WO2014191989A1 - Conjugate of a taxane and biotin and uses thereof - Google Patents

Conjugate of a taxane and biotin and uses thereof Download PDF

Info

Publication number
WO2014191989A1
WO2014191989A1 PCT/IL2014/050470 IL2014050470W WO2014191989A1 WO 2014191989 A1 WO2014191989 A1 WO 2014191989A1 IL 2014050470 W IL2014050470 W IL 2014050470W WO 2014191989 A1 WO2014191989 A1 WO 2014191989A1
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
mice
treatment
cancer
idd
Prior art date
Application number
PCT/IL2014/050470
Other languages
French (fr)
Inventor
Anwar Rayan
Original Assignee
Idd Therapeutics Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Idd Therapeutics Ltd. filed Critical Idd Therapeutics Ltd.
Publication of WO2014191989A1 publication Critical patent/WO2014191989A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • 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
    • A61K47/51Medicinal 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 non-active ingredient being a modifying agent
    • A61K47/54Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • 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
    • A61K47/51Medicinal 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 non-active ingredient being a modifying agent
    • A61K47/54Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/555Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells
    • A61K47/557Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells the modifying agent being biotin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Taxanes such as paclitaxel and docetaxel are widely used in cancer chemotherapy.
  • the primary mode of action of taxanes is to promote microtubulin assembly and stabilize them, preventing their depolymerization and thereby inhibiting microtubule dynamics which causes impaired mitosis, leading to cell cycle arrest and finally to apoptosis.
  • the taxane paclitaxel exhibits a significant antitumor activity against non- small-cell lung cancer (NSCLC), ovarian, head and neck tumors, prostate cancer and breast cancer and is also known as a useful anti-proliferative agent for preventing restenosis.
  • NSCLC non- small-cell lung cancer
  • Exemplary taxanes include, but are not limited to, paclitaxel, docetaxel, cephalomannine, 10-deacetyl cephalomannine, 10-deacetyl taxol, 7-epi- 10-deacetyl taxol, 7-epi- 10-deacetyl cephalomannine, 10-deacetyl baccatin III and other derivatives of paclitaxel.
  • Docetaxel (marketed under the trade name Taxotere®) is a clinically well- established anti-mitotic chemotherapy medication, used mainly for the treatment of breast, ovarian, prostate, and non-small cell lung cancer.
  • Dosage amount and interval may be adjusted individually to provide levels of the active ingredient sufficient to induce or suppress the biological effect (minimal effective concentration, MEC).
  • MEC minimum effective concentration
  • the MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • treating refers to inhibiting, preventing or arresting the development of a pathology (disease, disorder or condition) and/or causing the reduction, remission, or regression of a pathology.
  • pathology disease, disorder or condition
  • Those of skill in the art will understand that various methodologies and assays can be used to assess the development of a pathology, and similarly, various methodologies and assays may be used to assess the reduction, remission or regression of a pathology.
  • the conjugate, as described herein can be utilized in combination (e.g., combined therapy, co-administration and/or co-formulation) with an additional active ingredient usable for treating the indicated medical condition (e.g., cancer).
  • an additional active ingredient can be, as non- limiting examples, an additional anti-cancer agent, anti-angio genie agent and/or anti- proliferation agent.
  • Formulation solution 1 ml solution is comprised of 140 ⁇ Cremophor®EL: 35 ⁇ EtOH: 825 ⁇ Saline. Preparation: IDD-1010 is dissolved by stirring in Cremophor®EL:Ethanol solution until the solution is clear. Saline is thereafter added dropwise to form a white stable emulsion, as described hereinabove.
  • the whole tumor mass is harvested, weight, sized by caliper and calculated, photographed with cm, and kept in 4% formaldehyde solution.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

A conjugate comprising docetaxel and biotin covalently linked to one another is disclosed, as well as uses thereof in the treatment of proliferative diseases and disorders such as cancer.

