WO2021233534A1 - Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof - Google Patents

Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof Download PDF

Info

Publication number
WO2021233534A1
WO2021233534A1 PCT/EP2020/064039 EP2020064039W WO2021233534A1 WO 2021233534 A1 WO2021233534 A1 WO 2021233534A1 EP 2020064039 W EP2020064039 W EP 2020064039W WO 2021233534 A1 WO2021233534 A1 WO 2021233534A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
substance
composition
administration
use according
Prior art date
Application number
PCT/EP2020/064039
Other languages
French (fr)
Inventor
David Berglund
Thomas Engstrand
Original Assignee
Pvac Medical Technologies 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 Pvac Medical Technologies Ltd filed Critical Pvac Medical Technologies Ltd
Priority to PCT/EP2020/064039 priority Critical patent/WO2021233534A1/en
Priority to PCT/EP2021/056698 priority patent/WO2021185844A1/en
Publication of WO2021233534A1 publication Critical patent/WO2021233534A1/en

Links

Classifications

    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to the use of a pharmaceutical composition comprising a substance for prevention and treatment, as well as a method of prevention and treatment of cancer comprising administering said pharmaceutical composition.
  • BACKGROUND TECHNOLOGY Melanoma although a rare form of cancer is one of the most difficult to treat diseases once metastasized with increasing incidence and mortality world-wide. Treatment for the disease, once spread, was traditionally chemotherapy and radiotherapy but progress has been made and new targeted treatments as well as immunotherapy are in use and prognosis, although bleak, has improved.
  • SSAO Semicarbazide amine oxidase
  • VAP-1 Semicarbazide amine oxidase
  • SSAO is an enzyme expressed for example by endothelial cells and smooth muscle cells in blood vessels.
  • SSAO is normally found in intracellular granulae. The enzyme translocates to the cell membrane upon activation and can be solubilized by proteolytic cleavage from the membrane.
  • SSAO exerts dual functions including leukocyte recruitment and also functions as ectoenzyme by the oxidation and conversion of primary amines to aldehydes, hydrogen peroxide and ammonium.
  • Enhanced SSAO activity has been shown to be associated with development of tumour growth. Deletion or reduction of SSAO enzyme activity reduces new blood vessel formation, lower the recruitment of immuno- suppressive Gr-l+CDl lb+ myeloid cells into tumours and reduces tumour growth as shown in melanoma and lymphoma animal models. A variety of different experimental SSAO inhibitor have been evaluated preclinically, but few are clinically tested and approved.
  • the object of the present invention is to provide an efficient substance acting as an substance or composition comprising the substance for treating cancer and especially semi-carbazide sensitive amine oxidase (SSAO) related cancer. Further, the present invention provides a substance and a composition that may easily be administrated for example topically for treating skin cancer such as melanoma or squamous cell carcinoma. Surprisingly, the present inventors have surprisingly found that administration of a polymeric substance comprising multiple nucleophilic centers has an anti-cancer effect.
  • SSAO semi-carbazide sensitive amine oxidase
  • the molecular size of the substances according to the present invention effect pharmacokinetic and distribution profiles in a way that may be advantageous for certain indications where a prolonged and directed effect is needed.
  • Other potential benefits include excellent in vivo tolerance and low manufacturing costs.
  • Example 2 The inhibiting effect of the present substance on SSAO is shown in Example 2.
  • Example 4 topical administration of the present substance showed a treating effect on early suspected squamous cell carcinoma.
  • the present inventors have surprisingly found that administration of the present substance or the present pharmaceutical composition has an inhibitory effect on SSAO.
  • the present substance blocks or inhibits SSAO activity and thereby the complications caused by the expression of SSAO may be prevented, inhibited or treated (Treatment of retinopathy, Example 3 and 4).
  • the present invention relates to a substance, composition and method which may be used to treat cancer or metastasis especially those related to enhanced SSAO expression or activity.
  • the present invention relates to a substance comprising a carrier exhibiting at least one scavenger structure, said scavenger structure comprising a nucleophilic centre complying with the formula
  • X 1 is a single-bonded heteroatom selected amongst N, O and S and exhibits a free electron pair
  • m is 0 or 1
  • c) -R”- is a bivalent organic group providing attachment to the carrier via one of its free valences and direct attachment to the heteroatom X 1 at the other one of its free valences
  • R’ - is a monovalent organic group directly attached to the heteroatom X 1 via its free valence; for use as a medicament for treating cancer and/or cancer metastasis.
  • the present invention relates to a pharmaceutical composition for use as a medicament for treating cancer and/or cancer metastasis, comprising the substance according to the present invention and pharmaceutically acceptable adjuvants.
  • the composition may further comprise a buffer, saline solution, and/or other pharmaceutically suitable adjuvants.
  • the pharmaceutical composition may be combined with other known cytostatic agent and/or agents that will ameliorate or facilitate the cancer treatment.
  • the present invention relates to a method of treating cancer and/or cancer metastasis in a subject in need thereof comprising administering a first dose or dosage of the substance according to the present invention or administering a first dose or dosage of the pharmaceutical composition according to the present invention to the subject. All embodiments disclosed herein may be combined and all embodiments are applicable to all aspects unless stated otherwise.
  • Figure 1 illustrating the effect of the substance of the present invention
  • Figure 2 graph illustrating the effect of the present substance at different concentrations.
  • Figure 3a and b graphs from CD3 positive scoring from histological sections collected at both central and peripheral tumour sites indicates a statistical difference (p ⁇ 0.05) in comparison with the NaCl group.
  • the present disclosure is generally directed to methods of treating cancer or cancer metastasis and to methods of reducing the proliferation of cancer cells.
  • administering refers to providing, contacting, and/or delivering a compound or compounds by any appropriate route to achieve the desired effect.
  • Administration may include, but is not limited to, oral, sublingual, parenteral (e.g., intravenous, subcutaneous, intracutaneous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional or intracranial injection), transdermal, topical, buccal, rectal, vaginal, nasal, ophthalmic, via inhalation, and implants.
  • Co-administered refers to simultaneous or sequential administration of multiple compounds or agents.
  • a first compound or agent may be administered before, concurrently with, or after administration of a second compound or agent.
  • the first compound or agent and the second compound or agent may be simultaneously or sequentially administered on the same day, or may be sequentially administered within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks or one month of each other.
  • compounds or agents are co-administered during the period in which each of the compounds or agents are exerting at least some physiological effect and/or has remaining efficacy.
  • Contacting refers to contacting a cell directly or indirectly in vitro, ex vivo, or in vivo (i.e. within a subject, such as a mammal, including humans, mice, rats, rabbits, cats, and dogs). Contacting a cell, which also includes “reacting" a cell, can occur as a result of administration to a subject. Contacting encompasses administration to a cell, tissue, mammal, subject, patient, or human. Further, contacting a cell includes adding an agent to a cell culture. Other suitable methods may include introducing or administering an agent to a cell, tissue, mammal, subject, or patient using appropriate procedures and routes of administration as defined herein.
  • Effective amount refers to a dosage of compounds or compositions effective for eliciting a desired effect. This term as used herein may also refer to an amount effective at bringing about a desired in vivo effect in an animal, mammal, or human, such as reducing proliferation of a cancer cell or treating cancer.
  • “Pharmaceutically acceptable”, as used herein, pertains to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • a subject e.g. human
  • Each carrier, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • Reducing proliferation of a cell refers to reducing, inhibiting, or preventing the survival, growth, or differentiation of a cell, including killing a cell.
  • a cell can be derived from any organism or tissue type and includes, for example, a cancer cell (e.g., neoplastic cells, tumour cells, and the like).
  • Subject is intended to include human and non-human animals.
  • the subject is a human.
  • Exemplary human subjects include a human patient having a disorder, e.g., cancer.
  • non-human animals includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals (such as sheep, dogs, cats, cows, pigs, etc.), and rodents (such as mice, rats, hamsters, guinea pigs, etc.).
  • “Susceptibility,” as used herein regarding a cancer cell, refers to the degree to which a cancer cell is affected by a chemotherapeutic agent. The cancer cell may not be affected at all, it may have its growth or proliferation slowed or halted without its being killed, or it may be killed. Susceptibility also refers to the degree a population of cancer cells, such as a tumour, is affected by a chemotherapeutic agent. "Increasing the susceptibility" of a cancer cell to a chemotherapeutic following contact or treatment with an agent, e.g., an inhibitor of an enzyme or an inhibitor of a protein-protein interaction, indicates that the cell is more affected by the chemotherapeutic agent than a corresponding cancer cell that has not been exposed to the agent.
  • an agent e.g., an inhibitor of an enzyme or an inhibitor of a protein-protein interaction
  • Treating refers to administering a regimen to the subject, e.g., the administration of a platinum-based therapeutic and/or another agent, such that at least one symptom of the disorder is healed, alleviated, relieved, altered, remedied, ameliorated, or improved. Treating includes administering an amount effective to alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder or the symptoms of the disorder.
  • the treatment may inhibit deterioration or worsening of a symptom of a disorder.
  • the substance according to the present invention comprises a carrier exhibiting the plurality of the scavenger structure. Every scavenger structure is firmly attached to the carrier, for instance covalently.
  • the substance as well as the carrier as such may be soluble or insoluble in aqueous liquids such as water, body fluids, such as blood, serum, plasma, urine, lymph, lachrymal fluid, intestinal juice, gastric juice, saliva, synovial fluid, etc.
  • the substance comprises a carrier which exhibits a plurality of a scavenger structure, said scavenger structure comprising a nucleophilic centre complying with the formula
  • the substance blocks or inhibits SSAO and thereby facilitates reduced new blood vessel formation.
  • Specific and preferred cancer types to be treated are listed herein.
  • Preferred scavenger structures thus have a nucleophilic centre which contain the first heteroatom X 1 together with a structure complying with formula II.
  • the nucleophilic center is or is part of a group selected amongst: a) amino groups preferably primary or secondary amino groups b) hydrazide groups such as -NH-NH2, e.g.
  • a semicarbazide group such as-NHCONHNH2, a carbazate group such as- OCONHNH2, a thiosemicarbazide group such as-NHCSNHNH2, a thiocarbazate group such as-OCSNHNH2 (formation of hydrazone, semicarbazone, thiocarbazone linkages/groups, etc when undergoing addition/elimination reactions with an aldehyde group) c) aminooxy groups, such as-ONH2, etc (formation oxime linkages/groups, etc when undergoing addition/elimination reactions with an aldehyde group), d) a thiol group e.g.-SH (Michael addition products are formed when the thiol group reacts with a C,C-double bond. The product may undergo keto-enol tautomerisation when the double bond is a,b to a keto-or aldehyde-carbonyl
  • These scavenger structures are believed to be effective when it comes to binding or interact with SSAO and thereby blocking or inhibiting SSAO. Therefore, these scavenger structures are believed to be effective when it comes to treating cancer.
  • the carrier is a macromolecular carrier and/or is water-soluble or water-insoluble and preferably exhibits polymer structure.
  • the carrier may be water-insoluble and define a support and/or the substance is attached to a water-insoluble support.
  • the carrier is a water- soluble polymer preferably polyvinyl alcohol, polysaccharides preferably glucose amino glucan, hyaluronic acid or dextran.
  • the substance has a scavenger structure capable of undergoing an addition reaction with a carbonyl group of an aldehyde group and/or with a carbon-carbon multiple bond to which is directly attached a carbonyl group, such as an aldehyde group.
  • the substance is carbazate-functionalized polyvinyl alcohol or a semicarbazide-functionalized polyvinyl alcohol. In on preferred embodiment the substance is carbazate-functionalized polyvinyl alcohol.
  • the substance is preferably soluble aqueous liquids such as water, body fluids, such as blood, serum, plasma, urine, lymph, lachrymal fluid, intestinal juice, gastric juice, saliva, synovial fluid, etc.
  • the selection of suitable carriers depends on the requirements of a particular use.
  • the typical carrier is selected amongst macromolecular compounds, i.e. is a compound which has a molecular weight of > 2000 dalton, preferably > 10000 dalton or > 50000 dalton, and preferably exhibits a polymeric structure, i.e. is a polymer which may be a homopolymer, copolymer or a chemical adduct between two or more polymers of different polymeric structure.
  • the carrier has a molecular weight of 10.000-30. OOOdalton.
  • Other suitable carriers may have molecular weights ⁇ 2000 dalton and exhibit polymeric structure as indicated by the possibility of the low numbers of monomeric units discussed below, e.g.
  • the scavenger structure including the first, the optional second nucleophilic centre and the various heteroatoms discussed for the scavenger structures are typically part of one and the same organic group/substituent attached to the macromolecular carrier. In certain variants different parts of a scavenger structure may be part of different group s/substituents attached to the carrier and/or part of the carrier.
  • suitable carrier polymers will among others depend on the actual application/use of the composition/method of the invention.
  • suitable polymeric carriers with respect to a particular polymeric structure and/or size may vary within a wide interval.
  • the number of monomeric subunits (mean value) of a polymer present in the carrier may be > 20 or > 100 or > 200 or > 300 or > 500 or > 1000 or > 2000 or > 20 000 or > 50 000 and/or ⁇ 50000 or ⁇ 20000 or ⁇ 2000 or ⁇ 1000 or ⁇ 500 or ⁇ 300 or ⁇ 200, or ⁇ 100 (with the proviso that >-limit always is lower than a ⁇ -limit when these values are combined to define intervals).
  • the carrier is a water soluble polymer, preferably polyvinyl alcohol, having a molecular weight of 10.000-30. OOOdalton.
  • Suitable numbers of scavenger structures or nucleophilic centres per monomeric unit of a polymer of the carrier will also depend on the use, the scavenger structure, etc, and may thus be found within a wide interval, such as ⁇ 80%, preferably ⁇ 50%, ⁇ 40% or ⁇ 30% with lower limits being preferably 0.01% or 0.1% or 1% or 5% or 10% or 15% where 100% corresponds to one scavenger structure or nucleophilic centre per monomeric unit.
  • the number of nucleophilic centres per monomeric unit may exceed 100%, such as > 100% or > 125% or > 150%.
  • the number of scavenger structures per carrier is 15- 50%, more preferably 20-40%.
  • the substance according to the present invention may be synthesized according to well- known protocols, for instance of the kinds given in WO 2009108100 (IPR-Sy stems AB) and references cited therein which are hereby incorporated by reference.
  • Second main aspect Pharmaceutical composition
  • a pharmaceutical composition for use as a medicament in the treatment of cancer or metastasis comprising the substance mentioned above and pharmaceutically acceptable adjuvants for use in the prevention or treatment of cancer metastasis.
