WO2023245248A1 - Treatment of melanoma - Google Patents

Treatment of melanoma Download PDF

Info

Publication number
WO2023245248A1
WO2023245248A1 PCT/AU2023/050566 AU2023050566W WO2023245248A1 WO 2023245248 A1 WO2023245248 A1 WO 2023245248A1 AU 2023050566 W AU2023050566 W AU 2023050566W WO 2023245248 A1 WO2023245248 A1 WO 2023245248A1
Authority
WO
WIPO (PCT)
Prior art keywords
bisantrene
immune checkpoint
checkpoint inhibitor
melanoma
agent
Prior art date
Application number
PCT/AU2023/050566
Other languages
French (fr)
Inventor
Daniel Tillett
Original Assignee
Race Oncology 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
Priority claimed from AU2022901702A external-priority patent/AU2022901702A0/en
Application filed by Race Oncology Ltd filed Critical Race Oncology Ltd
Publication of WO2023245248A1 publication Critical patent/WO2023245248A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41681,3-Diazoles having a nitrogen attached in position 2, e.g. clonidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • This invention is directed to the use of bisantrene in combination with immune checkpoint inhibitors to treat melanoma.
  • the present invention also contemplates pharmaceutical compositions and kits for such treatments, as well as the use of such compositions for the manufacture of medicaments for such treatments.
  • BACKGROUND OF THE INVENTION [0002] Melanoma is a dangerous form of skin cancer that begins in cells called melanocytes. While melanoma is less frequent than other skin cancers such as basal cell carcinoma (BCC) or squamous cell carcinoma (SCC), melanoma is typically more dangerous because of its ability to metastasize to other organs if not detected and treated at an early stage.
  • BCC basal cell carcinoma
  • SCC squamous cell carcinoma
  • Risk factors that are associated with a higher incidence of melanoma include: a fair complexion (including fair skin that burns or freckles easily, blue or green eyes, and blonde or red hair); exposure to sunlight and other sources of ultraviolet (UV) energy; a prior history of sunburns that caused blistering (especially in childhood); having large moles, many small moles, or moles that look different from normal moles; a family history of unusual moles or melanoma; a personal history of skin cancer; genetic syndromes such as xeroderma pigmentosum; other environmental exposures, including to radiation and some chemicals; a weakened immune system; older age; and male sex.
  • a fair complexion including fair skin that burns or freckles easily, blue or green eyes, and blonde or red hair
  • UV energy ultraviolet
  • the present invention provides a new paradigm for treating melanoma by the administration of bisantrene in combination with a targeted agent, for example an immune checkpoint inhibitor or immunomodulator. This meets the need for new treatments that improve the clinical outcomes of patients with melanoma, especially patients who show resistance to current targeted therapies.
  • a targeted agent for example an immune checkpoint inhibitor or immunomodulator.
  • Bisantrene is an antineoplastic agent that has multiple mechanisms of action, including DNA intercalation, inhibition of topoisomerase and the fat mass and obesity-associated protein (FTO), and activation of the immune system.
  • One aspect of the invention therefore provides a method of treating a patient with melanoma, said method comprising administering to said patient a therapeutically effective amount of a targeted agent, which may be an immune checkpoint inhibitor, and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
  • a targeted agent which may be an immune checkpoint inhibitor
  • the immune checkpoint inhibitor inhibits one or more of PD-1, PD-L1, CTLA4 and LAG3.
  • the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) ipilimumab (b) nivolumab (c) cemiplimab (c) nivolumab-relatlimab (d) pembrolizumab (e) dostarlimab [0014]
  • the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) nivolumab (b) nivolumab-relatlimab (c) pembrolizumab (d) cemiplimab [0015]
  • the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) nivolumab (b) pembrolizumab (c) cemiplimab
  • the method comprises administering to a patient a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof.
  • the method comprises treatment of immune checkpoint inhibitor resistant melanoma.
  • the method may further comprise administration of at least one additional therapeutic agent, optionally a protein kinase inhibitor for the treatment melanoma.
  • the at least one additional therapeutic agent may be selected from the group consisting of: trametinib; encorafenib; binimetinib; vemurafenib; talimogene laherparepvec; tebentafusp-tebn; interleukin-2; and proleukin.
  • the method may comprise administering said at least one immune checkpoint inhibitor to said patient prior to, simultaneously with, or after administration of said second agent to said patient.
  • the method may comprise administering said at least one immune checkpoint inhibitor and the second agent to said patient at the same time, optionally in a single composition.
  • the treatment has enhanced efficacy results against melanoma compared to a method wherein said at least one immune checkpoint inhibitor or said second agent is administered alone.
  • the dose of said at least one immune checkpoint inhibitor is at least 20% lower than the dose required of said at least one immune checkpoint inhibitor agent when administered without said second agent to achieve the same targeted outcome.
  • Another aspect of the invention provides a pharmaceutical composition for the treatment of melanoma by embodiments of treatment methods as described above, said compositions comprising at least one immune checkpoint inhibitor, which may be a PD-1 inhibitor, as described above, and a second agent as described above comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
  • compositions may comprise further active agents as described above.
  • Another aspect of the invention provides the use of a composition according to the invention, as described above, for the manufacture of a medicament for the treatment of advanced melanoma, optionally immune checkpoint inhibitor resistant melanoma in a patient.
  • Yet another aspect of the invention provides a kit for the treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma by embodiments of methods of the present invention, as described above, said kit comprising at least one immune checkpoint inhibitor, optionally a PD-1 inhibitor, as described above, and at least a second agent as described above, said second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
  • Kits according to the present invention may comprise instructions for administering said at least one immune checkpoint inhibitor to said patient prior to, simultaneously with, or after administering said second agent to said patient.
  • kits according to the present invention may comprise instructions for administering said at least one immune checkpoint inhibitor and the second agent to said patient at the same time.
  • Bisantrene was administered by daily IV injections.
  • RMP1-14 was administered by IP injection 3 times per week.
  • Data points represent the mean of each group at each time point +/- SEM.
  • Figure 4 shows the percentage of CTLA4 + T-cells as a proportion of cytotoxic T-cells (CD8 + ) isolated from excised subcutaneous tumors taken at Day 7 or Day 13 after randomization. Data points represent samples from individual tumors, bars represent mean +/- SEM.
  • Figure 5 shows the use of bisantrene plus a targeted agent, for example, an immune checkpoint inhibitor to overcome treatment resistance in melanoma.
  • “treating” means affecting a subject/patient, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes inhibiting a condition, i.e. slowing or arresting its development; or relieving or ameliorating the effects of the condition i.e., cause reversal or regression of the effects of the condition.
  • preventing means preventing a condition from occurring in a cell, tissue or subject that may be at risk of having the condition, but does not necessarily mean that condition will not eventually develop, or that a subject will not eventually develop a condition. Preventing includes delaying the onset of a condition in a cell, tissue or subject.
  • the term “subject” or “patient” refers to a mammal. The mammal may be a human or a non-human. Examples of non-humans include primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g.
  • composition encompasses compositions and formulations comprising the active pharmaceutical ingredient (for example, “immune checkpoint inhibitor” and “second agent comprising bisantrene or a derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or cardioprotective derivative of bisantrene”) with excipients or carriers, and also compositions and formulations with a carrier.
  • active pharmaceutical ingredient for example, “immune checkpoint inhibitor” and “second agent comprising bisantrene or a derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or cardioprotective derivative of bisantrene”
  • the excipient or carrier is “pharmaceutically acceptable” meaning that it is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • Supplementary active ingredients can also be incorporated into the compositions.
