US20170333341A1 - Nanoparticles loaded with chemotherapeutic antitumoral drug - Google Patents

Nanoparticles loaded with chemotherapeutic antitumoral drug Download PDF

Info

Publication number
US20170333341A1
US20170333341A1 US15/673,661 US201715673661A US2017333341A1 US 20170333341 A1 US20170333341 A1 US 20170333341A1 US 201715673661 A US201715673661 A US 201715673661A US 2017333341 A1 US2017333341 A1 US 2017333341A1
Authority
US
United States
Prior art keywords
nanoparticles
doxorubicin
cancer
infusion
group
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
US15/673,661
Inventor
Emilia Pisani
Sophie Lebel-Binay
Valérie Polard
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.)
Valerio Therapeutics SA
Original Assignee
Onxeo 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 Onxeo SA filed Critical Onxeo SA
Priority to US15/673,661 priority Critical patent/US20170333341A1/en
Assigned to BIOALLIANCE PHARMA SA reassignment BIOALLIANCE PHARMA SA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEBEL-BINAY, SOPHIE, POLARD, Valérie, PISANI, Emilia
Assigned to ONXEO S.A. reassignment ONXEO S.A. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BIOALLIANCE PHARMA S.A.
Publication of US20170333341A1 publication Critical patent/US20170333341A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • A61K47/6951Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes using cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5138Organic macromolecular compounds; Dendrimers obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82BNANOSTRUCTURES FORMED BY MANIPULATION OF INDIVIDUAL ATOMS, MOLECULES, OR LIMITED COLLECTIONS OF ATOMS OR MOLECULES AS DISCRETE UNITS; MANUFACTURE OR TREATMENT THEREOF
    • B82B3/00Manufacture or treatment of nanostructures by manipulation of individual atoms or molecules, or limited collections of atoms or molecules as discrete units
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions

Definitions

  • the invention relates to the field of medicine, and in particular of oncology. It relates to a treatment of cancer, in particular hepatocellular carcinoma, with Nanoparticles loaded with at least one chemotherapeutic antitumoral drug.
  • Cancer is characterized by uncontrolled growth of cells coupled with malignant behavior: invasion and metastasis. It is a major cause of mortality in most industrialized countries. Different ways of cancer treatment can be used: chemotherapy, radiotherapy, surgery, immunotherapy and hormonotherapy.
  • Chemotherapy can be defined as the use of chemotherapeutic antitumoral agents to treat cancer. Broadly, most chemotherapeutic antitumoral agents work by impairing mitosis (cell division) or DNA synthesis, effectively targeting fast-dividing cells.
  • Chemotherapeutic antitumoral agents are delivered most often parenterally, noteworthy intravenously (IV) or intra-arterially (IA). IV or IA chemotherapy can be given over different amounts of time, depending on the drug and the type of cancer to be treated.
  • IV or IA chemotherapy can be given over different amounts of time, depending on the drug and the type of cancer to be treated.
  • Hepatocellular carcinoma is the fifth most common cancer in men (523,000 cases worldwide) and the seventh in women (226,000 cases worldwide), and most of the burden is in developing countries, where almost 85% of the cases occur, and particularly in men: the overall male/female ratio is about 4/1.
  • the regions of high incidence are Eastern and South-Eastern Asia, Middle and Western Africa, but also Melanesia and Micronesia/Polynesia. Low rates are estimated in developed regions, with the exception of Southern Europe where the incidence in men (ASR 10.5 per 100,000) is significantly higher than in other developed regions (Globocan 2008, WHO, International Agency for Research on Cancer-IARC-, Cancer Incidence and Mortality Worldwide 2008).
  • liver cancer is the third most common cause of death from cancer worldwide.
  • the geographical distribution of the mortality rates is similar to that observed for incidence (Globocan 2008, WHO, International Agency for Research on Cancer-IARC-, Cancer Incidence and Mortality Worldwide 2008).
  • HCC usually occurs in people suffering from cirrhosis or chronic liver disease (CLD). All factors favoring the development of CLD or cirrhosis are consequently risk factors for HCC.
  • the main etiologic factors are hepatitis B virus infection (HBV; 53%), hepatitis C virus infection (HCV; 25%), alcoholic liver diseases (ALD; 15-43%) or dysmetabolic disorders such as Non Alcoholic Steato-Hepatitis (NASH; 20%) obesity and diabetes.
  • HBV hepatitis B virus infection
  • HCV hepatitis C virus infection
  • ALD alcoholic liver diseases
  • NASH Non Alcoholic Steato-Hepatitis
  • Other factors are less frequent: haemochromatosis, other chronic biliary or inflammatory liver diseases.
  • Aflatoxins produced by the fungi Aspergillus flavus and Aspergillus parasiticus grown on grains, peanuts, and other food products, are hepatotoxic agents and chronic exposure to these mycotoxins leads to development of HCC. The effects of chemicals are still debated and not prominent.
  • TACE chemoembolization
  • sorafenib is the standard of care for advanced stage HCC and is registered for the treatment of HCC without restrictions by the European Medicines Agency (EMA) and the Food and Drug Administration (FDA), the narrow inclusion criteria of the clinical trials leave many patients without proven efficacious treatment with regard to their disease stage. Moreover, since treatment failure happens in some patients on sorafenib, there still remains a medical need for those patients to improve treatment efficacy, drug regimen and overall tolerance, and to overcome resistance (M. Peck-Radosavljevic, Therap. Adv. Gastroenterol. 2010, 3(4), 259).
  • HCC hypervascular solid cancer characterized by a high degree of drug resistance (T. Wakamatsu, Y. Nakahashi, D. Hachimine, T. Seki, K. Okazaki, Int. J. Oncol., 2007, 31(6), 1465).
  • MDR multidrug resistance
  • Chemoresistance related to the MDR phenotype may be intrinsic or be acquired during chemotherapy. Chemoresistance, whether spontaneous or acquired is a serious concern in cancer treatment. HCC is often intrinsically chemoresistant which is the major cause for failure of its therapy (R. Pérez-Tomas, Curr Med Chem., 2006, 13(16), 1859, Review). This poses a great obstacle in chemotherapy for cancer because higher doses of drugs need to be administered and in turn may cause severe adverse effects (F. Yan, X. M. Wang, Z. C. Liu, C. Pan, S.
  • Chemoresistance affects major chemotherapeutic agents and especially anthracyclins (like doxorubicin), vinca-alkaloids, epipodophyllotoxins or taxanes.
  • anthracyclins like doxorubicin
  • vinca-alkaloids like doxorubicin
  • epipodophyllotoxins or taxanes.
  • the poor efficacy of chemotherapeutic agents attributed to the overexpression of the MDR gene underlines the need to develop new treatment strategies for HCC, which could take into account the resistance issues.
  • Doxorubicin is a chemotherapeutic compound, efficacy of which has been shown in several cancers including HCC.
  • IV infusion of doxorubicin in HCC is modest with objective response rate of 5-10%.
  • a large, randomized, multicenter clinical trial compared the efficacy of doxorubicin 60 mg/m2 administered through the IV route and thymitaq a direct thymidilate synthase inhibitor (Porta C., 2006).
  • the effect of doxorubicin was found modest.
  • the modest effect of doxorubicin in HCC patients is assumed to result from multidrug resistance (MDR) mechanisms related to overactivity of PgP and MRP cellular pumps.
  • MDR multidrug resistance
  • Many strategies have been evaluated to overcome the resistance issues, including the use of Pgp and MRP inhibitors. The development of these drugs has been stopped due to their safety profile.
  • cytotoxic agents were developed by hepatic intra-arterial (IA) injection in order to reduce the systemic toxicity, to induce important hepatic tumor necrosis and to save the healthy hepatic parenchyma.
  • IA hepatic intra-arterial
  • Nanoparticles comprising a pharmaceutically active ingredient, a polymer such as poly(alkylcyanoacrylate) and a complexing agent such cyclodextrins, are thus taught in EP1056477, and in its US equivalent U.S. Pat. No. 6,881,421.
  • Nanoparticles loaded with Doxorubicin is a drug formulation that associates a kind of PACA, polyisohexylcyanoacrylate (PIHCA), Nanoparticles with the chemotherapeutic agent doxorubicin.
  • Nanoparticles display original mechanisms to bypass MDR that can be summarized as follows:
  • Nanoparticles loaded with Doxorubicin adsorbs to the surface of tumor cells and releases the entrapped doxorubicin close to the cell membrane which leads to a high local gradient concentration (Colin de Verdiére A, Cancer Chemother. Pharmacol. 1994; 33(6):504-8).
  • Nanoparticles degrade and release soluble polycyanoacrylic acid which might interact with the plasma membrane and contribute to improve the intracellular delivering of doxorubicin (De Verdiére A C, Br J Cancer. 1997; 76(2):198-205).
  • the soluble polymer could also mask the positive charge of doxorubicin thus preventing its effluxing by the Pgp (De Verdiére 1997, Br J Cancer. 1997; 76(2):198-205), acting as an ion pair without any covalent linkage).
  • Pgp De Verdiére 1997, Br J Cancer. 1997; 76(2):198-205
  • MRP MDR pumps
  • the study BA2002/03/02 phase I/II study in patients with advanced Hepatocellular Carcinoma was carried out according to a multicenter, open, dose-escalation design in patients suffering from HCC.
  • Nanoparticles loaded with Doxorubicin was to be injected through hepatic intra-arterial (IA) route as a bolus.
  • IA hepatic intra-arterial
  • Successive cohorts of 3 patients were injected a single 10, 20 and 30 mg/m 2 dose of Nanoparticles loaded with Doxorubicin. As the 30 mg/m 2 dose was well tolerated, the protocol was amended to assess 40 mg/m 2 and then 35 mg/m 2 .
  • TEAEs Treatment Emergent Adverse Events
  • tolerance was acceptable; most of the TEAEs were short lasting and of mild severity. All were reversible without sequellae.
  • 50% of the non-serious TEAEs were expected as already reported with free doxorubicin.
  • Nanoparticles loaded with Doxorubicin was injected through IA route as a 15-minute infusion at the dose of 30 mg/m 2 , preceded and followed by an oral premedication with methylprednisone. Fifty patients were to be included in the 1st part of the study. Three Nanoparticles loaded with Doxorubicin infusions were to be received by 33 patients at 4-week intervals and each of the other 17 patients were to receive the best standard of care treatment adapted to the severity of the disease. At the end of this 1st phase, if Nanoparticles loaded with Doxorubicin was considered active in 2 ⁇ 3 of patients (patients free of local progression at 3 months), then 150 additional patients were to be enrolled.
  • Nanoparticles loaded with chemotherapeutic drug such as Nanoparticles loaded with Doxorubicin
  • cancer treatment and in particular HCC treatment
  • New approaches allowing safer use, reducing the probability of occurrence of pulmonary adverse events and their severity under acceptable limits owing to the benefit/risk ratio are then needed.
  • the inventors succeeded in obtaining a suitable animal model for the severe pulmonary adverse events and, then, have surprisingly found that severe toxicological side effects, in particular lung injuries, associated with intra-arterial or intravenous administration of Nanoparticles loaded with a chemotherapeutic agent, can be prevented by intravenous or intra-arterial infusion of said Nanoparticles for several hours.
  • the present invention concerns Nanoparticles comprising at least one chemotherapeutic antitumoral agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin, for use for treating a cancer, wherein the Nanoparticles are administered by intravenous or intra-arterial infusion for at least 2 hours.
  • Nanoparticles can be administered by intravenous or intra-arterial infusion for between 2 and 24 hours, more particularly for between 4 and 12 hours, even more particularly for about 6 hours.
  • the administration is by intravenous infusion.
  • the poly(alkylcyanoacrylate) polymer comprised in the Nanoparticles is a polyisohexylcyanoacrylate polymer.
  • the Nanoparticles used in the invention may comprise said at least one chemotherapeutic agent at a concentration from 0.01 to 200 mg/g of Nanoparticles, from 0.1 to 70% w/w of said at least one cyclodextrin and from 1 to 25% w/w of said at least one poly(alkylcyanoacrylate).
  • the chemotherapeutic antitumoral agent comprised in the Nanoparticles may be selected from the group consisting of anthracyclines, topoisomerase inhibitors, spindle poison plant alkaloids, alkylating agents, anti-metabolites, ellipticine and harmine, and any combination thereof.
  • the chemotherapeutic antitumoral agent is an anthracycline.
  • the chemotherapeutic agent is selected from the group consisting of doxorubicin, daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, any pharmaceutically acceptable salt thereof, and any combination thereof.
  • the chemotherapeutic antitumoral agent is doxorubicin or any pharmaceutically acceptable salt thereof.
  • the dosage of doxorubicin may be from about 10 to about 75 mg/m 2 , from about 10 to about 60 mg/m 2 , from about 10 to about 45 mg/m 2 , from about 10 to about 30 mg/m 2 , from about 20 to about 30 mg/m 2 . In a particular embodiment, the dosage of doxorubicin may be about 20 mg/m 2 or 30 mg/m 2 .
  • the cancer treated with the Nanoparticles may be is a solid tumor or a hematopoietic tumor, preferably selected from the group consisting of hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, small cell lung cancer, colorectal cancer, pancreas cancer, breast cancer, ovary cancer, uterine cancer, cervix cancer, head and neck cancer, brain cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma.
  • the cancer is a hepatocellular carcinoma.
  • the present invention concerns a method for treating cancer, wherein the method comprises administering by intravenous or intra-arterial infusion for at least 2 hours, a therapeutically effective amount of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin to a patient in need of such treatment.
  • Nanoparticles are administered in the form of a pharmaceutical composition as described above. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect.
  • the administration is by intravenous infusion.
  • the present invention concerns the use of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin for the treatment of cancer, said Nanoparticles being administered by intravenous or intra-arterial infusion for at least 2 hours. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect. In a particular embodiment, the administration is by intravenous infusion.
  • the present invention concerns the use of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin, for the manufacture of a pharmaceutical preparation for the treatment of cancer, said pharmaceutical composition being administered by intravenous or intra-arterial infusion for at least 2 hours. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect. In a particular embodiment, the administration is by intravenous infusion.
  • Nanoparticles loaded with Doxorubicin As preliminary work to the present invention, the inventors have established and assessed a rat model to investigate respiratory adverse events observed in previous clinical trials for treatment of hepatocellular carcinoma by hepatic intra-arterial infusion of doxorubicin formulated into Nanoparticles comprising a poly(alkylcyanoacrylate) polymer and a cyclodextrin (hereinafter referred to as “Nanoparticles loaded with Doxorubicin”). Using this model, they have herein demonstrated that intravenous infusion of said Nanoparticles for at least 2 hours allows to strongly reduce toxicological side effects, in particular lung injuries, compared to bolus intravenous injection at the same dose. This new therapeutic approach allows safer use of Nanoparticles and then dramatically increases the benefit/risk ratio.
  • the present invention concerns Nanoparticles comprising at least one chemotherapeutic antitumoral agent for use for treating a cancer, wherein the Nanoparticles are to be administered by intravenous or intra-arterial infusion for at least 2 hours.
  • the Nanoparticles used in the present invention comprise at least one chemotherapeutic antitumoral agent, at least one poly(alkylcyanoacrylate) polymer, and at least one compound able to form a complex with the chemotherapeutic antitumoral agent, preferably chosen from among the cyclical oligosaccharides, in particular from among the cyclodextrins.
  • chemotherapeutic antitumoral agent preferably chosen from among the cyclical oligosaccharides, in particular from among the cyclodextrins.
  • the poly(alkylcyanoacrylate) polymer may be linear or branched, preferably branched.
  • the alkyl group of the poly(alkylcyanoacrylate) may be linear or branched, preferably branched.
  • the poly(alkylcyanoacrylate) polymer is a poly(C 1 -C 12 alkylcyanoacrylate), preferably a poly(C 4 -C 10 alkylcyanoacrylate), more preferably a poly(C 6 -C 8 alkylcyanoacrylate).
  • the poly(alkylcyanoacrylate) polymer is a polyisohexylcyanoacrylate.
  • the monomer corresponding to the latter polymer is available, for instance under the trademark Monorex® by Bioalliance Pharma (France).
  • the cyclodextrin may be neutral or charged, native (cyclodextrins ⁇ , ⁇ , ⁇ , ⁇ , ⁇ ), branched or polymerized, or even chemically modified, for example, by substitution of one or more hydroxypropyls by groups such as alkyls, aryls, arylalkyls, glycosidics, or by etherification, esterification with alcohols or aliphatic acids.
  • groups particular preference is given to those chosen from the group consisting of hydroxypropyl, methyl-m and sulfobutylether groups, and mixtures thereof.
  • the cyclodextrin is selected from the group consisting of hydroxypropyl-beta-cyclodextrin and/or randomly methylated-beta cyclodextrin, and mixtures thereof.
  • chemotherapeutic agent or “chemotherapeutic antitumoral agent” refers to any chemical compound or drug with anti-cancer activity that inhibits or halts the growth of cancerous cells or immature pre-cancerous cells, kills cancerous cells or immature pre-cancerous cells, increases the susceptibility of cancerous or pre-cancerous cells to other chemotherapeutic agents, and/or inhibits metastasis of cancerous cells.
  • the chemotherapeutic agent is selected from the group consisting of anthracyclines, topoisomerase I and/or II inhibitors, spindle poison plant alkaloids, alkylating agents, anti-metabolites, ellipticine and harmine.
  • Anthracyclines are derived from Streptomyces bacteria. These compounds are used to treat a wide range of cancers, including for example hepatocellular carcinoma, leukemias, lymphomas, and breast, uterine, ovarian, and lung cancers.
  • Anthracyclines have three mechanisms of action: (i) inhibition of DNA and RNA synthesis by intercalating between base pairs of the DNA/RNA strand, thus preventing the replication of rapidly-growing cancer cells; (ii) inhibition of topoisomerase II enzyme, preventing the relaxing of supercoiled DNA and thus blocking DNA transcription and replication; and (iii) creation of iron-mediated free oxygen radicals that damage the DNA and cell membranes.
  • Anthracyclines include, but are not limited to doxorubicin (also named adriamycin), daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, and their pharmaceutically acceptable salts thereof.
  • doxorubicin also named adriamycin
  • daunorubicin also named adriamycin
  • epirubicin idarubicin
  • valrubicin pirarubicin
  • zorubicin aclarubicin
  • detorubicin carminomycin
  • morpholinodoxorubicin morpholinodaunorubicin
  • methoxymorpholinyldoxorubicin methoxy
  • the anthracycline is selected from the group consisting of doxorubicin, daunorubicin, epirubicin, pirarubicin, zorubicin and aclarubicin, and any pharmaceutically acceptable salt thereof.
  • the anthracycline is doxorubicin or any pharmaceutically acceptable salt thereof.
  • Topoisomerases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisomerases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling.
  • Some type I topoisomerase inhibitors include camptothecins derivatives Camptothecin derivatives refer to camptothecin analogs such as irinotecan, topotecan, hexatecan, silatecan, lutortecan, karenitecin (BNP1350), gimatecan (ST1481), belotecan (CKD602), or their pharmaceutically acceptable salts.
  • topoisomerase inhibitors examples include, but are not limited to, amsacrine, etoposide, etoposide phosphate and teniposide They are semisynthetic derivatives of epipodophyllotoxins, alkaloids naturally occurring in the root of American Mayapple ( Podophyllum peltatum ).
  • the topoisomerase inhibitor is selected from the group consisting of irinotecan, its active metabolite SN38 and topotecan, and any pharmaceutically acceptable salt thereof.
  • the topoisomerase inhibitor is irinotecan.
  • Taxanes include, but are not limited to, paclitaxel, docetaxel, larotaxel, cabazitaxel, ortataxel, tesetaxel, and their pharmaceutically acceptable salts.
  • the taxane is selected from the group consisting of paclitaxel and docetaxel, and any pharmaceutically acceptable salt thereof.
  • Paclitaxel was originally derived from the Pacific yew tree.
  • Docetaxel is a semi-synthetic analogue of paclitaxel.
  • the vinca alkaloids destroy mitotic spindles. Both taxanes and vinca alkaloids are therefore named spindle poisons or mitosis poisons, but they act in different ways.
  • Alkylating agents are so named because of their ability to add alkyl groups to many electronegative groups under conditions present in cells. They impair cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules. Noteworthy, their cytotoxicity is thought to result from inhibition of DNA synthesis.
  • Alkylating agents include, but are not limited to, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and platinum compounds such as oxaliplatin, cisplatin or carboplatin.
  • An anti-metabolite is a chemical that inhibits the use of a metabolite, which is part of normal metabolism. Such substances are often similar in structure to the metabolite that they interfere with. The presence of anti-metabolites halters cell growth and cell division.
  • Purine or pyrimidine analogues prevent the incorporation of nucleotides into DNA, stopping DNA synthesis and thus cell divisions. They also affect RNA synthesis.
  • purine analogues include azathioprine, mercaptopurine, thioguanine, fludarabine, pentostatin and cladribine.
  • pyrimidine analogues include 5-fluorouracil (5FU), which inhibits thymidylate synthase, floxuridine (FUDR) and cytosine arabinoside (Cytarabine).
  • Antifolates are drugs which impair the function of folic acids. Many are used in cancer chemotherapy, some are used as antibiotics or antiprotozoal agents. A well known example is Methotrexate. This is a folic acid analogue, and owing to structural similarity with it binds and inhibits the enzyme dihydrofolate reductase (DHFR), and thus prevents the formation of tetrahydrofolate. Tetrahydrofolate is essential for purine and pyrimidine synthesis, and this leads to inhibited production of DNA, RNA and proteins (as tetrahydrofolate is also involved in the synthesis of amino acids serine and methionine). Other antifolates include, but are not limited to, trimethoprim, raltitrexed, pyrimethamine and pemetrexed.
  • chemotherapeutic agents examples are not limiting and other agents can be loaded in Nanoparticles. Among others, ellipticine and harmine can be cited.
  • Ellipticine is a natural plant alkaloid product which was isolated from the evergreen tree of the Apocynaceae family. Ellipticine was found to have cytotoxic and anticancer activity (Dalton et al., Aust. J. Chem., 1967. 20, 2715). The ellipticine derivative hydroxylated in position 9 (9-hydroxyellipticinium) was found to have greater antitumoural activity than ellipticine on many experimental tumours (Le Pecq et al., Proc. Natl. Acad, Sci., USA, 1974, 71, 5078-5082).
  • ellipticine derivatives appropriate for human therapeutics and lead to the preparation of Celiptium, or N2-methyl-9-hydroxyellipticinium (NMHE), which has been used for the treatment of some human cancers, in particular for the treatment of bone metastasis of breast cancers.
  • NMHE N2-methyl-9-hydroxyellipticinium
  • Other 9-hydroxy ellipticine derivatives such as 2-(diethyiamino-2-ethyl)9-hydroxyellipticinium acetate, 2-(diisopropylamino-ethyl)9-hydroxy-ellipticinium acetate and 2-(beta piperidino-2-ethyl)9-hydroxyellipticinium, had been described for instance in the U.S. Pat. No. 4,310,667.
  • Harmine is a natural plant alkaloid product which was isolated from the Peganum harmala seeds.
  • Peganum harmala (Zygophyllaceae) is a plant widely distributed in semi arid rangelands in the Central Asia, North Africa, Middle East and Australia.
  • the pharmacologically active compounds of P. harmala are several alkaloids that are found especially in the seeds and the roots. These include 3-carbolines such as harmine, harmaline, harmol, harmalol and harman, and quinazoline derivatives: vasicine and vasicinone.
  • Peganum harmala alkaloids were found to possess significant antitumor potential (Lamchouri and al., Therapie, 1999, 54(6):753-8).
  • the chemotherapeutic agent is an anthracycline, preferably selected from the group consisting of doxorubicin, daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, any pharmaceutically acceptable salt thereof, and any combination thereof, more preferably doxorubicin or any pharmaceutically acceptable salt thereof.
  • an anthracycline preferably selected from the group consisting of doxorubicin, daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubi
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, tartaric, citric, methanesulfonic, benzenesulfonic, glucoronic, glutamic, benzoic, salicylic, toluenesulfonic, oxalic, fumaric, maleic, lactic and the like.
  • Further addition salts include ammonium salts such as tromethamine, meglumine or epolamine, metal salts such as sodium, potassium, calcium, zinc or magnesium.
  • a suitable salt of doxorubicin is the doxorubicin hydrochloride.
  • the Nanoparticles used in the invention comprise at least one chemotherapeutic agent, preferably selected from among anthracyclines, at least one poly(C 1 -C 12 alkylcyanoacrylate), preferably a polyisohexylcyanoacrylate, and at least one cyclodextrin, preferably selected from the group consisting of hydroxypropyl-beta-cyclodextrin and randomly methylated-beta cyclodextrin, and mixtures thereof.
  • the Nanoparticles comprise doxorubicin, a polyisohexylcyanoacrylate and a hydroxypropyl-beta-cyclo dextrin.
  • the chemotherapeutic agent is generally present at a concentration from about 0.01 to about 200 mg/g of Nanoparticles, preferably from about 1 to about 50 mg/g.
  • the proportion of cyclodextrin is in general from about 0.1 to about 70% by weight of Nanoparticles, preferably from about 1 to about 30%, more preferably from about 5 to about 20%.
  • the proportion of the chemotherapeutic agent and the proportion of cyclodextrin are independent from one another.
  • the proportion of poly(alkylcyanoacrylate) polymer is in general from about 1 to about 25% by weight of Nanoparticles, preferably from about 5 to about 15%.
  • the Nanoparticles comprise a chemotherapeutic agent at a concentration from 0.01 to 200 mg/g of Nanoparticles, from 0.1 to 70% w/w of cyclodextrin and from 1 to 25% w/w of poly(alkylcyanoacrylate), preferably of polyisohexylcyanoacrylate.
  • the term “about” refers to a range of values ⁇ 10% of the specified value. For instance, “about 1” means from 0.9 to 1.1 when 10% is considered and from 0.95 to 1.05 when 5% is considered. Where “about” is used in connection with numeric ranges, for example “about 1 to about 3”, or “between about one and about three”, preferably the definition of “about” given above for a number is applied to each number defining the start and the end of a range separately. Preferably, where “about” is used in connection with any numerical values, the “about” can be deleted.
  • Nanoparticles used in the present invention may be prepared according any method known by the skilled person. Such a method is disclosed, for example, in the European patent EP 1 056 477 and its US equivalent U.S. Pat. No. 6,881,421.
  • Nanoparticles may be prepared by a method comprising the steps of
  • the polymerization is anionic but may also be inducible by other agents, in particular by photochemical agents.
  • the polymerization is conducted in presence of a surfactant agent such as poloxamer or dextran (such as dextran 70,000) or other non ionic surfactive agents (like polysorbate, sorbitan esters or others).
  • Poloxamers are preferred, such as poloxamer 407, poloxamer 401, poloxamer 237, poloxamer 338, poloxamer 331, poloxamer 231, or poloxamer 188 (also named Pluronic® F68). Poloxamer 188 is more preferred.
  • the size of the Nanoparticles is from about 50 to about 300 nm, preferably from about 100 to about 300 nm, more preferably about 200 nm.
  • the size of these Nanoparticles is essentially related to the concentration of the cyclodextrin.
  • Nanoparticles as described above are administered in the form of a pharmaceutical composition comprising said Nanoparticles and at least one pharmaceutically acceptable excipient.
  • the term “pharmaceutically acceptable” refers to those compounds, materials, excipients, compositions or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response or other problem complications commensurate with a reasonable benefit/risk ratio.
  • the pharmaceutical composition comprising said Nanoparticles is formulated in accordance with standard pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York) known by a person skilled in the art.
  • possible pharmaceutical compositions include those suitable for intravenous, intra-arterial and intra-tumoral administration.
  • conventional excipient can be used according to techniques well known by those skilled in the art.
  • compositions for parenteral administration are generally physiologically compatible sterile solutions or suspensions which can optionally be prepared immediately before use from solid or lyophilized form.
  • Adjuvants such as a local anaesthetic, preservative and buffering agents can be dissolved in the vehicle and a surfactant or wetting agent can be included in the composition to facilitate uniform distribution of the Nanoparticles.
  • the pharmaceutical composition may comprise one or several types of Nanoparticles, comprising one or several different chemotherapeutic agents.
  • the pharmaceutical composition may further comprise at least one additional active substance, such as another chemotherapeutic agent included in Nanoparticles.
  • Nanoparticles used in the present invention are to be administered to a patient in need thereof to provide a therapeutically effective amount of the chemotherapeutic agent(s).
  • the term “patient” refers to either an animal, such as a valuable animal for breeding, company or preservation purposes, or preferably a human or a human child, which is afflicted with, or has the potential to be afflicted cancer.
  • a “therapeutically effective amount” refers to an amount of a compound which is effective in preventing, reducing, eliminating, treating or controlling the symptoms of the herein-described diseases and conditions.
  • the term “controlling” is intended to refer to all processes wherein there may be a slowing, interrupting, arresting, or stopping of the progression of the diseases and conditions described herein, but does not necessarily indicate a total elimination of all disease and condition symptoms, and is intended to include prophylactic treatment.
  • the amount of Nanoparticles to be administered has to be determined by standard procedure well known by those of ordinary skill in the art. In particular, physiological data of the patient (e.g. age, size, and weight), the type and localisation of cancer, the nature of the chemotherapeutic agent have to be taken into account to determine the appropriate dosage.
  • the chemotherapeutic agent is doxorubicin and the Nanoparticles are to be administered in an amount providing a dosage of doxorubicin from about 10 to about 75 mg/m 2 , preferably from about 10 to about 60 mg/m 2 , preferably from about 10 to about 45 mg/m 2 , more preferably from about 10 to about 30 mg/m 2 , from about 20 to about 30 mg/m 2 . More preferably, the dosage of doxorubicin may be about 20 mg/m 2 or 30 mg/m 2 .
  • the body surface area of a patient can easily be calculated by the skilled person from the body weight and height of the patient (e.g. body weight of about 65 kg corresponds to a body surface of about 1.8 m 2 ).
  • the Nanoparticles as described above are to be administered by intravenous or intra-arterial infusion for at least 2 hours, preferably for between 2 and 24 hours, more preferably for between 4 and 12 hours, and even more preferably for about 6 hours.
  • the volume to be administered and the flow rate may be easily determined by the skilled person in order to infuse a therapeutically effective amount of Nanoparticles in at least two hours.
  • the Nanoparticles may be administered in more than one treatment. They may be administered in one or several further treatment cycles (after initial treatment, i.e. the first infusion) with an interval of about two to about eight weeks between treatments, preferably about three to about four weeks, more preferably about four weeks. In an embodiment, administration of the Nanoparticles occurs on the first day of a 2-weekly to 8-weekly cycle. In a particular embodiment, the Nanoparticles are administered in two or three treatments with an interval between each treatment of two to about eight weeks, preferably about four weeks. Doses administered in each treatment cycle may be identical or different. The number of treatment cycles and the doses to be administered may be determined by the physician according to the physiological state of the patient, and the evolution of the disease. In another preferred embodiment, one or several further treatment cycles as described above are given during one to 28 month.
  • IV intravenous administration
  • This term refers to any type of intravenous access devices.
  • this term refers to hypodermic needle. It is the simplest form of intravenous access by passing a hollow needle through the skin directly into the vein. This needle can be connected directly to a syringe or may be connected to a length of tubing and thence whichever collection or infusion system is desired.
  • the most convenient site is often the arm, especially the veins on the back of the hand, or the median cubital vein at the elbow, but any identifiable vein can be used.
  • a Peripheral IV line (PVC or PIV) consists of a short catheter (a few centimeters long) inserted through the skin into a peripheral vein (any vein not inside the chest or abdomen). This is usually in the form of a cannula-over-needle device, in which a flexible plastic cannula comes mounted on a metal trocar. Once the tip of the needle and cannula are located in the vein, the trocar is withdrawn and discarded and the cannula advanced inside the vein to the appropriate position and secured.
  • Any accessible vein can be used although arm and hand veins are used most commonly, with leg and foot veins used to a much lesser extent.
  • This term can also refer to central IV lines flow through a catheter with its tip within a large vein, usually the superior vena cava or inferior vena cava, or within the right atrium of the heart. This has several advantages over a peripheral IV:
  • PICC peripherally inserted central catheter
  • the PICC line is inserted through a sheath into a peripheral vein, usually in the arm, and then carefully advanced upward until the catheter is in the superior vena cava or the right atrium.
  • the intravenous access devices can also be central venous lines.
  • catheters that take a more direct route into central veins. These are collectively called central venous lines.
  • central venous lines In the simplest type of central venous access, a catheter is inserted into a subclavian, internal jugular, or (less commonly) a femoral vein and advanced toward the heart until it reaches the superior vena cava or right atrium.
  • This term also refers to implantable ports (often referred to by brand names such as Port-a-Cath or MediPort) which are a central venous line that does not have an external connector; instead, it has a small reservoir that is covered with silicone rubber and is implanted under the skin. Medication is administered intermittently by placing a small needle through the skin, piercing the silicone, into the reservoir. It is possible to leave the ports in the patient's body for years; if this is done however, the port must be accessed monthly.
  • Implantable ports often referred to by brand names such as Port-a-Cath or MediPort
  • the delivery of therapeutic agents intra-arterially requires selective vascular catheterization, which is accomplished by tailoring the catheter configuration to the vascular anatomy.
  • intra-arterial administration refers to the infusion of liquid substances directly into an artery. This term refers to any type of intra-arterial access devices.
  • This term can refers to an arterial line, or art-line, or a-line, which is a thin catheter inserted into an artery.
  • An arterial line is usually inserted in the wrist (radial artery); but can also be inserted into the elbow (brachial artery), groin (femoral artery), foot ( dorsalis pedis artery).
  • This term can refers to syringe driver or syringe pump which is a small infusion pump (some include infuse and withdraw capability), used to gradually administer small amounts of fluid to a patient or for use in chemical and biomedical research.
  • Syringe drivers are also useful for delivering medications over several minutes. In the case of a medication which should be slowly pushed in over the course of several minutes, this device saves staff time and reduces errors.
  • Catheter-Port Systems To facilitate the long-term administration of chemotherapeutic agents, percutaneoulsy implantable catheter-port systems have been developed for long-term use. These systems allow easy and repetitive puncture in infusion therapy without causing much harm to the vessels, and their use is comfortable for the patient.
  • Catheter system can be surgically implanted in the gastro duodenal artery or via the subclavian, axillary, or brachial arteries into the common hepatic artery or in the femoral artery. The configuration of the catheter used depended on the vascular anatomy of the patient.
  • the standard catheter-port device consisted of a titanium port reservoir with a silicone rubber membrane at the puncture site and a lateral stem to slip the silicone catheter over.
  • the connection between the silicone catheter and the diagnostic angiographic catheter was reinforced with a small metallic cannula (Liver Intraarterial Chemotherapy: Use of the Femoral Artery for Percutaneous Implantation of Catheter-Port Systems, Karin Anna Herrmann et al. April 2000 Radiology, 215, 294-299).
  • IAC intra-arterial chemotherapy
  • a thin catheter is inserted through the femoral artery in the right leg.
  • An angiogram is performed to obtain a “roadmap” of the arteries.
  • This roadmap is then used to insert a line into the hepatic artery, which is the main blood vessel that delivers blood to the liver. Using dye from a syringe to make sure the line is in the correct position, the chemotherapy drug or drugs are injected directly into the artery.
  • cancer refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. This term refers to any type of malignancy (primary or metastases). In particular, this term refers to any malignant proliferative cell disorders such as solid tumor or hematopoietic tumor, including carcinoma, sarcoma, lymphoma, stem cell tumor, blastoma.
  • the cancer is selected from the group consisting of hepatic cancer, in particular hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, lung cancer, in particular small cell lung cancer, colorectal cancer, pancreas cancer, breast cancer, ovary cancer, uterine cancer, cervix cancer, head and neck cancer, brain cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma.
  • hepatic cancer in particular hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease
  • the cancer is selected from the group consisting of hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, small cell lung cancer, breast cancer, ovary cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma.
  • the cancer is a hepatic cancer, preferably a hepatocellular carcinoma.
  • the Nanoparticles comprise doxorubicin, polyisohexylcyanoacrylate, hydroxypropyl-beta-cyclodextrin and/or randomly methylated-beta cyclodextrin, for use for treating a hepatocellular carcinoma, wherein the Nanoparticles are to be administered by intravenous or intra-arterial infusion for at least 2 hours.
  • Example 1 Manufacturing of Nanoparticles Loaded with Doxorubicin (Nanoparticles Loaded with Doxorubicin)
  • Nanoparticles loaded with Doxorubicin product developed is presented as a sterile lyophilisate for injectable suspension that contains doxorubicin hydrochloride as active ingredient and other excipients including the Nanoparticles polymer, PIHCA.
  • the drug-loaded Nanoparticles are obtained by aqueous emulsion polymerization of isohexylcyanoacrylate (IHCA) monomer dropped in the bulk solution containing the active ingredient doxorubicin and the other excipients (P. Couvreur, B. Kanté, M. Roland, P. Guiot, P. Baudhuin, P. Amsterdamr, J. Pharm. Pharmacol., 1979, 31, 331).
  • IHCA isohexylcyanoacrylate
  • Nanoparticles entrapping doxorubicin is obtained.
  • the Nanoparticles mean size is comprised between 100 nm to 300 nm.
  • the Nanoparticles suspension is then filtered and aseptically filled in glass vials before freeze-drying. Nanoparticles loaded with Doxorubicin freeze-dried product must be kept protected from light and humidity and stored in a refrigerator between 2° C.-8° C., for stability purposes.
  • the aim of the present in vivo study was to establish and assess a rat model able to induce toxicological signs and lung injuries similar to severe pulmonary adverse events observed in said clinical trial.
  • Inventors studied the toxicological effects of a single bolus IV injection of a clinical batch of Nanoparticles loaded with Doxorubicin on healthy Wistar rats at early (24 h, 48 h, 72 h) and late (7 days) monitoring times.
  • the dose levels of 5, 7.5 and 10 mg/kg were chosen according to previous in vivo studies, they respectively correspond to 30 mg/m2, 45 mg/m2 and 60 mg/m2 human doses.
  • Rats were weighted and distributed according to their individual body weight to form 5 groups of 24 rats the day of IV treatment.
  • the IV dose was administered as a bolus at a rate of about 1000 ⁇ l/min or 3700 ⁇ g Doxorubicin-HCl/min in the penis vein under light isoflurane anaesthesia.
  • the dose of 7.5 mg/kg was chosen as the dose allowing to follow the lung injury until Day 2 with limited unscheduled deaths in comparison to the 10 mg/kg dose.
  • the aim of the present study was to compare the tolerance of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at the same dose level of 7.5 mg/kg injected intravenously by a single bolus injection (1000 ⁇ l/min or 3660 ⁇ g Doxorubicin-HCl/min) or by a 2 hours perfusion (4.3 ⁇ l/min or 15.6 ⁇ g Doxorubicin-HCl/min).
  • the dose level of 5 mg/kg (corresponding to 30 mg/m2 in human) administered by a single bolus IV injection (1000 ⁇ l/min or 3660 ⁇ g Doxorubicin-HCl/min) was chosen as safe bolus dose based on results of the previous study: no mortality and no lung injuries were observed at this dose. Rat euthanasia was performed 48 h after the treatment, according to scheme appearance of toxicological effects observed in previous studies.
  • Rats were weighted and distributed according to their individual body weight to form 5 groups of 6 rats for IV bolus or 8 rats for IV perfusion the day of the first IV treatment.
  • the IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of perfusion at a rate of 4.3 ⁇ l/min (corresponding to 15.6 ⁇ g (Dox-HCl)/min) for a 250 g rat (3.66 mg/ml suspension) and by the penis vein in the case of bolus injection at a rate of about 1000 ⁇ l/min (corresponding to 3660 ⁇ g (Dox-HCl)/min).
  • each syringe and catheter was weighted before and after treatment.
  • Nanoparticles loaded with Doxorubicin IV bolus group two rats over 6 were found dead at D1 after bolus injection and two rats were found dead at D2, corresponding to mortality rate of 4/6 treated rats (66.7%) at the end of the study.
  • Nanoparticles loaded with Doxorubicin IV perfusion group the first treated rat was found dead at D2 after treatment, corresponding to a mortality rate of 1/8 treated rats (12.5%) at the end of the study.
  • Example 4 Histological Evaluation of Lung and Heart of Healthy Rats after a Single Intravenous Injection of Nanoparticles Loaded with Doxorubicin. Comparative Study Between IV Bolus and Perfusion Administration
  • Histological lesions consist in pulmonary changes such as alveolar damage, fibrosis and inflammatory infiltrate (Pereverzeva E et al., Influence of the formulation on the tolerance profile of nanoparticle-bound doxorubicin in healthy rats: focus on cardio- and testicular toxicity. Int J Pharm. 2007, 337(1-2):346-56.
  • Perivascular oedema is known to be a major parameter in the assessment of lung toxicity, particularly in hypersensitivity (Tigani B et al., Resolution of the oedema associated with allergic pulmonary inflammation in rats assessed noninvasively by magnetic resonance imaging. British Journal of Pharmacology. 2003, 140, 2, 239-246). Few data are available in the literature for pulmonary toxicity of doxorubicin.
  • HES Hematoxyline-Erythrosine-Safran
  • the IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of perfusion at a rate of 4.3 ⁇ l/min and by the penis vein in the case of bolus injection at a rate of about 1000 ⁇ l/min.
  • each syringe and catheter was weighted before and after treatment.
  • the first experiment involved 16 male Wistar rats. Rats were weighted and distributed according to their individual body weight to form 5 groups of 3 rats for IV bolus, and 4 rats for IV perfusion.
  • the second experiment involved 14 male Wistar rats. Rats were weighted and distributed according to their individual body weight to form 4 groups of 4 rats for IV bolus, and 3 rats for IV perfusion.
  • rats were euthanatized 48 hours after administration of the product by IV intravenous injection of an overdose of pentobarbital.
  • the 7.5 mg/kg dose when administered through a perfusion procedure at a rate of about 4.3 ⁇ L/min (or 15.6 ⁇ g Doxorubicin-HCl/min), induced mild or moderate pulmonary histological lesions and no cardiac lesions as opposed to the same dose level administered as a bolus injection.
  • the aim of the present study is to investigate potential delayed toxicological effects after a 2 h-perfusion of Nanoparticles loaded with Doxorubicin.
  • rats treated by a perfusion of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg will be euthanatized seven days after the treatment, compared to rats euthanatized after 48 h as performed in previous studies.
  • Rats were weighted and distributed to form 5 groups of 6 to 8 rats.
  • the IV injection was performed under light isoflurane anaesthesia via the femoral vein in the case of a 2 h perfusion of a suspension at 3.69 mg/mL at a rate of 4.2 ⁇ L/min or 15.6 ⁇ g Doxorubicin HCl/min for a 250 g rat.
  • the injection rate was about 1000 ⁇ L/min or 3660 ⁇ g Doxorubicin HCl/min.
  • each syringe and catheter was weighted before and after treatment.
  • the mean lung weight of each Nanoparticles loaded with Doxorubicin treated group was compared to the mean lung weight of Excipient control group.
  • the aim of the present study was to compare the impact on tolerance and lung toxicity of perfusion speed of Nanoparticles loaded with Doxorubicin suspension at the dose level of 7.5 mg/kg.
  • Nanoparticles loaded with Doxorubicin were administered by IV perfusion at the rates of 12.5 ⁇ g/min (corresponding to a perfusion of 150 min for a 250 g rat and for a suspension of Nanoparticles loaded with Doxorubicin at 3.7 mg/mL Doxo HCl equivalent). Rat euthanasia was performed 48 h after the treatment, according to the scheme appearance of toxicological observed effects.
  • the IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of a 150 min perfusion of at a rate of 12.5 ⁇ g Doxo HCl/min or 3.4 ⁇ L/min for a 250 g rat and a suspension at 3.70 mg/mL.
  • the injection rate was about 1000 ⁇ L/min or 3660 ⁇ g Doxo HCl/min.
  • each syringe and catheter was weighted before and after treatment.
  • the mean lung weight of each Nanoparticles loaded with Doxorubicin was compared to the mean lung weight of Excipient bolus control group.
  • the present study showed that decreasing the perfusion speed from 15.6 ⁇ g Doxo HCl/min to 12.5 ⁇ g/min still reduced the impact of the treatment on lungs, as no mortality was observed in perfusion group and the mean lung weight was similar to control excipient group. In comparison, the mean lung weight was increased of 124% in 7.5 mg/kg bolus group.
  • the study primarily aims at demonstrating the efficacy of slow IV infusions of Nanoparticles loaded with Doxorubicin in patients with advanced HCC.
  • the study is a multicenter, randomized, controlled, open-label study comparing the efficacy and safety of slow repeated intravenous infusion of 2 doses of Nanoparticles loaded with Doxorubicin (20 mg/m 2 and 30 mg/m 2 ) to those of Best Supportive Care (BSC) in patients suffering from advanced Hepatocellular Carcinoma (HCC) after failure or intolerance to Sorafenib.
  • BSC Best Supportive Care
  • the study will be carried out at multiple hepatology or oncology centers that manage patients with HCC with or without cirrhosis.
  • the study duration for each patient includes a screening period of 28 days maximum before randomization.
  • the patient will then be randomized and either infused Nanoparticles loaded with Doxorubicin 20 mg/m 2 or Nanoparticles loaded with Doxorubicin 30 mg/m 2 over 6 hours through the IV route or given BSC.
  • Patients will receive as many cycles as possible up to unequivocal tumor progression (assessed by the investigator), cure or occurrence of unacceptable toxicity. They will be evaluated for efficacy assessment every 2 months.
  • Nanoparticles loaded with Doxorubicin groups will be hospitalized from Day 0 of each cycle of treatment and will be discharged on Day 3.
  • Study treatment (20 or 30 mg/m 2 of Nanoparticles loaded with Doxorubicin) will be administered by infusion over 6 hours through intravenous route at Day 1 and will be repeated every 4 weeks until unequivocal progression (assessed by the investigator) or toxicity.
  • a study visit will be performed on Day 14 of each cycle. Two months after last study drug administration, a End of Study visit will be performed.
  • the total dose of Doxorubicin will not exceed 450 mg/m 2 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Dermatology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Manufacturing & Machinery (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention relates to new therapeutic approaches for treating cancer, in particular hepatocellular carcinoma, with Nanoparticles loaded with a chemotherapeutic antitumoral agent. In particular, it relates to the treatment of cancer by administration of said Nanoparticles by intravenous infusion for at least 2 hours in order to prevent toxicological side effects and increase the benefit/risk ratio of the treatment.