  • the pharmaceutical composition may be combined with other known cytostatic agent and/or agents that will ameliorate or facilitate the cancer treatment.
  • the pharmaceutical composition may be used for the same type of anti-cancer prevention and treatment as mentioned for the substance above.
  • the patient in need of the pharmaceutical composition may be an animal or a human.
  • the pharmaceutical composition may for example be used for cancer treatment of cancer forms selected from the group consisting of a head cancer, neck cancer, ovarian cancer, breast cancer, pancreatic cancer, testicular cancer, melanoma, bladder cancer, lung cancer, sarcoma, squamous cell carcinoma, or small cell lung cancer cell.
  • the cancer may be primary cancer and/or cancer metastasis.
  • the substance is present in the composition as a pharmaceutical composition in which the substance is: a) in dry form, for instance as free particles, b) in dissolved form, typically in an aqueous liquid medium, and c) in suspended/dispersed form, i.e. as water-insoluble particles suspended in an aqueous liquid medium, d) attached to a support which is insoluble in aqueous liquid media.
  • the term “dissolved” in this context means that the substance is present as a solute.
  • the substance particles comprise substance in a pure form or diluted with some solid material. Useful concentrations of substance in formulations according to (b) can be found within a broad interval.
  • the composition may in addition to the substance contain buffers, salts, etc required for enabling acceptable conditions in vivo for the patient and for the reaction of the substance with the cancer. These constituents may be co-formulated with the substance in pharmaceutical composition.
  • compositions or formulations according to the present invention may conveniently be presented in unit dosage form and may be prepared by any methods known in the art of pharmacy. Such methods include the step of bringing into association the substance(s) with the carrier of the composition which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the substance with liquid carriers or finely divided solid carriers or both, and then if necessary, shaping the product.
  • Formulations may be in the form of liquids, solutions, suspensions, emulsions, elixirs, syrups, tablets, lozenges, granules, powders, capsules, cachets, pills, ampoules, suppositories, pessaries, ointments, gels, pastes, creams, sprays, mists, foams, lotions, oils, boluses, electuaries, or aerosols.
  • Formulations suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the substance; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion; as a bolus; as an electuary; or as a paste.
  • a tablet may be made by conventional means, e.g., compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the substance in a free-flowing form such as a powder or granules, optionally mixed with one or more binders (e.g. povidone, gelatin, acacia, sorbitol, tragacanth, hydroxypropylmethyl cellulose); fillers or diluents (e.g. lactose, microcrystalline cellulose, calcium hydrogen phosphate); lubricants (e.g. magnesium stearate, talc, silica); disintegrants (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose); surface-active or dispersing or wetting agents (e.g.
  • binders e.g. povidone, gelatin, acacia, sorbitol, tragacanth, hydroxypropylmethyl cellulose
  • fillers or diluents e.g. lactose, microcrystalline cellulose, calcium hydrogen
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be ftod so as to provide slow or controlled release of the substance therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile.
  • Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic, pyrogen-free, sterile injection solutions which may contain antioxidants, buffers, preservatives, stabilizers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs.
  • Suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.
  • Formulations may be in the form of liposomes or other microparticulate systems which are designed to target the substance to blood components or one or more organs.
  • Formulations suitable for administration by inhalation include those presented as an aerosol spray from a pressurized pack, with the use of a suitable propellant, such as dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane, carbon dioxide, or other suitable gases. Further formulations suitable for inhalation include those presented as a nebulizer.
  • a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane, carbon dioxide, or other suitable gases.
  • Further formulations suitable for inhalation include those presented as a nebulizer.
  • composition suitable for topical administration is preferably formulated as an ointment, cream, suspension, lotion, powder, solution, past, gel, spray, aerosol, or oil.
  • a formulation may comprise a patch or a dressing such as a bandage or adhesive plaster impregnated with the substance or the composition according to the present invention and optionally one or more excipients or diluents.
  • the concentration of the substance in the composition or formulation depends on the way of administration. In a general embodiment the concentration of the substance in the composition is 20-3000pg/mg, preferably 30-2500pg/mg, more preferably 100- 2500pg/mg, more preferably 500-2000 pg/ml.
  • the concentration of the substance in the composition is 20- 500 pg/ml, preferably 30-150pg/ml, more preferably 40-120pg/ml.
  • the composition is for parenteral administration preferably intravenous infusion and the concentration of the substance in the composition is 100- 2500pg/ml, preferably 500-2000pg/ml.
  • the composition is for topical application preferably formulated in a patch, bandage, dressing or as a spray or a cream, ointment or gel and where the concentration of the substance in the composition is 100-2500pg/ml, preferably 500- 2000pg/ml.
  • Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects of the treatments described herein.
  • the selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, and the age, sex, weight, condition, general health, and prior medical history of the patient.
  • the amount of compound and route of administration will ultimately be at the discretion of the physician, although generally the dosage will be to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.
  • Administration in vivo can be effected in one dose, continuously or intermittently (e.g. in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • a suitable dose of an substance is in the range of about 100 pg to about 1.5 mg per kilogram body weight of the subject per day.
  • the clinician may utilize preferred dosages as warranted by the condition of the subject being treated.
  • the substance according to the invention may be administered at a dosing schedule described herein, e.g., once every one, two, three, four, five or six weeks.
  • the first dose or dosage is 0.005 to 15mg/kg weight of the subject, preferably 0.05 to 5mg/kg weight of the subject, more preferably 0.5 to 3 mg/kg weight of the subject.
  • the first dose or dosage is administrated 1-3 times/7 days.
  • the substance according to the invention and an optional additional chemotherapeutic agent(s) or other agents do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, have to be administered by different routes.
  • the determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician.
  • the initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
  • the actual dosages of the compounds employed may be varied depending upon the requirements of the subject and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small amounts until the optimum effect under the circumstances is reached.
  • the order of administration may be varied.
  • the determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the disease being treated and the condition of the subject.
  • the practicing physician can modify each protocol for the administration of a component of the treatment according to the individual subject's needs, as the treatment proceeds.
  • the attending clinician in judging whether treatment is effective at the dosage administered, will consider the general well-being of the subject as well as more definite signs such as relief of disease-related symptoms, inhibition of tumour growth, actual shrinkage of the tumour, or inhibition of metastasis. Size of the tumour can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether the growth of the tumour has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
  • the methods described herein can be used with any cancer cell or in a subject having any type of cancer.
  • the cancer can be a carcinoma, a sarcoma, a myeloma, a leukemia, a lymphoma or a mixed type.
  • An exemplary list of cancers include but are not limited to:
  • Digestive/gastrointestinal cancers such as anal cancer; bile duct cancer; extrahepatic bile duct cancer; appendix cancer; carcinoid tumour, gastrointestinal cancer; colon cancer; colorectal cancer including childhood colorectal cancer; esophageal cancer including childhood esophageal cancer; gallbladder cancer; gastric (stomach) cancer including childhood gastric (stomach) cancer; hepatocellular (liver) cancer including adult (primary) hepatocellular (liver) cancer and childhood (primary) hepatocellular (liver) cancer; pancreatic cancer including childhood pancreatic cancer; sarcoma, rhabdomyosarcoma; islet cell pancreatic cancer; rectal cancer; and small intestine cancer;
  • Endocrine cancers such as islet cell carcinoma (endocrine pancreas); adrenocortical carcinoma including childhood adrenocortical carcinoma; gastrointestinal carcinoid tumour; parathyroid cancer; pheochromocytoma; pituitary tumour; thyroid cancer including childhood thyroid cancer; childhood multiple endocrine neoplasia syndrome; and childhood carcinoid tumour;
  • Eye cancers such as intraocular melanoma; and retinoblastoma;
  • Musculoskeletal cancers such as Ewing's family of tumours; osteosarcoma/malignant fibrous histiocytoma of the bone; childhood rhabdomyosarcoma; soft tissue sarcoma including adult and childhood soft tissue sarcoma; clear cell sarcoma of tendon sheaths; and uterine sarcoma;
  • Breast cancer such as breast cancer including childhood and male breast cancer and breast cancer in pregnancy;
  • Neurologic cancers such as childhood brain stemglioma; brain tumour; childhood cerebellar astrocytoma; childhood cerebral astrocytoma/malignant glioma; childhood ependymoma; childhood medulloblastoma; childhood pineal and supratentorial primitive neuroectodermal tumours; childhood visual pathway and hypothalamic glioma; other childhood brain cancers; adrenocortical carcinoma; central nervous system lymphoma, primary; childhood cerebellar astrocytoma; neuroblastoma; craniopharyngioma; spinal cord tumours; central nervous system atypical teratoid/rhabdoid tumour; central nervous system embryonal tumours; and childhood supratentorial primitive neuroectodermal tumours and pituitary tumour;
  • Genitourinary cancers such as bladder cancer including childhood bladder cancer; renal cell (kidney) cancer; ovarian cancer including childhood ovarian cancer; ovarian epithelial cancer; ovarian low malignant potential tumour; penile cancer; prostate cancer; renal cell cancer including childhood renal cell cancer; renal pelvis and ureter, transitional cell cancer; testicular cancer; urethral cancer; vaginal cancer; vulvar cancer; cervical cancer; Wilms tumour and other childhood kidney tumours; endometrial cancer; and gestational trophoblastic tumour; Germ cell cancers such as childhood extracranial germ cell tumour; extragonadal germ cell tumour; ovarian germ cell tumour;
  • Head and neck cancers such as lip and oral cavity cancer; oral cancer including childhood oral cancer; hypopharyngeal cancer; laryngeal cancer including childhood laryngeal cancer; metastatic squamous neck cancer with occult primary; mouth cancer; nasal cavity and paranasal sinus cancer; nasopharyngeal cancer including childhood nasopharyngeal cancer; oropharyngeal cancer; parathyroid cancer; pharyngeal cancer; salivary gland cancer including childhood salivary gland cancer; throat cancer; and thyroid cancer;
  • Hematologic/blood cell cancers such as a leukemia (e.g., acute lymphoblastic leukemia including adult and childhood acute lymphoblastic leukemia; acute myeloid leukemia including adult and childhood acute myeloid leukemia; chronic lymphocytic leukemia; chronic myelogenous leukemia; and hairy cell leukemia); a lymphoma (e.g., AIDS-related lymphoma; cutaneous T-cell lymphoma; Hodgkin's lymphoma including adult and childhood Hodgkin's lymphoma and Hodgkin's lymphoma during pregnancy; non-Hodgkin's lymphoma including adult and childhood non-Hodgkin's lymphoma and non-Hodgkin's lymphoma during pregnancy; mycosis fungoides; Sezary syndrome;
  • a lymphoma e.g., acute lymphoblastic leukemia including adult and childhood acute lymphoblastic leukemia; acute myeloid
  • Respiratory cancers such as adult malignant mesothelioma; childhood malignant mesothelioma; malignant thymoma; childhood thymoma; thymic carcinoma; bronchial adenomas/carcinoids including childhood bronchial adenomas/carcinoids; pleuropulmonary blastoma; non-small cell lung cancer; and small cell lung cancer;
  • Skin cancers such as Kaposi's sarcoma; Merkel cell carcinoma; melanoma; and childhood skin cancer;
  • AIDS-related malignancies Other childhood cancers, unusual cancers of childhood and cancers of unknown primary site; and metastases of the aforementioned cancers can also be treated or prevented in accordance with the methods described herein.
  • the methods described herein may be suited for bladder, testicular, ovarian, head and neck, cervical, lung (e.g., small cell lung), mesothelioma, esophageal, melanoma, brain tumour, neuroblastoma, colorectal, Wilms' tumour, retinoblastoma, breast, endometrial, adrenocortical, anal, biliary tract, carcinoid tumours, choriocarcinoma, gastric, liver cancer, non-Hodgkin's lymphoma, osteosarcoma, soft- tissue sarcomas, penile, malignant thymoma, anaplastic thyroid cancer, rhabdoid tumour of the kidney, advanced medullary thyroid cancer, carcinoid, mesothelioma, bone, gliomas, squamous cell carcinoma, pancratic or prostate cancers.
  • the method is for treating breast cancer or melanoma
  • the methods may be used for bladder cancer (e.g., muscle- invasive bladder carcinoma, advanced or metastatic bladder carcinoma), testicular cancer (e.g., nonseminomatous testicular carcinoma, disseminated seminoma testis or extragonadal germ-cell tumours), ovarian cancer (e.g., ovarian epithelial cancer or ovarian germ-cell tumours), head and neck cancer (e.g., squamous cell carcinoma), breast cancer, pancreatic cancer, sarcomas, cervical cancer (e.g., invasive, metastatic or recurrent cervical cancer), lung cancer (e.g., small cell lung cancer or non-small cell lung cancer), Wilms' tumour, brain tumours (e.g., gliomas, medulloblastoma or germ cell tumours), neuroblastoma, retinoblastoma, mesothelioma (e.g., malignant pleural mesothelioma), es
  • the cancer is a SSAO associated or related cancer such as skin cancer, colorectal cancer, renal cancer, urothelial cancer (Example 2 and 3).
  • the cancer is skin cancer, colorectal cancer, renal cancer, urothelial cancer.
  • the skin cancer is selected from melanoma or squamous cell carcinoma. Combination with other drugs or therapies
  • Administered "in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous" or "concurrent delivery”.
  • the delivery of one treatment ends before the delivery of the other treatment begins.
  • the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • the additional therapeutic agent can be administered simultaneously with the present substance, e.g. PVAC, in the same or in separate compositions, or sequentially.
  • the present substance is administrated in combination with an immunotherapeutic substance such as an immunomodulator.
  • the substance eg. PVAC
  • other therapeutic treatment modalities including surgery, radiation, cryosurgery, and/or thermotherapy.
  • combination therapies may advantageously utilize lower dosages of the administered agent and/or other chemotherapeutic agent, thus avoiding possible toxicities or complications associated with the various therapies.
  • radiation includes, but is not limited to, external-beam therapy which involves three dimensional, conformal radiation therapy where the field of radiation is designed to conform to the volume of tissue treated; interstitial-radiation therapy where seeds of radioactive compounds are implanted using ultrasound guidance; and a combination of external -beam therapy and interstitial -radiation therapy.
  • the substance according to the present invention is administered with at least one additional therapeutic agent, such as a chemotherapeutic agent.
  • the substance is administered in combination with one or more additional chemotherapeutic agents, e.g., with one or more chemotherapeutic agents described herein.