  • a pharmaceutically acceptable such as in the recitation of a “pharmaceutically acceptable salt” or a “pharmaceutically acceptable excipient or carrier” is meant herein a material that is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • the term “effective amount” or “therapeutically effective amount” refers to the quantity of an active pharmaceutical ingredient that is sufficient to yield a desired therapeutic response.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, bulking agents, carrier solutions, suspensions, colloids, and forming and binding agents, any or all of which may include other pharmaceutical excipients as known in the art, including lubricants, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, antioxidants, other stabilisers, including physical stabilisers such as thickeners and viscosity enhancers, colouring agents, flavouring and sweetening agents, and the like.
  • administering refers to dispensing, applying, or tendering one or more agents to a subject. Administration can be performed using any of a number of methods known in the art. For example, “administering” as used herein is meant via infusion (intravenous administration (i.v.)), parenteral administration. By “parenteral” is meant intravenous, subcutaneous and intramuscular administration.
  • the present disclosure relates to a method for the treatment of melanoma, especially immune checkpoint inhibitor resistant melanoma, comprising administering a therapeutically active amount of bisantrene in combination with an immune checkpoint inhibitor, especially a PD-1 inhibitor.
  • Immune checkpoint inhibitors [0041] Programmed cell death 1 ligand 1 (PD-L1) and its receptor programmed cell death 1 (PD-1) regulate the activation of immune cells. Checkpoint inhibitors can assist overcoming this immune regulation, allowing immune cells to destroy tumors.
  • checkpoint inhibitors include antibodies that target PD-1, PD-L1, CTLA4 or LAG3.
  • the FDA has approved two PD-1 inhibitors (nivolumab and pembrolizumab), one CTLA-4 inhibitor (ipilimumab) and one PD-1/LAG3 inhibitor combination (nivolumab and relatlimab) for use in melanoma.
  • PD-1/ PD-L1 immune checkpoint inhibitors of potential consideration include avelumab, atezolizumab, bevacizumab, cemiplimab, dostarlimab, and durvalumab.
  • immunomodulators such as cytokines that regulate immune cell maturation, growth and activation, including interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and Granulocyte-macrophage colony-stimulating factor (GM-CSF) have also been approved for treatment of various cancers via immunostimulation.
  • Interleukin-2 (or the recombinant human version Aldesleukin/ Proleukin) has been approved for the treatment of melanoma.
  • Bisantrene and derivatives or analogs thereof, and pharmaceutically acceptable salts [0044] Bisantrene has immunologic properties that might be responsible for some of its activities, and which may make this agent a useful tool in combination with immune checkpoint inhibitors. Intravenously (i.v.), intraperitoneally (i.p.), or subcutaneously (s.c.), bisantrene was effective in cancer models using colon 26, Lewis lung, Ridgway osteosarcoma, B16, Lieberman plasma cell, P388 or L1210 cancer cells.
  • Anti-cancer activity was observed in clonogenic assays using tumor samples isolated from 684 patients with breast, small cell lung, large cell lung, squamous cell lung, ovarian, pancreatic, renal, adrenal, head and neck, sarcoma, gastric, lymphoma and melanoma cancers, but not in colorectal cancer. Importantly, a lack of cross resistance with adriamycin and mitoxantrone was found. Subsequent to treatment with bisantrene, and for 4 weeks thereafter, macrophages could be isolated from peritoneal exudate that had cytostatic anti-proliferative activity in cultures of P815 (mastocytoma) tumor cells.
  • Bisantrene has multiple anti-cancer mechanisms of activity including direct cytotoxic action, immunologic, and as a potent inhibitor (IC50142 nM) of the fat mass and obesity-associated protein (FTO), an RNA N6-methyladenosine (m6A) demethylase (Su, R., Dong, L., Li, Y., Gao, M., Han, L., Wunderlich, M., et al. (2020), “Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion”, Cancer Cell, 38(1): 79–96.e11).
  • FTO fat mass and obesity-associated protein
  • m6A RNA N6-methyladenosine
  • bisantrene The chemical name for bisantrene is 9, 10-anthracenedicarboxaldehyde-bis [(4, 5-dihydro-1H-imidazole-2-yl) hydrazine] dihydrochloride, and it was originally classed as an anthracycline chemotherapeutic agent.
  • the term “bisantrene” refers to bisantrene or any pharmacologically compatible salt form, not only bisantrene dihydrochloride, unless the dihydrochloride or another specific pharmacologically compatible salt form is specifically indicated.
  • the pharmacologically compatible salt of bisantrene is bisantrene dihydrochloride for most pharmacological applications.
  • Stabilization of the G-quadruplex can interfere with telomere- telomerase interaction and thus inhibit the activity of telomerase in various ways, including the displacement of telomerase binding proteins. Since the level of topoisomerase II inhibition does not always correlate with cytotoxic efficacy, alternative mechanisms may play a role in the actions of bisantrene. Analogs of bisantrene have been made in an attempt to improve upon the anti-telomerase activity; these analogs are described further below. Human melanoma (SK-Mel5) and colon cancer (LoVo) tumor cells were observed to lose their proliferative ability in the presence of these agents.
  • bisantrene in addition to direct antineoplastic effects related to the activity of bisantrene as a DNA intercalator, bisantrene also possesses other mechanisms of action, including immunopotentiation. These mechanisms are described in: (i) N.R. West et al. (2011), “Tumor-Infiltrating Lymphocytes Predict Response to Anthracycline-Based Chemotherapy in Estrogen-Resistant Breast Cancer”, Breast Canc.
  • TIL tumor-infiltrating lymphocytes
  • the markers associated with tumor-infiltrating lymphocytes include CD19, CD3D, CD48, GZMB, LCK, MS4A1, PRF1, and SELL;
  • Immunol.8 59-73, which states that DNA damage, such as that produced by intercalating agents such as bisantrene, induces the expression of NKG2D ligands on tumor cells in an ATM-dependent and CHK1- dependent (but p53-independent) manner; NKG2D is an activating receptor that is involved in tumor immunosurveillance by NK cells, NK T cells, ⁇ T cells and resting (in mice) and/or activated (in humans) CD8+ T cells, and also states that anthracycline-based agents may act as immunostimulators, particularly in combination with IL-12; such agents also promote HMGB1 release and activate T cells; (iii) D.V. Krysko et al.
  • TLR2 and TLR9 are Sensors of Apoptosis in a Mouse Model of Doxorubicin-Induced Acute Inflammation”, Cell Death Different.18: 1316-1325, which states that anthracycline-based antibiotics induce an immunogenic form of apoptosis that has immunostimulatory properties mediated by MyD88, TLR2, and TLR9; (iv) C. Ferraro et al.
  • HIF-1 also activates transcription of genes encoding glucose transporter GLUT1 and hexokinases HK1 and HK2, which are required for the high level of glucose uptake and phosphorylation that is observed in metastatic cancer cells, and pyruvate dehydrogenase kinase 1 (PDK1), which shunts pyruvate away from the mitochondria, thereby increasing lactate production; patients with HiF-1a overexpression based on immunohistochemical resuits were suggested to be good candidates for treatment with anthracyciine-based antibiotics.
  • PDK1 dehydrogenase kinase 1
  • bisantrene dihydrochloride is a tricyclic aromatic compound with the chemical name, 9,10-anthracenedicarboxaldehyde bis[(4,5-dihydro-1H-imidazol-2- yl)hydrazine] dihydrochloride.
  • the molecular formula of bisantrene dihydrochloride is C22H22N8 ⁇ 2HCl and the molecular weight, 471.4 g/mol.
  • the alkylimidazole side chains are very basic and, at physiologic pH, are positively charged. This is believed to facilitate electrostatic attractions to negatively charged ribose phosphate groups in DNA.
  • Bisantrene is typically administered intravenously, either centrally or peripherally.
  • Various formulations suitable for use in the administration of bisantrene or derivatives or analogs thereof are known in the art. United States Patent No. 4,784,845 to Desai et al.