Description

    FIELD OF THE INVENTION
  • The invention relates to the field of medicine, and in particular of oncology. It relates to a treatment of cancer, in particular hepatocellular carcinoma, with Nanoparticles loaded with at least one chemotherapeutic antitumoral drug.
  • BACKGROUND OF THE INVENTION
  • Cancer is characterized by uncontrolled growth of cells coupled with malignant behavior: invasion and metastasis. It is a major cause of mortality in most industrialized countries. Different ways of cancer treatment can be used: chemotherapy, radiotherapy, surgery, immunotherapy and hormonotherapy.
  • Chemotherapy can be defined as the use of chemotherapeutic antitumoral agents to treat cancer. Broadly, most chemotherapeutic antitumoral agents work by impairing mitosis (cell division) or DNA synthesis, effectively targeting fast-dividing cells.
  • Chemotherapeutic antitumoral agents are delivered most often parenterally, noteworthy intravenously (IV) or intra-arterially (IA). IV or IA chemotherapy can be given over different amounts of time, depending on the drug and the type of cancer to be treated.
  • Hepatocellular carcinoma (HCC) is the fifth most common cancer in men (523,000 cases worldwide) and the seventh in women (226,000 cases worldwide), and most of the burden is in developing countries, where almost 85% of the cases occur, and particularly in men: the overall male/female ratio is about 4/1. The regions of high incidence are Eastern and South-Eastern Asia, Middle and Western Africa, but also Melanesia and Micronesia/Polynesia. Low rates are estimated in developed regions, with the exception of Southern Europe where the incidence in men (ASR 10.5 per 100,000) is significantly higher than in other developed regions (Globocan 2008, WHO, International Agency for Research on Cancer-IARC-, Cancer Incidence and Mortality Worldwide 2008).
  • There were an estimated 694,000 deaths from liver cancer in 2008 (477,000 in men, 217,000 in women), and because of its high fatality (overall ratio of mortality to incidence of 0.93), liver cancer is the third most common cause of death from cancer worldwide. The geographical distribution of the mortality rates is similar to that observed for incidence (Globocan 2008, WHO, International Agency for Research on Cancer-IARC-, Cancer Incidence and Mortality Worldwide 2008).
  • HCC usually occurs in people suffering from cirrhosis or chronic liver disease (CLD). All factors favoring the development of CLD or cirrhosis are consequently risk factors for HCC. The main etiologic factors are hepatitis B virus infection (HBV; 53%), hepatitis C virus infection (HCV; 25%), alcoholic liver diseases (ALD; 15-43%) or dysmetabolic disorders such as Non Alcoholic Steato-Hepatitis (NASH; 20%) obesity and diabetes. Other factors are less frequent: haemochromatosis, other chronic biliary or inflammatory liver diseases. Aflatoxins, produced by the fungi Aspergillus flavus and Aspergillus parasiticus grown on grains, peanuts, and other food products, are hepatotoxic agents and chronic exposure to these mycotoxins leads to development of HCC. The effects of chemicals are still debated and not prominent.
  • Current therapeutic strategies for HCC can be divided into curative treatments such as surgical interventions (tumor resection and liver transplantation), percutaneous interventions (ethanol injection, radiofrequency thermal ablation), or palliative treatments such as transarterial interventions (mainly transarterial chemoembolization or TACE), systemic therapies and experimental strategies (H. C. Spangenberg, R. Thimine, H. E. Blum, Biologics: Targets & Therapy, 2008, 2(3), 453). In carefully selected patients, resection and liver transplantation, allow a 5-years survival from 60% to 70%. Unfortunately, most patients in Western countries present an intermediate or advanced HCC at diagnosis, with the consequent inability to use these curative treatments (L. Faloppi, M. Scartozzi, E. Maccaroni, P. M. Di Pietro, R. Berardi, M. Del Prete, S. Cascinu, Cancer Treat. Rev., 2011, 37(3), 169).
  • Among palliative treatments, intra-arterial approach with chemoembolization (TACE) has shown to induce objective responses in 16-55% of patients, although many randomized trials did not show any survival benefit. Unfortunately, TACE is known to be often accompanied by severe side effects like hepatic failure or renal dysfunction (K. Kamada, T. Nakanishi, M. Kitamoto, H. Aikata, Y. Kawakami, K. Ito, T. Asahara, G. Kajiyama, J. Vasc. Interv. Radiol., 2001, 12(7), 847).
  • Until recently, for patients with advanced HCCs no therapy was available that prolonged overall survival (OS), indicating the need for new targeted-therapies (H. C. Spangenberg, R. Thimine, H. E. Blum, Biologics:Targets & Therapy, 2008, 2(3), 453). In 2007 and for the first time, sorafenib (Nexavar®), a multikinase inhibitor, showed an increase even modest, of the overall survival over placebo in patients with unresectable HCC. Beside this agent, various different molecules are currently tested in advanced stage HCC among which Brivanib, another oral multikinase inhibitor being currently tested in several phase III studies (H. C. Spangenberg, R. Thimine, H. E. Blum, Biologics:Targets & Therapy, 2008, 2(3), 453-K. Almhanna, P A Philip, Onco. Targets Ther., 2009, 18(2), 261).
  • Even though sorafenib is the standard of care for advanced stage HCC and is registered for the treatment of HCC without restrictions by the European Medicines Agency (EMA) and the Food and Drug Administration (FDA), the narrow inclusion criteria of the clinical trials leave many patients without proven efficacious treatment with regard to their disease stage. Moreover, since treatment failure happens in some patients on sorafenib, there still remains a medical need for those patients to improve treatment efficacy, drug regimen and overall tolerance, and to overcome resistance (M. Peck-Radosavljevic, Therap. Adv. Gastroenterol. 2010, 3(4), 259).
  • HCC is known as hypervascular solid cancer characterized by a high degree of drug resistance (T. Wakamatsu, Y. Nakahashi, D. Hachimine, T. Seki, K. Okazaki, Int. J. Oncol., 2007, 31(6), 1465). The mechanisms of this chemoresistance in HCC are multiple. However, the more common mechanism is related to the multidrug resistance (MDR) transporters, P-gp and MRP pumps (D. M. Bradshaw, R. J. Arceci, J. Clin. Oncol., 1998, 16(11), 3674. Review. Erratum in: J. Clin. Oncol., 1999, 17(4), 1330). These pumps allow tumor cells to efflux different types of chemotherapeutic agents into the extracellular environment (Y. Chen, S. M. Simon, J. Cell Biol. 2000, 148(5), 863). Chemoresistance related to the MDR phenotype may be intrinsic or be acquired during chemotherapy. Chemoresistance, whether spontaneous or acquired is a serious concern in cancer treatment. HCC is often intrinsically chemoresistant which is the major cause for failure of its therapy (R. Pérez-Tomas, Curr Med Chem., 2006, 13(16), 1859, Review). This poses a great obstacle in chemotherapy for cancer because higher doses of drugs need to be administered and in turn may cause severe adverse effects (F. Yan, X. M. Wang, Z. C. Liu, C. Pan, S. B. Yuan, Q. M. Ma, Hepatobiliary Pancreat. Dis. Int. 2010, 9(3), 287). Chemoresistance affects major chemotherapeutic agents and especially anthracyclins (like doxorubicin), vinca-alkaloids, epipodophyllotoxins or taxanes. The poor efficacy of chemotherapeutic agents attributed to the overexpression of the MDR gene underlines the need to develop new treatment strategies for HCC, which could take into account the resistance issues.
  • Doxorubicin is a chemotherapeutic compound, efficacy of which has been shown in several cancers including HCC. However, IV infusion of doxorubicin in HCC is modest with objective response rate of 5-10%. A large, randomized, multicenter clinical trial compared the efficacy of doxorubicin 60 mg/m2 administered through the IV route and thymitaq a direct thymidilate synthase inhibitor (Porta C., 2006). In the 446 randomized patients with unresectable HCC, the effect of doxorubicin was found modest. The modest effect of doxorubicin in HCC patients is assumed to result from multidrug resistance (MDR) mechanisms related to overactivity of PgP and MRP cellular pumps. Many strategies have been evaluated to overcome the resistance issues, including the use of Pgp and MRP inhibitors. The development of these drugs has been stopped due to their safety profile.
  • In HCC, new therapeutic strategies using cytotoxic agents were developed by hepatic intra-arterial (IA) injection in order to reduce the systemic toxicity, to induce important hepatic tumor necrosis and to save the healthy hepatic parenchyma.
  • The technology described in patents EP1056477, and its US equivalent U.S. Pat. No. 6,881,421, use polyalkylcyanoacrylate (PACA) polymer to formulate active ingredients into Nanoparticles. EP1056477, and its US equivalent U.S. Pat. No. 6,881,421, indicate the use of a complexing agent to complex the active ingredient during preparation of the nanoparticle so as to protect the active ingredient against chemical reactions that are necessary for the formation of the particle. Therefore, the active ingredient is advantageously associated in a non-covalent manner with the particle and protected from reactions or degradation. Nanoparticles comprising a pharmaceutically active ingredient, a polymer such as poly(alkylcyanoacrylate) and a complexing agent such cyclodextrins, are thus taught in EP1056477, and in its US equivalent U.S. Pat. No. 6,881,421.
  • Doxorubicin loaded in said Nanoparticles (hereinafter referred to as “Nanoparticles loaded with Doxorubicin”) is a drug formulation that associates a kind of PACA, polyisohexylcyanoacrylate (PIHCA), Nanoparticles with the chemotherapeutic agent doxorubicin.
  • Said Nanoparticles display original mechanisms to bypass MDR that can be summarized as follows:
  • Nanoparticles loaded with Doxorubicin adsorbs to the surface of tumor cells and releases the entrapped doxorubicin close to the cell membrane which leads to a high local gradient concentration (Colin de Verdiére A, Cancer Chemother. Pharmacol. 1994; 33(6):504-8).
  • The Nanoparticles degrade and release soluble polycyanoacrylic acid which might interact with the plasma membrane and contribute to improve the intracellular delivering of doxorubicin (De Verdiére A C, Br J Cancer. 1997; 76(2):198-205).
  • The soluble polymer could also mask the positive charge of doxorubicin thus preventing its effluxing by the Pgp (De Verdiére 1997, Br J Cancer. 1997; 76(2):198-205), acting as an ion pair without any covalent linkage). The direct interaction with MDR pumps (Pgp and MRP) is thus avoided.
  • In man, the efficacy and safety of Nanoparticles loaded with Doxorubicin_through the hepatic intra-arterial route have been evaluated in 2 clinical trials: in one open phase I-II and one randomized phase II clinical trial (Table 1).
  • TABLE 1
    Summary of Study Designs for Clinical development
    of Nanoparticles loaded with
    Phase/ Patients
    Study Descrip- Route Popula- planned/
    number tion Dosage tion complete Status
    Phase Open Hepatic IA Ad-  21/20 Completed
    I/II MC 10 mg/m2 vanced
    BA/2002/ SD 20 mg/m2 HCC
    03/02 Esc 30 mg/m2
    dose 35 mg/m2
    40 mg/m2
    Phase Open, Hepatic IA Ad- 200/28 Stopped
    II/III MC 30 mg/m2 vanced
    BA/2006/ R every 4 HCC
    03/03 PG weeks
    max 3
    injections
  • Doxorubicin
  • The study BA2002/03/02 (phase I/II study in patients with advanced Hepatocellular Carcinoma) was carried out according to a multicenter, open, dose-escalation design in patients suffering from HCC. Nanoparticles loaded with Doxorubicin was to be injected through hepatic intra-arterial (IA) route as a bolus. Successive cohorts of 3 patients were injected a single 10, 20 and 30 mg/m2 dose of Nanoparticles loaded with Doxorubicin. As the 30 mg/m2 dose was well tolerated, the protocol was amended to assess 40 mg/m2 and then 35 mg/m2. As these 2 doses were considered toxic, additional patients were given 30 mg/m2 Nanoparticles loaded with Doxorubicin dose in 15 minutes. Two patients received a second IA dose of Nanoparticles loaded with Doxorubicin and one patient received 3 IA infusions.
  • Twenty patients were included in this study. Apart from the serious respiratory TEAEs (Treatment Emergent Adverse Events) reported at 35 and 40 mg/m2, tolerance was acceptable; most of the TEAEs were short lasting and of mild severity. All were reversible without sequellae. Overall, 50% of the non-serious TEAEs were expected as already reported with free doxorubicin. The most frequent TEAE were leukopenia (n=13; 65%), neutropenia (n=12; 60%), nausea (n=10; 50%), anaemia and abdominal pain (n=9; 45%), asthenia, fever, alopecia (n=6; 30%) and cough (n=5; 25%). Increase in transaminases was reported in 11 patients (55%) and was expected as likely related to treatment efficacy. Two patients had serious Acute Respiratory Distress Syndrome (ARDS) at the Nanoparticles loaded with Doxorubicin dose of 35 mg/m2.
  • Efficacy data clearly demonstrated a signal of efficacy with a mean and median survival of 548 and 315 days respectively, an objective response rate of 65 to 80% according to clinical study criteria.
  • On the basis of these data, the efficacy and safety of an hepatic IA 15-minute infusion of Nanoparticles loaded with Doxorubicin was compared to those of standard of care treatment.
  • The second study BA2006/03/03 (Phase II/III study in patients with advanced HCC) was carried out according to a multicenter, comparative, open, randomized (with a 2/1 ratio) design in patients suffering from advanced HCC. Nanoparticles loaded with Doxorubicin was injected through IA route as a 15-minute infusion at the dose of 30 mg/m2, preceded and followed by an oral premedication with methylprednisone. Fifty patients were to be included in the 1st part of the study. Three Nanoparticles loaded with Doxorubicin infusions were to be received by 33 patients at 4-week intervals and each of the other 17 patients were to receive the best standard of care treatment adapted to the severity of the disease. At the end of this 1st phase, if Nanoparticles loaded with Doxorubicin was considered active in ⅔ of patients (patients free of local progression at 3 months), then 150 additional patients were to be enrolled.
  • This study was prematurely discontinued when 28 patients had been enrolled because of the occurrence of ARDS leading to death in 2 patients treated with Nanoparticles loaded with Doxorubicin. 17 patients had received 39 Nanoparticles loaded with Doxorubicin infusions and 11 were randomized in the control group. No patients died of ARDS in the control group.
  • Despite many patients in the Nanoparticles loaded with Doxorubicin group did not complete treatment according to the protocol (3 infusions 4-week apart) because of the premature discontinuation of the trial, 63% of patients in the Nanoparticles loaded with Doxorubicin group were free of local progression at month 3 (versus 75% showing local progression in the control group). The patients enrolled were monitored and survival was recorded up to February 2011. Overall survival was significantly longer in the Nanoparticles loaded with Doxorubicin group than in the control group. At this time point, the mean and median overall survival was 952 days for Nanoparticles loaded with Doxorubicin group versus 449 days for control.
  • In addition, survival was significantly much longer in patients having completed 3 courses of Nanoparticles loaded with Doxorubicin as requested in the protocol. The mean and median overall survival was twice as long in the Nanoparticles loaded with Doxorubicin group having received 3 IA injections as in the control group. Likewise survival was much longer in patients having completed 3 courses of Nanoparticles loaded with Doxorubicin than in those having received only one or 2 IA injections of 30 mg/m2 Nanoparticles loaded with Doxorubicin. These data confirmed the strong signal of efficacy of Nanoparticles loaded with Doxorubicin in the treatment of patients suffering from advanced HCC.
  • Hence, albeit very promising in term of efficacy, use of Nanoparticles loaded with chemotherapeutic drug, such as Nanoparticles loaded with Doxorubicin, in cancer treatment, and in particular HCC treatment, is not currently possible because of its severe pulmonary adverse events. New approaches allowing safer use, reducing the probability of occurrence of pulmonary adverse events and their severity under acceptable limits owing to the benefit/risk ratio are then needed. In particular, it would be very advantageous to decrease the pulmonary adverse events induced by these Nanoparticles and at the same time to maintain the good efficacy already observed.
  • SUMMARY OF THE INVENTION
  • The inventors succeeded in obtaining a suitable animal model for the severe pulmonary adverse events and, then, have surprisingly found that severe toxicological side effects, in particular lung injuries, associated with intra-arterial or intravenous administration of Nanoparticles loaded with a chemotherapeutic agent, can be prevented by intravenous or intra-arterial infusion of said Nanoparticles for several hours.
  • On this basis, the present invention concerns Nanoparticles comprising at least one chemotherapeutic antitumoral agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin, for use for treating a cancer, wherein the Nanoparticles are administered by intravenous or intra-arterial infusion for at least 2 hours. In particular, Nanoparticles can be administered by intravenous or intra-arterial infusion for between 2 and 24 hours, more particularly for between 4 and 12 hours, even more particularly for about 6 hours. In a particular embodiment, the administration is by intravenous infusion.
  • Preferably, the poly(alkylcyanoacrylate) polymer comprised in the Nanoparticles is a polyisohexylcyanoacrylate polymer.
  • The Nanoparticles used in the invention may comprise said at least one chemotherapeutic agent at a concentration from 0.01 to 200 mg/g of Nanoparticles, from 0.1 to 70% w/w of said at least one cyclodextrin and from 1 to 25% w/w of said at least one poly(alkylcyanoacrylate).
  • The chemotherapeutic antitumoral agent comprised in the Nanoparticles may be selected from the group consisting of anthracyclines, topoisomerase inhibitors, spindle poison plant alkaloids, alkylating agents, anti-metabolites, ellipticine and harmine, and any combination thereof. Preferably, the chemotherapeutic antitumoral agent is an anthracycline. More preferably, the chemotherapeutic agent is selected from the group consisting of doxorubicin, daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, any pharmaceutically acceptable salt thereof, and any combination thereof. In a particularly preferred embodiment, the chemotherapeutic antitumoral agent is doxorubicin or any pharmaceutically acceptable salt thereof. In this embodiment, the dosage of doxorubicin may be from about 10 to about 75 mg/m2, from about 10 to about 60 mg/m2, from about 10 to about 45 mg/m2, from about 10 to about 30 mg/m2, from about 20 to about 30 mg/m2. In a particular embodiment, the dosage of doxorubicin may be about 20 mg/m2 or 30 mg/m2.
  • The cancer treated with the Nanoparticles may be is a solid tumor or a hematopoietic tumor, preferably selected from the group consisting of hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, small cell lung cancer, colorectal cancer, pancreas cancer, breast cancer, ovary cancer, uterine cancer, cervix cancer, head and neck cancer, brain cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma. In a particularly preferred embodiment, the cancer is a hepatocellular carcinoma.
  • In another aspect, the present invention concerns a method for treating cancer, wherein the method comprises administering by intravenous or intra-arterial infusion for at least 2 hours, a therapeutically effective amount of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin to a patient in need of such treatment. Preferably, Nanoparticles are administered in the form of a pharmaceutical composition as described above. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect. In a particular embodiment, the administration is by intravenous infusion.
  • In further aspect, the present invention concerns the use of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin for the treatment of cancer, said Nanoparticles being administered by intravenous or intra-arterial infusion for at least 2 hours. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect. In a particular embodiment, the administration is by intravenous infusion.
  • In a last aspect, the present invention concerns the use of Nanoparticles comprising at least one chemotherapeutic agent, at least one poly(alkylcyanoacrylate) and at least one cyclodextrin, for the manufacture of a pharmaceutical preparation for the treatment of cancer, said pharmaceutical composition being administered by intravenous or intra-arterial infusion for at least 2 hours. All embodiments disclosed above for the Nanoparticles are encompassed in this aspect. In a particular embodiment, the administration is by intravenous infusion.
  • DETAILED DESCRIPTION OF THE INVENTION
  • As preliminary work to the present invention, the inventors have established and assessed a rat model to investigate respiratory adverse events observed in previous clinical trials for treatment of hepatocellular carcinoma by hepatic intra-arterial infusion of doxorubicin formulated into Nanoparticles comprising a poly(alkylcyanoacrylate) polymer and a cyclodextrin (hereinafter referred to as “Nanoparticles loaded with Doxorubicin”). Using this model, they have herein demonstrated that intravenous infusion of said Nanoparticles for at least 2 hours allows to strongly reduce toxicological side effects, in particular lung injuries, compared to bolus intravenous injection at the same dose. This new therapeutic approach allows safer use of Nanoparticles and then dramatically increases the benefit/risk ratio.
  • Accordingly, in a first aspect, the present invention concerns Nanoparticles comprising at least one chemotherapeutic antitumoral agent for use for treating a cancer, wherein the Nanoparticles are to be administered by intravenous or intra-arterial infusion for at least 2 hours.
  • The one skilled in the art, considering the decrease of pulmonary toxicity induced by said Nanoparticles thanks to their intravenous infusion for at least 2 hours would have expected a similar decrease with their intra-arterial infusion for at least 2 hours.
  • Indeed, an analytical model for intra-arterial versus intravenous infusion of doxorubicin is known in the state of the art, and shows that the two doxorubicin concentration decay curves are similar. Hence, the same theoretical curve is fitted and used for both cases (“An analytical model for intra-arterial versus intravenous infusion of Adriamycin”, Logan S E et al., Biomed Sci Instrum. 1989; 25:239-46). This article indicates that even if these observations were made for bolus injections, similar relationships exist for continuous infusion. Indeed, the continuous infusion can be understood as a series of infinitesimal bolus injections, each one behaving as described in said analytical model.
  • Moreover, the state of the art confirms that in patients who received doxorubicin through a systemic vein, the drug level in the peripheral blood were comparable to those obtained from the peripheral blood of patient receiving doxorubicin intra-arterially. (comparison of Regional versus Systemic Chemotherapy with Adriamycin, Didolkar et al., Ann. Surg., March 1978, Vol. 187, No. 3, p. 332-336).