  • chemotherapeutic agents include, but are not limited to, e.g., the following: alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): uracil mustard (Aminouracil Mustard®, Chlorethaminacil®, Demethyldopan®, Desmethyldopan®, Haemanthamine®, Nordopan®, Uracil nitrogen Mustard®, Uracillost®, Uracilmostaza®, Uramustin®, Uramustine®), chlormethine (Mustargen®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®, Procytox®, RevimmuneTM), ifosfamide (Mitoxana®), melphalan (Alkeran®), Chlorambucil (Leukeran®), pipobroman (Amedel®
  • Anti-EGFR antibodies e.g., cetuximab (Erbitux®), panitumumab (Vectibix®), and gefitinib (Iressa®)).
  • Anti-Her-2 antibodies e.g., trastuzumab (Herceptin®) and other antibodies from Genentech.
  • Antimetabolites including, without limitation, folic acid antagonists (also referred to herein as antifolates), pyrimidine analogs, purine analogs and adenosine deaminase inhibitors: methotrexate (Rheumatrex®, Trexall®), 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), cytarabine (Cytosar-U®, Tarabine PFS), 6-mercaptopurine (Puri-Nethol®)), 6-thioguanine (Thioguanine Tabloid®), fludarabine phosphate (Fludara®), pentostatin (Nipent®), pemetrexed (Alimta®), raltitrexed (Tomudex®), cladribine (Leustatin®), clofarabine (Clofarex®, Clolar®), mercaptopurine (Puri-
  • Preferred antimetabolites include, e.g., 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), capecitabine (Xeloda®), pemetrexed (Alimta®), raltitrexed (Tomudex®) and gemcitabine (Gemzar®).
  • vinca alkaloids vinblastine (Velban®, Velsar®), vincristine (Vincasar®, Oncovin®), vindesine (Eldisine®), vinorelbine (Navelbine®).
  • platinum-based agents carboplatin (Paraplat®, Paraplatin®), cisplatin (Platinol®), oxaliplatin (Eloxatin®).
  • Anthracy clines daunorubicin (Cerubidine®, Rubidomycin®), doxorubicin (Adriamycin®), epirubicin (Ellence®), idarubicin (Idamycin®), mitoxantrone (Novantrone®), valrubicin (Valstar®).
  • Preferred anthracyclines include daunorubicin (Cerubidine®, Rubidomycin®) and doxorubicin (Adriamycin®).
  • Topoisom erase inhibitors topotecan (Hycamtin®), irinotecan (Camptosar®), etoposide (Toposar®, VePesid®), teniposide (Vumon®), lamellarin D, SN-38, camptothecin (e.g., GG-101).
  • Taxanes paclitaxel (Taxol®), docetaxel (Taxotere®), larotaxel, cabazitaxel.
  • Epothilones ixabepilone, epothilone B, epothilone D, BMS310705, dehydelone, ZK-Epothilone (ZK-EPO).
  • Antibiotics actinomycin (Cosmegen®), bleomycin (Blenoxane®), hydroxyurea (Droxia®, Hydrea®), mitomycin (Mitozytrex®, Mutamycin®).
  • Immunomodulators lenalidomide (Revlimid®), thalidomide (Thalomid®).
  • Immune cell antibodies alemtuzamab (Campath®), gemtuzumab (Myelotarg®), rituximab (Rituxan®), tositumomab (Bexxar®).
  • Interferons e.g., IFN-alpha (Alferon®, Roferon-A®) Intron®-A) or IFN- gamma (Actimmune®)
  • IFN-alpha Alferon®, Roferon-A®
  • IFN-gamma Actimmune®
  • Interleukins IL-1, IL-2 (Proleukin®), IL-24, IL-6 (Sigosix®), IL-12.
  • HSP90 inhibitors e.g., geldanamycin or any of its derivatives.
  • the HSP90 inhibitor is selected fromgeldanamycin, 17-alkylamino-17- desmethoxygeldanamycin (" 17-AAG”) or 17-(2-dimethylaminoethyl)amino-17- desmethoxygeldanamycin (" 17-DMAG").
  • Anti-androgens which include, without limitation nilutamide (Nilandron®) and bicalutamide (Caxodex®).
  • Antiestrogens which include, without limitation tamoxifen (Nolvadex®), toremifene (Fareston®), letrozole (Femara®), testolactone (Teslac®), anastrozole (Arimidex®), bicalutamide (Casodex®), exemestane (Aromasin®), flutamide (Eulexin®), fulvestrant (Faslodex®), raloxifene (Evista®) Keoxifene®) and raloxifene hydrochloride.
  • Anti-hypercalcaemia agents which include without limitation gallium (III) nitrate hydrate (Ganite®) and pamidronate disodium (Aredia®).
  • Apoptosis inducers which include without limitation ethanol, 2-[[3-(2,3- dichlorophenoxy)propyl]amino]-(9Cl), gambogic acid, embelin and arsenic trioxide (Trisenox®).
  • Aurora kinase inhibitors which include without limitation binucleine 2.
  • Bruton's tyrosine kinase inhibitors which include without limitation terreic acid.
  • Calcineurin inhibitors which include without limitation cypermethrin, deltamethrin, fenvalerate and tyrphostin 8.
  • CaM kinase II inhibitors which include without limitation 5- Isoquinolinesulfonic acid, 4-[ ⁇ 2S)-2-[(5-isoquinolinylsulfonyl)methylamino]-3-oxo-3- (4-phenyl-l-pipe-razinyl)propyl]phenyl ester and benzenesulfonamide.
  • CD45 tyrosine phosphatase inhibitors which include without limitation phosphonic acid.
  • CDC25 phosphatase inhibitors which include without limitation 1 ,4- naphthalene dione, 2,3-bis[(2-hydroxyethyl)thio]-(9Cl).
  • CHK kinase inhibitors which include without limitation debromohymenialdisine.
  • Cyclooxygenase inhibitors which include without limitation lH-indole-3- acetamide, l-(4-chlorobenzoyl)-5-methoxy-2-methyl-N-(2-phenylethyl)-(9Cl), 5-alkyl substituted 2-arylaminophenylacetic acid and its derivatives (e.g., celecoxib (Celebrex®), rofecoxib (Vioxx®), etoricoxib (Arcoxia®), lumiracoxib (Prexige®), valdecoxib (Bextra®) or 5-alkyl-2-arylaminophenylacetic acid).
  • celecoxib Celebrex®
  • rofecoxib Vioxx®
  • etoricoxib etoricoxib
  • lumiracoxib Prexige®
  • valdecoxib Bextra®
  • 5-alkyl-2-arylaminophenylacetic acid e.g., cele
  • cRAF kinase inhibitors which include without limitation 3-(3,5-dibromo-4- hydroxybenzylidene)-5-iodo-l,3-dihydroindol-2-one and benzamide, 3-(dimethylamino)- N-[3-[(4-hydroxybenzoyl)ami no]-4-methyl phenyl ]-(9Cl).
  • Cyclin dependent kinase inhibitors which include without limitation olomoucine and its derivatives, purvalanol B, roascovitine (Seliciclib®), indirubin, kenpaullone, purvalanol A and indirubin-3 '-monooxime.
  • Cysteine protease inhibitors which include without limitation 4- morpholinecarboxamide, N-[lS)-3-fluoro-2-oxo-l-(2-phenylethyl)propyl]amino]-2-oxo-l- (phenylmethy4)ethyl]-(9Cl).
  • DNA intercalators which include without limitation plicamycin (Mithracin®) and daptomycin (Cubicin®).
  • DNA strand breakers which include without limitation bleomycin (Blenoxane®).
  • E3 ligase inhibitors which include without limitation N-((3,3,3-trifruoro-2- trifluoromethyl)propionyl)sulfanilamide.
  • EGF Pathway Inhibitors which include, without limitation tyrphostin 46, EKB- 569, erlotinib (Tarceva®), gefitinib (Iressa®), lapatinib (Tykerb®) and those compounds that are generically and specifically disclosed in WO 97/02266, EP 0 564409, WO 99/03854, EP 0 520722, EP 0 566226, EP 0787722, EP 0 837 063, U.S. Pat. No. 5,747,498, WO98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and WO 96/33980.
  • Farnesyltransferase inhibitors which include without limitation A- hydroxyfamesylphosphonic acid, butanoic acid, 2-[(2S)-2-[[(2S,3S)-2-[[(2R)-2-amino-3- mercaptopropyl] amino] -3 -methylpent-yl] oxy ] -l-oxo-3 -phenylpropyl] amino-l -4- (methylsulfonyl)-l-methylethylester (2S)-(9C1), and manumycin A.
  • Flk-1 kinase inhibitors which include without limitation 2-propenamide, 2- cyano-3-[4-hydroxy-3,5-bis(l-methylethyl)phenyl]-N-(3-phenylpropyl)-(2E-)-(9Cl).
  • Glycogen synthase kinase-3 (GSK3) inhibitors which include without limitation indirubin-3 '-monooxime.
  • Histone deacetylase (HD AC) inhibitors which include without limitation suberoylanilide hydroxamic acid (SAHA), [4-(2-amino-phenylcarbamoyl)-benzyl]- carbamic acid pyridine-3 -ylmethylester and its derivatives, butyric acid, pyroxamide, trichostatin A, oxamflatin, apicidin, depsipeptide, depudecin, trapoxin and compounds disclosed in WO 02/22577.
  • SAHA suberoylanilide hydroxamic acid
  • pyroxamide pyroxamide
  • trichostatin A oxamflatin
  • apicidin apicidin
  • depsipeptide depudecin
  • trapoxin and compounds disclosed in WO 02/22577.
  • I-kappa B-alpha kinase inhibitors which include without limitation 2- propenenitrile, 3-[(4-methylphenyl)sulfonyl]-(2E)-(9Cl).
  • Imidazotetrazinones which include without limitation temozolomide (Methazolastone®, Temodar® and its derivatives (e.g., as disclosed generically and specifically in U.S. Pat. No. 5,260,291) and Mitozolomide.
  • Insulin tyrosine kinase inhibitors which include without limitation hydroxyl-2 - naphthalenylmethylphosphonic acid.
  • c-Jun-N-terminal kinase (JNK) inhibitors which include without limitation pyrazoleanthrone and epigallocatechin gallate.
  • Mitogen-activated protein kinase (MAP) inhibitors which include without limitation benzenesulfonamide, N-[2-[[[3-(4-chlorophenyl)-2- propenyl]methyl]amino]methyl]phenyl]-N-(2-hydroxyethyl)-4-methoxy-(9Cl).
  • MDM2 inhibitors which include without limitation trans-4-iodo, 4'-boranyl- chalcone.
  • MEK inhibitors which include without limitation butanedinitrile, bis[amino[2-aminophenyl)thio]methylene]-(9Cl).
  • MMP inhibitors which include without limitation Actinonin, epigallocatechin gallate, collagen peptidomimetic and non-peptidomimetic inhibitors, tetracycline derivatives marimastat (Marimastat®), prinomastat, incyclinide (Metastat®), shark cartilage extract AE-941 (eovastat®), Tanomastat, TAA21 1, MMI270B or AAJ996.
  • mTor inhibitors which include without limitation rapamycin (Rapamune®), and analogs and derivatives thereof, AP23573 (also known as ridaforolimus, deforolimus, or MK-8669), CCI-779 (also known as temsirolimus) (Torisel®) and SDZ-RAD.
  • NGFR tyrosine kinase inhibitors which include without limitation tyrphostin
  • p38 MAP kinase inhibitors which include without limitation Phenol, 4-[4-(4- fluorophenyl)-5-(4-pyridinyl)-lH-imidazol-2-yl]-(9Cl), and benzamide, 3- (dimethylamino)-N-[3-[(4-hydroxylbenzoyl)amino]-4-methylphenyl]-(9Cl).
  • p56 tyrosine kinase inhibitors which include without limitation damnacanthal and tyrphostin 46.
  • PDGF pathway inhibitors which include without limitation tyrphostin AG
  • Phosphatase inhibitors which include without limitation cantharidic acid, cantharidin, and L-leucinamide.
  • Protein phosphatase inhibitors which include without limitation cantharidic acid, cantharidin, L-P-bromotetramisole oxalate, 2(5H)-furanone, 4-hydroxy-5- (hydroxymethyl)-3-(l-oxohexadecyl)-(5R)-(9Cl) and benzylphosphonic acid.
  • PKC inhibitors which include without limitation l-H-pyrollo-2,5-dione,3-l-[[3- (dimethylamino)propyl]-lH-indol-3-yl]-4-(lH— indol-3-yl)-(9Cl), Bisindolylmaleimide IX, Sphinogosine, staurosporine, and Hypericin.
  • PKC delta kinase inhibitors which include without limitation rottlerin.
  • Polyamine synthesis inhibitors which include without limitation DMFO.
  • Proteasome inhibitors which include, without limitation aclacinomycin A, gliotoxin and bortezomib (Velcade®).
  • PTP1B inhibitors which include without limitation L-leucinamide.
  • protein tyrosine kinase inhibitors which include, without limitation tyrphostin Ag 216, tyrphostin Ag 1288, tyrphostin Ag 1295, geldanamycin, genistein and 7H-pyrollo[2,3-d]pyrimidine derivatives as generically and specifically described in PCT Publication No. WO 03/013541 and U.S. Publication No. 2008/0139587.
  • SRC family tyrosine kinase inhibitors which include without limitation PP 1 and PP2.
  • Syk tyrosine kinase inhibitors which include without limitation piceatannol.
  • Janus (JAK-2 and/or JAK-3) tyrosine kinase inhibitors which include without limitation tyrphostin AG 490 and 2-naphthyl vinyl ketone.
  • Retinoids which include without limitation isotretinoin (Accutane®, Amnesteem®, Cistane®, Claravis®, Sotret®) and tretinoin (Aberel®, Aknoten®,
  • RNA polymerase II elongation inhibitors which include without limitation 5,6- di chi oro- 1 -b eta-D -rib ofuranosy lb enzimi dazol e .
  • Serine/Threonine kinase inhibitors which include without limitation 2- aminopurine.
  • Sterol biosynthesis inhibitors which include without limitation squalene epoxidase and CYP2D6.
  • VEGF pathway inhibitors which include without limitation anti-VEGF antibodies, e.g., bevacizumab, and small molecules, e.g., sunitinib (Sutent®), sorafmib (Nexavar®), ZD6474 (also known as vandetanib) (ZactimaTM), SU6668, CP-547632 and AZD2171 (also known as cediranib) (RecentinTM).
  • sunitinib sunitinib
  • sorafmib sorafmib
  • ZactimaTM ZD6474
  • SU6668 cyclotanib
  • CP-547632 also known as cediranib
  • AZD2171 also known as cediranib
  • the substance according to the invention may be administered in combination with an immunosuppressive agent.
  • Immunosuppressive agents suitable for the combination include, but are not limited to natalizumab (Tysabri®), azathioprine (Imuran®), mitoxantrone (Novantrone®), mycophenolate mofetil (Cellcept®), cyclosporins (e.g., Cyclosporin A (Neoral®, Sandimmun®, Sandimmune®, SangCya®), calcineurin inhibitors (e.g., Tacrolimus (Prograf®, Protopic®), sirolimus (Rapamune®), everolimus (Afmitor®), cyclophosphamide (Cytoxan®, Neosar®), or methotrexate (Abitrexate®, Folex®, Methotrexate®, Mexate®)), fmgolimod, mycophenolate mofetil (CellCept®), myellC
  • the substance according to the invention is administered in combination with a CYP3 A4 inhibitor (e.g., ketoconazole (Nizoral®, Xolegel®), itraconazole (Sporanox®), clarithromycin (Biaxin®), atazanavir (Reyataz®), nefazodone (Serzone®, Nefadar®), saquinavir (Invirase®), telithromycin (Ketek®), ritonavir (Norvir®), amprenavir (also known as Agenerase, a prodrug version is fosamprenavir (Lexiva®, Telzir®), indinavir (Crixivan®), nelfmavir (Viracept®), delavirdine (Rescriptor®) or voriconazole (Vfend®)).
  • a CYP3 A4 inhibitor e.g., ketoconazole (Nizoral®, Xolegel®),
  • agents used in the modulation of tumour growth or metastasis in a clinical setting such as antiemetics, can also be administered as desired.
  • a first dose or dosage of the substance or the pharmaceutical composition mentioned above would be administered to a patient in need thereof.
  • the patient may be an animal or a human.
  • the administration ways or routes may vary according to the specific cancer or medical situation and are part of the knowledge of a medical practitioner.
  • a first dose or dosage is administrated to the subject and the first dose or dosage is preferably administrated 1-3 times/7 days, preferably 1 or 2 times/7 days.
  • the first dose or dosage is preferably 0.005 to 15mg/kg weight of the subject, preferably 0.05 to 5mg/kg weight of the subject, more preferably 0.5 to 3 mg/kg weight of the subject.
  • the total amount of the substance administrated to a subject per 7 days is preferably 5-20 mg/kg body weight of the subject, preferably 10-15mg/kg.
  • the administration may be done locally or systemically or in combination. Some are given below as illustrative examples of such administration ways or routes.
  • the composition is for parenteral administration preferably intravenous infusion and the concentration of the substance in the composition is 100- 2500pg/ml.
  • the total amount of substance per seven days is preferably 5-20mg/kg body weight of the subject, preferably 10-15 mg/kg body weight.
  • the cancer is skin cancer and preferably melanoma or squamous cell carcinoma.
  • the composition is for topical application preferably formulated in a patch, bandage, dressing, adhesive plaster or as a spray or a cream, ointment or gel and where the concentration of the substance in the composition is 100-2500pg/ml.
  • the substance or the composition is administrated to the site of treatment and preferably the administration is done 1-3 times per 7 days and preferably for 2 weeks or more.
  • the composition is formulated as a patch or a dressing such as a bandage or adhesive plaster impregnated with the substance or the composition according to the present invention and optionally one or more excipients or diluents.
  • EXAMPLE 3 - ANTI CANCER EFFECT A study was conducted to determine the cancerostatic effect of PVAC on B16.F10 and MDA-MB-231 tumours grown in mice. The study was conducted by Adlego Biomedical AB (Report AB-17-56-01).
  • tumours Following injection of B16.F10 cells into the C57BL/6J mice, the animals were monitored twice weekly with regard to weight, general health and tumour size. Once the tumour volumes reached 50m1 the animals were stratified into three experimental groups based on tumour size and were subjected to intra/peri tumoural injections three times per week for the duration of the study period. One experimental group received NaCl injections, while the remaining two groups received either 50 or 1000 pg of PVAC per injection. Tumour size and health status were recorded three times weekly during the treatment period. The animals were euthanized when the average tumour size of the control group reached 2ml in volume. The tumours were then excised and divided into two parts. One part was frozen on dry ice and the second part was placed in fixative for histological processing and analysis.
  • Body weights of both C57BL/6J and athymic nude mice increased at a normal rate throughout the study. There were no apparent effects on the treatments on body weight in either mouse strain during the study period.
  • a study of treating an early suspected skin cancer on the forehead of a subject was done by applying a composition according to the present invention comprising PVAC in water (2mg/ml).
  • the site of treatment had scaly red patches and a wart like appearance and was diagnosed by a physician as an early stage squamous cell carcinoma.
  • the composition was applied to the site using a cotton ball to which the composition had been added. The application was repeated every second day for two weeks.