  • composition of matter for delivery of a hydrophobic drug comprising: (i) the hydrophobic drug; (ii) an oleaginous vehicle or oil phase that is substantially free of butylated hydroxyanisole (BHA) or butylated hydroxytoluene (BHT); (iii) a co- surfactant or emulsifier; (iv) a co-surfactant or auxiliary emulsifier; and (v) benzyl alcohol as a co-solvent.
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluene
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluene
  • co-surfactant or emulsifier iv
  • benzyl alcohol as a co-solvent.
  • compositions of matter for delivery by intravenous, intramuscular, or intraarticular routes of hydrophobic drugs comprising: (i) the hydrophobic drug; (ii) a pharmaceutically acceptable oleaginous vehicle or oil selected from the group consisting of: (a) naturally occurring vegetable oils and (b) semisynthetic mono-, di-, and triglycerides, wherein the oleaginous vehicle or oil is free of BHT or BHA; (iii) a surfactant or emulsifier; (iv) a co-surfactant or emulsifier; (v) an ion-pair former selected from C6-C20 saturated or unsaturated aliphatic acids when the hydrophobic drug is basic and a pharmaceutically acceptable aromatic amine when the hydrophobic drug is acidic; and (vi) water.
  • hydrophobic drugs such as bisantrene or a derivative or analog thereof
  • United States Patent No.5,000,886 to Lawter et al. and United States Patent No. 5,143,661 to Lawter et al. disclose compositions for delivery of pharmaceutical agents such as bisantrene or a derivative or analog thereof comprising a microcapsule, wherein the microcapsule includes a hardening agent that is a volatile silicone fluid.
  • United States Patent No.5,070,082 to Murdock et al., United States Patent No.5,077,282 to Murdock et al., and United States Patent No.5,077,283 to Murdock et al. disclose prodrug forms of poorly soluble hydrophobic drugs, including bisantrene and derivatives and analogs, that are salts of a phosphoramidic acid.
  • United States Patent No.5,116,827 to Murdock et al. and United States Patent No. 5,212,291 to Murdock et al. disclose prodrug forms of poorly soluble hydrophobic drugs, including bisantrene and derivatives and analogs, that are quinolinecarboxylic acid derivatives.
  • United States Patent No.5,378,456 to Tsou includes compositions containing an anthracene antitumor agent, such as bisantrene or a derivative or analog thereof, in which the bisantrene or derivative or analog thereof is conjugated to or admixed with a divinyl ether-maleic acid (MVE) copolymer.
  • MVE divinyl ether-maleic acid
  • United States Patent No.5,609,867 to Tsou discloses polymeric 1,4-bis derivatives of bisantrene and copolymers of bisantrene and another monomer, such as a dianhydride.
  • Methods and compositions described herein can use a derivative or analog of bisantrene in place of bisantrene itself.
  • Derivatives and analogs of bisantrene are described in in US Patents 10,500,19 and 10,548,876, by Garner et al. Methods for treating melanoma
  • the present studies have shown that immune checkpoint inhibitors and bisantrene (or pharmaceutically acceptable salts thereof) show beneficial antitumor effects in a mouse model of melanoma.
  • bisantrene was able to increase the number of antitumorigenic M1 macrophages when combined with a murine PD-1 inhibitor relative to mice treated with the PD-1 inhibitor alone.
  • bisantrene/Anti-PD-1 combination produced a significant decrease in protumorigenic M2b macrophages in tumours relative to PD-1 inhibition alone, indicating that bisantrene is able to elicit an antitumor immunophenotype through selective effects on myeloid cell populations.
  • Bisantrene/anti-PD-1 combinations also potently downregulated the expression of CTLA4, a validated immunosuppressive checkpoint target, in CD8 + T-cells, priming these key mediators of antitumor immunity for effective destruction of tumor cells.
  • the FDA has approved three PD-1 inhibitors (pembrolizumab [Keytruda], nivolumab [Opdivo], ostarlimab [Jemperli], and cemiplimab [Libtayo]), one PD-L1 inhibitor/LAG3 inhibitor combination (nivolumab-relatlimab), and one CTLA-4 inhibitor (ipilimumab) for use.
  • PD-1/ PD-L1 immune checkpoint inhibitors of potential consideration include atezolizumab, bevacizumab, dostarlimab, and durvalumab.
  • one aspect of the invention provides a method of treating a patient with melanoma, especially immune checkpoint inhibitor resistant melanoma, said method comprising administering to said patient a therapeutically effective amount of an immune checkpoint inhibitor, such as a PD-1 inhibitor, and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
  • an immune checkpoint inhibitor such as a PD-1 inhibitor
  • the immune checkpoint inhibitor may inhibit one or more potential immune checkpoint targets associated with melanoma.
  • the immune checkpoint inhibitor or inhibitor combination inhibits PD-1, PD-L1, CTLA- 4, LAG3 or a combination of at least any two of these targets.
  • the immune checkpoint inhibitor inhibits at least one or more of PD-1, PD-L1, LAG3. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1 or PD-L1. According to certain embodiments, the immune checkpoint inhibitor inhibits PD-1. [0060] According to certain embodiments the at least one immune checkpoint inhibitor or inhibitor combination is selected from the group consisting of: ipilimumab; nivolumab; cemiplimab; nivolumab-relatlimab; and pembrolizumab.
  • the at least one immune checkpoint inhibitor is selected from the group consisting of: nivolumab; cemiplimab; nivolumab- relatlimab; and pembrolizumab.
  • the at least one immune checkpoint inhibitor is selected from the group consisting of nivolumab, cemiplimab or pembrolizumab.
  • the second agent is bisantrene or a pharmaceutically acceptable salt thereof.
  • the bisantrene can be administered as a drug compound or as a component of a pharmaceutical composition, as discussed further below.
  • the method comprises administering to said patient a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical compositions and medicaments of the present invention may be administered to a subject by standard enteral or parenteral routes, including, but not limited to, injection (such as intravenous, subcutaneous, intramuscular, bolus, etc.), or by, for example, topical, oral, sublingual, nasal, pulmonary, otic, rectal or vaginal administration routes.
  • pharmaceutical compositions according to the invention may be administered to a subject by themselves or in combination with other pharmaceutical composition(s).
  • the administration may be simultaneous or sequential, or administration of the pharmaceutical composition(s) may be independent of one another.
  • bisantrene or derivative, or pharmaceutically acceptable salt of either
  • the pharmaceutical compositions and medicaments of the present invention can be administered in a manner compatible with the route of administration and physical characteristics of the subject (including health status) and in such a way that the desired effect(s) are induced (i.e. therapeutically effective and/or preventative).
  • the appropriate dosage may depend on a variety of factors including, but not limited to, a subject’s physical characteristics (e.g.
  • Suitable dosages, dosage frequencies, dosage durations, and routes of administration for chemotherapeutic agents are known in the art.
  • bisantrene, derivatives of bisantrene, or pharmaceutically acceptable salts of either can be administered in the same pharmaceutical composition as an immune checkpoint inhibitor, in a separate composition as, but simultaneously with the immune checkpoint inhibitor, or at a different time.
  • the bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of bisantrene or derivative of bisantrene is administered at a different time than the immune checkpoint inhibitor, it can either be administered before or after the immune checkpoint inhibitor and/or at different timings, being administered according to different timing and/or frequency regimes.
  • One of ordinary skill in the art can determine a suitable schedule for administration based on variables such as the age, weight, and sex of the subject, the susceptibility of the subject to side effects of the active agents, genetic markers, the dose of active agent(s), the subjects history with prior active agent(s), and other pharmacokinetic parameters such as heart, liver or kidney function.
  • Figure 5 shows a prophetic example of a bisantrene/immune checkpoint inhibitor trial to overcome treatment resistance in a model of melanoma.
  • the methods and compositions provided herein enable a subject to receive a therapy more frequently without having the dosage regimen significantly altered by the risk of side-effects, such as cardiotoxicity.