Description

CONJUGATE OF A TAXANE AND BIOTIN AND USES THEREOF
FIELD AND BACKGROUND OF THE INVENTION
The present invention, in some embodiments thereof, relates to pharmacology and, more particularly, but not exclusively, to novel compounds which are useful in the treatment of proliferative diseases and disorders such as cancer.
Taxanes such as paclitaxel and docetaxel are widely used in cancer chemotherapy. The primary mode of action of taxanes is to promote microtubulin assembly and stabilize them, preventing their depolymerization and thereby inhibiting microtubule dynamics which causes impaired mitosis, leading to cell cycle arrest and finally to apoptosis. The taxane paclitaxel exhibits a significant antitumor activity against non- small-cell lung cancer (NSCLC), ovarian, head and neck tumors, prostate cancer and breast cancer and is also known as a useful anti-proliferative agent for preventing restenosis.
Despite its strong anticancer activity, paclitaxel is poorly water-soluble and exhibits serious dose-limiting toxicities and hypersensitivity reactions, some of which originate from the vehicle cremophorEL® required for its formulation and the absence of selectivity for target tissue.
WO 2009/090614 and corresponding US Patent No. 8,586,625 describe esters of paclitaxel and acidic moieties such as lipoic acid and pharmaceutical compositions comprising them.
SUMMARY OF THE INVENTION
In accordance with the present invention, a novel conjugate of a taxane (e.g., docetaxel) and biotin has been designed and successfully prepared, which was shown to exhibit a potent anti-cancer activity and reduced adverse effects, which therefore enable to utilize docetaxel within a wider therapeutic window.
According to an aspect of some embodiments of the present invention there is provided a conjugate comprising a taxane and biotin covalently coupled to one another via an ester bond.
Exemplary taxanes include, but are not limited to, paclitaxel, docetaxel, cephalomannine, 10-deacetyl cephalomannine, 10-deacetyl taxol, 7-epi- 10-deacetyl taxol, 7-epi- 10-deacetyl cephalomannine, 10-deacetyl baccatin III and other derivatives of paclitaxel. According to some embodiments of the invention, the taxane is docetaxel. According to some embodiments of the invention, the conjugate has the structure:
Figure imgf000003_0001
According to an aspect of some embodiments of the present invention there is provided a pharmaceutical composition comprising the conjugate as described herein and a pharmaceutically acceptable carrier.
According to some embodiments of the invention, the pharmaceutical composition is for use in the treatment of a proliferative disease or disorder.
According to an aspect of some embodiments of the present invention there is provided a conjugate as described herein, for use in the treatment of a proliferative disease or disorder such as cancer.
According to an aspect of some embodiments of the present invention there is provided a use of a conjugate as described herein, in the manufacture of a medicament for the treatment of a proliferative disease or disorder.
According to an aspect of some embodiments of the present invention there is provided a method of treating a proliferative disease or disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the conjugate as described herein.
According to some embodiments of the invention, the proliferative disease or disorder is cancer. Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.
BRIEF DESCRIPTION OF THE DRAWINGS
Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.
In the drawings:
FIG. 1 is a bar graph presenting the in vitro drug efficacy using cell cycle analysis.
PC3-3M-Luc cells were treated with IDD-1010 at the indicated concentrations for 72 hours, and were thereafter fixed, stained with propidium iodide and analyzed by flow cytometry.
FIG. 2 is a plot presenting mice weights on week 8 following cancer cells injection, in the different treatment groups. Black lines represent median. P values in comparison to vehicle for Taxol and IDD-1010 are >0.05, i.e., 0.3768 and 0.1837, respectively.
FIGs. 3A-3B present the tumor volume estimation curve built by blotting the luciferase intensity of some tumors against their tumor volume at the beginning of the experiment (FIG. 3A) and the estimated relative tumor volume as determined by making a correlation between tumor volume and luciferase signal obtained from 3 mice scarified before starting the treatments (FIG. 3B).
FIG. 4 presents the tumor volume in each tested treatment group, as calculated by measuring the short and the long dimensions of the tumor by the end of the experiment according to the following equation: V = (the shortest diameter) x (the longest diameter)/2. Black lines represent median. FIG. 5 presents comparative plots demonstrating the animal survival as determined by counting the dead animal in each treatment group throughout the experiment.
FIG. 6 are images presenting histopathological analysis obtained by hematoxylin and eosin (H&E) staining of representative tissue sample sections. Panels A, B, D and G are images of a liver tissue t; panels L and M are images of primary tumor samples treated, with N denoting Normal tissue, T denoting Tumor and Nec denoting Necrosis.
DETAILED DESCRIPTION OF THE INVENTION
The present invention, in some embodiments thereof, relates to pharmacology and, more particularly, but not exclusively, to novel compounds which are useful in the treatment of proliferative diseases and disorders such as cancer.
Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.
According to certain embodiments, the present invention provides a conjugate of a taxane and biotin.
The present inventor has designed and successfully prepared and practiced a novel conjugate which is suitable for targeted delivery of taxanes to proliferating cells, and hence is useful as an anti-cancer agent.
The novel conjugate, according to some embodiments of the present invention, comprises a taxane (e.g., docetaxel) and biotin.
Exemplary taxanes include, but are not limited to, paclitaxel, docetaxel, cephalomannine, 10-deacetyl cephalomannine, 10-deacetyl taxol, 7-epi- 10-deacetyl taxol, 7-epi- 10-deacetyl cephalomannine, 10-deacetyl baccatin III and other derivatives of paclitaxel.
The taxane and the biotin can be linked to one another either directly, e.g., upon formation of a covalent bond between two compatible functional groups thereof, as is exemplified hereinbelow, or via a linking moiety.
In some embodiments, the taxane has at least one hydroxy group that is capable of forming an ester bond with a carboxylic acid functionality of biotin. Paclitaxel (isolated from the bark of the rare Pacific yew tree, Taxus brevifolia, and named Taxol), for example, has three hydroxyl groups to which the biotin can be attached. These hydroxyl groups are located at the C-2', C-7 and C-1 positions, with their relative order of reactivity generally believed to be C-2'>C-7>C-1 (from most reactive to least reactive).
In some embodiments, the taxane is docetaxel.
Docetaxel (marketed under the trade name Taxotere®) is a clinically well- established anti-mitotic chemotherapy medication, used mainly for the treatment of breast, ovarian, prostate, and non-small cell lung cancer.