Abstract

A substance comprising a carrier exhibiting at least one scavenger structure, said scavenger structure comprising a nucleophilic centre complying with formula (I): X1(-R''-)(-R')mHn, wherein a) X1 is a single-bonded heteroatom selected amongst N, O and S and exhibits a free electron pair, b) m is 0 or 1 and n is 1 or 2 with the sum of m plus n being 2 for X1= N and 1 for X1=S and O, c) -R''- is a bivalent organic group providing attachment to the carrier via one of its free valences and direct attachment to the heteroatom X1 at the other one of its free valences, and d) R'- is a monovalent organic group directly attached to the heteroatom X1 via its free valence; for use as a medicament for treating cancer and/or cancer metastasis.

Description

USE OF SUBSTANCE AND PHARMACEUTICAL COMPOSITION THEREOF, AND MEDICAL TREATMENTS OR USES THEREOF
TECHNICAL FIELD The present invention relates to the use of a pharmaceutical composition comprising a substance for prevention and treatment, as well as a method of prevention and treatment of cancer comprising administering said pharmaceutical composition.
BACKGROUND TECHNOLOGY Melanoma although a rare form of cancer is one of the most difficult to treat diseases once metastasized with increasing incidence and mortality world-wide. Treatment for the disease, once spread, was traditionally chemotherapy and radiotherapy but progress has been made and new targeted treatments as well as immunotherapy are in use and prognosis, although bleak, has improved.
Semicarbazide amine oxidase (SSAO or VAP-1) is an enzyme expressed for example by endothelial cells and smooth muscle cells in blood vessels. SSAO is normally found in intracellular granulae. The enzyme translocates to the cell membrane upon activation and can be solubilized by proteolytic cleavage from the membrane. SSAO exerts dual functions including leukocyte recruitment and also functions as ectoenzyme by the oxidation and conversion of primary amines to aldehydes, hydrogen peroxide and ammonium.
Enhanced SSAO activity has been shown to be associated with development of tumour growth. Deletion or reduction of SSAO enzyme activity reduces new blood vessel formation, lower the recruitment of immuno- suppressive Gr-l+CDl lb+ myeloid cells into tumours and reduces tumour growth as shown in melanoma and lymphoma animal models. A variety of different experimental SSAO inhibitor have been evaluated preclinically, but few are clinically tested and approved.
SUMMARY OF THE INVENTION
The object of the present invention is to provide an efficient substance acting as an substance or composition comprising the substance for treating cancer and especially semi-carbazide sensitive amine oxidase (SSAO) related cancer. Further, the present invention provides a substance and a composition that may easily be administrated for example topically for treating skin cancer such as melanoma or squamous cell carcinoma. Surprisingly, the present inventors have surprisingly found that administration of a polymeric substance comprising multiple nucleophilic centers has an anti-cancer effect.
Also, as compared to smaller agents, the molecular size of the substances according to the present invention effect pharmacokinetic and distribution profiles in a way that may be advantageous for certain indications where a prolonged and directed effect is needed. Other potential benefits include excellent in vivo tolerance and low manufacturing costs.
The inhibiting effect of the present substance on SSAO is shown in Example 2.
In the present specification substances according to the present invention which are capable of blocking or inhibiting or treating cancer are presented. In the experimental part, this kind of binding, blocking or treating effect of the cancer is shown in Example 3.
In Example 4 topical administration of the present substance showed a treating effect on early suspected squamous cell carcinoma.
The present inventors have surprisingly found that administration of the present substance or the present pharmaceutical composition has an inhibitory effect on SSAO. As shown in the examples (Example 2) the present substance blocks or inhibits SSAO activity and thereby the complications caused by the expression of SSAO may be prevented, inhibited or treated (Treatment of retinopathy, Example 3 and 4). Hence by presenting supporting data (Example 2) in combination with a confirmed mechanism (Inhibition of SSAO, Example 3) it is evident that the present invention relates to a substance, composition and method which may be used to treat cancer or metastasis especially those related to enhanced SSAO expression or activity.
In a first aspect the present invention relates to a substance comprising a carrier exhibiting at least one scavenger structure, said scavenger structure comprising a nucleophilic centre complying with the formula
Xl(-R”-)(-R’)mHn (formula I) wherein a) X1 is a single-bonded heteroatom selected amongst N, O and S and exhibits a free electron pair, b) m is 0 or 1 and n is 1 or 2 with the sum of m plus n being 2 for X1 = N and 1 for X1 = S and O, c) -R”- is a bivalent organic group providing attachment to the carrier via one of its free valences and direct attachment to the heteroatom X1 at the other one of its free valences, and d) R’ - is a monovalent organic group directly attached to the heteroatom X1 via its free valence; for use as a medicament for treating cancer and/or cancer metastasis.
In a second aspect the present invention relates to a pharmaceutical composition for use as a medicament for treating cancer and/or cancer metastasis, comprising the substance according to the present invention and pharmaceutically acceptable adjuvants. The composition may further comprise a buffer, saline solution, and/or other pharmaceutically suitable adjuvants. The pharmaceutical composition may be combined with other known cytostatic agent and/or agents that will ameliorate or facilitate the cancer treatment. In a third aspect the present invention relates to a method of treating cancer and/or cancer metastasis in a subject in need thereof comprising administering a first dose or dosage of the substance according to the present invention or administering a first dose or dosage of the pharmaceutical composition according to the present invention to the subject. All embodiments disclosed herein may be combined and all embodiments are applicable to all aspects unless stated otherwise.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 graph illustrating the effect of the substance of the present invention Figure 2 graph illustrating the effect of the present substance at different concentrations. Figure 3a and b graphs from CD3 positive scoring from histological sections collected at both central and peripheral tumour sites indicates a statistical difference (p<0.05) in comparison with the NaCl group. DETAILED DESCRIPTION OF THE INVENTION
The present disclosure is generally directed to methods of treating cancer or cancer metastasis and to methods of reducing the proliferation of cancer cells.
Definitions:
"Administration" or "administering", as used herein, refers to providing, contacting, and/or delivering a compound or compounds by any appropriate route to achieve the desired effect. Administration may include, but is not limited to, oral, sublingual, parenteral (e.g., intravenous, subcutaneous, intracutaneous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional or intracranial injection), transdermal, topical, buccal, rectal, vaginal, nasal, ophthalmic, via inhalation, and implants.
"Co-administered", as used herein, refers to simultaneous or sequential administration of multiple compounds or agents. A first compound or agent may be administered before, concurrently with, or after administration of a second compound or agent. The first compound or agent and the second compound or agent may be simultaneously or sequentially administered on the same day, or may be sequentially administered within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks or one month of each other. Suitably, compounds or agents are co-administered during the period in which each of the compounds or agents are exerting at least some physiological effect and/or has remaining efficacy.
"Contacting", as used herein as in "contacting a cell," refers to contacting a cell directly or indirectly in vitro, ex vivo, or in vivo (i.e. within a subject, such as a mammal, including humans, mice, rats, rabbits, cats, and dogs). Contacting a cell, which also includes "reacting" a cell, can occur as a result of administration to a subject. Contacting encompasses administration to a cell, tissue, mammal, subject, patient, or human. Further, contacting a cell includes adding an agent to a cell culture. Other suitable methods may include introducing or administering an agent to a cell, tissue, mammal, subject, or patient using appropriate procedures and routes of administration as defined herein.
"Effective amount", as used herein, refers to a dosage of compounds or compositions effective for eliciting a desired effect. This term as used herein may also refer to an amount effective at bringing about a desired in vivo effect in an animal, mammal, or human, such as reducing proliferation of a cancer cell or treating cancer.
"Pharmaceutically acceptable", as used herein, pertains to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, excipient, etc. must also be "acceptable" in the sense of being compatible with the other ingredients of the formulation.
"Reducing proliferation of a cell", as used herein, refers to reducing, inhibiting, or preventing the survival, growth, or differentiation of a cell, including killing a cell. A cell can be derived from any organism or tissue type and includes, for example, a cancer cell (e.g., neoplastic cells, tumour cells, and the like).
"Subject", as used herein, is intended to include human and non-human animals. In embodiments, the subject is a human. Exemplary human subjects include a human patient having a disorder, e.g., cancer. The term "non-human animals" includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals (such as sheep, dogs, cats, cows, pigs, etc.), and rodents (such as mice, rats, hamsters, guinea pigs, etc.).
"Susceptibility," as used herein regarding a cancer cell, refers to the degree to which a cancer cell is affected by a chemotherapeutic agent. The cancer cell may not be affected at all, it may have its growth or proliferation slowed or halted without its being killed, or it may be killed. Susceptibility also refers to the degree a population of cancer cells, such as a tumour, is affected by a chemotherapeutic agent. "Increasing the susceptibility" of a cancer cell to a chemotherapeutic following contact or treatment with an agent, e.g., an inhibitor of an enzyme or an inhibitor of a protein-protein interaction, indicates that the cell is more affected by the chemotherapeutic agent than a corresponding cancer cell that has not been exposed to the agent.
"Treat" or "treating", as used herein, the term a subject having a disorder refers to administering a regimen to the subject, e.g., the administration of a platinum-based therapeutic and/or another agent, such that at least one symptom of the disorder is healed, alleviated, relieved, altered, remedied, ameliorated, or improved. Treating includes administering an amount effective to alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder or the symptoms of the disorder. The treatment may inhibit deterioration or worsening of a symptom of a disorder.
First main aspect - A substance for use as a medicament for treating cancer or cancer metastasis and embodiments thereof
The substance according to the present invention comprises a carrier exhibiting the plurality of the scavenger structure. Every scavenger structure is firmly attached to the carrier, for instance covalently. The substance as well as the carrier as such may be soluble or insoluble in aqueous liquids such as water, body fluids, such as blood, serum, plasma, urine, lymph, lachrymal fluid, intestinal juice, gastric juice, saliva, synovial fluid, etc.
The substance comprises a carrier which exhibits a plurality of a scavenger structure, said scavenger structure comprising a nucleophilic centre complying with the formula
X'f-R’ -jC-R’j Hn (formula I) wherein a) X1 is a single-bonded heteroatom selected amongst N, O and S and exhibits a free electron pair, b) m is 0 or 1 and n is 1 or 2 with the sum of m plus n being 2 for X1 = N and 1 for X1 = S and O, c)-R”-is a bivalent organic group providing attachment to the carrier via one of its free valences and direct attachment to the heteroatom X1 at the other one of its free valences, and d) R’-is a monovalent organic group directly attached to the heteroatom X1 via its free valence for use as medicament for treating cancer and/or cancer metastasis.
The substance blocks or inhibits SSAO and thereby facilitates reduced new blood vessel formation. Specific and preferred cancer types to be treated are listed herein.
According to an embodiment of the present substance, either one or both of the organic groups R’-and-R”-, with preference for -R”-, comprise a structure of the formula: -CH2(X4)o-(C=X'V(X2)m- (formula II) wherein a) each of m’, n’ and o’ is 0 or 1, with preference for m’ being 1 with further preference for either one or both of n’ and o’ also being 1, b) each of X2 , X3, and X4, is selected amongst NH and a heteroatom S or O, with preference for either one or both of X2 and X4 being selected amongst NH and O with further preference for X3 being selected amongst NH, O and S, c) the left free valence provides binding to a monovalent alkyl group R*-or to the carrier via at least a bivalent alkylene group -R**-, each of which two groups comprises the methylene group-CTk-shown in formula II, and d) the right free valence binds directly to the first heteroatom X1.
The scavenger structure comprises a first nucleophilic centre which preferably is capable of participating in an addition reaction with the carbonyl group (C=0) of an aldehyde group, -and/or with a C,C-multiple bond to which one or more electron-withdrawing substituents preferably are directly attached.
Preferred scavenger structures thus have a nucleophilic centre which contain the first heteroatom X1 together with a structure complying with formula II. According to one embodiment of the present invention, the nucleophilic center is or is part of a group selected amongst: a) amino groups preferably primary or secondary amino groups b) hydrazide groups such as -NH-NH2, e.g. as part of a-CONHNH2 group, a semicarbazide group such as-NHCONHNH2, a carbazate group such as- OCONHNH2, a thiosemicarbazide group such as-NHCSNHNH2, a thiocarbazate group such as-OCSNHNH2 (formation of hydrazone, semicarbazone, thiocarbazone linkages/groups, etc when undergoing addition/elimination reactions with an aldehyde group) c) aminooxy groups, such as-ONH2, etc (formation oxime linkages/groups, etc when undergoing addition/elimination reactions with an aldehyde group), d) a thiol group e.g.-SH (Michael addition products are formed when the thiol group reacts with a C,C-double bond. The product may undergo keto-enol tautomerisation when the double bond is a,b to a keto-or aldehyde-carbonyl, see above).
These scavenger structures are believed to be effective when it comes to binding or interact with SSAO and thereby blocking or inhibiting SSAO. Therefore, these scavenger structures are believed to be effective when it comes to treating cancer.