  • The Nanoparticles used in the present invention comprise at least one chemotherapeutic antitumoral agent, at least one poly(alkylcyanoacrylate) polymer, and at least one compound able to form a complex with the chemotherapeutic antitumoral agent, preferably chosen from among the cyclical oligosaccharides, in particular from among the cyclodextrins. Such Nanoparticles have been previously described in the European patent EP 1 056 477 and its US equivalent U.S. Pat. No. 6,881,421, which are herein enclosed by reference.
  • The poly(alkylcyanoacrylate) polymer may be linear or branched, preferably branched. The alkyl group of the poly(alkylcyanoacrylate) may be linear or branched, preferably branched. In a particular embodiment, the poly(alkylcyanoacrylate) polymer is a poly(C1-C12 alkylcyanoacrylate), preferably a poly(C4-C10 alkylcyanoacrylate), more preferably a poly(C6-C8 alkylcyanoacrylate). In a preferred embodiment, the poly(alkylcyanoacrylate) polymer is a polyisohexylcyanoacrylate. The monomer corresponding to the latter polymer is available, for instance under the trademark Monorex® by Bioalliance Pharma (France).
  • The cyclodextrin may be neutral or charged, native (cyclodextrins α, β, γ, δ, ε), branched or polymerized, or even chemically modified, for example, by substitution of one or more hydroxypropyls by groups such as alkyls, aryls, arylalkyls, glycosidics, or by etherification, esterification with alcohols or aliphatic acids. Among the above groups, particular preference is given to those chosen from the group consisting of hydroxypropyl, methyl-m and sulfobutylether groups, and mixtures thereof. In a preferred embodiment, the cyclodextrin is selected from the group consisting of hydroxypropyl-beta-cyclodextrin and/or randomly methylated-beta cyclodextrin, and mixtures thereof.
  • As used herein, the term “chemotherapeutic agent” or “chemotherapeutic antitumoral agent” refers to any chemical compound or drug with anti-cancer activity that inhibits or halts the growth of cancerous cells or immature pre-cancerous cells, kills cancerous cells or immature pre-cancerous cells, increases the susceptibility of cancerous or pre-cancerous cells to other chemotherapeutic agents, and/or inhibits metastasis of cancerous cells.
  • In a particular embodiment, the chemotherapeutic agent is selected from the group consisting of anthracyclines, topoisomerase I and/or II inhibitors, spindle poison plant alkaloids, alkylating agents, anti-metabolites, ellipticine and harmine.
  • Anthracyclines (or anthracycline antibiotics) are derived from Streptomyces bacteria. These compounds are used to treat a wide range of cancers, including for example hepatocellular carcinoma, leukemias, lymphomas, and breast, uterine, ovarian, and lung cancers. Anthracyclines have three mechanisms of action: (i) inhibition of DNA and RNA synthesis by intercalating between base pairs of the DNA/RNA strand, thus preventing the replication of rapidly-growing cancer cells; (ii) inhibition of topoisomerase II enzyme, preventing the relaxing of supercoiled DNA and thus blocking DNA transcription and replication; and (iii) creation of iron-mediated free oxygen radicals that damage the DNA and cell membranes. Anthracyclines include, but are not limited to doxorubicin (also named adriamycin), daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, and their pharmaceutically acceptable salts thereof. Preferably, the anthracycline is selected from the group consisting of doxorubicin, daunorubicin, epirubicin, pirarubicin, zorubicin and aclarubicin, and any pharmaceutically acceptable salt thereof. In a preferred embodiment, the anthracycline is doxorubicin or any pharmaceutically acceptable salt thereof.
  • Topoisomerases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisomerases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling. Some type I topoisomerase inhibitors include camptothecins derivatives Camptothecin derivatives refer to camptothecin analogs such as irinotecan, topotecan, hexatecan, silatecan, lutortecan, karenitecin (BNP1350), gimatecan (ST1481), belotecan (CKD602), or their pharmaceutically acceptable salts. Examples of type II topoisomerase inhibitors include, but are not limited to, amsacrine, etoposide, etoposide phosphate and teniposide They are semisynthetic derivatives of epipodophyllotoxins, alkaloids naturally occurring in the root of American Mayapple (Podophyllum peltatum). In a particular embodiment, the topoisomerase inhibitor is selected from the group consisting of irinotecan, its active metabolite SN38 and topotecan, and any pharmaceutically acceptable salt thereof. In a preferred embodiment, the topoisomerase inhibitor is irinotecan.
  • Spindle poison plant alkaloids are derived from plants and block cell division by preventing microtubule function, essential for cell division. These alkaloids include, but are not limited to, vinca alkaloids (like vinblastine, vincristine, vindesine, vinorelbine and vinpocetine) and taxanes. Taxanes include, but are not limited to, paclitaxel, docetaxel, larotaxel, cabazitaxel, ortataxel, tesetaxel, and their pharmaceutically acceptable salts. Preferably, the taxane is selected from the group consisting of paclitaxel and docetaxel, and any pharmaceutically acceptable salt thereof. Paclitaxel was originally derived from the Pacific yew tree. Docetaxel is a semi-synthetic analogue of paclitaxel. In contrast to the taxanes, the vinca alkaloids destroy mitotic spindles. Both taxanes and vinca alkaloids are therefore named spindle poisons or mitosis poisons, but they act in different ways.
  • Alkylating agents are so named because of their ability to add alkyl groups to many electronegative groups under conditions present in cells. They impair cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules. Noteworthy, their cytotoxicity is thought to result from inhibition of DNA synthesis. Alkylating agents include, but are not limited to, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and platinum compounds such as oxaliplatin, cisplatin or carboplatin.
  • An anti-metabolite is a chemical that inhibits the use of a metabolite, which is part of normal metabolism. Such substances are often similar in structure to the metabolite that they interfere with. The presence of anti-metabolites halters cell growth and cell division.
  • Purine or pyrimidine analogues prevent the incorporation of nucleotides into DNA, stopping DNA synthesis and thus cell divisions. They also affect RNA synthesis. Examples of purine analogues include azathioprine, mercaptopurine, thioguanine, fludarabine, pentostatin and cladribine. Examples of pyrimidine analogues include 5-fluorouracil (5FU), which inhibits thymidylate synthase, floxuridine (FUDR) and cytosine arabinoside (Cytarabine).
  • Antifolates are drugs which impair the function of folic acids. Many are used in cancer chemotherapy, some are used as antibiotics or antiprotozoal agents. A well known example is Methotrexate. This is a folic acid analogue, and owing to structural similarity with it binds and inhibits the enzyme dihydrofolate reductase (DHFR), and thus prevents the formation of tetrahydrofolate. Tetrahydrofolate is essential for purine and pyrimidine synthesis, and this leads to inhibited production of DNA, RNA and proteins (as tetrahydrofolate is also involved in the synthesis of amino acids serine and methionine). Other antifolates include, but are not limited to, trimethoprim, raltitrexed, pyrimethamine and pemetrexed.
  • Examples of chemotherapeutic agents above are not limiting and other agents can be loaded in Nanoparticles. Among others, ellipticine and harmine can be cited.
  • Ellipticine is a natural plant alkaloid product which was isolated from the evergreen tree of the Apocynaceae family. Ellipticine was found to have cytotoxic and anticancer activity (Dalton et al., Aust. J. Chem., 1967. 20, 2715). The ellipticine derivative hydroxylated in position 9 (9-hydroxyellipticinium) was found to have greater antitumoural activity than ellipticine on many experimental tumours (Le Pecq et al., Proc. Natl. Acad, Sci., USA, 1974, 71, 5078-5082). Researches were performed to identify an ellipticine derivative appropriate for human therapeutics and lead to the preparation of Celiptium, or N2-methyl-9-hydroxyellipticinium (NMHE), which has been used for the treatment of some human cancers, in particular for the treatment of bone metastasis of breast cancers. Other 9-hydroxy ellipticine derivatives, such as 2-(diethyiamino-2-ethyl)9-hydroxyellipticinium acetate, 2-(diisopropylamino-ethyl)9-hydroxy-ellipticinium acetate and 2-(beta piperidino-2-ethyl)9-hydroxyellipticinium, had been described for instance in the U.S. Pat. No. 4,310,667.
  • Harmine is a natural plant alkaloid product which was isolated from the Peganum harmala seeds. Peganum harmala (Zygophyllaceae) is a plant widely distributed in semi arid rangelands in the Central Asia, North Africa, Middle East and Australia. The pharmacologically active compounds of P. harmala are several alkaloids that are found especially in the seeds and the roots. These include 3-carbolines such as harmine, harmaline, harmol, harmalol and harman, and quinazoline derivatives: vasicine and vasicinone. Peganum harmala alkaloids were found to possess significant antitumor potential (Lamchouri and al., Therapie, 1999, 54(6):753-8). Proliferation of tumoral cells lines was significantly reduced. Harmine was reported to exhibit strong cytotoxicity against a number of human tumor cell lines (Ishida and al, Bioorg Mad Chem Lett, 1999, 9(23):3319-24). Anticancer activity of harmol dimers has also been described for instance in the international patent WO2009047298.
  • In a preferred embodiment, the chemotherapeutic agent is an anthracycline, preferably selected from the group consisting of doxorubicin, daunorubicin, epirubicin, idarubicin, valrubicin, pirarubicin, zorubicin, aclarubicin, detorubicin, carminomycin, morpholinodoxorubicin, morpholinodaunorubicin, methoxymorpholinyldoxorubicin, any pharmaceutically acceptable salt thereof, and any combination thereof, more preferably doxorubicin or any pharmaceutically acceptable salt thereof.
  • As used herein, “pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, tartaric, citric, methanesulfonic, benzenesulfonic, glucoronic, glutamic, benzoic, salicylic, toluenesulfonic, oxalic, fumaric, maleic, lactic and the like. Further addition salts include ammonium salts such as tromethamine, meglumine or epolamine, metal salts such as sodium, potassium, calcium, zinc or magnesium. For instance, a suitable salt of doxorubicin is the doxorubicin hydrochloride.
  • In a particular embodiment, the Nanoparticles used in the invention comprise at least one chemotherapeutic agent, preferably selected from among anthracyclines, at least one poly(C1-C12 alkylcyanoacrylate), preferably a polyisohexylcyanoacrylate, and at least one cyclodextrin, preferably selected from the group consisting of hydroxypropyl-beta-cyclodextrin and randomly methylated-beta cyclodextrin, and mixtures thereof. In a particularly preferred embodiment, the Nanoparticles comprise doxorubicin, a polyisohexylcyanoacrylate and a hydroxypropyl-beta-cyclo dextrin.
  • The chemotherapeutic agent is generally present at a concentration from about 0.01 to about 200 mg/g of Nanoparticles, preferably from about 1 to about 50 mg/g.
  • The proportion of cyclodextrin is in general from about 0.1 to about 70% by weight of Nanoparticles, preferably from about 1 to about 30%, more preferably from about 5 to about 20%. The proportion of the chemotherapeutic agent and the proportion of cyclodextrin are independent from one another.
  • The proportion of poly(alkylcyanoacrylate) polymer is in general from about 1 to about 25% by weight of Nanoparticles, preferably from about 5 to about 15%.
  • In a particular embodiment, the Nanoparticles comprise a chemotherapeutic agent at a concentration from 0.01 to 200 mg/g of Nanoparticles, from 0.1 to 70% w/w of cyclodextrin and from 1 to 25% w/w of poly(alkylcyanoacrylate), preferably of polyisohexylcyanoacrylate.
  • As used in this specification, the term “about” refers to a range of values ±10% of the specified value. For instance, “about 1” means from 0.9 to 1.1 when 10% is considered and from 0.95 to 1.05 when 5% is considered. Where “about” is used in connection with numeric ranges, for example “about 1 to about 3”, or “between about one and about three”, preferably the definition of “about” given above for a number is applied to each number defining the start and the end of a range separately. Preferably, where “about” is used in connection with any numerical values, the “about” can be deleted.
  • The Nanoparticles used in the present invention may be prepared according any method known by the skilled person. Such a method is disclosed, for example, in the European patent EP 1 056 477 and its US equivalent U.S. Pat. No. 6,881,421.
  • In particular, the Nanoparticles may be prepared by a method comprising the steps of
  • a) preparing, in an acid aqueous, a complex of the chemotherapeutic agent with cyclodextrin;
  • b) gradually adding the alkylcyanoacrylate monomer, preferably the isohexylcyanoacrylate monomer, in the solution obtained at step (a); and
  • c) conducting polymerization of this monomer, optionally in the presence of one or more surfactant and/or stabilising agents.
  • Preferably, the polymerization is anionic but may also be inducible by other agents, in particular by photochemical agents. In a particular embodiment, the polymerization is conducted in presence of a surfactant agent such as poloxamer or dextran (such as dextran 70,000) or other non ionic surfactive agents (like polysorbate, sorbitan esters or others). Poloxamers are preferred, such as poloxamer 407, poloxamer 401, poloxamer 237, poloxamer 338, poloxamer 331, poloxamer 231, or poloxamer 188 (also named Pluronic® F68). Poloxamer 188 is more preferred.
  • The size of the Nanoparticles is from about 50 to about 300 nm, preferably from about 100 to about 300 nm, more preferably about 200 nm. The size of these Nanoparticles is essentially related to the concentration of the cyclodextrin.
  • Nanoparticles as described above are administered in the form of a pharmaceutical composition comprising said Nanoparticles and at least one pharmaceutically acceptable excipient.
  • As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, excipients, compositions or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response or other problem complications commensurate with a reasonable benefit/risk ratio.
  • The pharmaceutical composition comprising said Nanoparticles is formulated in accordance with standard pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York) known by a person skilled in the art. In particular, possible pharmaceutical compositions include those suitable for intravenous, intra-arterial and intra-tumoral administration. For these formulations, conventional excipient can be used according to techniques well known by those skilled in the art. Such compositions for parenteral administration are generally physiologically compatible sterile solutions or suspensions which can optionally be prepared immediately before use from solid or lyophilized form. Adjuvants such as a local anaesthetic, preservative and buffering agents can be dissolved in the vehicle and a surfactant or wetting agent can be included in the composition to facilitate uniform distribution of the Nanoparticles.
  • The pharmaceutical composition may comprise one or several types of Nanoparticles, comprising one or several different chemotherapeutic agents.
  • In addition to Nanoparticles as described above, the pharmaceutical composition may further comprise at least one additional active substance, such as another chemotherapeutic agent included in Nanoparticles.
  • The Nanoparticles used in the present invention are to be administered to a patient in need thereof to provide a therapeutically effective amount of the chemotherapeutic agent(s).
  • As used herein, the term “patient” refers to either an animal, such as a valuable animal for breeding, company or preservation purposes, or preferably a human or a human child, which is afflicted with, or has the potential to be afflicted cancer.
  • As used herein, a “therapeutically effective amount” refers to an amount of a compound which is effective in preventing, reducing, eliminating, treating or controlling the symptoms of the herein-described diseases and conditions. The term “controlling” is intended to refer to all processes wherein there may be a slowing, interrupting, arresting, or stopping of the progression of the diseases and conditions described herein, but does not necessarily indicate a total elimination of all disease and condition symptoms, and is intended to include prophylactic treatment. The amount of Nanoparticles to be administered has to be determined by standard procedure well known by those of ordinary skill in the art. In particular, physiological data of the patient (e.g. age, size, and weight), the type and localisation of cancer, the nature of the chemotherapeutic agent have to be taken into account to determine the appropriate dosage.
  • In a particular embodiment, the chemotherapeutic agent is doxorubicin and the Nanoparticles are to be administered in an amount providing a dosage of doxorubicin from about 10 to about 75 mg/m2, preferably from about 10 to about 60 mg/m2, preferably from about 10 to about 45 mg/m2, more preferably from about 10 to about 30 mg/m2, from about 20 to about 30 mg/m2. More preferably, the dosage of doxorubicin may be about 20 mg/m2 or 30 mg/m2.
  • The body surface area of a patient can easily be calculated by the skilled person from the body weight and height of the patient (e.g. body weight of about 65 kg corresponds to a body surface of about 1.8 m2).
  • The inventors have herein demonstrated that intravenous or intra-arterial infusion of Nanoparticles for at least 2 hours allows to strongly reduce toxicological side effects of the treatment then dramatically increases the benefit/risk ratio.
  • Consequently, the Nanoparticles as described above are to be administered by intravenous or intra-arterial infusion for at least 2 hours, preferably for between 2 and 24 hours, more preferably for between 4 and 12 hours, and even more preferably for about 6 hours. The volume to be administered and the flow rate may be easily determined by the skilled person in order to infuse a therapeutically effective amount of Nanoparticles in at least two hours.
  • The Nanoparticles may be administered in more than one treatment. They may be administered in one or several further treatment cycles (after initial treatment, i.e. the first infusion) with an interval of about two to about eight weeks between treatments, preferably about three to about four weeks, more preferably about four weeks. In an embodiment, administration of the Nanoparticles occurs on the first day of a 2-weekly to 8-weekly cycle. In a particular embodiment, the Nanoparticles are administered in two or three treatments with an interval between each treatment of two to about eight weeks, preferably about four weeks. Doses administered in each treatment cycle may be identical or different. The number of treatment cycles and the doses to be administered may be determined by the physician according to the physiological state of the patient, and the evolution of the disease. In another preferred embodiment, one or several further treatment cycles as described above are given during one to 28 month.
  • As used herein, the term “intravenous administration” (“IV”) refers to the infusion of liquid substances directly into a vein.
  • This term refers to any type of intravenous access devices. In particular, this term refers to hypodermic needle. It is the simplest form of intravenous access by passing a hollow needle through the skin directly into the vein. This needle can be connected directly to a syringe or may be connected to a length of tubing and thence whichever collection or infusion system is desired.
  • The most convenient site is often the arm, especially the veins on the back of the hand, or the median cubital vein at the elbow, but any identifiable vein can be used.
  • It also refers to peripheral cannula. A Peripheral IV line (PVC or PIV) consists of a short catheter (a few centimeters long) inserted through the skin into a peripheral vein (any vein not inside the chest or abdomen). This is usually in the form of a cannula-over-needle device, in which a flexible plastic cannula comes mounted on a metal trocar. Once the tip of the needle and cannula are located in the vein, the trocar is withdrawn and discarded and the cannula advanced inside the vein to the appropriate position and secured.
  • Any accessible vein can be used although arm and hand veins are used most commonly, with leg and foot veins used to a much lesser extent.
  • This term can also refer to central IV lines flow through a catheter with its tip within a large vein, usually the superior vena cava or inferior vena cava, or within the right atrium of the heart. This has several advantages over a peripheral IV:
      • It can deliver fluids and medications that would be overly irritating to peripheral veins because of their concentration or chemical composition. These include some chemotherapy drugs and total parenteral nutrition.
      • Medications reach the heart immediately, and are quickly distributed to the rest of the body.
        There are several types of central IVs, depending on the route that the catheter takes from the outside of the body to the vein.
  • It also can refer to peripherally inserted central catheter (PICC) which are used when intravenous access is required over a prolonged period of time or when the material to be infused would cause quick damage and early failure of a peripheral IV and when a conventional central line may be too dangerous to attempt. Typical uses for a PICC include: long chemotherapy regimens, extended antibiotic therapy, or total parenteral nutrition.
  • The PICC line is inserted through a sheath into a peripheral vein, usually in the arm, and then carefully advanced upward until the catheter is in the superior vena cava or the right atrium.
  • The intravenous access devices can also be central venous lines. There are several types of catheters that take a more direct route into central veins. These are collectively called central venous lines. In the simplest type of central venous access, a catheter is inserted into a subclavian, internal jugular, or (less commonly) a femoral vein and advanced toward the heart until it reaches the superior vena cava or right atrium.
  • This term also refers to implantable ports (often referred to by brand names such as Port-a-Cath or MediPort) which are a central venous line that does not have an external connector; instead, it has a small reservoir that is covered with silicone rubber and is implanted under the skin. Medication is administered intermittently by placing a small needle through the skin, piercing the silicone, into the reservoir. It is possible to leave the ports in the patient's body for years; if this is done however, the port must be accessed monthly.
  • The delivery of therapeutic agents intra-arterially requires selective vascular catheterization, which is accomplished by tailoring the catheter configuration to the vascular anatomy.
  • As used herein, the term “intra-arterial administration” refers to the infusion of liquid substances directly into an artery. This term refers to any type of intra-arterial access devices.
  • This term can refers to an arterial line, or art-line, or a-line, which is a thin catheter inserted into an artery. An arterial line is usually inserted in the wrist (radial artery); but can also be inserted into the elbow (brachial artery), groin (femoral artery), foot (dorsalis pedis artery).
  • This term can refers to syringe driver or syringe pump which is a small infusion pump (some include infuse and withdraw capability), used to gradually administer small amounts of fluid to a patient or for use in chemical and biomedical research. Syringe drivers are also useful for delivering medications over several minutes. In the case of a medication which should be slowly pushed in over the course of several minutes, this device saves staff time and reduces errors.