Docetaxel is a semi-synthetic analogue of paclitaxel, which was designed due to the scarcity of paclitaxel. Docetaxel is an esterified product of 10-deacetyl baccatin III, which is extracted from the renewable and readily available European yew tree.
Docetaxel differs from paclitaxel at two positions in its chemical structure. It has a hydroxyl functional group on carbon 10, whereas paclitaxel has an acetate ester group, and a tert-butyl carbamate ester group on the phenylpropionate side chain instead of the benzyl amide group in paclitaxel. The carbon 10 hydroxyl functional group change causes docetaxel to be more water-soluble than paclitaxel.
Figure imgf000006_0001
Docetaxel
Docetaxel is a white powder and is typically formulated in polyoxyethylene (20) sorbitan monooleate (polysorbate 80) :ethanol: saline carrier and administered as an aqueous intravenous solution.
Biotin, also known as Vitamin B7 or Vitamin H or Coenzyme R, is a water-soluble B-complex vitamin, which is composed of a ureido ring fused with a tetrahydrothiophene ring, and has a valeric acid substituent attached to one of the carbon atoms of the tetrahydrothiophene ring. Biotin is a coenzyme in the synthesis of fatty acids, isoleucine, and valine, and it plays a role in gluconeogenesis.
Figure imgf000007_0001
As is known in the art, since proliferating cells require vitamins for their growth, vitamin receptors are typically overexpressed in such cells, so as to allow vitamins internalization into the cells. Avidin (or streptavidin) is an exemplary such receptor which is known by its high binding affinity to biotin.
In the conjugates of the present invention, the biotin is covalently coupled/linked to docetaxel at the carbon atom at position 1, 7, 10 or 2' via an ester bond.
The chemical structure of an exemplary conjugate of the invention which comprises docetaxel having biotin coupled thereto via an ester bond, is depicted in Scheme 1 in the Examples section that follows and is identified herein as IDD-1010.
The present inventor has envisioned that conjugating biotin to a taxane such as docetaxel would act as a delivery system for targeted delivery of the conjugate to proliferating cells such as cancer cells, where the docetaxel will be released by hydrolysis of the ester bond coupling the biotin thereto. The hydrolysis can be enzymatically- triggered hydrolysis, effected in the presence of hydrolases or esterases at the diseased cells, tissue or organ, or can be simply effected at the aqueous environment of the diseased tissue, organ or cells.
As demonstrated in the Examples section that follows, while reducing the present invention to practice it was indeed demonstrated that such a conjugate exhibits anti-cancer activity and reduced toxicity, at least as compared to Taxol. Its maximal tolerated dose was determined to be 150 mg/kg body in mice, which is equivalent to about 12.2 mg/Kg body in humans, or to about 850 mg dose for a human weighing 70 Kg. It is noted that the acceptable doses for administering docetaxel to humans are much lower.
The conjugates described herein are therefore useful in the treatment of proliferative diseases and disorders.
As used herein, a proliferative disease or disorder encompasses any disease or disorder that is associated with abnormal cell proliferation, including, but not limited to, restenosis, cancer and other malignant, pre-malignant and benign tumors. Examples of types of cancers that can be treated with the conjugate of the invention include, but are not limited to, carcinoma (e.g., ductal carcinoma, squamous-cell carcinoma, renal cell carcinoma, carcinoma in situ, adenocarcinomaetc), lymphoma (e.g., Hodgkin's, Non-Hodgkin's, Burkitt's lymphoma, etc.), blastoma (e.g., pleuropulmonary, neuroblastoma, retinoblastoma, glioblatoma, etc), sarcoma (e.g. liposarcoma, osteosarcomasynovial sarcoma, rhabdomyosarcoma (alveolar), , Ewing's sarcoma), leukemia (e.g., myeloid leukemia such as chronic myelogenous leukemia; acute myelogenous leukemia with maturation; acute promyelocytic leukemia; acute nonlymphocytic leukemia with increased basophils; acute monocytic leukemia; acute myelomonocytic leukemia with eosinophilia; ; lymphocytic leukemia, such as acute lumphoblastic leukemia; chronic lymphocytic leukemia, etc) and solid tumor cancers such as salivary gland, small cell lung cancer, brain, kidney, uterus, prostate, bladder, ovary, colon, skin cancer such as melanoma and breast cancer.
In some embodiments, the cancer is breast cancer, prostate cancer, small cell lung cancer, ovarian cancer or metastases thereof.
The conjugate as described herein can be used either per se or, preferably, as a pharmaceutical composition that further comprises a pharmaceutically acceptable carrier.
As used herein a "pharmaceutical composition" refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of an active ingredient to an organism.
Herein the term "active ingredient" refers to a conjugate as described herein, as well as to any other ingredient combined therewith which is accountable for the biological effect.
Hereinafter, the phrases "physiologically acceptable carrier" and "pharmaceutically acceptable carrier", which may be interchangeably used, refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.
Herein the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intraocular injections. Alternately, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a desired bodily site (e.g., tissue or organ) of a patient.
The term "tissue" refers to part of an organism consisting of cells designed to perform a function or functions. Examples include, but are not limited to, brain tissue, retina, skin tissue, hepatic tissue, pancreatic tissue, bone cartilage, connective tissue, blood tissue, muscle tissue, cardiac tissue, vascular tissue, renal tissue, pulmonary tissue, gonadal tissue, hematopoietic tissue.
Organs include, for example, breast, liver, heart, ovary, prostate, pancreas, brain, stomach, intestine, lungs, etc.
Pharmaceutical compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
Pharmaceutical compositions for use in accordance with some embodiments of the invention thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically.
Proper formulation is dependent upon the route of administration chosen.
For injection, the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as
Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. For oral administration, the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, PVP, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical compositions which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by nasal inhalation, the active ingredients for use according to some embodiments of the invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro- ethane or carbon dioxide. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The pharmaceutical composition described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.
The pharmaceutical composition of some embodiments of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