According to a still further embodiment of the substance, the carrier is a macromolecular carrier and/or is water-soluble or water-insoluble and preferably exhibits polymer structure. The carrier may be water-insoluble and define a support and/or the substance is attached to a water-insoluble support. In a preferred embodiment the carrier is a water- soluble polymer preferably polyvinyl alcohol, polysaccharides preferably glucose amino glucan, hyaluronic acid or dextran. By using a water soluble carrier the substance is easier dissolved making it easier to prepare and it is also believed to increase the efficiency of the substance when administered. According to another embodiment, the substance has a scavenger structure capable of undergoing an addition reaction with a carbonyl group of an aldehyde group and/or with a carbon-carbon multiple bond to which is directly attached a carbonyl group, such as an aldehyde group. In embodiment, the substance is carbazate-functionalized polyvinyl alcohol or a semicarbazide-functionalized polyvinyl alcohol. In on preferred embodiment the substance is carbazate-functionalized polyvinyl alcohol.
The substance is preferably soluble aqueous liquids such as water, body fluids, such as blood, serum, plasma, urine, lymph, lachrymal fluid, intestinal juice, gastric juice, saliva, synovial fluid, etc.
The selection of suitable carriers depends on the requirements of a particular use. The typical carrier is selected amongst macromolecular compounds, i.e. is a compound which has a molecular weight of > 2000 dalton, preferably > 10000 dalton or > 50000 dalton, and preferably exhibits a polymeric structure, i.e. is a polymer which may be a homopolymer, copolymer or a chemical adduct between two or more polymers of different polymeric structure. In a preferred embodiment the carrier has a molecular weight of 10.000-30. OOOdalton. Other suitable carriers may have molecular weights < 2000 dalton and exhibit polymeric structure as indicated by the possibility of the low numbers of monomeric units discussed below, e.g. > 20 and < 100. The term “adduct polymer” in this context means a product formed by reacting two polymers exhibiting mutually groups capable of forming covalent bonds that link the two polymers together upon reaction of the two mutually groups with each other. See for instance WO 2009108100 and references cited therein. Suitable macromolecular carriers may thus be selected amongst synthetic polymers (= man-made polymers), biopolymers (nature-made polymers such as polysaccharides, polypeptides, proteins, etc) and biosynthetic polymers where “biosynthetic polymer” refers to a macromolecular carrier or compound exhibiting both a synthetic polymeric structure and a biopolymeric structure.
The scavenger structure including the first, the optional second nucleophilic centre and the various heteroatoms discussed for the scavenger structures are typically part of one and the same organic group/substituent attached to the macromolecular carrier. In certain variants different parts of a scavenger structure may be part of different group s/substituents attached to the carrier and/or part of the carrier.
Sizes/molecular weights of suitable carrier polymers will among others depend on the actual application/use of the composition/method of the invention. Thus suitable polymeric carriers with respect to a particular polymeric structure and/or size may vary within a wide interval. Thus as a rule the number of monomeric subunits (mean value) of a polymer present in the carrier may be > 20 or > 100 or > 200 or > 300 or > 500 or > 1000 or > 2000 or > 20 000 or > 50 000 and/or < 50000 or < 20000 or < 2000 or < 1000 or < 500 or < 300 or < 200, or < 100 (with the proviso that >-limit always is lower than a <-limit when these values are combined to define intervals). Preferred numbers of monomeric units may in some cases be found in the interval of 200 - 600. In a preferred embodiment the carrier is a water soluble polymer, preferably polyvinyl alcohol, having a molecular weight of 10.000-30. OOOdalton.
Suitable numbers of scavenger structures or nucleophilic centres per monomeric unit of a polymer of the carrier will also depend on the use, the scavenger structure, etc, and may thus be found within a wide interval, such as < 80%, preferably < 50%, <40% or < 30% with lower limits being preferably 0.01% or 0.1% or 1% or 5% or 10% or 15% where 100% corresponds to one scavenger structure or nucleophilic centre per monomeric unit. For scavenger structures containing two or more nucleophilic centres the number of nucleophilic centres per monomeric unit may exceed 100%, such as > 100% or > 125% or > 150%. In a preferred embodiment the number of scavenger structures per carrier is 15- 50%, more preferably 20-40%.
The substance according to the present invention may be synthesized according to well- known protocols, for instance of the kinds given in WO 2009108100 (IPR-Sy stems AB) and references cited therein which are hereby incorporated by reference.
All variants and examples of the first main aspect can be combined with the second and third main aspects unless expressly stated otherwise.
Second main aspect - Pharmaceutical composition comprising the substance for use as a medicament for treating cancer and/or cancer metastasis and embodiments thereof According to a second aspect, there is provided a pharmaceutical composition for use as a medicament in the treatment of cancer or metastasis, comprising the substance mentioned above and pharmaceutically acceptable adjuvants for use in the prevention or treatment of cancer metastasis. The pharmaceutical composition may be combined with other known cytostatic agent and/or agents that will ameliorate or facilitate the cancer treatment.
The pharmaceutical composition may be used for the same type of anti-cancer prevention and treatment as mentioned for the substance above. The patient in need of the pharmaceutical composition may be an animal or a human.
The pharmaceutical composition may for example be used for cancer treatment of cancer forms selected from the group consisting of a head cancer, neck cancer, ovarian cancer, breast cancer, pancreatic cancer, testicular cancer, melanoma, bladder cancer, lung cancer, sarcoma, squamous cell carcinoma, or small cell lung cancer cell.
The cancer may be primary cancer and/or cancer metastasis.
Formulation
The substance is present in the composition as a pharmaceutical composition in which the substance is: a) in dry form, for instance as free particles, b) in dissolved form, typically in an aqueous liquid medium, and c) in suspended/dispersed form, i.e. as water-insoluble particles suspended in an aqueous liquid medium, d) attached to a support which is insoluble in aqueous liquid media.
The term “dissolved” in this context means that the substance is present as a solute. The substance particles comprise substance in a pure form or diluted with some solid material. Useful concentrations of substance in formulations according to (b) can be found within a broad interval. The composition may in addition to the substance contain buffers, salts, etc required for enabling acceptable conditions in vivo for the patient and for the reaction of the substance with the cancer. These constituents may be co-formulated with the substance in pharmaceutical composition. Pharmaceutical compositions
The compositions or formulations according to the present invention may conveniently be presented in unit dosage form and may be prepared by any methods known in the art of pharmacy. Such methods include the step of bringing into association the substance(s) with the carrier of the composition which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the substance with liquid carriers or finely divided solid carriers or both, and then if necessary, shaping the product.
Formulations may be in the form of liquids, solutions, suspensions, emulsions, elixirs, syrups, tablets, lozenges, granules, powders, capsules, cachets, pills, ampoules, suppositories, pessaries, ointments, gels, pastes, creams, sprays, mists, foams, lotions, oils, boluses, electuaries, or aerosols.
Formulations suitable for oral administration (e.g. by ingestion) may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the substance; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion; as a bolus; as an electuary; or as a paste. A tablet may be made by conventional means, e.g., compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the substance in a free-flowing form such as a powder or granules, optionally mixed with one or more binders (e.g. povidone, gelatin, acacia, sorbitol, tragacanth, hydroxypropylmethyl cellulose); fillers or diluents (e.g. lactose, microcrystalline cellulose, calcium hydrogen phosphate); lubricants (e.g. magnesium stearate, talc, silica); disintegrants (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose); surface-active or dispersing or wetting agents (e.g. sodium lauryl sulfate); and preservatives (e.g. methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, sorbic acid). Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be ftod so as to provide slow or controlled release of the substance therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
Formulations suitable for parenteral administration (e.g. by injection, including cutaneous, subcutaneous, intramuscular, intravenous and intradermal), include aqueous and nonaqueous isotonic, pyrogen-free, sterile injection solutions which may contain antioxidants, buffers, preservatives, stabilizers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs. Examples of suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets. Formulations may be in the form of liposomes or other microparticulate systems which are designed to target the substance to blood components or one or more organs.
Formulations suitable for administration by inhalation include those presented as an aerosol spray from a pressurized pack, with the use of a suitable propellant, such as dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane, carbon dioxide, or other suitable gases. Further formulations suitable for inhalation include those presented as a nebulizer.
Pharmaceutical composition suitable for topical administration (e.g. dermal, intranasal, ocular) is preferably formulated as an ointment, cream, suspension, lotion, powder, solution, past, gel, spray, aerosol, or oil. Alternatively, a formulation may comprise a patch or a dressing such as a bandage or adhesive plaster impregnated with the substance or the composition according to the present invention and optionally one or more excipients or diluents.
The concentration of the substance in the composition or formulation depends on the way of administration. In a general embodiment the concentration of the substance in the composition is 20-3000pg/mg, preferably 30-2500pg/mg, more preferably 100- 2500pg/mg, more preferably 500-2000 pg/ml.
In another embodiment the the concentration of the substance in the composition is 20- 500 pg/ml, preferably 30-150pg/ml, more preferably 40-120pg/ml.
In one embodiment the composition is for parenteral administration preferably intravenous infusion and the concentration of the substance in the composition is 100- 2500pg/ml, preferably 500-2000pg/ml.
In one embodiment the composition is for topical application preferably formulated in a patch, bandage, dressing or as a spray or a cream, ointment or gel and where the concentration of the substance in the composition is 100-2500pg/ml, preferably 500- 2000pg/ml.
Dosages It will be appreciated that appropriate dosages of the substance or substances, and compositions comprising the substance substances, can vary from patient to patient.
Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects of the treatments described herein. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, and the age, sex, weight, condition, general health, and prior medical history of the patient. The amount of compound and route of administration will ultimately be at the discretion of the physician, although generally the dosage will be to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects. Administration in vivo can be effected in one dose, continuously or intermittently (e.g. in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
In general, a suitable dose of an substance is in the range of about 100 pg to about 1.5 mg per kilogram body weight of the subject per day.
When formulating the pharmaceutical compositions described herein, the clinician may utilize preferred dosages as warranted by the condition of the subject being treated. For example, in one embodiment, the substance according to the invention may be administered at a dosing schedule described herein, e.g., once every one, two, three, four, five or six weeks. In one embodiment the first dose or dosage is 0.005 to 15mg/kg weight of the subject, preferably 0.05 to 5mg/kg weight of the subject, more preferably 0.5 to 3 mg/kg weight of the subject. In one embodiment the first dose or dosage is administrated 1-3 times/7 days.
Also, in general, the substance according to the invention and an optional additional chemotherapeutic agent(s) or other agents do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, have to be administered by different routes. The determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician. The initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
The actual dosages of the compounds employed may be varied depending upon the requirements of the subject and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small amounts until the optimum effect under the circumstances is reached.
The particular choice of additional anti-proliferative cytotoxic agent(s) or radiation will depend upon the diagnosis of the attending physicians and their judgment of the condition of the subject and the appropriate treatment protocol.
If the substance according to the invention, and the additional chemotherapeutic agent(s) or agents and/or radiation, are not administered simultaneously or essentially simultaneously, then the order of administration may be varied. The determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the disease being treated and the condition of the subject.
Thus, in accordance with experience and knowledge, the practicing physician can modify each protocol for the administration of a component of the treatment according to the individual subject's needs, as the treatment proceeds.
The attending clinician, in judging whether treatment is effective at the dosage administered, will consider the general well-being of the subject as well as more definite signs such as relief of disease-related symptoms, inhibition of tumour growth, actual shrinkage of the tumour, or inhibition of metastasis. Size of the tumour can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether the growth of the tumour has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
Cancer forms
The methods described herein can be used with any cancer cell or in a subject having any type of cancer. The cancer can be a carcinoma, a sarcoma, a myeloma, a leukemia, a lymphoma or a mixed type. An exemplary list of cancers, include but are not limited to:
Digestive/gastrointestinal cancers such as anal cancer; bile duct cancer; extrahepatic bile duct cancer; appendix cancer; carcinoid tumour, gastrointestinal cancer; colon cancer; colorectal cancer including childhood colorectal cancer; esophageal cancer including childhood esophageal cancer; gallbladder cancer; gastric (stomach) cancer including childhood gastric (stomach) cancer; hepatocellular (liver) cancer including adult (primary) hepatocellular (liver) cancer and childhood (primary) hepatocellular (liver) cancer; pancreatic cancer including childhood pancreatic cancer; sarcoma, rhabdomyosarcoma; islet cell pancreatic cancer; rectal cancer; and small intestine cancer;