  • This term also refers to the Catheter-Port Systems. To facilitate the long-term administration of chemotherapeutic agents, percutaneoulsy implantable catheter-port systems have been developed for long-term use. These systems allow easy and repetitive puncture in infusion therapy without causing much harm to the vessels, and their use is comfortable for the patient. Catheter system can be surgically implanted in the gastro duodenal artery or via the subclavian, axillary, or brachial arteries into the common hepatic artery or in the femoral artery. The configuration of the catheter used depended on the vascular anatomy of the patient. The standard catheter-port device consisted of a titanium port reservoir with a silicone rubber membrane at the puncture site and a lateral stem to slip the silicone catheter over. The connection between the silicone catheter and the diagnostic angiographic catheter was reinforced with a small metallic cannula (Liver Intraarterial Chemotherapy: Use of the Femoral Artery for Percutaneous Implantation of Catheter-Port Systems, Karin Anna Herrmann et al. April 2000 Radiology, 215, 294-299).
  • This term can also refers to intra-arterial chemotherapy (IAC) which is an innovative chemotherapy method used to treat liver cancer, as well as cancers that have spread to the liver, such as metastatic pancreatic cancer. IAC is used to send chemotherapy directly into a tumor through a catheter placed in the artery. The goal of IAC is to concentrate the drug inside the tumor and minimize the exposure to healthy tissues. During IAC, a thin catheter is inserted through the femoral artery in the right leg. An angiogram is performed to obtain a “roadmap” of the arteries. This roadmap is then used to insert a line into the hepatic artery, which is the main blood vessel that delivers blood to the liver. Using dye from a syringe to make sure the line is in the correct position, the chemotherapy drug or drugs are injected directly into the artery.
  • As used herein, the term “cancer” refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. This term refers to any type of malignancy (primary or metastases). In particular, this term refers to any malignant proliferative cell disorders such as solid tumor or hematopoietic tumor, including carcinoma, sarcoma, lymphoma, stem cell tumor, blastoma. Preferably, the cancer is selected from the group consisting of hepatic cancer, in particular hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, lung cancer, in particular small cell lung cancer, colorectal cancer, pancreas cancer, breast cancer, ovary cancer, uterine cancer, cervix cancer, head and neck cancer, brain cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma. More preferably, the cancer is selected from the group consisting of hepatocellular carcinoma, acute lymphoblastic leukemia, acute myeloblastic leukemia, chronic myelogenous leukemia, Hodgkin's disease, diffuse large B-cell lymphoma, small cell lung cancer, breast cancer, ovary cancer, blade cancer, multiple myeloma, neuroblastoma, Edwing's sarcoma, osteosarcoma, soft tissue sarcoma, thyroid cancer, prostate cancer, stomach cancer, nephroblastoma, Kaposi's sarcoma, and non-Hodgkins lymphoma. In a preferred embodiment, the cancer is a hepatic cancer, preferably a hepatocellular carcinoma.
  • In a particular embodiment, the Nanoparticles comprise doxorubicin, polyisohexylcyanoacrylate, hydroxypropyl-beta-cyclodextrin and/or randomly methylated-beta cyclodextrin, for use for treating a hepatocellular carcinoma, wherein the Nanoparticles are to be administered by intravenous or intra-arterial infusion for at least 2 hours.
  • The following examples are given for purposes of illustration and not by way of limitation.
  • EXAMPLES Example 1: Manufacturing of Nanoparticles Loaded with Doxorubicin (Nanoparticles Loaded with Doxorubicin)
  • Nanoparticles loaded with Doxorubicin product developed is presented as a sterile lyophilisate for injectable suspension that contains doxorubicin hydrochloride as active ingredient and other excipients including the Nanoparticles polymer, PIHCA. The drug-loaded Nanoparticles are obtained by aqueous emulsion polymerization of isohexylcyanoacrylate (IHCA) monomer dropped in the bulk solution containing the active ingredient doxorubicin and the other excipients (P. Couvreur, B. Kanté, M. Roland, P. Guiot, P. Baudhuin, P. Speiser, J. Pharm. Pharmacol., 1979, 31, 331). At the end of polymerization, a stable suspension of Nanoparticles entrapping doxorubicin is obtained. The Nanoparticles mean size is comprised between 100 nm to 300 nm. The Nanoparticles suspension is then filtered and aseptically filled in glass vials before freeze-drying. Nanoparticles loaded with Doxorubicin freeze-dried product must be kept protected from light and humidity and stored in a refrigerator between 2° C.-8° C., for stability purposes.
  • Raw Materials
  • For 100 ml of total volume of polymerization media:
  • Excipients Quantity (gram)
    Anhydrous glucose 5
    Lactose 0.4
    Poloxamer 188 1
    Hydroxypropyl-beta Cyclodextrin 0.5
    H2O qsp 100 ml
    Anhydrous citric acid (1M) qsp ph = 3 to 4
  • Method for Preparing 100 ml of Polymerization Medium (pH Comprises Between 3 and 4)
      • Excipients are added to about 75 ml of H2O.
      • Excipients are dissolved with magnetic stirring bar
      • Completion with H2O until obtaining a 100 ml volume.
      • pH is eventually adjusted with citric acid 1M
        Method for Preparing Nanoparticles Loaded with Doxorubicin (5 ml Batches)
      • In a 10 ml flask:
      • Add 4,625 ml of polymerization media
      • Add 375 μl of doxorubicin solution at 10 mg/ml
      • Mix with magnetic stirring bar
      • Add 50 μl of pure IHCA solution (density of IHCA=0.980)—Let polymerization occurs up to 2 h30 period of time or more, under magnetic stirring and room temperature
      • Filtration onto 2 μm filters
    Example 2: Assessment of a Model of Adverse Effects Induced after a Single iv Bolus Injection of Nanoparticles Loaded with Doxorubicin in Healthy Wistar Rats
  • Described above randomized, multicenter phase 2-3 evaluating Nanoparticles loaded with Doxorubicin in Advanced HepatoCellular Carcinoma by intraarterial route, has been stopped in 2008 for severe to fatal respiratory distress in treated patients.
  • The aim of the present in vivo study was to establish and assess a rat model able to induce toxicological signs and lung injuries similar to severe pulmonary adverse events observed in said clinical trial. Inventors studied the toxicological effects of a single bolus IV injection of a clinical batch of Nanoparticles loaded with Doxorubicin on healthy Wistar rats at early (24 h, 48 h, 72 h) and late (7 days) monitoring times.
  • The dose levels of 5, 7.5 and 10 mg/kg were chosen according to previous in vivo studies, they respectively correspond to 30 mg/m2, 45 mg/m2 and 60 mg/m2 human doses.
  • Drug Administration
  • The study involved finally 123 male Wistar rats.
  • Rats were weighted and distributed according to their individual body weight to form 5 groups of 24 rats the day of IV treatment.
  • The IV dose was administered as a bolus at a rate of about 1000 μl/min or 3700 μg Doxorubicin-HCl/min in the penis vein under light isoflurane anaesthesia.
      • The rats from groups 1 to 4 received a single IV injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 340 μl of a 3.71 mg/ml suspension to a 250 g rat.
      • The rats from groups 5 to 8 received a single IV injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 510 μl of a 3.71 mg/ml suspension to a 250 g rat.
      • The rats from groups 9 to 12 received a single IV injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 10 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 670 μl of a 3.71 mg/ml suspension to a 250 g rat.
      • The rats from groups 13 to 16 received a single IV injection of Doxorubicin at 7.5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Solution was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 530 μl of a 3.55 mg/ml suspension to a 250 g rat.
      • The rats from groups 17 to 20 (Excipient control group) received a single IV administration of the same volume of Excipient solution that 10 mg/kg of Nanoparticles loaded with Doxorubicin treated group. Solution was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 670 μl solution to a 250 g rat.
      • The rats from group 21 (Untreated control group) received no treatment.
        • Treatment allocation was decided at random and was as disclosed in Table 2.
  • TABLE 2
    Treatment allocation of rats at D0
    Quantity of
    equivalent
    No of Dose Dox-HCl Dose
    animals mg/kg, (mg, rat mg/kg, Scheduled
    per equivalent weight = equivalent Treatment day of
    Group group Treatment Route Dox-HCl 250 g) NP-PIHCA schedule euthanasia
    1 6 Nanoparticles IV 5 1.250 66.50 Q1D × 1 D1
    2 6 Nanoparticles IV 5 1.250 66.50 Q1D × 1 D2
    3 6 Nanoparticles IV 5 1.250 66.50 Q1D × 1 D3
    4 6 Nanoparticles IV 5 1.250 66.50 Q1D × 1 D7
    5 6 Nanoparticles IV 7.5 1.875 99.75 Q1D × 1 D1
    6 6 Nanoparticles IV 7.5 1.875 99.75 Q1D × 1 D2
    7 6 Nanoparticles IV 7.5 1.875 99.75 Q1D × 1 D3
    8 6 Nanoparticles IV 7.5 1.875 99.75 Q1D × 1 D7
    9 6 Nanoparticles IV 10 2.500 133.00 Q1D × 1 D1
    10 6 Nanoparticles IV 10 2.500 133.00 Q1D × 1 D2
    11 6 Nanoparticles IV 10 2.500 133.00 Q1D × 1 D3
    12 5 Nanoparticles IV 10 2.500 133.00 Q1D × 1 D7
    13 6 Doxorubicin IV 7.5 1.875 Q1D × 1 D1
    14 6 Doxorubicin IV 7.5 1.875 Q1D × 1 D2
    15 6 Doxorubicin IV 7.5 1.875 Q1D × 1 D3
    16 6 Doxorubicin IV 7.5 1.875 Q1D × 1 D7
    17 6 Excipient IV Q1D × 1 D1
    Control
    18 6 Excipient IV Q1D × 1 D2
    Control
    19 6 Excipient IV Q1D × 1 D3
    Control
    20 6 Excipient IV Q1D × 1 D7
    Control
    21 4 Untreated
    Control
  • Results
  • First, inventors observed at the highest doses of Nanoparticles loaded with Doxorubicin (7.5 mg/kg and 10 mg/kg) an important number of deaths during the first two days of follow up. These deaths were closely associated with lung injuries characterized by:
      • macroscopic modifications of the lungs,
      • presence of exudates
      • increase in lung weight
      • oedemas, alveolitis
  • Neither mortality, nor lung injuries were observed in the group treated with IV bolus injection of Nanoparticles loaded with Doxorubicin 5 mg/kg, of free doxorubicin at 7.5 mg/kg or with the Nanoparticles excipients.
  • Thus, this study allows to establish rat model of lung injury after IV bolus administration of Nanoparticles loaded with Doxorubicin and to study the prevention of these serious adverse events.
  • For further investigation in the prevention of these adverse events, the dose of 7.5 mg/kg was chosen as the dose allowing to follow the lung injury until Day 2 with limited unscheduled deaths in comparison to the 10 mg/kg dose.
  • Example 3: Investigation of the Impact of Administration Speed on the Tolerance of a Single IV Injection of Nanoparticles Loaded with Doxorubicin in Healthy Wistar Rats
  • To investigate respiratory adverse events observed in clinical trials with Nanoparticles loaded with Doxorubicin, a rat model of lung injury has been set up. Previous studies (Example 2) showed that IV bolus administration of Nanoparticles loaded with Doxorubicin at the dose of 7.5 mg/kg to healthy Wistar rats induced major toxicological effects, with lung injury and respiratory distress. Moreover, 48 h after the injection, in 63% of injected rats, mortality was observed correlated with major lung injury.
  • The aim of the present study was to compare the tolerance of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at the same dose level of 7.5 mg/kg injected intravenously by a single bolus injection (1000 μl/min or 3660 μg Doxorubicin-HCl/min) or by a 2 hours perfusion (4.3 μl/min or 15.6 μg Doxorubicin-HCl/min). The dose level of 5 mg/kg (corresponding to 30 mg/m2 in human) administered by a single bolus IV injection (1000 μl/min or 3660 μg Doxorubicin-HCl/min) was chosen as safe bolus dose based on results of the previous study: no mortality and no lung injuries were observed at this dose. Rat euthanasia was performed 48 h after the treatment, according to scheme appearance of toxicological effects observed in previous studies.
  • Drug Administration
  • The study involved 32 male Wistar rats.
  • Rats were weighted and distributed according to their individual body weight to form 5 groups of 6 rats for IV bolus or 8 rats for IV perfusion the day of the first IV treatment.
  • The IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of perfusion at a rate of 4.3 μl/min (corresponding to 15.6 μg (Dox-HCl)/min) for a 250 g rat (3.66 mg/ml suspension) and by the penis vein in the case of bolus injection at a rate of about 1000 μl/min (corresponding to 3660 μg (Dox-HCl)/min).
  • To determine the real administered dose, each syringe and catheter was weighted before and after treatment.
      • The rats from group 1 received a single IV perfusion injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.05 ml/kg of a 3.66 mg/ml suspension. The perfusion rate was defined as 4.3 μl/min (15.6 (Dox-HCl)/min) for a 250 g rat (2 h perfusion).
      • The rats from group 2 received a single bolus IV injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.05 ml/kg of a 3.66 mg/ml suspension. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min),
      • The rats from group 3 received a single IV bolus injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 5 mg/kg equivalent Doxorubicin HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 1.37 mL/kg of a 3.66 mg/mL suspension. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min).
      • The rats from group 4 (Excipient control group) received a single IV administration of the same volume of excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group. Solution was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.05 ml/kg.
      • The rats from group 5 (Anesthesia/surgery control) were anesthetized and a catheter filled with saline solution and connected to a syringe of saline solution was inserted in the femoral vein. The rats were maintained under anesthesia during 2 hours after the surgery. Treatment allocation was decided according to their individual body weight as follows:
  • TABLE 3
    Treatment allocation of rats at the day of administration
    Quantity of
    equivalent
    Dose Doxo HCl Dose
    No of mg/kg, (mg, rat mg/kg,
    animals/ equivalent weight = equivalent Treatment
    Group group Treatment Route Doxo HCl 250 g) NP-PIHCA schedule
    1 8 Nanoparticles IV, 7.5 1.875 99.75 Q1D × 1
    BA003- perfusion
    07C001PH
    2 6 Nanoparticles IV, 7.5 1.875 99.75 Q1D × 1
    BA003- bolus
    07C001PH
    3 6 Nanoparticles IV, 5 1.250 66.50 Q1D × 1
    BA003- bolus
    07C001PH
    4 6 Excipient IV, Q1D × 1
    Control bolus
    5 6 Anesthesia/
    Surgery
    control
  • Results
  • Mortality
  • In 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus group, two rats over 6 were found dead at D1 after bolus injection and two rats were found dead at D2, corresponding to mortality rate of 4/6 treated rats (66.7%) at the end of the study.
  • In 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV perfusion group, the first treated rat was found dead at D2 after treatment, corresponding to a mortality rate of 1/8 treated rats (12.5%) at the end of the study.
  • In 5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus group, no mortality was observed until the end of the study.
  • In excipient control group and in anesthesia/surgery control group, no mortality was observed until the end of the study.
  • TABLE 4
    Mortality follow-up
    At the end
    Treatment D 1 D 2 of study
    Nanoparticles 7.5 Rat No 6 4 2
    mg/Kg Death 2/6 2/4 4/6
    IV Bolus Mortality 33.3%   50.0%   66.7%  
    Nanoparticles 7.5 Rat No 8 8 7
    mg/Kg Death 0/8 1/8 1/8
    IV perfusion Mortality 0% 12.5%   12.5%  
    Nanoparticles 5 Rat No 6 6 6
    mg/Kg Death 0/6 0/6 0/6
    IV Bolus Mortality 0% 0% 0%
    Anesthesia/ Rat No 6 6 6
    Surgery Control Death 0/6 0/6 0/6
    Mortality 0% 0% 0%
    Excipient Rat No 6 6 6
    Control Death 0/6 0/6 0/6
    IV Bolus Mortality 0% 0% 0%
  • Lungs Examination
  • Major lung injuries were observed in 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus treated group whether dead rats (2/2) or euthanatized rats (3/4). These physiological alterations were characterized by the presence of exudate in the thoracic cavity, the increase of lung weight and hemorrhagic lungs with dark dots.
  • In the 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV perfusion treated group, the inventors observed exudate in the thoracic cavity of one rat: the rat No 1 that was found dead at D2. Moreover, this rat showed hemorrhagic lungs. The inventors did not observe exudate in the thoracic cavity for the others rats of this group. However, they observed hemorrhagic lungs for rat No 2 that showed a decrease of its general health status before euthanasia. It is to note that the color of lungs of rats No 3 and No 4 was not uniform. The last 4 rats of this perfusion group of 8 animals did not show any lung alterations.
  • Inventors did not observe any exudate in the thoracic cavity and any macroscopic lung alterations in 5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus treated group, in Excipient control group and anesthesia/surgery control group.
  • TABLE 5
    Summary of macroscopic lung injury
    Treatment Rat No Incidence of macroscopic injuries
    Nanoparticles7.5 6 5/6
    mg/Kg
    IV Bolus
    Nanoparticles 7.5 8 2/8
    mg/Kg
    IV Perfusion
    Nanoparticles 5 6 0/6
    mg/Kg
    IV Bolus
    Anesthesia/ 6 0/6
    Surgery Control
    Excipient Control 6 0/6
  • Conclusion
  • Inventors previously described a rat model of lung injury after an IV bolus injection (1000 μl/min or 3660 μg Doxorubicin-HCl/min) of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg (Equivalent Human Dose—EHD-=45 mg/m2). Inventors observed, 48 h after the treatment a high mortality (62.5% of treated animals) and strong lung injury with exudates in the thoracic cavity, hemorrhagic lungs with dark dots and a significant increase of lung weight (98% vs control group).
  • In this study, Inventors first confirmed the pulmonary toxicity of an IV bolus injection (1000 μl/min or 3660 μg Doxorubicin-HCl/min) of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg, with similar lung macroscopic observations (exudates in the thoracic cavity, hemorrhagic lungs with dark dots), an increase of lung weight (116.6% vs control group, p=0.0038) and an associated mortality (66.7% of treated animals).
  • Moreover, the present study surprisingly showed that a slow perfusion rate of Nanoparticles loaded with Doxorubicin (2 h perfusion, 4.3 μl/min or 15.6 μg Doxorubicin-HCl/min) at the dose level of 7.5 mg/kg strongly reduced these toxicological side effects compared to a bolus IV injection at the same dose, 48 h after administration. Indeed, mortality was markedly reduced in perfusion group as 87.5% of rats were still alive 48 h after the treatment compared to 33.3% in bolus group at the same dose level. Macroscopic lung injuries were also reduced in perfusion group as only 2/8 rats showed hemorrhagic lungs whereas 5/6 rats showed normal lungs in bolus group at the same dose level. In the same way, the increase of lung weight was weaker and no significant in perfusion group compared to bolus group (75.4%).
  • Example 4: Histological Evaluation of Lung and Heart of Healthy Rats after a Single Intravenous Injection of Nanoparticles Loaded with Doxorubicin. Comparative Study Between IV Bolus and Perfusion Administration
  • Previous studies showed that IV bolus administration of Nanoparticles loaded with Doxorubicin at the dose of 7.5 mg/kg to healthy Wistar rats induced major toxicological effects, with lung injury and respiratory distress. Moreover, in 63-67% of treated rats, mortality was observed within 48 h after the injection and was correlated with major lung injuries (example 2). Further investigations demonstrated that a slow IV administration of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg strongly reduced toxicological side effects compared to a bolus IV injection at the same dose. Neither mortality nor macroscopic lung injury were observed in perfusion group 48 h after the treatment compared to bolus group (87.5% of mortality and lung injury). The increase of lung weight was also significantly weaker in perfusion group (12%) compared to bolus group (166%). These studies also demonstrated that IV bolus injection of Nanoparticles loaded with Doxorubicin at a lower dose level of 5 mg/kg is safe and well tolerated without mortality or lung injury (example 3).
  • The major adverse effects reported in the field of drug-induced pulmonary toxicity are histological changes in the pulmonary parenchyma, the pleura, the airways, the pulmonary vascular system, and the mediastinum. Histological lesions consist in pulmonary changes such as alveolar damage, fibrosis and inflammatory infiltrate (Pereverzeva E et al., Influence of the formulation on the tolerance profile of nanoparticle-bound doxorubicin in healthy rats: focus on cardio- and testicular toxicity. Int J Pharm. 2007, 337(1-2):346-56. Perivascular oedema is known to be a major parameter in the assessment of lung toxicity, particularly in hypersensitivity (Tigani B et al., Resolution of the oedema associated with allergic pulmonary inflammation in rats assessed noninvasively by magnetic resonance imaging. British Journal of Pharmacology. 2003, 140, 2, 239-246). Few data are available in the literature for pulmonary toxicity of doxorubicin.
  • To further investigate the observed lung injuries, a histological evaluation after HES (Hematoxyline-Erythrosine-Safran) staining of lung and heart of Nanoparticles loaded with Doxorubicin injected intravenously in healthy Wistar rats was performed. Histological changes studied were in particular pulmonary perivascular oedema, alveolar damage, pleural inflammation and cellular infiltrates.
  • Treatment/Drug Administration
  • Two experiments have been conducted for this histological study.
  • For the two experiments, the IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of perfusion at a rate of 4.3 μl/min and by the penis vein in the case of bolus injection at a rate of about 1000 μl/min. To determine the real administered dose, each syringe and catheter was weighted before and after treatment.
  • First Experiment (BA003-PKT-010Rv01)
  • The first experiment involved 16 male Wistar rats. Rats were weighted and distributed according to their individual body weight to form 5 groups of 3 rats for IV bolus, and 4 rats for IV perfusion.
      • The rats from group 1 received a single bolus IV injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min),
      • The rats from group 2 received a single IV perfusion injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. The perfusion rate was defined as 4.3 μl/min (15.6 μg (Dox-HCl)/min) (2 h perfusion).
      • The rats from group 3 received a single IV bolus injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 5 mg/kg equivalent Doxorubicin HCl. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min).
      • The rats from group 4 (Anesthesia/surgery control) were anesthetized and a catheter filled with saline solution and connected to a syringe of saline solution was inserted in the femoral vein. The rats were maintained under anesthesia during 2 hours after the surgery.
      • The rats from group 5 (Excipient control group) received a single IV administration of the same volume of excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group.