  • the dose(s) of a immune checkpoint inhibitor and bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of bisantrene or derivative of bisantrene may be administered to a subject in one or more doses per day.
  • the daily dose of the chemotherapeutic agent may be administered together with bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of either, in a single dose.
  • the pharmaceutical compositions described herein may be administered to a patient one or more times per day. In some cases, the pharmaceutical composition may be administered to a patient once per day. In some cases, the pharmaceutical composition may be administered to a patient at least 2 times, 3 times, 4 times 5 times, or 6 times per day. For example, a pharmaceutical composition may be administered to a patient 3 times per day. [0073] In methods described herein, suitable dosages of bisantrene (or a derivative of bisantrene, or a pharmaceutically acceptable salt of either) can be determined by one of ordinary skill in the art.
  • the selected dosage level depends upon a variety of pharmacokinetic factors including the amount of active agent(s) being administered, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the severity of the condition, other health considerations affecting the subject, and the status of liver and kidney function of the subject. It also depends on the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular therapeutic agent employed, as well as the age, weight, condition, general health and prior medical history of the subject being treated, and like factors.
  • administration of bisantrene is performed at a dosage of from about 0.1 mg/m 2 /day to about 400 mg/m 2 /day, such as from about 0.2 mg/m 2 /day to about 300 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 200 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 100 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 50 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 30 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 20 mg/m 2 /day, from about 1.0 mg/m 2 /day to about 10 mg/m 2 /day, from about 1.0 mg/m 2 /day to about 8 mg/m 2 /day, about 1 mg/m 2 /day, about 2 mg/m 2 /day, about 3 mg/m 2 /day, about 4
  • bisantrene is administered daily or weekly, once every two weeks, once every three weeks, once every four weeks, over a period of, for example, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 21 days, 28 days, 35 days, 42 days, 49 days, 56 days or 63 days.
  • bisantrene is administered once or multiple times over a period of 28 days, optionally once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, at a dosage of about 20-50 mg/m 2 /28 days.
  • compositions of bisantrene or derivatives or pharmaceutically acceptable salts thereof may be performed at similar dosage rates, adjusted for molar equivalence.
  • Dosing regimens for immune checkpoint inhibitors will depend on the particular immune checkpoint inhibitor, and have been published for known immune checkpoint inhibitors, including for treatment of melanoma.
  • suitable dosages of a given immune checkpoint inhibitor can be readily determined by one of ordinary skill in the art using known approaches and techniques as discussed above in relation to bisantrene.
  • a therapeutic outcome may be achieved more effectively for a given dose rate for that immune checkpoint inhibitor.
  • This biologic activity of the bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either) may also allow for use, or expanded use of immune checkpoint inhibitor which at normal dosages may have deleterious side-effects, such as immune-related adverse events.
  • the immune checkpoint inhibitor when administered in combination with bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either), the immune checkpoint inhibitor may be administered to a patient at a lower dosage than it would normally be administered, over a longer period while maintaining a comparable ongoing therapeutic outcome. This is particularly important for active agents with deleterious side-effects, such as immune-related adverse events.
  • the immune checkpoint inhibitor when administered in combination with bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either), may be administered at a rate which is significantly lower than the recommended dosage rate for that immune checkpoint inhibitor when administered alone, such as at a rate which is at least 10%, 20%, 30%, 40%, 50%, 60% or 70% lower than the dosage rate for that immune checkpoint inhibitor when administered alone.
  • Additional active agents [0078] Programmed cell death 1 ligand 1 (PD-L1) and its receptor programmed cell death 1 (PD-1) regulate the activation of immune cells. Checkpoint inhibitors can assist in suppressing this regulation, allowing immune cells to destroy tumors.
  • checkpoint inhibitors include antibodies that target PD-1, PD-L1, CTLA4 and LAG3.
  • immunomodulators such as cytokines that regulate immune cell maturation, growth and activation, including interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and Granulocyte-macrophage colony-stimulating factor (GM-CSF) have also been approved for treatment of various cancers via immunostimulation.
  • Interleukin-2 or the recombinant human version Aldesleukin/ Proleukin
  • targeted therapeutic agents that have been FDA approved for treating melanoma include: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib.
  • methods according to the present invention may further comprise administration of at least one additional therapeutic agent, such as immunomodulators, protein kinase inhibitors, or combinations of immunomodulators, protein kinase inhibitors for the treatment of melanoma.
  • the protein kinase inhibitor may inhibit one or more potential protein kinase targets associated with melanoma.
  • the protein kinase inhibitor inhibits at least one or more of RAS, MEK and BRAF. According to certain embodiments, the protein kinase inhibitor inhibits BRAF.
  • at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib.
  • at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and trametinib.
  • At least one protein kinase inhibitor is selected from the group consisting of: dabrafenib, encorafenib and vemurafenib. [0086] According to still more specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and vemurafenib. [0087] According to even more specific embodiments the protein kinase inhibitor is vemurafenib.
  • the additional therapeutic agent is an immunomodulator selected from interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and GM-CSF.
  • the at least one additional therapeutic agent comprises interleukin-2 (Aldesleukin/ Proleukin).
  • Suitable dosages, dosage frequencies, dosage durations, and routes of administration for these additional agents are known in the art. As suggested by Figure 5, these additional agents can either be administered simultaneously with the bisantrene or the derivative or analog of bisantrene, or at a different time than the bisantrene or the derivative or analog of bisantrene.
  • the additional agent is administered at a different time than the bisantrene or the derivative or analog of bisantrene, it can either be administered before or after the bisantrene or the derivative or analog of bisantrene and/or at different timings, being administered according to different timing and/or frequency regimes.
  • these additional agents can either be administered simultaneously with the immune checkpoint inhibitor, or at a different time than the immune checkpoint inhibitor. If the additional agent is administered at a different time than the immune checkpoint inhibitor, it can either be administered before or after the immune checkpoint inhibitor and/or at different timings, being administered according to different timing and/or frequency regimes.
  • compositions for the treatment of melanoma comprising at least one immune checkpoint inhibitor, as described above, and a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
  • the compositions of the invention may comprise an additional agent selected from immunomodulators, protein kinase inhibitors or combinations thereof.
  • the immune checkpoint inhibitor or inhibitor combination inhibits PD-1, PD-L1, CTLA-4, LAG3 or a combination of at least any two of these targets. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1, PD-L1 or CTLA-4. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1 or PD-L1. According to certain embodiments, the immune checkpoint inhibitor inhibits PD-1.
  • the at least one immune checkpoint inhibitor or inhibitor combination is selected from the group consisting of: ipilimumab; nivolumab; nivolumab-relatlimab; cemiplimab; dostarlimab; and pembrolizumab.
  • the at least one immune checkpoint inhibitor is selected from the group consisting of: nivolumab; nivolumab-relatlimab; and pembrolizumab.
  • the at least one immune checkpoint inhibitor is selected from the group consisting of nivolumab or pembrolizumab.
  • the second agent is bisantrene or a pharmaceutically acceptable salt thereof.
  • the composition may comprise a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof.
  • Compositions according to the present invention may further comprise administration of at least one additional therapeutic agent for the treatment melanoma.
  • the additional therapeutic agent is a checkpoint inhibitor or an immunomodulator such as those discussed above.
  • the at least one additional therapeutic agent comprises a an immunomodulator selected from interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and GM-CSF.
  • the at least one additional therapeutic agent comprises interleukin-2 (Aldesleukin/ Proleukin).
  • the additional therapeutic agent is at least one protein kinase inhibitor selected from the group consisting of: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib.
  • the at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and trametinib. According to other specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: dabrafenib, encorafenib and vemurafenib. According to still more specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and vemurafenib. According to even more specific embodiments the protein kinase inhibitor is vemurafenib.
  • compositions according to the present invention may be administered by any route, and in a form suitable for that route, as known in the art.