Endocrine cancers such as islet cell carcinoma (endocrine pancreas); adrenocortical carcinoma including childhood adrenocortical carcinoma; gastrointestinal carcinoid tumour; parathyroid cancer; pheochromocytoma; pituitary tumour; thyroid cancer including childhood thyroid cancer; childhood multiple endocrine neoplasia syndrome; and childhood carcinoid tumour;
Eye cancers such as intraocular melanoma; and retinoblastoma;
Musculoskeletal cancers such as Ewing's family of tumours; osteosarcoma/malignant fibrous histiocytoma of the bone; childhood rhabdomyosarcoma; soft tissue sarcoma including adult and childhood soft tissue sarcoma; clear cell sarcoma of tendon sheaths; and uterine sarcoma;
Breast cancer such as breast cancer including childhood and male breast cancer and breast cancer in pregnancy;
Neurologic cancers such as childhood brain stemglioma; brain tumour; childhood cerebellar astrocytoma; childhood cerebral astrocytoma/malignant glioma; childhood ependymoma; childhood medulloblastoma; childhood pineal and supratentorial primitive neuroectodermal tumours; childhood visual pathway and hypothalamic glioma; other childhood brain cancers; adrenocortical carcinoma; central nervous system lymphoma, primary; childhood cerebellar astrocytoma; neuroblastoma; craniopharyngioma; spinal cord tumours; central nervous system atypical teratoid/rhabdoid tumour; central nervous system embryonal tumours; and childhood supratentorial primitive neuroectodermal tumours and pituitary tumour;
Genitourinary cancers such as bladder cancer including childhood bladder cancer; renal cell (kidney) cancer; ovarian cancer including childhood ovarian cancer; ovarian epithelial cancer; ovarian low malignant potential tumour; penile cancer; prostate cancer; renal cell cancer including childhood renal cell cancer; renal pelvis and ureter, transitional cell cancer; testicular cancer; urethral cancer; vaginal cancer; vulvar cancer; cervical cancer; Wilms tumour and other childhood kidney tumours; endometrial cancer; and gestational trophoblastic tumour; Germ cell cancers such as childhood extracranial germ cell tumour; extragonadal germ cell tumour; ovarian germ cell tumour;
Head and neck cancers such as lip and oral cavity cancer; oral cancer including childhood oral cancer; hypopharyngeal cancer; laryngeal cancer including childhood laryngeal cancer; metastatic squamous neck cancer with occult primary; mouth cancer; nasal cavity and paranasal sinus cancer; nasopharyngeal cancer including childhood nasopharyngeal cancer; oropharyngeal cancer; parathyroid cancer; pharyngeal cancer; salivary gland cancer including childhood salivary gland cancer; throat cancer; and thyroid cancer;
Hematologic/blood cell cancers such as a leukemia (e.g., acute lymphoblastic leukemia including adult and childhood acute lymphoblastic leukemia; acute myeloid leukemia including adult and childhood acute myeloid leukemia; chronic lymphocytic leukemia; chronic myelogenous leukemia; and hairy cell leukemia); a lymphoma (e.g., AIDS-related lymphoma; cutaneous T-cell lymphoma; Hodgkin's lymphoma including adult and childhood Hodgkin's lymphoma and Hodgkin's lymphoma during pregnancy; non-Hodgkin's lymphoma including adult and childhood non-Hodgkin's lymphoma and non-Hodgkin's lymphoma during pregnancy; mycosis fungoides; Sezary syndrome;
Waldenstrom's macroglobulinemia; and primary central nervous system lymphoma); and other hematologic cancers (e.g., chronic myeloproliferative disorders; multiple myeloma/plasma cell neoplasm; myelodysplasia syndromes; and myelodysplastic/myeloproliferative disorders); Lung cancer such as non-small cell lung cancer; and small cell lung cancer;
Respiratory cancers such as adult malignant mesothelioma; childhood malignant mesothelioma; malignant thymoma; childhood thymoma; thymic carcinoma; bronchial adenomas/carcinoids including childhood bronchial adenomas/carcinoids; pleuropulmonary blastoma; non-small cell lung cancer; and small cell lung cancer;
Skin cancers such as Kaposi's sarcoma; Merkel cell carcinoma; melanoma; and childhood skin cancer;
AIDS-related malignancies; Other childhood cancers, unusual cancers of childhood and cancers of unknown primary site; and metastases of the aforementioned cancers can also be treated or prevented in accordance with the methods described herein.
The methods described herein may be suited for bladder, testicular, ovarian, head and neck, cervical, lung (e.g., small cell lung), mesothelioma, esophageal, melanoma, brain tumour, neuroblastoma, colorectal, Wilms' tumour, retinoblastoma, breast, endometrial, adrenocortical, anal, biliary tract, carcinoid tumours, choriocarcinoma, gastric, liver cancer, non-Hodgkin's lymphoma, osteosarcoma, soft- tissue sarcomas, penile, malignant thymoma, anaplastic thyroid cancer, rhabdoid tumour of the kidney, advanced medullary thyroid cancer, carcinoid, mesothelioma, bone, gliomas, squamous cell carcinoma, pancratic or prostate cancers. In one preferred embodiment the method is for treating breast cancer or melanoma.
In embodiments, the methods may be used for bladder cancer (e.g., muscle- invasive bladder carcinoma, advanced or metastatic bladder carcinoma), testicular cancer (e.g., nonseminomatous testicular carcinoma, disseminated seminoma testis or extragonadal germ-cell tumours), ovarian cancer (e.g., ovarian epithelial cancer or ovarian germ-cell tumours), head and neck cancer (e.g., squamous cell carcinoma), breast cancer, pancreatic cancer, sarcomas, cervical cancer (e.g., invasive, metastatic or recurrent cervical cancer), lung cancer (e.g., small cell lung cancer or non-small cell lung cancer), Wilms' tumour, brain tumours (e.g., gliomas, medulloblastoma or germ cell tumours), neuroblastoma, retinoblastoma, mesothelioma (e.g., malignant pleural mesothelioma), esophageal cancer (e.g., localized or advanced esophageal cancer), melanoma, and colorectal cancer.
In a preferred embodiment the cancer is a SSAO associated or related cancer such as skin cancer, colorectal cancer, renal cancer, urothelial cancer (Example 2 and 3). In one embodiment the cancer is skin cancer, colorectal cancer, renal cancer, urothelial cancer. In a preferred embodiment the skin cancer is selected from melanoma or squamous cell carcinoma. Combination with other drugs or therapies
Methods described herein may be used in further combination with other known therapies. Administered "in combination", as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as "simultaneous" or "concurrent delivery". In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
The additional therapeutic agent can be administered simultaneously with the present substance, e.g. PVAC, in the same or in separate compositions, or sequentially. Preferably the present substance is administrated in combination with an immunotherapeutic substance such as an immunomodulator.
In some embodiments the substance, eg. PVAC, is administered in combination with other therapeutic treatment modalities, including surgery, radiation, cryosurgery, and/or thermotherapy. Such combination therapies may advantageously utilize lower dosages of the administered agent and/or other chemotherapeutic agent, thus avoiding possible toxicities or complications associated with the various therapies. The phrase "radiation" includes, but is not limited to, external-beam therapy which involves three dimensional, conformal radiation therapy where the field of radiation is designed to conform to the volume of tissue treated; interstitial-radiation therapy where seeds of radioactive compounds are implanted using ultrasound guidance; and a combination of external -beam therapy and interstitial -radiation therapy.
In some embodiments, the substance according to the present invention is administered with at least one additional therapeutic agent, such as a chemotherapeutic agent. In certain embodiments, the substance is administered in combination with one or more additional chemotherapeutic agents, e.g., with one or more chemotherapeutic agents described herein.
Exemplary classes of chemotherapeutic agents include, but are not limited to, e.g., the following: alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): uracil mustard (Aminouracil Mustard®, Chlorethaminacil®, Demethyldopan®, Desmethyldopan®, Haemanthamine®, Nordopan®, Uracil nitrogen Mustard®, Uracillost®, Uracilmostaza®, Uramustin®, Uramustine®), chlormethine (Mustargen®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®, Procytox®, Revimmune™), ifosfamide (Mitoxana®), melphalan (Alkeran®), Chlorambucil (Leukeran®), pipobroman (Amedel®, Vercyte®), triethylenemelamine (Hem el®, Hexylen®, Hexastat®), triethylenethiophosphoramine, Temozolomide (Temodar®), thiotepa (Thioplex®), busulfan (Busilvex®, Myleran®), carmustine (BiCNU®), lomustine (CeeNU®), streptozocin (Zanosar®), and Dacarbazine (DTIC- Dome®).
Anti-EGFR antibodies (e.g., cetuximab (Erbitux®), panitumumab (Vectibix®), and gefitinib (Iressa®)).
Anti-Her-2 antibodies (e.g., trastuzumab (Herceptin®) and other antibodies from Genentech).
Antimetabolites (including, without limitation, folic acid antagonists (also referred to herein as antifolates), pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): methotrexate (Rheumatrex®, Trexall®), 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), cytarabine (Cytosar-U®, Tarabine PFS), 6-mercaptopurine (Puri-Nethol®)), 6-thioguanine (Thioguanine Tabloid®), fludarabine phosphate (Fludara®), pentostatin (Nipent®), pemetrexed (Alimta®), raltitrexed (Tomudex®), cladribine (Leustatin®), clofarabine (Clofarex®, Clolar®), mercaptopurine (Puri-Nethol®), capecitabine (Xeloda®), nelarabine (Arranon®), azacitidine (Vidaza®) and gemcitabine (Gemzar®). Preferred antimetabolites include, e.g., 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), capecitabine (Xeloda®), pemetrexed (Alimta®), raltitrexed (Tomudex®) and gemcitabine (Gemzar®).
[00106] vinca alkaloids: vinblastine (Velban®, Velsar®), vincristine (Vincasar®, Oncovin®), vindesine (Eldisine®), vinorelbine (Navelbine®).
[00107] additional platinum-based agents: carboplatin (Paraplat®, Paraplatin®), cisplatin (Platinol®), oxaliplatin (Eloxatin®).
Anthracy clines: daunorubicin (Cerubidine®, Rubidomycin®), doxorubicin (Adriamycin®), epirubicin (Ellence®), idarubicin (Idamycin®), mitoxantrone (Novantrone®), valrubicin (Valstar®). Preferred anthracyclines include daunorubicin (Cerubidine®, Rubidomycin®) and doxorubicin (Adriamycin®).
Topoisom erase inhibitors: topotecan (Hycamtin®), irinotecan (Camptosar®), etoposide (Toposar®, VePesid®), teniposide (Vumon®), lamellarin D, SN-38, camptothecin (e.g., GG-101).
Taxanes: paclitaxel (Taxol®), docetaxel (Taxotere®), larotaxel, cabazitaxel.
Epothilones: ixabepilone, epothilone B, epothilone D, BMS310705, dehydelone, ZK-Epothilone (ZK-EPO).
Antibiotics: actinomycin (Cosmegen®), bleomycin (Blenoxane®), hydroxyurea (Droxia®, Hydrea®), mitomycin (Mitozytrex®, Mutamycin®).
Immunomodulators: lenalidomide (Revlimid®), thalidomide (Thalomid®).
Immune cell antibodies: alemtuzamab (Campath®), gemtuzumab (Myelotarg®), rituximab (Rituxan®), tositumomab (Bexxar®).
Interferons (e.g., IFN-alpha (Alferon®, Roferon-A®) Intron®-A) or IFN- gamma (Actimmune®))
Interleukins: IL-1, IL-2 (Proleukin®), IL-24, IL-6 (Sigosix®), IL-12.
HSP90 inhibitors (e.g., geldanamycin or any of its derivatives). In certain embodiments, the HSP90 inhibitor is selected fromgeldanamycin, 17-alkylamino-17- desmethoxygeldanamycin (" 17-AAG") or 17-(2-dimethylaminoethyl)amino-17- desmethoxygeldanamycin (" 17-DMAG").
Anti-androgens which include, without limitation nilutamide (Nilandron®) and bicalutamide (Caxodex®).
Antiestrogens which include, without limitation tamoxifen (Nolvadex®), toremifene (Fareston®), letrozole (Femara®), testolactone (Teslac®), anastrozole (Arimidex®), bicalutamide (Casodex®), exemestane (Aromasin®), flutamide (Eulexin®), fulvestrant (Faslodex®), raloxifene (Evista®) Keoxifene®) and raloxifene hydrochloride.
Anti-hypercalcaemia agents which include without limitation gallium (III) nitrate hydrate (Ganite®) and pamidronate disodium (Aredia®).
Apoptosis inducers which include without limitation ethanol, 2-[[3-(2,3- dichlorophenoxy)propyl]amino]-(9Cl), gambogic acid, embelin and arsenic trioxide (Trisenox®).
Aurora kinase inhibitors which include without limitation binucleine 2.
Bruton's tyrosine kinase inhibitors which include without limitation terreic acid.
Calcineurin inhibitors which include without limitation cypermethrin, deltamethrin, fenvalerate and tyrphostin 8.
CaM kinase II inhibitors which include without limitation 5- Isoquinolinesulfonic acid, 4-[ {2S)-2-[(5-isoquinolinylsulfonyl)methylamino]-3-oxo-3- (4-phenyl-l-pipe-razinyl)propyl]phenyl ester and benzenesulfonamide.
CD45 tyrosine phosphatase inhibitors which include without limitation phosphonic acid.
CDC25 phosphatase inhibitors which include without limitation 1 ,4- naphthalene dione, 2,3-bis[(2-hydroxyethyl)thio]-(9Cl).
CHK kinase inhibitors which include without limitation debromohymenialdisine.
Cyclooxygenase inhibitors which include without limitation lH-indole-3- acetamide, l-(4-chlorobenzoyl)-5-methoxy-2-methyl-N-(2-phenylethyl)-(9Cl), 5-alkyl substituted 2-arylaminophenylacetic acid and its derivatives (e.g., celecoxib (Celebrex®), rofecoxib (Vioxx®), etoricoxib (Arcoxia®), lumiracoxib (Prexige®), valdecoxib (Bextra®) or 5-alkyl-2-arylaminophenylacetic acid). cRAF kinase inhibitors which include without limitation 3-(3,5-dibromo-4- hydroxybenzylidene)-5-iodo-l,3-dihydroindol-2-one and benzamide, 3-(dimethylamino)- N-[3-[(4-hydroxybenzoyl)ami no]-4-methyl phenyl ]-(9Cl).
Cyclin dependent kinase inhibitors which include without limitation olomoucine and its derivatives, purvalanol B, roascovitine (Seliciclib®), indirubin, kenpaullone, purvalanol A and indirubin-3 '-monooxime. Cysteine protease inhibitors which include without limitation 4- morpholinecarboxamide, N-[lS)-3-fluoro-2-oxo-l-(2-phenylethyl)propyl]amino]-2-oxo-l- (phenylmethy4)ethyl]-(9Cl).
DNA intercalators which include without limitation plicamycin (Mithracin®) and daptomycin (Cubicin®).
DNA strand breakers which include without limitation bleomycin (Blenoxane®).
E3 ligase inhibitors which include without limitation N-((3,3,3-trifruoro-2- trifluoromethyl)propionyl)sulfanilamide.
EGF Pathway Inhibitors which include, without limitation tyrphostin 46, EKB- 569, erlotinib (Tarceva®), gefitinib (Iressa®), lapatinib (Tykerb®) and those compounds that are generically and specifically disclosed in WO 97/02266, EP 0 564409, WO 99/03854, EP 0 520722, EP 0 566226, EP 0787722, EP 0 837 063, U.S. Pat. No. 5,747,498, WO98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and WO 96/33980.
Farnesyltransferase inhibitors which include without limitation A- hydroxyfamesylphosphonic acid, butanoic acid, 2-[(2S)-2-[[(2S,3S)-2-[[(2R)-2-amino-3- mercaptopropyl] amino] -3 -methylpent-yl] oxy ] -l-oxo-3 -phenylpropyl] amino-l -4- (methylsulfonyl)-l-methylethylester (2S)-(9C1), and manumycin A.
Flk-1 kinase inhibitors which include without limitation 2-propenamide, 2- cyano-3-[4-hydroxy-3,5-bis(l-methylethyl)phenyl]-N-(3-phenylpropyl)-(2E-)-(9Cl).
Glycogen synthase kinase-3 (GSK3) inhibitors which include without limitation indirubin-3 '-monooxime.
Histone deacetylase (HD AC) inhibitors which include without limitation suberoylanilide hydroxamic acid (SAHA), [4-(2-amino-phenylcarbamoyl)-benzyl]- carbamic acid pyridine-3 -ylmethylester and its derivatives, butyric acid, pyroxamide, trichostatin A, oxamflatin, apicidin, depsipeptide, depudecin, trapoxin and compounds disclosed in WO 02/22577.