In some embodiments, the pharmaceutical composition is formulated for intravenous injection, e.g., intravenous bolus injection.
In some embodiments, the carrier comprises saline and a carrier suitable for use in administration of taxanes. These include, for example, a mixture of ethanol and a polymer such as, but not limited to, ChremophorEL® and/or polysorbate.
Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients (e.g., a conjugate as described herein) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., cancer) or prolong the survival of the subject being treated.
Determination of a therapeutically effective amount is well known within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
For any preparation used in the methods of the invention, the therapeutically effective amount or dose (also referred to as therapeutic window) can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l).
Dosage amount and interval may be adjusted individually to provide levels of the active ingredient sufficient to induce or suppress the biological effect (minimal effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc. Compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed herein.
The conjugates described herein can be utilized in a method of treating a proliferative disease or disorder, as defined herein, in a subject in need thereof.
The term "treating" refers to inhibiting, preventing or arresting the development of a pathology (disease, disorder or condition) and/or causing the reduction, remission, or regression of a pathology. Those of skill in the art will understand that various methodologies and assays can be used to assess the development of a pathology, and similarly, various methodologies and assays may be used to assess the reduction, remission or regression of a pathology.
As used herein, the term "preventing" refers to keeping a disease, disorder or condition from occurring in a subject who may be at risk for the disease, but has not yet been diagnosed as having the disease.
As used herein, the term "subject" includes mammals, preferably human beings at any age which suffer from the pathology. In some embodiments, this term encompasses individuals who are at risk to develop the pathology.
As used herein the phrase "treatment regimen" refers to a treatment plan that specifies the type of treatment, dosage, schedule and/or duration of a treatment provided to a subject in need thereof (e.g., a subject diagnosed with a pathology). The selected treatment regimen can be an aggressive one which is expected to result in the best clinical outcome (e.g., complete cure of the pathology) or a more moderate one which may relief symptoms of the pathology yet results in incomplete cure of the pathology. It will be appreciated that in certain cases the more aggressive treatment regimen may be associated with some discomfort to the subject or adverse side effects (e.g., a damage to healthy cells or tissue). The type of treatment can include a surgical intervention (e.g., removal of lesion, diseased cells, tissue, or organ), a cell replacement therapy, an administration of a therapeutic drug (e.g., receptor agonists, antagonists, hormones, chemotherapy agents) in a local or a systemic mode, an exposure to radiation therapy using an external source (e.g., external beam) and/or an internal source (e.g., brachytherapy) and/or any combination thereof. The dosage, schedule and duration of treatment can vary, depending on the severity of pathology and the selected type of treatment, and those of skills in the art are capable of adjusting the type of treatment with the dosage, schedule and duration of treatment.
In any of the compositions, uses and methods described herein, the conjugate, as described herein, can be utilized in combination (e.g., combined therapy, co-administration and/or co-formulation) with an additional active ingredient usable for treating the indicated medical condition (e.g., cancer). Such an additional active ingredient can be, as non- limiting examples, an additional anti-cancer agent, anti-angio genie agent and/or anti- proliferation agent.
As used herein the term "about" refers to ± 10 %.
The terms "comprises", "comprising", "includes", "including", "having" and their conjugates mean "including but not limited to".
The term "consisting of means "including and limited to".
The term "consisting essentially of" means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
The word "exemplary" is used herein to mean "serving as an example, instance or illustration". Any embodiment described as "exemplary" is not necessarily to be construed as preferred or advantageous over other embodiments and/or to exclude the incorporation of features from other embodiments.
The word "optionally" is used herein to mean "is provided in some embodiments and not provided in other embodiments". Any particular embodiment of the invention may include a plurality of "optional" features unless such features conflict. As used herein, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a compound" or "at least one compound" may include a plurality of compounds, including mixtures thereof. Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases "ranging/ranges between" a first indicate number and a second indicate number and "ranging/ranges from" a first indicate number "to" a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
As used herein the term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
As used herein, the term "treating" includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.
Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples.
EXAMPLES
Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non-limiting fashion.
EXAMPLE 1
Chemical Synthesis of a docetaxel-hiotin conjugate
Biotin (36 mg; 0.00015 mol), 4-dimethyl amino pyridine (DMAP) and Ν,Ν'- dicyclohexylcarbodiimide (DCC) (0.029 gr; 1 eq) were added to a solution of docetaxel (100 mg) in dry dichloromethane (10 ml) under nitrogen, and the reaction mixture was stirred at room temperature for 24 hours. Thereafter, the mixture was diluted with diethyl ether, and the docetaxel-biotin conjugate (also referred to herein as IDD-1010) precipitated from the mixture. The precipitate was further purified by column chromatography, so as to afford the product at a yield of about 20 % and a purity, as measured by HPLC at 230 nm, higher than 99 %).
The chemical structure of IDD-1010, as confirmed by nuclear magnetic resonance (NMR) and mass spectrometry (MS) measurements, is depicted in Scheme 1, along with the MS and elemental analysis data obtained for this product.
Scheme 1
Figure imgf000017_0001
C53H67N3016S
Exact Mass: 1033.4242
Mol. Wt: 1034.1746
m/e: 1033.4242 (100.0%), 1034.4276 (58.9%), 1035.4309 (17.0%), 1035.4200 (4.4%), 1036.4343 (3.2%), 1035.4285 (3.2%), 1036.4234 (2.6%), 1036.4318 (1.9%),
1034.4212 (1.1 %), 1034.4304 (1.0%)
C, 61.55; H, 6.53; N, 4.06; O, 24.75; S, 3.10
EXAMPLE 2
Activity Assays