  • compositions according to the present invention may be adapted for administration by enteral or parenteral routes, including by injection (such as intravenous, subcutaneous, intramuscular, bolus, etc.), or by, for example, topical, oral, sublingual, nasal, pulmonary, otic, rectal or vaginal administration routes.
  • the pharmaceutical compositions described herein include at least one pharmaceutically acceptable carrier or excipient and/or diluents.
  • pharmaceutically acceptable carriers can be either solid or liquid.
  • Liquid form preparations include solutions, suspensions and emulsions, for example water or water-propylene glycol solutions for parenteral injection. Also included are solid form preparations, such as tablets, or amorphous or crystalline powders, including lyophilized preparations, that are intended to be converted, shortly before use, to liquid form preparations for either oral or injection administration. Such liquid forms include solutions, suspensions and emulsions.
  • Pharmaceutically acceptable carriers and excipients include: (i) a liquid carrier; (ii) an isotonic agent; (iii) a wetting or emulsifying agent; (iv) a preservative; (v) a buffer; (vi) an acidifying agent; (vii) an antioxidant; (viii) an alkalinizing agent; (ix) a carrying agent; (x) a chelating agent; (xi) a coloring agent; (xii) a complexing agent; (xiii) a solvent; (xiv) a suspending and/or viscosity-increasing agent; (xv) an oil; (xvi) a penetration enhancer; (xvii) a polymer; (xviii) a stiffening agent; (xix) a protein; (xx) a carbohydrate; (xxi) a bulking agent; and (xxii) a lubricating agent.
  • Non-limiting examples of pharmaceutically acceptable carriers or diluents are demineralised or distilled water; saline solution; vegetable-based oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil; sesame oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oil, arachis oil or coconut oil; silicone oils, including polysiloxanes, such as methyl polysiloxane, phenyl polysiloxane and methylphenyl polysolpoxane; volatile silicones; mineral oils such as liquid paraffin, soft paraffin or squalane; cellulose derivatives such as methyl cellulose, ethyl cellulose, carboxymethylcellulose, sodium carboxymethylcellulose or hydroxylpropyl- methylcellulose; lower alkanols, for example ethanol or isopropanol; lower aralkanols; lower polyalkylene glycols or lower alkylene glycols,
  • non-toxic parenterally acceptable diluents or carriers can include, Ringer’s solution, isotonic saline, phosphate buffered saline, ethanol and 1,2 propylene glycol.
  • suitable carriers include peanut oil, liquid paraffin, sodium carboxymethylcellulose, methylcellulose, sodium alginate, gum acacia, gum tragacanth, dextrose, sucrose, sorbitol, mannitol, gelatine and lecithin.
  • these oral formulations may contain suitable flavouring and colourings agents.
  • the capsules When used in capsule form the capsules may be coated with compounds such as glyceryl monostearate or glyceryl stearate which delay disintegration.
  • Solid forms for oral administration may contain binders acceptable in human and veterinary pharmaceutical practice, sweeteners, disintegrating agents, diluents, flavourings, coating agents, preservatives, lubricants and/or time delay agents.
  • Suitable binders include gum acacia, gelatine, corn starch, gum tragacanth, sodium alginate, carboxymethylcellulose or polyethylene glycol.
  • Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
  • Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, guar gum, xanthan gum, bentonite, alginic acid or agar.
  • Suitable diluents include lactose, sorbitol, mannitol, dextrose, kaolin, cellulose, calcium carbonate, calcium silicate or dicalcium phosphate.
  • Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring.
  • Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten.
  • Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • Liquid forms for oral administration may contain, in addition to the above agents, a liquid carrier.
  • Suitable liquid carriers include water, oils such as olive oil, peanut oil, sesame oil, sunflower oil, safflower oil, arachis oil, coconut oil, liquid paraffin, ethylene glycol, propylene glycol, polyethylene glycol, ethanol, propanol, isopropanol, glycerol, fatty alcohols, triglycerides or mixtures thereof.
  • Suspensions for oral administration may further comprise dispersing agents and/or suspending agents.
  • Suitable suspending agents include sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, poly-vinyl- pyrrolidone, sodium alginate or acetyl alcohol.
  • Suitable dispersing agents include lecithin, polyoxyethylene esters of fatty acids such as stearic acid, polyoxyethylene sorbitol mono- or di-oleate, -stearate or- laurate, polyoxyethylene sorbitan mono- or di-oleate, -stearate or -laurate and the like.
  • the pharmaceutical composition may comprise a liposome.
  • a liposomal formulation suitable for bisantrene or a cardioprotective derivative thereof comprises small unilamellar or multilamellar liposomes of size range between 0.01 and 100 ⁇ M, and between about 50-95% liposome-entrapped bisantrene, composed of hydrogenated soy phosphatidylcholine, distearoyl phosphatidylglycerol, and cholesterol of natural or synthetic origin lipids, in aqueous solution which can be reconstituted from a lyophilized form to an injectable liposome suspension.
  • the composition is prepared by reconstituting a lyophilized bisantrene/liposome composition to a liposome concentrate, then diluting the concentrate for parenteral administration for the treatment of melanoma.
  • the pharmaceutical composition may comprise a complex with a beta-cyclodextrin.
  • a liposomal formulation suitable for bisantrene, a therapeutically active derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or derivative thereof comprises a complex formed in aqueous solution which may be reconstituted from a lyophilized form to an injectable suspension.
  • One such composition is prepared by reconstituting a lyophilized bisantrene/beta- cyclodextrin composition to a concentrate, then diluting the concentrate for parenteral administration.
  • Beta-cyclodextrin complexes and methods for preparing such complexes are known in the art and are described in., for example, WO 2019/073296 by Rothman.
  • Various formulations suitable for use in the administration of bisantrene, a therapeutically active derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or derivative thereof, are known in the art.
  • compositions for delivery of a hydrophobic drug comprising: (i) the hydrophobic drug; (ii) an oleaginous vehicle or oil phase that is substantially free of butylated hydroxyanisole (BHA) or butylated hydroxytoluene (BHT); (iii) a co- surfactant or emulsifier; (iv) a co-surfactant or auxiliary emulsifier; and (v) benzyl alcohol as a co-solvent.
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluene
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluene
  • co-surfactant or emulsifier iv
  • benzyl alcohol as a co-solvent.
  • compositions for delivery by intravenous, intramuscular, or intraarticular routes of hydrophobic drugs comprising: (i) the hydrophobic drug; (ii) a pharmaceutically acceptable oleaginous vehicle or oil selected from the group consisting of: (a) naturally occurring vegetable oils and (b) semisynthetic mono-, di-, and triglycerides, wherein the oleaginous vehicle or oil is free of BHT or BHA; (iii) a surfactant or emulsifier; (iv) a co-surfactant or emulsifier; (v) an ion-pair former selected from C6-C20 saturated or unsaturated aliphatic acids when the hydrophobic drug is basic and a pharmaceutically acceptable aromatic amine when the hydrophobic drug is acidic; and (vi) water.
  • hydrophobic drugs such as bisantrene or a cardioprotective derivative or analog thereof
  • compositions according to the present invention may be adapted for administration of bisantrene at a dosage of from about 0.1 mg/m 2 /day to about 100 mg/m 2 /day, such as from about 0.2 mg/m 2 /day to about 50 mg/m 2 /day, from about 0.5 mg/m 2 /day to about 20 mg/m 2 /day, from about 1.0 mg/m 2 /day to about 10 mg/m 2 /day, from about 1.0 mg/m 2 /day to about 8 mg/m 2 /day, about 1 mg/m 2 /day, about 2 mg/m 2 /day, about 3 mg/m 2 /day, about 4 mg/m 2 /day, about 5 mg/m 2 /day, about 6 mg/m 2 /day, about 7 mg/m 2 /day, or about 10 mg/m 2 /day.
