602A1 (en) 2018-01-11

Similar Documents

Publication Publication Date Title
US20230404999A1 (en) Combination therapy with an antitumor alkaloid

Legal Events

Date Code Title Description
AS Assignment

Owner name: PHARMA MAR, S.A., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MONEO OCANA, VICTORIA;SANTAMARIA NUNEZ, GEMA;GARCIA FERNANDEZ, LUIS FRANCISCO;AND OTHERS;REEL/FRAME:030476/0696

Effective date: 20130425

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

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