The choice of the appropriate cancer mouse model in preclinical studies is a critical factor in the evaluation of potential anticancer drugs and the elucidation of molecular events associated with carcinogenesis. Orthotopic transplantation of tumor cells remains the most appropriate way of cancer modeling, if compared with ectopic implantation, since it mimics the interaction between the tumor cells and their stroma or microenvironment and allows accurate expression of the clinical features of human cancer in mice.
In the herein described study, PC3 prostate cancer cells were used in an orthotopic prostate cancer model designed for the efficacy evaluation of the tested antitumor agents. These cells express luciferase (Luc) in a stable manner which allows for real time in vivo monitoring of tumor response to drugs and to follow the progression of the tumor. Materials and Methods
In vitro drug activity:
PC3-Luc prostate carcinoma cells (5.0 x 105) were cultured in 60 mm plates in triplicates. One day after, cells were treated with different concentrations (10 ng/ml-50 g/ml) of IDD-1010. Seventy two hours post treatment, the cells were collected, fixed and stained with propidium iodide. To determine the efficacy of IDD-1010, cell cycle analysis was performed to determine the fraction of cells in each stage of the cell cycle.
Prostate orthotopic injection of PC3-Luc cells:
PC3-Luc cells (0.5 x 106) were injected into the prostate of 6-week male NOD/SCID mice. Mice were imaged weekly for 8 weeks to monitor tumor growth kinetics. At the end, mice were euthanized, and tumors were excised, weighed, and preserved for further histopathological analyses.
Preparation of tumor cells:
PC3-Luc cells (maintained in RPMI, 10 % fetal bovine serum (FBS) and supplements) were trypsinized and re-suspended in the same growth medium to achieve a concentration of 0.5 x 106 cells in 20 μΐ.
Injection:
Six-week old male, NOD/SCID mice were anesthetized by intramuscular injection of ketamine (120 mg/kg)/xylazine (6 mg/kg) body weight of mice. Once anesthetized, mice were placed in the supine position and the surgical area was sterilized by alternate wipes of 70 % ethanol and iodine three times. A 5-10 mm incision along the posterior midline of the abdomen was created, so as to expose the prostate just beneath. Another incision was made into the peritoneal wall. Bladder was retracted, and pressed lightly to expose the prostate. PC3-Luc cells (0.5 x 106 cells in 20 μΐ) were slowly injected into the dorsal prostatic lobe using a 30-gauge needle attached to a Hamilton syringe. A well-localized bleb within the injected prostatic lobe indicated a technically satisfactory injection. The peritoneal incision was sutured using 6-0 silk suture with C-22 reverse cutting needle and the skin was closed by 2-3 autoclips. During recovery, mice were given petroleum ointment over eyes and 500 μΐ of warm PBS subcutaneously on the back. Once recovered, mice were injected subcutaneously with 50 μΐ of buprenorphine (0.05-0.1 mg/kg) and placed back to the cages. Imaging:
Mice were injected with 100 μΐ of luciferin (15 mg/ml stock) per 10 grams of mouse body weight by intraperitoneal injection in the animal's lower left abdominal quadrant. After 8-10 minutes, mice were anesthetized by gas anesthesia (3 % isoflurane). Once anesthetized (2-3 minutes), mice were placed onto black paper and imaged ventrally. Animals were first imaged on day 7 after wounds are healed to ensure successful tumor cells implantation and then once a week for 8 weeks. At the end of the experiment, animals were euthanized and tumors and selected tissues were assessed by histopathological analyses. Drug treatment:
After assuring that the injections were successful and that the tumors are well established (tumor volume of about 200 mm ), the mice were divided randomly in two different groups and named.
IDD-1010 was dissolved completely in 175 μΐ solution of cremophonethanol 80 %:20 % v/v. Then, 825 μΐ of saline was added dropwise and mixed to form a 1 ml formulated solution. On days 28, 35, 42 and 49 after cells injection, the different groups were injected intravenously with the IDD-1010 solution (hereinafter "the drug") at a concentration of 10 mg/kg of mouse body weight. Total of 4 injections, once a week for 3 sequential weeks, were performed, and were followed by 2 weeks monitoring.
Results and Discussion
In vitro drug activity:
In order to determine the in vitro drug efficacy, IDD-1010 was dissolved in dimethyl sulfoxide (DMSO) and added to PC3-Luc prostate cancer cells to the final concentrations of 10 nM, 100 nM, 500 nM, 1 μΜ, 5 μΜ, 10 μΜ and 50 μΜ. Seventy two hours later, cells were fixed, stained with propidium iodide and cell cycle analysis was performed using flow cytometry. The results are presented in Figure 1 and indicate that IDD-1010 treatment was associated strongly with increased sub-Gl and G2/M populations, which may be indicative of increased apoptosis and mitotic arrest, respectively. In vivo assessment of drug activity using an orthotopic prostate cancer model:
To determine the efficacy of the compound IDD-1010, an in vivo orthotopic prostate cancer model was generated. Prostate cancer cell line (PC3-Luc) were injected into the dorsal loop of the mouse prostate. Four weeks after cell injection, drug treatments were initiated and continued for additional 3 weeks, following by two weeks monitoring, as described in Materials and Methods section hereinabove. The development of prostate tumors was followed by measuring the luciferase activity expressed by the injected cells. The effect of the different preparations on mice weight:
Mice weight loss results mainly from either drug toxicity or growth of tumors to a size that exceeds the capability of the body to cope with. To test the effect of IDD-1010 on mice weight, the surviving animals were weighed every week and at the end of the experiment. The recorded data is presented in Figure 2, and show that IDD-1010 treatment did not lead to any significant weight loss; P-value in comparison to vehicle was 0.1837. The average weight of the vehicle group was 27 grams.
Tumor volume kinetics:
To estimate the starting tumor volume and to follow the growth rate of the tumors throughout the study, 3 mice were sacrificed before treatment initiation and were measured for their tumor volume. The luciferase activity in these mice was also measured. From the tumor volume and the luciferase activity, a correlation curve was established, so as to estimate the tumor volume at any time point throughout the experiment, as presented in Figure 3A, according to the following equation:
Estimated tumor volume = 6 x 10 -"8 X + 138.46
where X = luciferase intensity reading.
The estimated tumor volume at start of drug injection was about 200 mm .
The obtained data is presented in Figure 3B. As can be seen in Figure 3B, the relative luciferase activity values show that IDD-1010 treatment was the only treatment that leads to a decrease in tumor volume and is more potent than Taxol. Tumor volume:
To determine the anti-cancer activity of IDD-1010, the tumor volume at the end of the study was measured. The average tumor volume for each treatment group was calculated. The obtained data is presented in Figure 4 and show that Taxol and IDD-1010 exhibit a statistically significant reduction in tumor volume compared to the vehicle group, with a final average tumor volume of 681 and 289.4 mm , respectively, compared to 1737 mm in the vehicle group. Tumor weight:
The effect of the tested compounds on tumor development was also assessed by measuring the tumor weight at the end of the experiment. Taxol and IDD-1010 treatments resulted in statistically significant reduction in tumor weight compared to the vehicle group. P-values in comparison to vehicle were 0.0192 and 0.0011 for Taxol and IDD-1010, respectively.
It is suggested that the inconsistency between tumor volume and tumor weight might result from the fact that tumor volume measurement itself is inaccurate because most of the tumors have irregular architecture. Mice survival:
Mice survival was also tested for determining treatment efficacy. Using Kaplan- Meier estimator, the number of dead animal throughout the experiment was plotted and the resulting percentages between the different groups were compared. The results are presented in Figure 5 and show that Taxol and IDD-1010 both resulted in 100 % survival whereby in the vehicle group 40 % survival was observed.
Histopathology:
Toxicity: To assess the toxicity of the tested drugs, the histology of the main tissues was tested. Figures 6A, B, D and G present sections of liver tissue of mice treated with vehicle, Taxol and IDD-1010 treatments, showing that all were associated with reduced liver abnormalities.
Primary tumors: In addition to drug toxicity confirmation, the primary tumors were tested for the effect of the different treatments. As shown in Figure 6L, increased necrosis areas were observed in the IDD-1010 treatment group. The same was observed also in the Taxol treatment group (data not shown). In some cases, it was observed that the normal prostate tissue occupied some areas in the primary tumors following IDD-1010 treatment, as presented in Figure 6M, a finding that may indicate a good response.
Overall, the data presented herein demonstrate that IDD-1010 showed a consistent anti-cancer effect which was demonstrated by all the tested parameters. IDD-1010 was also showed to be less toxic to the animals, at least as compared to Taxol, as indicated by mice weight, survival and histology. These findings indicate that IDD-1010 is a potent anticancer agent, which exhibits a wider therapeutic window, compared to Taxol. EXAMPLE 3
Single Dose Toxicity Studies
Studies were conducted to evaluate the maximum tolerated dose (MTD) of IDD- 1010 following a single intravenous (IV) injection to male and female mice, in consideration of its intended use as an anti cancer agent.
Materials and Animals
Preparation of IDD-1010 Dosing Solutions:
For preparation of 1 ml emulsion at the required concentration, on each day of dosing, the appropriate amount of IDD-1010 powder was weighed and dissolved in 175 μΐ of cremophor®EL:ethanol absolute (80 %:20% v/v; 140 μΐ of cremophor®EL and 35 μΐ of ethanol). The solution was then mixed until the IDD-1010 powder was completely dissolved. Subsequently, the solution was diluted with 825 μΐ of physiological saline to obtain 1 ml of stable emulsion at the appropriate concentration.
Different volumes of IDD-1010 emulsion are prepared in identical fashion as long as the ratio between all components remains.
Preparation of Vehicle Control:
On the day of dosing, 140 μΐ of Cremophor®EL was mixed with 35 μΐ ethanol absolute (80 %:20 % v/v). The solution was then diluted with 825 μΐ of physiological saline.
Different volumes of vehicle control are prepared in identical fashion as long as the ratio between all components remains.
Animals:
Young adults (male and female) Balb/cOlaHsd mice, 8-9 week-old at study initiation (obtained from Harlan Laboratories Israel, Ltd. (ISO 9001:2008 Certificate No.: GB06/68708)) were used. Weight variation of animals at the time of treatment initiation did not exceed ± 20 % of the mean weight for each sex.
Healthy mice were acclimated for at least 5 days. Animals were housed within a limited access rodent facility and kept in groups of 3 mice of the same sex in polypropylene cages fitted with solid bottoms and filled with wood shavings as bedding material. Environment was controlled automatically by a control computer and set to maintain temperature at 20-24 °C with a Relative Humidity (RH) of 30-70 %, a 12-hours light/dark cycle and 10-30 air changes/hour in the study room.
Animals were fed with certified commercial rodent diet ad libitum, together with free access to drinking water, supplied to each cage via polyethylene bottles with stainless steel sipper tubes. The water was filtered (0.1 μ filter), chlorinated and acidified.
Animals were given a unique animal identification ear number. This number also appeared on a cage card, visible at the front of each cage. The cage card also contained the study number, route of administration, sex, strain and all other relevant details as to treatment group and dose level.
On the day of arrival animals were randomly assigned to the various test groups.
At the end of the experiment, surviving animals were euthanized by C02 asphyxiation prior to the scheduled necropsy. Animals euthanized for humane reasons were sacrificed by the same method.
Test Groups and Dose Levels:
Mice were divided into 5 groups, 6 mice per group (3 animals of each sex). 4 groups received 3 appropriately spaced doses of IDD-1010. The other group received vehicle only.
The initial target dose level injected was 150 mg/kg. Additional appropriate dose levels were determined during study and according to reactions observed.
The volume dosage applied was 10 ml/kg.
Study Protocol
The study was aimed at determining the acute toxicity (MTD) of IDD-1010 using a stepwise procedure with the use of minimum number of animals per step. An initial single target dose of 150 mg/kg was intravenously injected to one group of 3 male and 3 female mice. Depending on the observed presence or absence of lethality incidence and/or severe adverse reactions to treatment, further groups were then injected either with higher or lower appropriately interspaced dose levels. Dosing was sequential and the time interval between the treated groups was determined according to the onset, duration and severity of toxic signs, and was delayed until one was confident of survival of the previously dosed animals. Additional group of 3 male and 3 female mice was injected with the vehicle control, at equal volume dosage and served as a control group. All dosed animals, which survived, were observed for a total duration of 14 days, after which they were subjected to sacrifice and gross necropsy.
The IDD-1010 formulation and the vehicle control formulation were injected by intravenous slow bolus injection over approximately 1 minute, into one of the tail veins, by the use of a suitable syringe and needle after warming the cage environment for about 3-5 minutes and/or warming the tail veins using soaked gauze pad.
Clinical Observations:
Individual clinical examinations were carried out immediately post-dosing and up to the first 30 minutes and at least twice more during the first 2 hours. Thereafter, animals were observed at least once daily or more frequently when indicated by the response of the animals to treatment, for a total duration of 14 days (on weekend cage side observations were performed). Observations include changes in skin, fur, eyes, mucous membranes, occurrence of secretions and excretions (e.g. diarrhea) and autonomic activity (e.g. lacrimation, salivation, piloerection, unusual respiratory pattern). Changes in gait, posture and response to handling, as well as the presence of bizarre behavior, tremors, convulsions, sleep and coma were also observed and recorded. Any local reaction at injection sites were recorded as well. All animals were observed for morbidity and mortality twice daily on weekdays and once daily during weekends and official holidays.
Body Weights:
Determination of individual body weights of all animals was initially carried out following arrival of animals, followed by body weight determination shortly before dosing and thereafter 2 and 7 days post-dosing and prior to termination (14 days post-dosing). In case of decedents, determination of individual body weights was carried out as close as possible to the time of death (when applicable). Necropsy Procedures and Macroscopic Examination:
All animals (including decedents or animals which were removed from the study for animal welfare reasons) were subjected to a full detailed necropsy and gross pathological examination following study termination. At necropsy, all animals were subjected to thorough examination, including the external surface of the body, all orifices, cranial, thoracic and abdominal cavities and their contents. Any abnormality or gross pathological changes observed in tissues and/or organs were recorded and collected accordingly. Histopathological examinations for any abnormalities, detected during necropsy, were performed as required.
Results
The potential acute toxicity of IDD-1010 was assessed following a single bolus intravenous (IV) injection over 1 minute to male and female Balb/cOlaHsd mice in order to establish the MTD (maximum tolerated dose) level.
IDD-1010 was injected as stable emulsion by a single IV bolus injection over 1 minute at four dose levels of 150, 175, 208, and 250 mg/kg in a sequential fashion. An additional group of 3 male and 3 female mice was injected with the vehicle control Item Cremophor® EL:Ethanol Absolute:physiological saline and served as a control group. All surviving animals were observed for a total duration of 14 days.
Incidence of mortality was observed in all animals injected with IDD-1010 at a higher dose level than 150 mg/kg, amounting to 1/1 female per each dose level of 175, 208 or 250 mg/kg. Mortality occurred during or immediately following dosing.
Mean group body weight gain at the end of the 14-day study period of the 150 mg/kg IDD-lOlO-treated group was not significantly different from that of the control group.
No gross pathological findings were noted at necropsy in any of the animals at the time of their scheduled necropsy, 14 days post-dosing.
These results suggest that the maximum tolerated dose (MTD) level of the IDD-
1010, when formulated as stable emulsion in Cremophor® EL:Ethanol Absolute: physiological saline following IV bolus injection over 1 minute to male or female Balb/cOlaHsd mice, is 150 mg/kg. EXAMPLE 4
Pharmacokinetic Studies
Animals: ICR (CD-I) female mice weighing 20-25 grams; 13 groups, 3 mice in each group, were acclimated for at least 5 days.
Formulation solution: 1 ml solution is comprised of 140 μΐ Cremophor®EL: 35 μΐ EtOH: 825 μΐ Saline. Preparation: IDD-1010 is dissolved by stirring in Cremophor®EL:Ethanol solution until the solution is clear. Saline is thereafter added dropwise to form a white stable emulsion, as described hereinabove.
Protocol: Mice were injected with a single dose of 40 mg/Kg body weight of a 4 mg/ml formulation solution, by intravenous bolus injection for 1 minute.
Blood samples are obtained 5 minutes, 1 hour, 3 hours, 6 hours, 9 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 120 hours (5 days), 168 hours (7 day), from 3 mice for each time point and from 3 mice for time zero.
100 μΐ aliquots of plasma are prepared from each mouse, by withdrawing maximum blood possible from each mouse (about 600 μΐ blood that would generate 300 μΐ of plasma to be aliquoted). Heparin is used as an anticoagulant.
Clinical signs observations are done frequently within the immediate post injection period and examination is carried out periodically and as signs warrant. Thereafter, observations are performed once daily until study termination.
Animal body weight is determined before commencement of the study and at the time of sacrifice.
EXAMPLE 5
IDD-1010 activity in in vivo xenograft model
2 x 106 A549 human non-small cell lung carcinoma cells suspended in 100 microliter PBS (grown and supplied by PharmaSeed Ltd., Israel) are injected subcutaneously (SC) to the dorsal side/ flank of each of the 60 NOD-SCID female mice of 6-8 weeks with an average weight of 20-25 gr.
Animal body weight is determined before commencement of the study, throughout treatment and till the end of the study once a week and before sacrifice. Data is recorded in an Excel file.
Tumor volume is monitored by caliper, by measuring the length and width of each tumor, from the beginning of the study, throughout treatment and till the end of the study twice weekly. Tumor volume is calculated and data is recorded in an Excel file.
When tumor volume reaches -70-100 mm , mice are randomly divided into 3 groups, 5-6 mice per group and the treatment is started. Tumor-bearing mice are injected intravenously (IV) in the tail vein (slow injection for 2 min) with the relevant test items solutions at the indicated schedule below.
Three experimental groups are treated as follows:
Group 1 (5 mice): Vehicle is administered at 10 ml/Kg unless otherwise stated. Schedule: once a week for 3 consecutive weeks, total of 4 injections. At the end of the treatment mice are monitored for two additional weeks.
Group 2 (5 mice): Docetaxel, dose: 7.4 mg/Kg. Schedule: once a week for 3 consecutive weeks, total of 4 injections. At the end of the treatment mice are monitored for two additional weeks.
Group 3 (9 mice): IDD-1010, dose 1: 10 mg/Kg. Schedule: once a week for 3 consecutive weeks, total of 4 injections. At the end of treatment mice are for two additional weeks.
Clinical signs observation and survival of mice are done within the immediate post cell line and treatment injection period and examination are carried out periodically as signs warrant during the entire experiment. Data are recorded.
At termination of the experiment, the whole tumor mass is harvested, weight, sized by caliper and calculated, photographed with cm, and kept in 4% formaldehyde solution.
Vital organs are preserved in 4% formaldehyde solution for histology and organ lysate preparation purposes. Macroscopic evaluation of inner organs is performed to detect any abnormalities or metastases. All data are recorded.
Results: inhibition of tumor growth in Group 3.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.
All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. To the extent that section headings are used, they should not be construed as necessarily limiting.