  • compositions according to the present invention may be adapted for administration of bisantrene once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, over a period of, for example, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 21 days, 28 days, 35 days, 42 days, 49 days, 56 days or 63 days.
  • compositions according to the present invention may be adapted for administration of bisantrene over a period of 28 days, optionally once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, at a dosage of about 20-50 mg/m 2 /28 days.
  • compositions according to the present invention comprising pharmaceutically acceptable salts of bisantrene or therapeutically active derivatives or pharmaceutically acceptable salts thereof may be adapted to deliver similar dosage rates, adjusted for molar equivalence.
  • Dosing regimens for immune checkpoint inhibitors will depend on the particular immune checkpoint inhibitor, and have been published for known immune checkpoint inhibitors, including for treatment of melanoma.
  • suitable dosages of a given immune checkpoint inhibitor can be readily determined by one of ordinary skill in the art using known approaches and techniques as discussed above in relation to bisantrene.
  • Compositions according to the invention may be formulated to include the immune checkpoint inhibitor(s) at their recommended dosage rates, or different dosage rates as discussed below.
  • the dosage of immune checkpoint inhibitor in a composition according to the present invention may be lower than it would be when administered to a patient by itself. This is particularly important for active agents with deleterious side-effects, such as immune related adverse events.
  • the immune checkpoint inhibitor may be included in the composition at a dosage which is significantly lower than the recommended dosage rate for that immune checkpoint inhibitor when administered alone, such as at a rate which is at least 10%, 20%, 30%, 40%, 50%, 60% or 70% lower than the dosage rate for that immune checkpoint inhibitor when administered alone.
  • a dosage which is significantly lower than the recommended dosage rate for that immune checkpoint inhibitor when administered alone such as at a rate which is at least 10%, 20%, 30%, 40%, 50%, 60% or 70% lower than the dosage rate for that immune checkpoint inhibitor when administered alone.
  • an additional therapeutic agent being an immunomodulatory or a protein kinase inhibitor
  • that additional therapeutic agent may be included in the composition at its known recommended rate(s) for the treatment of melanoma, or even lower rates if synergy amongst the components is determined.
  • compositions for the manufacture of medicaments for the treatment of melanoma are described above.
  • kits for the treatment of melanoma are described above.
  • kits with unit doses of one or more of the active agents described herein, usually in injectable doses, are provided. Such kits may include containers containing unit doses, informational package inserts describing the use and attendant benefits of the drugs in treating the disease, and optionally appliance(s) or device(s) for delivery of the composition. [0123] The kit may further comprise any device suitable for administration of the composition. For example, a kit may comprise a needle suitable for intravenous administration. [0124] In some cases, kits may be provided with instructions. The instructions may be provided in the kit or they may be accessed electronically.
  • the instructions may provide information on how to use the compositions of the present disclosure.
  • the instructions may further provide information on how to use the devices of the present disclosure.
  • the instructions may provide information on how to perform the methods of the disclosure.
  • the instructions may provide dosing information.
  • the instructions may provide drug information such as the mechanism of action, the formulation of the drug, adverse risks, contraindications, and the like.
  • the kit is purchased by a physician or health care provider for administration at a clinic or hospital. In some cases, the kit is purchased by a laboratory and used for screening candidate compounds.
  • Antibody control rat IgG2a and RMP1-14 (each 10 mg/kg) were administered via intraperitoneal injection (i.p.) every three days.
  • Vehicle (8% w/v Sulfobutyl-ether- ⁇ -Cyclodextrin (SBECD) and Zantrene (5 or 15 mg/kg) were administered via intravenous injection (i.v.) every second day.
  • the high dose of Zantrene was reduced from 15 mg/kg to 10 mg/kg from Study Day 8 onwards due to adverse events at the higher dose.
  • Combinations of rat IgG2a / Vehicle, RMP1- 14/Vehicle, rat IgG2a / 5 mg/kg Zantrene, rat IgG2a / 15 mg/kg Zantrene, RMP1-14 / 5 mg/kg Zantrene and RMP1-14 / 15 mg/kg Zantrene were administered.
  • Ten doses of Vehicle (8% w/v SBECD) and Zantrene, and seven doses of rat IgG2a and RMP1- 14 were administered to efficacy groups. Seven doses of Vehicle (8% w/v SBECD) and Zantrene, and five doses of rat IgG2a and RMP1-14 were administered to flow groups.
  • the present invention provides a new paradigm for treating of melanoma by the administration of bisantrene, an antineoplastic agent that has multiple mechanisms of action, including DNA intercalation, inhibition of topoisomerase, inhibition of the fat and obesity associated protein (FTO), and activation of the immune system.
  • Bisantrene is well tolerated and, in particular, lacks the cardiotoxicity that is characteristic of some other anthracene derivatives.
  • Bisantrene can be used together with other therapeutic agents that are used for the treatment of melanoma.
  • the inventions illustratively described herein can suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein.

Abstract

The present invention provides methods for treating patients with melanoma, the method comprising administering to the patient a therapeutically effective amount of an immune checkpoint inhibitor and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. Also provided by the present invention are pharmaceutical compositions and kits for the treatment of melanoma as well as the use of such compositions for the manufacture of medicaments for the treatment of melanoma.

Description

TREATMENT OF MELANOMA FIELD OF THE INVENTION [0001] This invention is directed to the use of bisantrene in combination with immune checkpoint inhibitors to treat melanoma. The present invention also contemplates pharmaceutical compositions and kits for such treatments, as well as the use of such compositions for the manufacture of medicaments for such treatments. BACKGROUND OF THE INVENTION [0002] Melanoma is a dangerous form of skin cancer that begins in cells called melanocytes. While melanoma is less frequent than other skin cancers such as basal cell carcinoma (BCC) or squamous cell carcinoma (SCC), melanoma is typically more dangerous because of its ability to metastasize to other organs if not detected and treated at an early stage. [0003] Risk factors that are associated with a higher incidence of melanoma include: a fair complexion (including fair skin that burns or freckles easily, blue or green eyes, and blonde or red hair); exposure to sunlight and other sources of ultraviolet (UV) energy; a prior history of sunburns that caused blistering (especially in childhood); having large moles, many small moles, or moles that look different from normal moles; a family history of unusual moles or melanoma; a personal history of skin cancer; genetic syndromes such as xeroderma pigmentosum; other environmental exposures, including to radiation and some chemicals; a weakened immune system; older age; and male sex. [0004] In 2020, an estimated 196,060 cases of melanoma are expected to be diagnosed in the U.S. While the estimated five-year survival rate for patients whose melanoma is detected early is about 99%, an estimated 6,850 people (4,610 men and 2,240 women) are expected to die of melanoma in the U.S. in 2020. Similar rates of melanoma and deaths are seen in other regions off the world with large populations of people with fair complexion include Australia, New Zealand, South America, South Africa and Europe. [0005] Melanoma can be treated very effectively by surgical removal if identified at an early stage (Stage 0), but once the melanoma has spread into the dermis or beyond (Stage I and greater), treatment becomes more difficult. While some melanomas respond to conventional cancer treatment options such as chemotherapy and radiation, many are resistant to such treatments especially those that are advanced (Stages III & IV). Newer targeted treatments such as the BRAF inhibitors (for example, trametinib, encorafenib, binimetinib, vemurafinib) have proven effective in melanoma, however, the majority of the patients treated with an inhibitor of mutant BRAF eventually suffer relapse, treatment resistance, and disease progression. [0006] Recent breakthroughs in immunotherapy, including anti-PD-1 and PD-L1 checkpoint blockade therapy, have benefitted a growing number of melanoma patients. These agents include ipililmumab, nivolumab, pembrolizumab and combinations of nivolumab and relatlimab. Despite this advance more than half of these patients do not