        Treatment allocation was performed as follows:
  • TABLE 6
    Treatment allocation of rats at the day of administration
    No of Dose mg/kg,
    animals/ equivalent
    Group group Treatment Route Doxo HCl
    1 3 Nanoparticles loaded IV 7.5
    with Doxorubicin bolus
    BA003-07C001PH
    2 4 Nanoparticles loaded IV 7.5
    with Doxorubicin perfusion
    BA003-07C001PH
    3 3 Nanoparticles loaded IV 5
    with Doxorubicin bolus
    BA003-07C001PH
    4 3 Anesthesia/
    Surgery Control
    5 3 Excipients Control IV
    bolus
  • Second Experiment (BA003-PKT-011Rv01)
  • The second experiment involved 14 male Wistar rats. Rats were weighted and distributed according to their individual body weight to form 4 groups of 4 rats for IV bolus, and 3 rats for IV perfusion.
      • The rats from group 1 received a single bolus IV injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min),
      • The rats from group 2 received a single IV perfusion injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. The perfusion rate was defined as 4.3 μl/min (15.6 μg (Dox-HCl)/min) (2 h perfusion).
      • The rats from group 3 received a single IV bolus injection of Nanoparticles loaded with Doxorubicin (clinical batch BA003-07C001PH) at 5 mg/kg equivalent Doxorubicin HCl. The injection rate was defined as 1000 μl/min (3660 μg (Dox-HCl)/min).
      • The rats from group 4 (Excipient control group) received a single IV administration of the same volume of excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group.
  • Treatment allocation was performed as follows:
  • TABLE 7
    Treatment allocation of rats at the day of administration
    No of Dose mg/kg,
    animals/ equivalent
    Group group Treatment Route Doxo HCl
    1 4 Nanoparticles loaded IV 7.5
    with Doxorubicin bolus
    BA003-07C001PH
    2 3 Nanoparticles loaded IV 7.5
    with Doxorubicin perfusion
    BA003-07C001PH
    3 4 Nanoparticles loaded IV 5
    with Doxorubicin bolus
    BA003-07C001PH
    4 3 Excipients Control IV
    bolus
  • Results
  • In both experiments, rats were euthanatized 48 hours after administration of the product by IV intravenous injection of an overdose of pentobarbital.
  • In the first experiment, 1/3 rat was found dead 24 h after 7.5 mg/kg IV bolus administration of Nanoparticles loaded with Doxorubicin and in the second experiment, 3/4 rats were found dead 24 hours or 48 hours after administration of 7.5 mg/kg IV bolus administration of Nanoparticles loaded with Doxorubicin. These observations have to be taken into account in the interpretation of the following histological observations.
  • In these two experiments testing intravenous injection of a clinical batch of Nanoparticles loaded with Doxorubicin in healthy Wistar rats, histological changes were observed, consisting in pulmonary perivascular oedema of variable intensity, occasional alveolar damage and slight pleural inflammation. The main feature observed in treated animals was an enlargement of the perivascular area consisting in an oedematous aspect.
  • Major pulmonary and cardiac damages were observed in the group receiving 7.5 mg/kg of Nanoparticles loaded with Doxorubicin at a bolus rate of about 1000 μL/min (or 3700 μg Doxorubicin-HCl/min) having caused the death of the animals. A major perivascular oedema was observed in 7.5 mg/kg bolus treated group: an increase of a ratio of 21.8 (p=0.0065) was observed in this group compared to control group.
  • The 7.5 mg/kg dose, when administered through a perfusion procedure at a rate of about 4.3 μL/min (or 15.6 μg Doxorubicin-HCl/min), induced mild or moderate pulmonary histological lesions and no cardiac lesions as opposed to the same dose level administered as a bolus injection.
  • The animals in the 5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus group showed only mild pulmonary lesions. Pulmonary perivascular oedema was similar in 5 mg/kg bolus group and 7.5 mg/kg perfusion group and strongly reduced compared to 7.5 mg/kg bolus treated group.
  • Altogether, these results demonstrated that the reduction of the infusion rate of Nanoparticles loaded with Doxorubicin markedly decreased the incidence of fatal respiratory distress in healthy rats mainly characterized by a pulmonary perivascular oedema.
  • Example 5: Investigation of Potential Delayed Toxicological Effects of a Single iv Perfusion of Nanoparticles Loaded with Doxorubicin in Healthy Wistar Rats
  • A 2-hour IV perfusion of Nanoparticles loaded with Doxorubicin (4.2 μL/min or 15.6 μg Doxorubicin-HCl/min for a 3.7 mg/mL suspension) was shown to induce an important decrease of the toxicological effects at this dose level, when comparing to a single bolus IV injection. A decrease of mortality was observed, with 79% of dead rats two days after the treatment in bolus group versus 6.7% in perfusion group. Moreover, a marked reduction of lung injuries was observed two days after the perfusion treatment (examples 3 and 4).
  • In these studies, euthanasia was always performed two days after the administration of Nanoparticles loaded with Doxorubicin whether in bolus treated group or perfusion treated group.
  • The aim of the present study is to investigate potential delayed toxicological effects after a 2 h-perfusion of Nanoparticles loaded with Doxorubicin. In this way, rats treated by a perfusion of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg will be euthanatized seven days after the treatment, compared to rats euthanatized after 48 h as performed in previous studies.
  • Treatment Drug/Administration
  • The study involved 34 male Wistar rats.
  • Rats were weighted and distributed to form 5 groups of 6 to 8 rats. The IV injection was performed under light isoflurane anaesthesia via the femoral vein in the case of a 2 h perfusion of a suspension at 3.69 mg/mL at a rate of 4.2 μL/min or 15.6 μg Doxorubicin HCl/min for a 250 g rat. In the case of the bolus injection via the penis vein, the injection rate was about 1000 μL/min or 3660 μg Doxorubicin HCl/min.
  • To determine the real administered dose, each syringe and catheter was weighted before and after treatment.
      • The rats from group 1 will receive a single IV bolus injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.04 mL/kg of a 3.69 mg/mL suspension. The injection rate was defined as 1000 μL/min or 3660 μg Doxorubicin HCl/min.
      • The rats from group 2 received a single IV perfusion of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.04 mL/kg of a 3.69 mg/mL suspension. The perfusion rate was defined as 4.2 μL/min or 15.6 μg Doxorubicin HCl/min for a 250 g rat (2 h perfusion). Rats were euthanatized 48 h after the treatment.
      • The rats from group 3 received a single IV perfusion of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxorubicin HCl. Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.04 mL/kg of a 3.69 mg/mL suspension. The perfusion rate was defined as 4.2 μL/min or 15.6 μg Doxorubicin HCl/min for a 250 g rat (2 h perfusion). Rats were euthanatized 7 days after the treatment.
      • The rats from group 4 (Excipient control group) received a single IV administration of the same volume of Excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group. Solution was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.04 mL/kg. The injection rate was defined as 1000 μL/min. Rats were euthanatized 48 h after the treatment.
      • The rats from group 5 (Excipient control group) received a single IV administration of the same volume of Excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group. Solution was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.04 mL/kg. The injection rate was defined as 1000 μL/min. Rats were euthanatized 7 days after the treatment.
  • Treatment allocation was performed as follows:
  • TABLE 8
    Treatment allocation of rats the day of administration
    Quantity of
    equivalent
    No of Dose Dox-HCl Dose
    animals mg/kg, (mg, rat mg/kg,
    per equivalent weight = equivalent
    Group group Treatment Route Dox-HCl 250 g) PIHCA Euthanasia
    1 6 Nanoparticles IV, 7.5 1.875 99.75 D2
    loaded with bolus
    Doxorubicin
    BA003-
    07C001PH
    2 8 Nanoparticles IV, 7.5 1.875 99.75 D2
    loaded with perfusion
    Doxorubicin
    BA003-
    07C001PH
    3 8 Nanoparticles IV, 7.5 1.875 99.75 D7
    loaded with perfusion
    Doxorubicin
    BA003-
    07C001PH
    4 6 Excipient IV, D2
    Control bolus
    5 6 Excipient IV, D7
    Control bolus
  • Mortality
  • In bolus group, three rats dead 24 h after the treatment (50% of injected rats) and one rat was found dead 48 h after the treatment, corresponding to a mortality rate of 66.7% at the end of the monitoring.
  • In the perfusion group, two rats dead at the end of the perfusion and one rat was found dead 24 h after the treatment. The mortality rate was about 20% at the end of the monitoring at D7 considering all rats infused at D0.
  • No mortality was observed in Excipient control group.
  • TABLE 9
    Mortality follow-up during the study for Nanoparticles loaded with
    Doxorubicin treated groups (NA = not applicable)
    At the
    end of the
    Treatment D0 D1 D2 D7 follow-up
    Nanoparticles loaded with Number of 6 6 3 NA 2
    Doxorubicin 7.5 mg/kg iv rats
    bolus Death 0/6 3/6 NA 4/6
    Mortality 0 50 33.3 NA 66.7
    (%)
    Nanoparticles loaded with Number of 16 13 12 7 7
    Doxorubicin 7.5 mg/kg iv rats
    perfusion Death 2/15* 1/13 0/12 0/7 3/15
    Mortality 13.3 7.7 0 0 20
    (%)
    *Withdrawal of rat n°13 of Nanoparticles loaded with Doxorubicin perfusion group from the follow-up because of a drug outflow from the catheter during the infusion time at D0.
  • Macroscopic Lungs Examination
  • Major lung injuries were observed in 7.5 mg/kg IV bolus treated group whether dead rats or one of the two surviving rats (5/6 rats). These physiological alterations were characterized by the presence of fluid in the thoracic cavity, the increase of lung weight, hemorrhagic lungs and/or lungs with dark dots.
  • We did not observe any exudate in the thoracic cavity in 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV perfusion group. Nevertheless, two rats showed changes in lung color, as rat No 12 dead at D1 (with dark and hemorrhagic lungs) and rat No 4 euthanatized at D7 (clear lungs with dark dots).
  • It is to note that rats in Excipient control group showed lungs with not uniform color compared to the previous studies.
  • TABLE 10
    Summary of incidence of macroscopic lung injuries
    Incidence of
    Number macroscopic
    Treatment of rats lung injuries
    Nanoparticles loaded with 6 5/6 
    Doxorubicin
    7.5 mg/kg iv bolus
    Nanoparticles loaded with 15 2/15
    Doxorubicin
    7.5 mg/kg iv perfusion
    Excipient control 12 0/12
  • Lung Weight
  • The mean lung weight of each Nanoparticles loaded with Doxorubicin treated group was compared to the mean lung weight of Excipient control group.
  • The increase of lung weight in 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus group was important and statistically significant compared to Excipient control group (131%, p=0.003).
  • The increase of lung weight compared to control group was lower and not significant in 7.5 mg/kg IV perfusion group at D2 (31%, p=0.154). At D7, this increase of lung weight in perfusion group compared to control group was similar to the one observed at D2 (40%, p=0.178).
  • Conclusion
  • In the present study, we showed that a two-hour perfusion of Nanoparticles loaded with Doxorubicin at the dose level of 7.5 mg/kg did not induce delayed toxicological effects 7 days after the treatment. Indeed, most of the rats did not show lungs injury (86% of injected rats) in perfusion group 7 days after the treatment. Moreover, no difference was observed on the various observed organs two days or seven days after the treatment administration by perfusion.
  • Example 6: Investigation of Toxicological Effects after a Single IV Perfusion of 150 Min of Nanoparticles Loaded with Doxorubicin in Healthy Wistar Rats
  • We previously described (example 3) that a slow perfusion rate of about 120 min of Nanoparticles loaded with Doxorubicin (4.3 μL/min or 15.6 μg Doxo HCl/min) strongly reduced toxicological side effects compared to IV bolus injection at the same dose level.
  • The aim of the present study was to compare the impact on tolerance and lung toxicity of perfusion speed of Nanoparticles loaded with Doxorubicin suspension at the dose level of 7.5 mg/kg.
  • In this way, Nanoparticles loaded with Doxorubicin were administered by IV perfusion at the rates of 12.5 μg/min (corresponding to a perfusion of 150 min for a 250 g rat and for a suspension of Nanoparticles loaded with Doxorubicin at 3.7 mg/mL Doxo HCl equivalent). Rat euthanasia was performed 48 h after the treatment, according to the scheme appearance of toxicological observed effects.
  • Drug Administration
  • The study involved 41 male Wistar rats.
  • Rats were distributed at random to form 6 groups of 6 rats (bolus injection groups), 12 rats (Nanoparticles loaded with Doxorubicin perfusion group) and 4 rats (excipient solution perfusion) the day of the first IV treatment.
  • The IV injection was performed under light isoflurane anesthesia via the femoral vein in the case of a 150 min perfusion of at a rate of 12.5 μg Doxo HCl/min or 3.4 μL/min for a 250 g rat and a suspension at 3.70 mg/mL. In the case of the bolus injection via the penis vein, the injection rate was about 1000 μL/min or 3660 μg Doxo HCl/min.
  • To determine the real administered dose, each syringe and catheter was weighted before and after treatment.
      • The rats from group 1 received a single IV perfusion injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxo HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.02 ml/kg of a 3.70 mg/ml suspension. The perfusion rate was defined as 3.4 μL/min or 12.5 Doxo HCl/min for a 250 g rat (150 min perfusion).
      • The rats from group 2 received a single bolus IV injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 7.5 mg/kg equivalent Doxo HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.02 ml/kg of a 3.70 mg/ml suspension. The injection rate was defined as 1000 μL/min or 3660 μg Doxo HCl/min.
      • The rats from group 3 received a single IV bolus injection of Nanoparticles loaded with Doxorubicin (batch BA003-07C001PH) at 5 mg/kg equivalent Doxo HCl (Q1D×1). Drug suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 1.34 ml/kg of a 3.70 mg/ml suspension. The injection rate was defined as 1000 μL/min or 3660 μg Doxo HCl/min.
      • The rats from group 4 (Nanoparticles group) received a single IV administration of the same volume of Nanoparticles that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group. Suspension was administered according to the body weight of rat determined just before the administration, so as to administer a dose volume of 2.02 ml/kg. The injection rate was defined as 1000 μL/min.
      • The rats from groups 5 and 6 received a single IV administration of the same volume of Excipient solution that 7.5 mg/kg Nanoparticles loaded with Doxorubicin treated group at a bolus rate of 1000 μL/min (Excipient bolus control group) and a perfusion rate of 3.4 and 4.2 μL/min (Excipient perfusion control group). Solution was administered according to the body weight of rat determined just before the administration.
  • Treatment allocation was performed as follows:
  • TABLE 11
    Treatment allocation of rats at the day of administration
    Quantity of Dose
    No of Dose mg/kg, equivalent Dox- mg/kg,
    animals equivalent HCl (mg, rat equivalent
    Group per group Treatment Route Dox-HCl weight = 250 g) PIHCA
    1 12 Nanoparticles IV, 7.5 1.875 99.75
    loaded with perfusion
    Doxorubicin
    BA003-07C001PH
    2 6 Nanoparticles IV, 7.5 1.875 99.75
    loaded with bolus
    Doxorubicin
    BA003-07C001PH
    3 6 Nanoparticles IV, 5 1.250 66.5
    loaded with bolus
    Doxorubicin
    BA003-07C001PH
    4 6 Nanoparticles IV, 99.75
    bolus
    5 6 Excipient Control IV,
    bolus
    6 4 Excipient Control IV,
    perfusion
  • Mortality
  • Mortality was observed only in 7.5 mg/kg IV bolus treated group. Four rats were found dead 48 h after the treatment (66.7% of injected rats).
  • There was no difference in survival between Excipient control group, 7.5 mg/kg IV perfusion group, 5 mg/kg bolus group and Nanoparticles group.
  • TABLE 12
    Mortality follow-up
    Treatment Mortality at D 1 Mortality at D 2
    Nanoparticles loaded with 0/6 (0%) 4/6 (66.7%)
    Doxorubicin BA003-07C001PH,
    7.5 mg/kg IV bolus
    Nanoparticles loaded with 0/12 (0%) 0/12 (0%)
    Doxorubicin BA003-07C001PH,
    7.5 mg/kg IV perfusion
    Nanoparticles loaded with 0/6 (0%) 0/6 (0%)
    Doxorubicin BA003-07C001PH,
    5 mg/kg IV bolus
    Nanoparticles IV bolus 0/6 (0%) 0/6 (0%)
    Excipient Control IV bolus 0/6 (0%) 0/6 (0%)
  • Macroscopic Lungs Examination
  • Major lung injuries were observed in 7.5 mg/kg IV bolus treated group. These physiological alterations were characterized by the presence of fluid in the thoracic cavity, the increase of lung weight and hemorrhagic lungs for dead rats. Lungs with non homogeneous color and with black dots were observed for one surviving rat/2. We did not observe any exudates in the thoracic cavity and no macroscopic lung alterations in 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV perfusion group, 5 mg/kg IV bolus treated group, Nanoparticles group and in Excipient control bolus group. It is to note that lungs could present non homogeneous color in these groups.
  • No macroscopic lung alteration was observed in Excipient control perfusion groups (3.4 or 4.2 μL/min) euthanatized just after the treatment.
  • TABLE 13
    Summary of incidence of macroscopic lung injuries
    Incidence of
    Number macroscopic
    Treatment of rats lung injuries
    Nanoparticles loaded with 6 5/6
    Doxorubicin BA003-07C001PH,
    7.5 mg/kg IV bolus
    Nanoparticles loaded with 12  0/12
    Doxorubicin BA003-07C001PH
    7.5 mg/kg IV perfusion
    150 min
    Nanoparticles loaded with 6 0/6
    Doxorubicin BA003-07C001PH
    5 mg/kg IV bolus
    Nanoparticles IV bolus 6 0/6
    Excipient bolus control 6 0/6
    Excipient perfusion control 4 0/4
  • Lung Weight
  • The mean lung weight of each Nanoparticles loaded with Doxorubicin was compared to the mean lung weight of Excipient bolus control group.
  • The increase of lung weight in 7.5 mg/kg Nanoparticles loaded with Doxorubicin IV bolus group was important compared to Excipient bolus control group (124.2%, p=0.007). No increase of lung weight was observed in 7.5 mg/kg Nanoparticles loaded with Doxorubicin perfusion group compared to control group (2.1%, p=0.472). Moreover, the increase of lung weight in 5 mg/kg and Nanoparticles groups was minor compared to excipient bolus control group with respectively 5.4% (p=0.349) and 7.9% (p=0.051).
  • Conclusion
  • The present study showed that decreasing the perfusion speed from 15.6 μg Doxo HCl/min to 12.5 μg/min still reduced the impact of the treatment on lungs, as no mortality was observed in perfusion group and the mean lung weight was similar to control excipient group. In comparison, the mean lung weight was increased of 124% in 7.5 mg/kg bolus group.
  • Example 7: Clinical Study Phase III
  • The study primarily aims at demonstrating the efficacy of slow IV infusions of Nanoparticles loaded with Doxorubicin in patients with advanced HCC.
  • Trial Design
  • The study is a multicenter, randomized, controlled, open-label study comparing the efficacy and safety of slow repeated intravenous infusion of 2 doses of Nanoparticles loaded with Doxorubicin (20 mg/m2 and 30 mg/m2) to those of Best Supportive Care (BSC) in patients suffering from advanced Hepatocellular Carcinoma (HCC) after failure or intolerance to Sorafenib. The study will be carried out at multiple hepatology or oncology centers that manage patients with HCC with or without cirrhosis. Based on the survival data obtained in the phase II clinical trial, it has been calculated that 390 patients have to be included to adequately compare the efficacy of Nanoparticles loaded with Doxorubicin 20 mg/m2 and Nanoparticles loaded with Doxorubicin 30 mg/m2 to that of BSC. A safety evaluation will be carried out on a regular basis at least twice a year by the Data Safety Monitoring Board (DSMB) and every 25 patients or every 50 infusions of Nanoparticles loaded with Doxorubicin whichever comes first. Patients who meet the inclusion/exclusion criteria will be randomized according to a 1:1:1 ratio to receive 20 mg/m2 of Nanoparticles loaded with Doxorubicin or 30 mg/m2 of Nanoparticles loaded with Doxorubicin, or Best Supportive Care (BSC). Only those patients whose survival expectations is longer than 2 months and could receive more than 1 Nanoparticles loaded with Doxorubicin infusion could be included.
  • The study duration for each patient includes a screening period of 28 days maximum before randomization. The patient will then be randomized and either infused Nanoparticles loaded with Doxorubicin 20 mg/m2 or Nanoparticles loaded with Doxorubicin 30 mg/m2 over 6 hours through the IV route or given BSC. Patients will receive as many cycles as possible up to unequivocal tumor progression (assessed by the investigator), cure or occurrence of unacceptable toxicity. They will be evaluated for efficacy assessment every 2 months.
  • The patients randomized in Nanoparticles loaded with Doxorubicin groups will be hospitalized from Day 0 of each cycle of treatment and will be discharged on Day 3. Study treatment (20 or 30 mg/m2 of Nanoparticles loaded with Doxorubicin) will be administered by infusion over 6 hours through intravenous route at Day 1 and will be repeated every 4 weeks until unequivocal progression (assessed by the investigator) or toxicity. A study visit will be performed on Day 14 of each cycle. Two months after last study drug administration, a End of Study visit will be performed.
  • The patients randomized in control group (“Best supportive care” group) will be receiving treatment as usual and will be followed according to the center's usual practices. Study visits at Day 1 and Day 14 of each cycle will be mandatory.
  • For patient randomized in Nanoparticles loaded with Doxorubicin group, the total dose of Doxorubicin will not exceed 450 mg/m2.
  • The Follow up survival status of all patients (including patients who prematurely withdraw) will be maintained every 3 months until death.
  • Subject Selection
  • Inclusion Criteria
  • All patients included in the study have to meet the following criteria for inclusion in the study:
  • 1. Male or non-pregnant, non-breast feeding female;
  • 2. Aged ≧18 years;
  • 3. Patient with
      • advanced HCC (BCLC-C according to BCLC staging classification) having progressed (RECIST criteria) under sorafenib therapy or intolerant to sorafenib, or; intermediate HCC (BCLC-B) non eligible or nonresponders to transarterial chemoembolization (TACE), and having progressed under or intolerant to sorafenib therapy
  • 4. HCC diagnosed according to the AASLD criteria:
      • Cyto-histology criteria;
      • Non-invasive criteria:
        • Nodule ≧20 mm: one imaging technique among MRI and CT scan showing arterial enhancement;
        • Nodule 10-20 mm: two imaging techniques showing arterial enhancement and portal wash-out;
  • 5. Without cirrhosis or with non-decompensated cirrhosis and a Child-Pugh score from A5 to B7 included
  • 6. ECOG Performance Status 0 or 1;
  • 7. Laboratory tests as follows:
      • Platelets ≧50,000/mm3
      • Neutrophil count ≧1000/mm3
      • Hemoglobin ≧10 g/dL
      • Serum transaminases <5 ULN (NCI/CTC grades 0, 1, or 2)
      • Alkaline phosphatases <5 ULN (NCI/CTC grades 0, 1, or 2)
      • Serum bilirubin <35 μM/L (or 2.0 mg/dL);
  • 8. Signed and dated written informed consent form.