Claims

CLAIMS:
1. A conjugate comprising a taxane and biotin covalently coupled to one another via an ester bond.
2. The conjugate of claim 1, wherein said taxane is selected from paclitaxel, docetaxel, cephalomannine, 10-deacetyl cephalomannine, 10-deacetyl taxol, 7-epi- 10-deacetyl taxol, 7-epi- 10-deacetyl cephalomannine, or 10-deacetyl baccatin III.
3. The conjugate of claim 2, wherein said taxane is docetaxel.
4. The conjugate of claim 3, wherein said biotin is covalently coupled to docetaxel at the carbon atom at position 1, 7, 10 or 2' via an ester bond.
5. The conjugate of claim 4, having the structure:
Figure imgf000029_0001
6. A pharmaceutical composition comprising the conjugate of any one of claims 1 to 5 and a pharmaceutically acceptable carrier.
7. A pharmaceutical composition comprising the conjugate of claim 5 and a pharmaceutically acceptable carrier.
8. The pharmaceutical composition of claim 6 or 7, for use in the treatment of a proliferative disease or disorder.
9. The pharmaceutical composition of claim 8, wherein said proliferative disease or disorder is cancer.
10. The conjugate of any one of claims 1 to 5, for use in the treatment of a proliferative disease or disorder.
11. The conjugate of claim 10, wherein said proliferative disease or disorder is cancer.
12. A method of treating a proliferative disease or disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the conjugate of any one of claims 1 to 5.
13. The method of claim 12, wherein said proliferative disease or disorder is cancer.
PCT/IL2014/050470 2013-05-26 2014-05-26 Conjugate of a taxane and biotin and uses thereof WO2014191989A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361827598P 2013-05-26 2013-05-26
US61/827,598 2013-05-26

Publications (1)

Publication Number Publication Date
WO2014191989A1 true WO2014191989A1 (en) 2014-12-04

Family

ID=51988108

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2014/050470 WO2014191989A1 (en) 2013-05-26 2014-05-26 Conjugate of a taxane and biotin and uses thereof

Country Status (1)

Country Link
WO (1) WO2014191989A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4000613A1 (en) 2020-11-19 2022-05-25 Polyphor Ag Pharmaceutical combinations comprising a peptide cxcr4 inhibitor and a taxane for treating cancer
WO2022106061A1 (en) 2020-11-19 2022-05-27 Polyphor Ag Pharmaceutical combinations comprising a peptide cxcr4 inhibitor and a taxane for treating cancer
EP4223292A1 (en) 2022-02-07 2023-08-09 Cellestia Biotech AG Pharmaceutical combinations for treating cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999009021A1 (en) * 1997-08-18 1999-02-25 Florida State University Process for selective derivatization of taxanes
US20090004118A1 (en) * 2004-10-07 2009-01-01 Shuming Nie Multifunctional Nanoparticle Conjugates And Their Use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999009021A1 (en) * 1997-08-18 1999-02-25 Florida State University Process for selective derivatization of taxanes
US20090004118A1 (en) * 2004-10-07 2009-01-01 Shuming Nie Multifunctional Nanoparticle Conjugates And Their Use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GUENARD D. ET AL.: "Fluorescent and biotinylated analogues of docetaxel: synthesis and biological evaluation", BIOORG MED CHEM., vol. 3, no. 10, 31 October 1995 (1995-10-31), pages 1357 - 1368 *
IWAO OJIMA ET AL.: "Design and synthesis of a novel photoaffinity taxoid as a potential probe for the study of paclitaxel - microtubules interactions", TETRAHEDRON LETTERS, vol. 41, 31 December 2000 (2000-12-31), pages 4287 - 4290, XP004205554, DOI: doi:10.1016/S0040-4039(00)00627-4 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4000613A1 (en) 2020-11-19 2022-05-25 Polyphor Ag Pharmaceutical combinations comprising a peptide cxcr4 inhibitor and a taxane for treating cancer
WO2022106061A1 (en) 2020-11-19 2022-05-27 Polyphor Ag Pharmaceutical combinations comprising a peptide cxcr4 inhibitor and a taxane for treating cancer
EP4223292A1 (en) 2022-02-07 2023-08-09 Cellestia Biotech AG Pharmaceutical combinations for treating cancer

Similar Documents

Publication Publication Date Title
KR20180054793A (en) Methods for the treatment of small cell carcinoma of the malignant squamous cell carcinoma or hypercalcemic type ovary
US20230225996A1 (en) Biomarkers of metap2 inhibitors and applications thereof
JP2006524289A (en) PEG-Wortmannin conjugate
WO2014191989A1 (en) Conjugate of a taxane and biotin and uses thereof
JP6462147B2 (en) HSP90 inhibitory peptide conjugate and its application in tumor therapy
Liu et al. Multi-action platinum (IV) prodrugs conjugated with COX-inhibiting NSAIDs
CN106176721A (en) The cancer prevention of organic selenium compounds and therapeutic use
US20090232906A1 (en) Treatment methods and compositions for lung cancer, adenocarcinoma, and other medical conditions
US8846768B2 (en) Use of compounds isolated from Euphorbia neriifolia for treating cancer and/or thrombocytopenia
TW202114682A (en) Combination of a mcl-1 inhibitor and a standard of care treatment for breast cancer, uses and pharmaceutical compositions thereof
KR20160003652A (en) Methods and compositions for gamma-glutamyl cycle modulation
CA2717181C (en) Treatment methods and compositions for lung cancer, adenocarcinoma, and other medical conditions
US20230405132A1 (en) Antitumor pharmaceutical composition and use thereof
JP7311177B2 (en) Combined use of A-NOR-5α androstane drugs with anticancer drugs
Hao et al. Novel fluorinated docetaxel analog for anti-hepatoma: Molecular docking and biological evaluation
CN101830819B (en) Phenylbutyryl curcumin derivate and application thereof in anti-tumor drug preparation
EP3127544B1 (en) Anti-tumor drug containing anti-tumor platinum complex, and anti-tumor effect enhancer
US11696904B2 (en) Prodrug for therapeutic applications
TWI727858B (en) Water-soluble polymer derivatives of Venetoclax
US20230381083A1 (en) Phytic Acid Ester Derivative
WO2021192726A1 (en) Choline uptake inhibitor, apoptosis inducer, anticancer drug, and use thereof
WO2015152409A1 (en) Anti-tumor drug containing taxane compound, and anti-tumor effect enhancer
RU2623034C1 (en) Antineoplastic agent
KR20030086276A (en) Metronomic dosing of taxanes
TW201513849A (en) Reduction of toxicities by synthetic PANAXYTRIOL analogs

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14804690

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14804690

Country of ref document: EP

Kind code of ref document: A1