I-kappa B-alpha kinase inhibitors (IKK) which include without limitation 2- propenenitrile, 3-[(4-methylphenyl)sulfonyl]-(2E)-(9Cl).
Imidazotetrazinones which include without limitation temozolomide (Methazolastone®, Temodar® and its derivatives (e.g., as disclosed generically and specifically in U.S. Pat. No. 5,260,291) and Mitozolomide. Insulin tyrosine kinase inhibitors which include without limitation hydroxyl-2 - naphthalenylmethylphosphonic acid. c-Jun-N-terminal kinase (JNK) inhibitors which include without limitation pyrazoleanthrone and epigallocatechin gallate. Mitogen-activated protein kinase (MAP) inhibitors which include without limitation benzenesulfonamide, N-[2-[[[3-(4-chlorophenyl)-2- propenyl]methyl]amino]methyl]phenyl]-N-(2-hydroxyethyl)-4-methoxy-(9Cl).
MDM2 inhibitors which include without limitation trans-4-iodo, 4'-boranyl- chalcone. [00147] MEK inhibitors which include without limitation butanedinitrile, bis[amino[2-aminophenyl)thio]methylene]-(9Cl).
MMP inhibitors which include without limitation Actinonin, epigallocatechin gallate, collagen peptidomimetic and non-peptidomimetic inhibitors, tetracycline derivatives marimastat (Marimastat®), prinomastat, incyclinide (Metastat®), shark cartilage extract AE-941 (eovastat®), Tanomastat, TAA21 1, MMI270B or AAJ996. mTor inhibitors which include without limitation rapamycin (Rapamune®), and analogs and derivatives thereof, AP23573 (also known as ridaforolimus, deforolimus, or MK-8669), CCI-779 (also known as temsirolimus) (Torisel®) and SDZ-RAD.
NGFR tyrosine kinase inhibitors which include without limitation tyrphostin
AG879. p38 MAP kinase inhibitors which include without limitation Phenol, 4-[4-(4- fluorophenyl)-5-(4-pyridinyl)-lH-imidazol-2-yl]-(9Cl), and benzamide, 3- (dimethylamino)-N-[3-[(4-hydroxylbenzoyl)amino]-4-methylphenyl]-(9Cl). p56 tyrosine kinase inhibitors which include without limitation damnacanthal and tyrphostin 46. PDGF pathway inhibitors which include without limitation tyrphostin AG
1296, tyrphostin 9,l,3-butadiene-l,l,3-tricarbonitrile, 2-amino-4-(lH-indol-5-yl)-(9Cl), imatinib (Gleevec®) and gefitinib (Iressa®) and those compounds generically and specifically disclosed in European Patent No. 0 564409 and PCT Publication No. WO 99/03854. Phosphatidylinositol 3 -kinase inhibitors which include without limitation wortmannin, and quercetin dihydrate.
Phosphatase inhibitors which include without limitation cantharidic acid, cantharidin, and L-leucinamide. Protein phosphatase inhibitors which include without limitation cantharidic acid, cantharidin, L-P-bromotetramisole oxalate, 2(5H)-furanone, 4-hydroxy-5- (hydroxymethyl)-3-(l-oxohexadecyl)-(5R)-(9Cl) and benzylphosphonic acid.
PKC inhibitors which include without limitation l-H-pyrollo-2,5-dione,3-l-[[3- (dimethylamino)propyl]-lH-indol-3-yl]-4-(lH— indol-3-yl)-(9Cl), Bisindolylmaleimide IX, Sphinogosine, staurosporine, and Hypericin.
PKC delta kinase inhibitors which include without limitation rottlerin.
Polyamine synthesis inhibitors which include without limitation DMFO.
Proteasome inhibitors which include, without limitation aclacinomycin A, gliotoxin and bortezomib (Velcade®). [00161] PTP1B inhibitors which include without limitation L-leucinamide. protein tyrosine kinase inhibitors which include, without limitation tyrphostin Ag 216, tyrphostin Ag 1288, tyrphostin Ag 1295, geldanamycin, genistein and 7H-pyrollo[2,3-d]pyrimidine derivatives as generically and specifically described in PCT Publication No. WO 03/013541 and U.S. Publication No. 2008/0139587.
SRC family tyrosine kinase inhibitors which include without limitation PP 1 and PP2.
Syk tyrosine kinase inhibitors which include without limitation piceatannol.
Janus (JAK-2 and/or JAK-3) tyrosine kinase inhibitors which include without limitation tyrphostin AG 490 and 2-naphthyl vinyl ketone.
Retinoids which include without limitation isotretinoin (Accutane®, Amnesteem®, Cistane®, Claravis®, Sotret®) and tretinoin (Aberel®, Aknoten®,
Avita®, Renova®, Retin-A®, Retin-A MICRO®, Vesanoid®).
RNA polymerase II elongation inhibitors which include without limitation 5,6- di chi oro- 1 -b eta-D -rib ofuranosy lb enzimi dazol e .
Serine/Threonine kinase inhibitors which include without limitation 2- aminopurine.
Sterol biosynthesis inhibitors which include without limitation squalene epoxidase and CYP2D6. VEGF pathway inhibitors, which include without limitation anti-VEGF antibodies, e.g., bevacizumab, and small molecules, e.g., sunitinib (Sutent®), sorafmib (Nexavar®), ZD6474 (also known as vandetanib) (Zactima™), SU6668, CP-547632 and AZD2171 (also known as cediranib) (Recentin™). Examples of chemotherapeutic agents are also described in the scientific and patent literature, see, e.g., Bulinski (1997) J. Cell Sci. 1 10:3055-3064; Panda (1997) Proc. Natl. Acad. Sci. USA 94: 10560-10564; Muhlradt (1997) Cancer Res. 57:3344- 3346; Nicolaou (1997) Nature 387:268-272; Vasquez (1997) Mol. Biol. Cell. 8:973-985; Panda (1996) J. Biol. Chem. 271 :29807-29812.
In some embodiments, the substance according to the invention may be administered in combination with an immunosuppressive agent. Immunosuppressive agents suitable for the combination include, but are not limited to natalizumab (Tysabri®), azathioprine (Imuran®), mitoxantrone (Novantrone®), mycophenolate mofetil (Cellcept®), cyclosporins (e.g., Cyclosporin A (Neoral®, Sandimmun®, Sandimmune®, SangCya®), calcineurin inhibitors (e.g., Tacrolimus (Prograf®, Protopic®), sirolimus (Rapamune®), everolimus (Afmitor®), cyclophosphamide (Cytoxan®, Neosar®), or methotrexate (Abitrexate®, Folex®, Methotrexate®, Mexate®)), fmgolimod, mycophenolate mofetil (CellCept®), mycophenolic acid (Myfortic®), anti-CD3 antibody, anti-CD25 antibody (e.g., Basiliximab (Simulect®) or daclizumab (Zenapax®)), and anti-TNF. alpha, antibody (e.g., Infliximab (Remicade®) or adalimumab (Humira®)).
In some embodiments, the substance according to the invention is administered in combination with a CYP3 A4 inhibitor (e.g., ketoconazole (Nizoral®, Xolegel®), itraconazole (Sporanox®), clarithromycin (Biaxin®), atazanavir (Reyataz®), nefazodone (Serzone®, Nefadar®), saquinavir (Invirase®), telithromycin (Ketek®), ritonavir (Norvir®), amprenavir (also known as Agenerase, a prodrug version is fosamprenavir (Lexiva®, Telzir®), indinavir (Crixivan®), nelfmavir (Viracept®), delavirdine (Rescriptor®) or voriconazole (Vfend®)).
When employing the methods or compositions, other agents used in the modulation of tumour growth or metastasis in a clinical setting, such as antiemetics, can also be administered as desired.
All variants and examples of the second main aspect can be combined with the first and third main aspects unless expressly stated otherwise.
Third main aspect - Method of treating cancer or cancer metastasis using the substance or pharmaceutical composition and embodiments thereof This aspect relates to the use of the above-mentioned substance or pharmaceutical composition for the treatment of cancer or metastasis as listed above in connection with the description of the substance and the pharmaceutical composition.
In the above-mentioned methods for treating of cancer or metastasis a first dose or dosage of the substance or the pharmaceutical composition mentioned above would be administered to a patient in need thereof. The patient may be an animal or a human. The administration ways or routes may vary according to the specific cancer or medical situation and are part of the knowledge of a medical practitioner.
A first dose or dosage is administrated to the subject and the first dose or dosage is preferably administrated 1-3 times/7 days, preferably 1 or 2 times/7 days. The first dose or dosage is preferably 0.005 to 15mg/kg weight of the subject, preferably 0.05 to 5mg/kg weight of the subject, more preferably 0.5 to 3 mg/kg weight of the subject.
The total amount of the substance administrated to a subject per 7 days is preferably 5-20 mg/kg body weight of the subject, preferably 10-15mg/kg.
The administration may be done locally or systemically or in combination. Some are given below as illustrative examples of such administration ways or routes.
In one embodiment the composition is for parenteral administration preferably intravenous infusion and the concentration of the substance in the composition is 100- 2500pg/ml. The total amount of substance per seven days is preferably 5-20mg/kg body weight of the subject, preferably 10-15 mg/kg body weight.
In one preferred embodiment the cancer is skin cancer and preferably melanoma or squamous cell carcinoma. In one embodiment the composition is for topical application preferably formulated in a patch, bandage, dressing, adhesive plaster or as a spray or a cream, ointment or gel and where the concentration of the substance in the composition is 100-2500pg/ml. The substance or the composition is administrated to the site of treatment and preferably the administration is done 1-3 times per 7 days and preferably for 2 weeks or more. In a preferred embodiment the composition is formulated as a patch or a dressing such as a bandage or adhesive plaster impregnated with the substance or the composition according to the present invention and optionally one or more excipients or diluents.
All variants and examples of the third main aspect can be combined with the first and second main aspects unless expressly stated otherwise. EXPERIMENTAL PART EXAMPLE 1
SYNTHESIS OF CARBAZATE-FUNCTIONALIZED POLYVINYL ALCOHOL (PVAC) Polyvinyl alcohol (5 g, 13-23 kDa) was dissolved in dimethyl sulfoxide (100 mL) while stirring for 1 hour at 80°C under argon gas. Carbonyl diimidazole (10 g) was added and stirring continued at room temperature overnight. Hydrazine hydrate (10 mL) was then added, the reaction stirred overnight, and the product collected and purified by repeated precipitation in ethanol. The degree of substitution was determined spectrophotometrically by performing a trinitrobenzene sulfonic acid assay described elsewhere (Stephen L. Snyder and Philip Z. Sobocinski; Analytical Biochemistry 64, 284- 288, 1975).
EXAMPLE 2 - INHIBITION OF SSAO A study performed by the use of Fluoro:SSAO kit (Cell Technology Inc. USA) was conducted according to instructions. Two different doses of PVAC were applied; 20 ug/ml and 80 ug/ml. The inhibitory effect, as assessed by the measurement of hydrogen peroxide-conjugated chromophore, was dose dependent showing an approximately 80 % reduction of enzyme activity with 20 ug/ml and 90 % reduction with 80 ug/mL PVAC. Thus, the PVAC effect was highly efficient and doses below 20 ug/ml serum are predicted to be suitable in an in vivo situation (corresponding to a dose of 60 mg in adult patient). The results are shown in Figure 1.
EXAMPLE 3 - ANTI CANCER EFFECT A study was conducted to determine the cancerostatic effect of PVAC on B16.F10 and MDA-MB-231 tumours grown in mice. The study was conducted by Adlego Biomedical AB (Report AB-17-56-01).
This study was performed in two parts. The first part encompassed 36 C57BL/6J mice injected with B16.F10 cells. The second part encompassed 36 athymic nude mice injected with MDA-MB-231 tumour cells. Figure 2, Figure 3a and b disclose the results from the study. Figure 2 discloses mean volumes of B16.F10 and MDA-MB-231 tumours treated with intra/peri tumoural injections of NaCl or PVAC during the study period indicates a statistical difference (p<0.05) between the NaCl group and both the 50 and the 1000 pg PVAC groups.
Following injection of B16.F10 cells into the C57BL/6J mice, the animals were monitored twice weekly with regard to weight, general health and tumour size. Once the tumour volumes reached 50m1 the animals were stratified into three experimental groups based on tumour size and were subjected to intra/peri tumoural injections three times per week for the duration of the study period. One experimental group received NaCl injections, while the remaining two groups received either 50 or 1000 pg of PVAC per injection. Tumour size and health status were recorded three times weekly during the treatment period. The animals were euthanized when the average tumour size of the control group reached 2ml in volume. The tumours were then excised and divided into two parts. One part was frozen on dry ice and the second part was placed in fixative for histological processing and analysis.
Following injection of MDA-MB-231 cells into the athymic nude mice the animals were monitored twice weekly with regard to weight, general health and tumour size. Once the average tumour volume reached IOOmI the animals were stratified into three experimental groups based on tumour size and were subjected to 28 days of twice-weekly intra/peri tumoural injections. One experimental group received NaCl injections, while the remaining two groups received either 50 or 1000 pg of PVAC per injection. Tumour size and health status were recorded twice weekly during the treatment period. The animals were euthanized on Day 29 of the treatment period after which the tumours were excised and divided into two parts. One part was frozen on dry ice and the second part was placed in fixative for histologic processing and analysis.
Body weights of both C57BL/6J and athymic nude mice increased at a normal rate throughout the study. There were no apparent effects on the treatments on body weight in either mouse strain during the study period.
Six B16.F10 inoculated mice died prior to the scheduled study endpoint as a result of the invasive tumour growth. There were no observed health effects observed as a result of the PVAC injections in B16/F10 tumour-inoculated mice. There were no spontaneous deaths in the MDA-MB-231 inoculated mice. Five of these mice were euthanized prior to the planned study endpoint due to abscess formation on the tumours. There were no observed health effects observed as a result of the PVAC injections in MDA-MB-231 tumour inoculated mice.
B16.F10 tumours grew rapidly and within two weeks of inoculation the tumours had reached the maximum allowable size in the control group. Intra/peri tumoural injections of PVAC at both 50pg and lOOOpg per injection doses led to a reduced mean tumour volume compared to NaCL injected mice at the last two time points measured. Both injections of 50 and 1000 pg also affected the tumour weight of excised tumours in a similar manner.
Even though a volume and size reduction of the tumours were seen in MDA-MB-231 the reduction not statistically significant compared to NaCl treated. No statistically significant volume reduction of the tumours was seen upon PVAC treatment in athymic mice with implanted MDA-MB-231 cells. The fact that these athymic mice are immunodeficient may indicate that PVAC is immunomodulating and requires intact immune response to induce anti -tumor effect, as seen in the B 16 melanoma implant study.
EXAMPLE 4 TOPICAL APPLICATION OF P AC
A study of treating an early suspected skin cancer on the forehead of a subject was done by applying a composition according to the present invention comprising PVAC in water (2mg/ml).
The site of treatment had scaly red patches and a wart like appearance and was diagnosed by a physician as an early stage squamous cell carcinoma. The composition was applied to the site using a cotton ball to which the composition had been added. The application was repeated every second day for two weeks.
After two weeks there was no sign of the patches or the wart like appearance.
While the invention has been described and pointed out with reference to operative embodiments thereof, it will be understood by those skilled in the art that various changes, modifications, substitutions and omissions can be made without departing from the spirit of the invention.