Claims (25)

1-15. (canceled)
16. A method for reducing, in a cancer patient, the occurrence or severity of macroscopic lung injuries induced by nanoparticles that comprise:
at least one chemotherapeutic antitumoral agent that is doxorubicin or a pharmaceutically acceptable salt thereof,
at least one poly(alkylcyanoacrylate), and
at least one cyclodextrin,
the method comprising administering the nanoparticles to the cancer patient by intravenous infusion for at least 2 hours; wherein
the macroscopic lung injuries induced by the nanoparticles do not occur at the time of infusion;
the dosage of doxorubicin or pharmaceutically acceptable salt thereof administered by the intravenous infusion is from about 10 to about 30 mg/m2; and
the method treats the cancer of the cancer patient.
17. The method according to claim 16, wherein the nanoparticles are administered by intravenous infusion for between 2 and 24 hours.
18. The method according to claim 16, wherein the nanoparticles are administered by intravenous infusion for between 4 and 12 hours.
19. The method according to claim 16, wherein the nanoparticles are administered by intravenous infusion for about 6 hours.
20. The method according to claim 16, wherein the at least one poly(alkylcyanoacrylate) is a poly(C6-C8 alkylcyanoacrylate).
21. The method according to claim 16, wherein the at least one poly(alkylcyanoacrylate) is a polyisohexylcyanoacrylate.
22. The method according to claim 16, wherein:
the at least one chemotherapeutic antitumoral agent is at a concentration from 0.01 to 200 mg/g of the nanoparticles,
the at least one cyclodextrin is in an amount of from 0.1% to 70% w/w of the nanoparticles, and
the at least one poly(alkylcyanoacrylate) is in an amount of from 1% to 25% w/w of the nanoparticles.
23. The method according to claim 22, wherein the at least one chemotherapeutic antitumoral agent is at a concentration from about 1 to about 50 mg/g of the nanoparticles.
24. The method according to claim 22, wherein the at least one cyclodextrin is in an amount of from about 1% to about 30% w/w of the nanoparticles.
25. The method according to claim 22, wherein the at least one poly(alkylcyanoacrylate) is in an amount of from about 5% to about 15% w/w of the nanoparticles.
26. The method according to claim 16, wherein the dosage of doxorubicin administered by the intravenous infusion is from about 20 to about 30 mg/m2.
27. The method according to claim 16, wherein the dosage of doxorubicin administered by the intravenous infusion is about 30 mg/m2.
28. The method according to claim 16, wherein the cancer is a hepatocellular carcinoma.
29. The method according to claim 28, wherein the cancer is an advanced hepatocellular carcinoma after failure or intolerance to sorafenib.
30. The method according to claim 16, wherein the cancer is a solid tumor.
31. The method according to claim 16, wherein the cancer is a hematopoietic tumor.
32. The method according to claim 16, wherein the macroscopic lung injuries induced by the nanoparticles are fatal or life-threatening macroscopic lung injuries.
33. The method according to claim 16, wherein
the nanoparticles are administered by intravenous infusion for about 6 hours;
the dosage of doxorubicin or pharmaceutically acceptable salt thereof administered by the intravenous infusion is from about 20 to about 30 mg/m2; and
the cancer is an advanced hepatocellular carcinoma after failure or intolerance to sorafenib.
34. The method according to claim 16, wherein the method reduces the occurrence of macroscopic lung injuries such that the macroscopic lung injuries are prevented.
35. The method according to claim 16, wherein the macroscopic lung injuries induced by the nanoparticles occur two or more days after infusion.
36. A method for reducing, in a cancer patient, the mortality associated with macroscopic lung injuries induced by nanoparticles that comprise:
at least one chemotherapeutic antitumoral agent that is doxorubicin or a pharmaceutically acceptable salt thereof,
at least one poly(alkylcyanoacrylate), and
at least one cyclodextrin,
the method comprising administering the nanoparticles to the cancer patient by intravenous infusion for at least 2 hours; wherein
the macroscopic lung injuries induced by the nanoparticles do not occur at the time of infusion;
the dosage of doxorubicin or pharmaceutically acceptable salt thereof administered by the intravenous infusion is from about 10 to about 30 mg/m2; and
the method treats the cancer of the cancer patient.
37. A method for reducing, in a cancer patient, the occurrence of fatal respiratory distress induced by nanoparticles that comprise:
at least one chemotherapeutic antitumoral agent that is doxorubicin or a pharmaceutically acceptable salt thereof,
at least one poly(alkylcyanoacrylate), and
at least one cyclodextrin,
the method comprising administering the nanoparticles to the cancer patient by intravenous infusion for at least 2 hours; wherein
the fatal respiratory distress induced by the nanoparticles does not occur at the time of infusion;
the dosage of doxorubicin or pharmaceutically acceptable salt thereof administered by the intravenous infusion is from about 10 to about 30 mg/m2; and
the method treats the cancer of the cancer patient.
38. A liquid pharmaceutical composition comprising a suspension of nanoparticles that comprise:
at least one chemotherapeutic antitumoral agent that is doxorubicin or a pharmaceutically acceptable salt thereof,
at least one poly(alkylcyanoacrylate), and
at least one cyclodextrin,
wherein the composition is formulated for administering a dosage of about 10 to about 30 mg/m2 of doxorubicin or a pharmaceutically acceptable salt thereof to a cancer patient by intravenous infusion for an infusion period of at least 2 hours;
wherein administering the dosage for the infusion period reduces, relative to bolus infusion, the occurrence or severity of macroscopic lung injuries induced by the nanoparticles that do not occur at the time of infusion.
39. A continuous infusion system comprising a liquid pharmaceutical composition that comprises a suspension of nanoparticles that comprise:
at least one chemotherapeutic antitumoral agent that is doxorubicin or a pharmaceutically acceptable salt thereof,
at least one poly(alkylcyanoacrylate), and
at least one cyclodextrin,
wherein the system is configured for administering a dosage of about 10 to about 30 mg/m2 of doxorubicin or a pharmaceutically acceptable salt thereof to a cancer patient by intravenous infusion for an infusion period of at least 2 hours;
wherein administering the dosage for the infusion period reduces, relative to bolus infusion, the occurrence or severity of macroscopic lung injuries induced by the nanoparticles that do not occur at the time of infusion.
US15/673,661 2011-03-31 2017-08-10 Nanoparticles loaded with chemotherapeutic antitumoral drug Abandoned US20170333341A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/673,661 US20170333341A1 (en) 2011-03-31 2017-08-10 Nanoparticles loaded with chemotherapeutic antitumoral drug