Claims

C L A I M S
1. A substance comprising a carrier exhibiting at least one scavenger structure, said scavenger structure comprising a nucleophilic centre complying with the formula
X1(-R”-)(-R,)mHn (formula I) wherein a) X1 is a single-bonded heteroatom selected amongst N, O and S and exhibits a free electron pair, b) m is 0 or 1 and n is 1 or 2 with the sum of m plus n being 2 for X1 = N and 1 for
X1 = S and O, c) -R”- is a bivalent organic group providing attachment to the carrier via one of its free valences and direct attachment to the heteroatom X1 at the other one of its free valences, and d) R’ - is a monovalent organic group directly attached to the heteroatom X1 via its free valence; for use as a medicament for treating cancer and/or cancer metastasis.
2. The substance for use according to claim 1 wherein the nucleophilic center is or is part of a group selected amongst a) amino groups, preferably primary or secondary amino groups, b) hydrazide groups, a semicarbazide group, a carbazate group, a thiosemicarbazide group, a thiocarbazate group, c) aminooxy groups, and d) thiol groups.
3. The substance for use according to claim 1 or 2 wherein the carrier is a polymer and wherein the number of scavengers per monomeric unit of the carrier is 15-50%, preferably 20-40%.
4. The substance for use according to any one of claims 1 to 3 wherein the carrier is a polymer having a molecular weight of 2.000 dalton or higher, preferably 10.000 dalton or higher, more preferably 10.000 to 30.000 dalton.
5. The substance for use according to any of the preceding claims, wherein that the carrier is a water-soluble polymer, preferably polyvinyl alcohol, polysaccharides preferably glucose amino glucan, hyaluronic acid or dextran.
6. The substance for use according to claim any of the preceding claims, wherein that the substance is carbazate-functionalized polyvinyl alcohol or semicarbazide functionalized polyvinyl alcohol.
7. The substance for use according to any one of claim 1 to 7 wherein the cancer is selected from head cancer, neck cancer, stomach cancer, ovarian cancer, breast cancer, pancreatic cancer, testicular cancer, skin cancer, bladder cancer, lung cancer, sarcoma, skin cancer, colorectal cancer, renal cancer, urothelial cancer or small cell lung cancer cell, preferably skin cancer preferably melanoma or squamous cell carcinoma.
8. A pharmaceutical composition comprising the substance defined in any of claims 1-7 and pharmaceutically acceptable adjuvants for use as a medicament for treating cancer and/or cancer metastasis.
9. The pharmaceutical composition for use according to claim 8 wherein the cancer is selected from head cancer, neck cancer, stomach cancer, ovarian cancer, breast cancer, pancreatic cancer, testicular cancer, skin cancer, bladder cancer, lung cancer, sarcoma, skin cancer, colorectal cancer, renal cancer, urothelial cancer or small cell lung cancer cell, preferably skin cancer preferably melanoma or squamous cell carcinoma.
10. The composition for use according to claim 8 or 9 wherein the composition is suitable for parenteral administration preferably intravenous injection, intramuscular or subcutaneous injection.
11. The composition for use according to claim 8 or 9 wherein the composition is suitable for oral administration.
12. The composition for use according to claim 8 or 9 wherein the composition is suitable for administration by inhalation including the use of a nebulizer.
13. The composition for use according to any one of claim 8 to 12, wherein the concentration of the substance is 20-3000 pg/ml, preferably 100-2500pg/ml, more preferably 500-2000 pg/ml.
14. The composition for use according to any one of claim 8 to 12, wherein the concentration of the substance is 20-500pg/mg, preferably 30-150pg/mg, more preferably 40-120pg/mg
15. A method of treating cancer and/or cancer metastasis in a subject in need thereof comprising administering a first dose or dosage of the substance according to anyone of claims 1-7 or administering a first dose or dosage of the pharmaceutical composition according to anyone of claims 8-14 to the subject.
16. The method according to claim 15 wherein the cancer is selected from head cancer, neck cancer, stomach cancer, ovarian cancer, breast cancer, pancreatic cancer, testicular cancer, skin cancer, bladder cancer, lung cancer, sarcoma, skin cancer, colorectal cancer, renal cancer, urothelial cancer or small cell lung cancer cell, preferably skin cancer preferably melanoma or squamous cell carcinoma.
17. The method according to any one of claim 15 to 16, wherein the first dose or dosage is 0.005 to 15mg/kg weight of the subject, preferably 0.05 to 5mg/kg weight of the subject, more preferably 0.5 to 3 mg/kg weight of the subject.
18. The method according to any one of claim 15 to 17 wherein the administration is done by parenteral administration preferably intravenous injection, topical application, intramuscular or subcutaneous injection.
19. The method according to any one of claim 15 to 17 wherein the administration is done by oral administration.
20. The method according to any one of claim 15 to 17 wherein the administration is done by inhalation including the use of a nebulizer.
21. The method according to claim 15 wherein the cancer is skin cancer preferably squamous cell carcinoma and wherein wherein the administration is done by topical application.
22. The method according to claim 21 wherein the composition is formulated in a patch, bandage, dressing, adhesive plaster or as a spray, cream, ointment or gel and where the concentration of the substance in the composition is 100-2500pg/ml.
23. The method according to any one of claim 15 to 21 wherein the first dose or dosage is administrated 1-3 times/7 days, preferably 1 or 2 times/7 days.
24. The method according to any one of claim 15 to 23 wherein the total amount of the substance is 5-20 mg/kg body weight of the subject and 7 days, preferably 10-
15 mg/kg.
PCT/EP2020/064039 2020-03-16 2020-05-20 Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof WO2021233534A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/EP2020/064039 WO2021233534A1 (en) 2020-05-20 2020-05-20 Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof
PCT/EP2021/056698 WO2021185844A1 (en) 2020-03-16 2021-03-16 Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2020/064039 WO2021233534A1 (en) 2020-05-20 2020-05-20 Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof

Publications (1)

Publication Number Publication Date
WO2021233534A1 true WO2021233534A1 (en) 2021-11-25

Family

ID=70775405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/064039 WO2021233534A1 (en) 2020-03-16 2020-05-20 Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof

Country Status (1)

Country Link
WO (1) WO2021233534A1 (en)

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0520722A1 (en) 1991-06-28 1992-12-30 Zeneca Limited Therapeutic preparations containing quinazoline derivatives
EP0564409A1 (en) 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
EP0566226A1 (en) 1992-01-20 1993-10-20 Zeneca Limited Quinazoline derivatives
US5260291A (en) 1981-08-24 1993-11-09 Cancer Research Campaign Technology Limited Tetrazine derivatives
WO1996033980A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivatives
WO1997002266A1 (en) 1995-07-06 1997-01-23 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
EP0787722A1 (en) 1996-02-05 1997-08-06 American Cyanamid Company Substituted quinazoline derivatives
WO1997030034A1 (en) 1996-02-14 1997-08-21 Zeneca Limited Quinazoline derivatives as antitumor agents
WO1997038983A1 (en) 1996-04-12 1997-10-23 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
WO1997049688A1 (en) 1996-06-24 1997-12-31 Pfizer Inc. Phenylamino-substituted tricyclic derivatives for treatment of hyperproliferative diseases
WO1998010767A2 (en) 1996-09-13 1998-03-19 Sugen, Inc. Use of quinazoline derivatives for the manufacture of a medicament in the treatment of hyperproliferative skin disorders
EP0837063A1 (en) 1996-10-17 1998-04-22 Pfizer Inc. 4-Aminoquinazoline derivatives
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
WO1999003854A1 (en) 1997-07-18 1999-01-28 Novartis Ag Crystal modification of a n-phenyl-2-pyrimidineamine derivative, processes for its manufacture and its use
WO2002022577A2 (en) 2000-09-01 2002-03-21 Novartis Ag Hydroxamate derivatives useful as deacetylase inhibitors
WO2003013541A1 (en) 2001-08-07 2003-02-20 Novartis Ag 4-amino-6-phenyl-pyrrolo[2,3-d]pyrimidine derivatives
US20080139587A1 (en) 2004-11-30 2008-06-12 Novartis Ag Combinations Comprising Epothilones and Protein Tyrosine Kinase Inhibitors and Pharmaceutical Uses Thereof
WO2009108100A1 (en) 2008-02-29 2009-09-03 Ipr-Systems Sweden Ab Composition for the formation of gels
EP2670439A1 (en) * 2011-01-31 2013-12-11 Bowden, Tim Active principle for mitigating undesired medical conditions
WO2018172422A1 (en) * 2017-03-21 2018-09-27 Pvac Medical Technologies Ltd Method of preserving erythrocytes using pvac
WO2019016189A1 (en) * 2017-07-18 2019-01-24 Pvac Medical Technologies Ltd Anti-bacterial substance and composition thereof, medical and non-medical uses using the same, and products comprising said substance and compositions

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5260291A (en) 1981-08-24 1993-11-09 Cancer Research Campaign Technology Limited Tetrazine derivatives
EP0520722A1 (en) 1991-06-28 1992-12-30 Zeneca Limited Therapeutic preparations containing quinazoline derivatives
EP0566226A1 (en) 1992-01-20 1993-10-20 Zeneca Limited Quinazoline derivatives
EP0564409A1 (en) 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
WO1996033980A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivatives
WO1997002266A1 (en) 1995-07-06 1997-01-23 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
EP0787722A1 (en) 1996-02-05 1997-08-06 American Cyanamid Company Substituted quinazoline derivatives
WO1997030034A1 (en) 1996-02-14 1997-08-21 Zeneca Limited Quinazoline derivatives as antitumor agents
WO1997038983A1 (en) 1996-04-12 1997-10-23 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
WO1997049688A1 (en) 1996-06-24 1997-12-31 Pfizer Inc. Phenylamino-substituted tricyclic derivatives for treatment of hyperproliferative diseases
WO1998010767A2 (en) 1996-09-13 1998-03-19 Sugen, Inc. Use of quinazoline derivatives for the manufacture of a medicament in the treatment of hyperproliferative skin disorders
EP0837063A1 (en) 1996-10-17 1998-04-22 Pfizer Inc. 4-Aminoquinazoline derivatives
WO1999003854A1 (en) 1997-07-18 1999-01-28 Novartis Ag Crystal modification of a n-phenyl-2-pyrimidineamine derivative, processes for its manufacture and its use
WO2002022577A2 (en) 2000-09-01 2002-03-21 Novartis Ag Hydroxamate derivatives useful as deacetylase inhibitors
WO2003013541A1 (en) 2001-08-07 2003-02-20 Novartis Ag 4-amino-6-phenyl-pyrrolo[2,3-d]pyrimidine derivatives
US20080139587A1 (en) 2004-11-30 2008-06-12 Novartis Ag Combinations Comprising Epothilones and Protein Tyrosine Kinase Inhibitors and Pharmaceutical Uses Thereof
WO2009108100A1 (en) 2008-02-29 2009-09-03 Ipr-Systems Sweden Ab Composition for the formation of gels
EP2670439A1 (en) * 2011-01-31 2013-12-11 Bowden, Tim Active principle for mitigating undesired medical conditions
WO2018172422A1 (en) * 2017-03-21 2018-09-27 Pvac Medical Technologies Ltd Method of preserving erythrocytes using pvac
WO2019016189A1 (en) * 2017-07-18 2019-01-24 Pvac Medical Technologies Ltd Anti-bacterial substance and composition thereof, medical and non-medical uses using the same, and products comprising said substance and compositions

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BULINSKI, J. CELL SCI., vol. 1, no. 10, 1997, pages 3055 - 3064
FELIX SELLBERG ET AL: "Polyvinylalcohol-carbazate (PVAC) reduces red blood cell hemolysis", PLOS ONE, vol. 14, no. 12, 6 December 2019 (2019-12-06), pages e0225777, XP055771755, DOI: 10.1371/journal.pone.0225777 *
FREDRIKSSON F ET AL: "Sutures impregnated with carbazate-activated polyvinyl alcohol reduce intraperitoneal adhesions", JOURNAL OF PEDIATRIC SURGERY, vol. 52, no. 11, 1 February 2017 (2017-02-01), pages 1853 - 1858, XP085277473, ISSN: 0022-3468, DOI: 10.1016/J.JPEDSURG.2017.01.058 *
LIZZIE RAFFAGHELLO ET AL: "In vitro and In vivo Antitumor Activity of the Novel Derivatized Polyvinyl Alcohol-Based Polymer P10(4)", CLINICAL CANCER RESEARCH, vol. 12, no. 11, 1 June 2006 (2006-06-01), US, pages 3485 - 3493, XP055771762, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-05-2318 *
MUHLRADT, CANCER RES., vol. 57, 1997, pages 3344 - 3346
NICOLAOU, NATURE, vol. 387, 1997, pages 268 - 272
PANDA, J. BIOL. CHEM., vol. 271, 1996, pages 29807 - 29812
PANDA, PROC. NATL. ACAD. SCI. USA, vol. 94, 1997, pages 10560 - 10564
ROBIN FRÖBOM: "Translational investigations of novel and current antitumoral therapies in gastrointestinal stromal tumors", 13 December 2019 (2019-12-13), XP055771621, ISBN: 978-91-7-831593-2, Retrieved from the Internet <URL:https://openarchive.ki.se/xmlui/bitstream/handle/10616/46933/Thesis_Robin_Fröbom.pdf?sequence=3&isAllowed=y> [retrieved on 20210203] *
ROBIN FRÖBOM: "Translational investigations of novel and current antitumoral therapies in gastrointestinal stromal tumors", 13 December 2019 (2019-12-13), XP055771626, Retrieved from the Internet <URL:https://openarchive.ki.se/xmlui/handle/10616/46933> [retrieved on 20210203] *
STEPHEN L. SNYDERPHILIP Z. SOBOCINSKI, ANALYTICAL BIOCHEMISTRY, vol. 64, 1975, pages 284 - 288
VASQUEZ, MOL. BIOL. CELL., vol. 8, 1997, pages 973 - 985

Similar Documents

Publication Publication Date Title
US20110287110A1 (en) Combination cancer treatment
AU2016281620B2 (en) Compositions and methods for inhibiting arginase activity
JP6220126B2 (en) Polymers based on cyclodextrins for therapeutic delivery
US20110160159A1 (en) Treatment of cancer
JP2016528174A (en) Cyclodextrin-based polymers for therapeutic delivery
US20200093932A1 (en) Treatment of cancer
US20160166567A1 (en) Treatment of cancer
AU2012229503B2 (en) Treatment of cancer
US20200323889A1 (en) Cyclodextrin-based polymers for therapeutic delivery
US20190275038A1 (en) Combination therapy with glutaminase inhibitors
US20220409612A1 (en) Treatment of cancer
US20110152512A1 (en) Cyclodextrin-based polymers for therapeutics delivery
US20110245201A1 (en) Treatment of cancer
WO2021233534A1 (en) Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof
JP2015505557A (en) Polymers based on cyclodextrins for therapeutic delivery
WO2021185844A1 (en) Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof
US20160175351A1 (en) Cancer treatment
WO2021185842A1 (en) Use of substance and pharmaceutical composition thereof, and medical treatments or uses thereof
JP2023536346A (en) Treatment of Cancer with Cyclodextrin-Containing Polymeric Topoisomerase Inhibitor Conjugates and PARP Inhibitors
US20160082111A1 (en) Cyclodextrin-based polymers for therapeutic delivery
US20140205594A1 (en) Treatment of cancer

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: 20726828

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: 20726828

Country of ref document: EP

Kind code of ref document: A1