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11305364.9A EP2508207B1 (en) 2011-03-31 2011-03-31 Nanoparticles loaded with chemotherapeutic antitumoral Drug
EP11305364.9 2011-03-31
PCT/EP2012/055756 WO2012131018A1 (en) 2011-03-31 2012-03-30 Nanoparticles loaded with chemotherapeutic antitumoral drug
US201314009021A 2013-09-30 2013-09-30
US15/673,661 US20170333341A1 (en) 2011-03-31 2017-08-10 Nanoparticles loaded with chemotherapeutic antitumoral drug

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2012/055756 Continuation WO2012131018A1 (en) 2011-03-31 2012-03-30 Nanoparticles loaded with chemotherapeutic antitumoral drug
US14/009,021 Continuation US9763874B2 (en) 2011-03-31 2012-03-30 Nanoparticles loaded with chemotherapeutic antitumoral drug

Publications (1)

Publication Number Publication Date
US20170333341A1 true US20170333341A1 (en) 2017-11-23

Family

ID=44513370

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/009,021 Expired - Fee Related US9763874B2 (en) 2011-03-31 2012-03-30 Nanoparticles loaded with chemotherapeutic antitumoral drug
US15/673,661 Abandoned US20170333341A1 (en) 2011-03-31 2017-08-10 Nanoparticles loaded with chemotherapeutic antitumoral drug

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/009,021 Expired - Fee Related US9763874B2 (en) 2011-03-31 2012-03-30 Nanoparticles loaded with chemotherapeutic antitumoral drug

Country Status (26)

Country Link
US (2) US9763874B2 (en)
EP (2) EP2508207B1 (en)
JP (1) JP5523638B2 (en)
KR (1) KR101578273B1 (en)
CN (2) CN103379920A (en)
AU (1) AU2012233666B2 (en)
BR (1) BR112013019105A2 (en)
CA (1) CA2822177C (en)
CY (1) CY1117868T1 (en)
DK (2) DK2508207T3 (en)
ES (2) ES2424476T3 (en)
HK (1) HK1192841A1 (en)
HR (2) HRP20130702T1 (en)
HU (1) HUE028465T2 (en)
IL (1) IL227466A (en)
ME (1) ME02470B (en)
MX (1) MX344704B (en)
MY (1) MY162339A (en)
PL (1) PL2508207T3 (en)
PT (1) PT2508207E (en)
RS (1) RS52880B (en)
RU (1) RU2616494C2 (en)
SG (1) SG193453A1 (en)
SI (1) SI2691116T1 (en)
SM (1) SMT201600197B (en)
WO (1) WO2012131018A1 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2508207T3 (en) 2011-03-31 2013-07-29 Bioalliance Pharma Nanoparticles charged with chemotherapeutic antitumor agent
CN104603194B (en) * 2012-07-17 2017-11-24 陶氏环球技术有限责任公司 Composition comprising organic liquid diluent and low-viscosity cellulose ether
WO2015130585A1 (en) * 2014-02-28 2015-09-03 Merck Sharp & Dohme Corp. Method for treating cancer
CA2946149A1 (en) * 2014-04-18 2015-10-22 Pfizer Inc. Methods of treating cancers with therapeutic nanoparticles
US9907786B2 (en) 2014-10-21 2018-03-06 Ions Pharmaceutical S.À R.L. Therapeutic compositions containing harmine and isovanillin components, and methods of use thereof
US10092550B2 (en) 2014-10-21 2018-10-09 Ions Pharmaceutical S.À R.L. Therapeutic compositions containing curcumin, harmine, and isovanillin components, and methods of use thereof
US20160106722A1 (en) 2014-10-21 2016-04-21 Life Plus, LLC Human therapeutic agents
AU2016298210B2 (en) 2015-07-28 2021-12-09 Board Of Regents, The University Of Texas System Implant compositions for the unidirectional delivery of therapeutic compounds to the brain
WO2017046037A1 (en) 2015-09-14 2017-03-23 Onxeo Nanoparticles loaded with active ingredients, their process of preparation and their uses
CA3015241A1 (en) * 2016-02-22 2017-08-31 Onxeo Combination therapies comprising immuno-oncology agents and belinostat
NO342271B1 (en) * 2016-09-29 2018-04-30 Sintef Tto As A new drug delivery system for treatment of cancer
US10646540B2 (en) * 2016-11-18 2020-05-12 City Of Hope Peptide inhibitors of twist
CN106511365A (en) * 2016-12-17 2017-03-22 郑州郑先医药科技有限公司 Western medicine composition for treating liver cirrhosis
CN111989087A (en) * 2018-03-27 2020-11-24 新泰福托特股份有限公司 Poly (alkyl cyanoacrylate) nanoparticles for the treatment of cancer
MX2019015508A (en) 2019-12-18 2021-06-21 Univ Guadalajara Nanoparticles for treating cancer.

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4310667A (en) 1976-04-22 1982-01-12 Agence Nationale De Valorisation De La Recherche (Anvar) 2-N Quaternary ammonium salt derivatives of 9-hydroxy ellipticine
FR2775435B1 (en) 1998-02-27 2000-05-26 Bioalliance Pharma NANOPARTICLES COMPRISING AT LEAST ONE POLYMER AND AT LEAST ONE COMPOUND CAPABLE OF COMPLEXING ONE OR MORE ACTIVE INGREDIENTS
IL165392A0 (en) * 2002-06-05 2006-01-15 Genentech Inc Compositions and methods for liver growth and liver protection
US20040202665A1 (en) * 2002-07-01 2004-10-14 Janette Lazarovits Compositions and methods for therapeutic treatment
JP5201763B2 (en) * 2007-02-28 2013-06-05 昇一 城武 Method for producing mixed fine particle capsules comprising particles having different average particle sizes
US20090061009A1 (en) * 2007-08-29 2009-03-05 Joseph Schwarz Composition and Method of Treatment of Bacterial Infections
EP2050747A1 (en) 2007-10-12 2009-04-22 BioAlliance Pharma Dimers of harmol or of its derivatives and uses thereof
CA2703149A1 (en) * 2007-10-26 2009-04-30 Pfizer Products Inc. Idarubicin for the treatment of lymphoma in a dog
US20110003710A1 (en) * 2008-02-28 2011-01-06 John Hopkins University Selectin ligands useful in the diagnosis and treatment of cancer
EP2153821A1 (en) * 2008-08-06 2010-02-17 BioAlliance Pharma Oral formulations of camptothecin derivatives
DK2508207T3 (en) 2011-03-31 2013-07-29 Bioalliance Pharma Nanoparticles charged with chemotherapeutic antitumor agent

Also Published As

Publication number Publication date
CA2822177C (en) 2016-02-16
JP5523638B2 (en) 2014-06-18
IL227466A (en) 2016-12-29
HRP20160708T1 (en) 2016-07-15
US20140024610A1 (en) 2014-01-23
PT2508207E (en) 2013-07-22
HK1192841A1 (en) 2014-09-05
EP2691116B8 (en) 2016-06-01
EP2508207B1 (en) 2013-05-08
EP2691116B1 (en) 2016-03-23
CN105233301A (en) 2016-01-13
ES2577955T3 (en) 2016-07-19
ES2424476T3 (en) 2013-10-02
HRP20130702T1 (en) 2013-09-30
SI2691116T1 (en) 2016-10-28
EP2508207A1 (en) 2012-10-10
KR101578273B1 (en) 2015-12-16
SMT201600197B (en) 2016-08-31
CY1117868T1 (en) 2017-05-17
US9763874B2 (en) 2017-09-19
EP2691116A1 (en) 2014-02-05
IL227466A0 (en) 2013-09-30
WO2012131018A1 (en) 2012-10-04
PL2508207T3 (en) 2013-09-30
MY162339A (en) 2017-06-15
JP2014503577A (en) 2014-02-13
CN103379920A (en) 2013-10-30
RU2616494C2 (en) 2017-04-17
RS52880B (en) 2014-02-28
MX344704B (en) 2017-01-03
RU2013128934A (en) 2015-01-10
DK2508207T3 (en) 2013-07-29
AU2012233666B2 (en) 2014-01-16
HUE028465T2 (en) 2016-12-28
KR20130114240A (en) 2013-10-16
CA2822177A1 (en) 2012-10-04
DK2691116T3 (en) 2016-07-04
BR112013019105A2 (en) 2019-04-02
AU2012233666A1 (en) 2013-07-11
ME02470B (en) 2017-02-20
MX2013007581A (en) 2013-11-01
SG193453A1 (en) 2013-10-30

Similar Documents

Publication Publication Date Title
US9763874B2 (en) Nanoparticles loaded with chemotherapeutic antitumoral drug
Zhang et al. Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats
KR101943230B1 (en) Nanoparticle comprising rapamycin and albumin as anticancer agent
KR20130109025A (en) Methods of treating bladder cancer
AU2017246316A1 (en) Methods for solid tumor treatment
CN109771663B (en) Preparation and application of acid-responsive anticancer nano-drug
IL160927A (en) Compositions comprising suspensions of nanoparticulates of at least one antimitotic drug and their use as medicaments
CN108495619A (en) Echinomycin preparation and preparation method thereof and application method
WO2013149538A1 (en) Pharmaceutical composition
US20170071866A1 (en) Nanoparticles loaded with active ingredients, their process of preparation, and their uses
EP4153180A1 (en) Combination therapy for treating cancer
JP4020256B2 (en) Local treatment for prostate cancer
Chawla et al. Longer term cardiac safety of aldoxorubicin
WO2017046037A1 (en) Nanoparticles loaded with active ingredients, their process of preparation and their uses

Legal Events

Date Code Title Description
AS Assignment

Owner name: ONXEO S.A., FRANCE

Free format text: CHANGE OF NAME;ASSIGNOR:BIOALLIANCE PHARMA S.A.;REEL/FRAME:043267/0228

Effective date: 20150302

Owner name: BIOALLIANCE PHARMA SA, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PISANI, EMILIA;LEBEL-BINAY, SOPHIE;POLARD, VALERIE;SIGNING DATES FROM 20131003 TO 20131011;REEL/FRAME:043267/0130

STCB Information on status: application discontinuation

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