show a durable response to immunotherapy. Multiple mechanisms, such as driver mutations, epigenetic changes, tumor plasticity, and immunosuppression, all mediate resistance to immunotherapy. [0007] At present, there are no approved treatments that can overcome immune checkpoint inhibitor or anti PD-1 resistance in melanoma patients. Therefore, there is a need for new treatments that can enhance the effectiveness of targeted agents and improve treatment outcomes. SUMMARY OF THE INVENTION [0008] The present invention provides a new paradigm for treating melanoma by the administration of bisantrene in combination with a targeted agent, for example an immune checkpoint inhibitor or immunomodulator. This meets the need for new treatments that improve the clinical outcomes of patients with melanoma, especially patients who show resistance to current targeted therapies. [0009] Bisantrene is an antineoplastic agent that has multiple mechanisms of action, including DNA intercalation, inhibition of topoisomerase and the fat mass and obesity-associated protein (FTO), and activation of the immune system. [0010] Surprisingly, it has been found through the course of these studies that bisantrene and pharmaceutically acceptable salts thereof act to enhance the formation of antitumor immune cell subtypes when combined with anti PD-1 inhibition in a mouse model of melanoma. [0011] One aspect of the invention therefore provides a method of treating a patient with melanoma, said method comprising administering to said patient a therapeutically effective amount of a targeted agent, which may be an immune checkpoint inhibitor, and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. [0012] According to certain embodiments the immune checkpoint inhibitor inhibits one or more of PD-1, PD-L1, CTLA4 and LAG3. [0013] According to certain embodiments, the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) ipilimumab (b) nivolumab (c) cemiplimab (c) nivolumab-relatlimab (d) pembrolizumab (e) dostarlimab [0014] According to certain embodiments, the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) nivolumab (b) nivolumab-relatlimab (c) pembrolizumab (d) cemiplimab [0015] According to certain embodiments, the at least one immune checkpoint inhibitor is selected from the group consisting of: (a) nivolumab (b) pembrolizumab (c) cemiplimab [0016] According to certain embodiments, the second agent is bisantrene or a pharmaceutically acceptable salt thereof. [0017] According to certain embodiments, the method comprises administering to a patient a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof. [0018] According to certain embodiments, the method comprises treatment of immune checkpoint inhibitor resistant melanoma. [0019] According to certain embodiments, the method may further comprise administration of at least one additional therapeutic agent, optionally a protein kinase inhibitor for the treatment melanoma. The at least one additional therapeutic agent may be selected from the group consisting of: trametinib; encorafenib; binimetinib; vemurafenib; talimogene laherparepvec; tebentafusp-tebn; interleukin-2; and proleukin. [0020] According to certain embodiments, the method may comprise administering said at least one immune checkpoint inhibitor to said patient prior to, simultaneously with, or after administration of said second agent to said patient. [0021] According to other embodiments, the method may comprise administering said at least one immune checkpoint inhibitor and the second agent to said patient at the same time, optionally in a single composition. [0022] According to certain embodiments, the treatment has enhanced efficacy results against melanoma compared to a method wherein said at least one immune checkpoint inhibitor or said second agent is administered alone. In certain embodiments, the dose of said at least one immune checkpoint inhibitor is at least 20% lower than the dose required of said at least one immune checkpoint inhibitor agent when administered without said second agent to achieve the same targeted outcome. [0023] Another aspect of the invention provides a pharmaceutical composition for the treatment of melanoma by embodiments of treatment methods as described above, said compositions comprising at least one immune checkpoint inhibitor, which may be a PD-1 inhibitor, as described above, and a second agent as described above comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. Said compositions may comprise further active agents as described above. [0024] Another aspect of the invention provides the use of a composition according to the invention, as described above, for the manufacture of a medicament for the treatment of advanced melanoma, optionally immune checkpoint inhibitor resistant melanoma in a patient. [0025] Yet another aspect of the invention provides a kit for the treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma by embodiments of methods of the present invention, as described above, said kit comprising at least one immune checkpoint inhibitor, optionally a PD-1 inhibitor, as described above, and at least a second agent as described above, said second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. Kits according to the present invention may comprise instructions for administering said at least one immune checkpoint inhibitor to said patient prior to, simultaneously with, or after administering said second agent to said patient. Alternatively, kits according to the present invention may comprise instructions for administering said at least one immune checkpoint inhibitor and the second agent to said patient at the same time. BRIEF DESCRIPTION OF THE DRAWINGS [0026] The following invention will become better understood with reference to the specification, appended claims, and accompanying drawings, where the terms “Zan” or “Zantrene” are alternative terms for bisantrene. [0027] Figure 1 shows the change in tumor volume over time in response to Bisantrene, Anti-PD-1 inhibitor (RMP1-14) or Bisantrene/RMP1-14 combinations. Bisantrene was administered by daily IV injections. RMP1-14 was administered by IP injection 3 times per week. Data points represent the mean of each group at each time point +/- SEM. [0028] Figure 2 shows the percentage of M2b macrophages (CD86+ CD206-) as a proportion of F4/80+ cells isolated from excised subcutaneous tumors taken at Day 7 or Day 13 after randomization. Data points represent samples from individual tumors, bars represent mean +/- SEM. Difference between groups was assessed via 2-way ANOVA using Šidák’s correction for multiple comparisons, threshold for significance = p <0.05. [0029] Figure 3 shows the percentage of M1 macrophages (CD86+ CD11b+) as a proportion of myeloid cells (MHCII+ CD11c+) isolated from excised spleens taken at Day 7 or Day 13 after randomization. Data points represent samples from individual mice, bars represent mean +/- SEM. Difference between groups was assessed via 2-way ANOVA using Šidák’s correction for multiple comparisons, threshold for significance = p <0.05. [0030] Figure 4 shows the percentage of CTLA4+ T-cells as a proportion of cytotoxic T-cells (CD8+) isolated from excised subcutaneous tumors taken at Day 7 or Day 13 after randomization. Data points represent samples from individual tumors, bars represent mean +/- SEM. Difference between groups was assessed via 2-way ANOVA using Šidák’s correction for multiple comparisons, threshold for significance = p <0.05. [0031] Figure 5 shows the use of bisantrene plus a targeted agent, for example, an immune checkpoint inhibitor to overcome treatment resistance in melanoma. DEFINITIONS [0032] As used herein, “treating” means affecting a subject/patient, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes inhibiting a condition, i.e. slowing or arresting its development; or relieving or ameliorating the effects of the condition i.e., cause reversal or regression of the effects of the condition. [0033] As used herein, “preventing” means preventing a condition from occurring in a cell, tissue or subject that may be at risk of having the condition, but does not necessarily mean that condition will not eventually develop, or that a subject will not eventually develop a condition. Preventing includes delaying the onset of a condition in a cell, tissue or subject. [0034] As used herein, the term “subject” or “patient” refers to a mammal. The mammal may be a human or a non-human. Examples of non-humans include primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Typically, the mammal is a human or a non- human primate. More typically, the mammal is a human. [0035] The term “composition” encompasses compositions and formulations comprising the active pharmaceutical ingredient (for example, “immune checkpoint inhibitor” and “second agent comprising bisantrene or a derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or cardioprotective derivative of bisantrene”) with excipients or carriers, and also compositions and formulations with a carrier. In pharmaceutical compositions, the excipient or carrier is “pharmaceutically acceptable” meaning that it is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Supplementary active ingredients can also be incorporated into the compositions. [0036] By “pharmaceutically acceptable” such as in the recitation of a “pharmaceutically acceptable salt” or a “pharmaceutically acceptable excipient or carrier” is meant herein a material that is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. [0037] The term “effective amount” or “therapeutically effective amount” refers to the quantity of an active pharmaceutical ingredient that is sufficient to yield a desired therapeutic response. The specific effective amount or therapeutically effective amount will vary with such factors as the particular condition being treated, the age, body weight, general health, physical condition, gender and diet of the subject, the duration of the treatment, the nature of concurrent therapy (if any), and the severity of the particular condition. [0038] As used herein, “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, bulking agents, carrier solutions, suspensions, colloids, and forming and binding agents, any or all of which may include other pharmaceutical excipients as known in the art, including lubricants, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, antioxidants, other stabilisers, including physical stabilisers such as thickeners and viscosity enhancers, colouring agents, flavouring and sweetening agents, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. [0039] As used herein, “administration” or “administer” or “administering” refers to dispensing, applying, or tendering one or more agents to a subject. Administration can be performed using any of a number of methods known in the art. For example, "administering" as used herein is meant via infusion (intravenous administration (i.v.)), parenteral administration. By "parenteral" is meant intravenous, subcutaneous and intramuscular administration. DETAILED DESCRIPTION OF THE INVENTION [0040] The present disclosure relates to a method for the treatment of melanoma, especially immune checkpoint inhibitor resistant melanoma, comprising administering a therapeutically active amount of bisantrene in combination with an immune checkpoint inhibitor, especially a PD-1 inhibitor. Immune checkpoint inhibitors [0041] Programmed cell death 1 ligand 1 (PD-L1) and its receptor programmed cell death 1 (PD-1) regulate the activation of immune cells. Checkpoint inhibitors can assist overcoming this immune regulation, allowing immune cells to destroy tumors. An emerging strategy is the combination of checkpoint inhibitors with cytotoxic chemotherapies or other anti-cancer agents or treatments, with the underlying rationale that the damage signals caused by cell death further stimulate immune activation, leading to enhanced killing of tumor cells by lymphocytes. Other strategies involve the combination of immune activation treatments to increase the immune response to a tumor. Known checkpoint inhibitors include antibodies that target PD-1, PD-L1, CTLA4 or LAG3. [0042] The FDA has approved two PD-1 inhibitors (nivolumab and pembrolizumab), one CTLA-4 inhibitor (ipilimumab) and one PD-1/LAG3 inhibitor combination (nivolumab and relatlimab) for use in melanoma. Other PD-1/ PD-L1 immune checkpoint inhibitors of potential consideration include avelumab, atezolizumab, bevacizumab, cemiplimab, dostarlimab, and durvalumab. [0043] Similarly, immunomodulators such as cytokines that regulate immune cell maturation, growth and activation, including interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and Granulocyte-macrophage colony-stimulating factor (GM-CSF) have also been approved for treatment of various cancers via immunostimulation. Interleukin-2 (or the recombinant human version Aldesleukin/ Proleukin) has been approved for the treatment of melanoma. Bisantrene and derivatives or analogs thereof, and pharmaceutically acceptable salts [0044] Bisantrene has immunologic properties that might be responsible for some of its activities, and which may make this agent a useful tool in combination with immune checkpoint inhibitors. Intravenously (i.v.), intraperitoneally (i.p.), or subcutaneously (s.c.), bisantrene was effective in cancer models using colon 26, Lewis lung, Ridgway osteosarcoma, B16, Lieberman plasma cell, P388 or L1210 cancer cells. Anti-cancer activity was observed in clonogenic assays using tumor samples isolated from 684 patients with breast, small cell lung, large cell lung, squamous cell lung, ovarian, pancreatic, renal, adrenal, head and neck, sarcoma, gastric, lymphoma and melanoma cancers, but not in colorectal cancer. Importantly, a lack of cross resistance with adriamycin and mitoxantrone was found. Subsequent to treatment with bisantrene, and for 4 weeks thereafter, macrophages could be isolated from peritoneal exudate that had cytostatic anti-proliferative activity in cultures of P815 (mastocytoma) tumor cells. Moreover, the supernatants from bisantrene activated macrophages also had a protective cytostatic effect in the tumor cell cultures. Further work revealed that macrophages activated with bisantrene and adoptively transferred to mice with EL-4 lymphomas more than doubled their median survival time, with 7 of 10 mice in the group being cured. Multiple administrations of activated macrophages were more effective than a single administration (reviewed in Rothman, J. (2017) “The Rediscovery of Bisantrene: A Review of the Literature”. Int J Cancer Res Ther 2, 1–10). [0045] Bisantrene has multiple anti-cancer mechanisms of activity including direct cytotoxic action, immunologic, and as a potent inhibitor (IC50142 nM) of the fat mass and obesity-associated protein (FTO), an RNA N6-methyladenosine (m6A) demethylase (Su, R., Dong, L., Li, Y., Gao, M., Han, L., Wunderlich, M., et al. (2020), “Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion”, Cancer Cell, 38(1): 79–96.e11). [0046] The chemical name for bisantrene is 9, 10-anthracenedicarboxaldehyde-bis [(4, 5-dihydro-1H-imidazole-2-yl) hydrazine] dihydrochloride, and it was originally classed as an anthracycline chemotherapeutic agent. As used herein, the term “bisantrene” refers to bisantrene or any pharmacologically compatible salt form, not only bisantrene dihydrochloride, unless the dihydrochloride or another specific pharmacologically compatible salt form is specifically indicated. Typically, the pharmacologically compatible salt of bisantrene is bisantrene dihydrochloride for most pharmacological applications. [0047] Recent studies have identified that bisantrene suppresses immune checkpoint gene expression and immune evasion via enzymatic inhibition of the FTO RNA demethylase as described in R. Su. et al. (2020), “Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion”, Cancer Cell, 38(1): 79– 96.e11. [0048] Bisantrene has also been found to have non-immunologic telomeric effects. Bisantrene binds to DNA at a site called a G-quadruplex, in which 4 guanines are associated by folding. Stabilization of the G-quadruplex can interfere with telomere- telomerase interaction and thus inhibit the activity of telomerase in various ways, including the displacement of telomerase binding proteins. Since the level of topoisomerase II inhibition does not always correlate with cytotoxic efficacy, alternative mechanisms may play a role in the actions of bisantrene. Analogs of bisantrene have been made in an attempt to improve upon the anti-telomerase activity; these analogs are described further below. Human melanoma (SK-Mel5) and colon cancer (LoVo) tumor cells were observed to lose their proliferative ability in the presence of these agents. Apoptosis was not observed, but a loss of immortality was seen; treated cells reacquiring the ability to senesce, age, and die. [0049] As detailed above, in addition to direct antineoplastic effects related to the activity of bisantrene as a DNA intercalator, bisantrene also possesses other mechanisms of action, including immunopotentiation. These mechanisms are described in: (i) N.R. West et al. (2011), “Tumor-Infiltrating Lymphocytes Predict Response to Anthracycline-Based Chemotherapy in Estrogen-Resistant Breast Cancer”, Breast Canc. Res.13: R126, which concludes that the level of tumor- infiltrating lymphocytes is correlated with a response to the administration of anthracycline-based agents; the markers associated with tumor-infiltrating lymphocytes (TIL) include CD19, CD3D, CD48, GZMB, LCK, MS4A1, PRF1, and SELL; (ii) L. Zitvogel et al. (2008), “Immunological Aspects of Cancer Chemotherapy”, Nature Rev. Immunol.8: 59-73, which states that DNA damage, such as that produced by intercalating agents such as bisantrene, induces the expression of NKG2D ligands on tumor cells in an ATM-dependent and CHK1- dependent (but p53-independent) manner; NKG2D is an activating receptor that is involved in tumor immunosurveillance by NK cells, NK T cells, γδ T cells and resting (in mice) and/or activated (in humans) CD8+ T cells, and also states that anthracycline-based agents may act as immunostimulators, particularly in combination with IL-12; such agents also promote HMGB1 release and activate T cells; (iii) D.V. Krysko et al. (2011), “TLR2 and TLR9 Are Sensors of Apoptosis in a Mouse Model of Doxorubicin-Induced Acute Inflammation”, Cell Death Different.18: 1316-1325, which states that anthracycline-based antibiotics induce an immunogenic form of apoptosis that has immunostimulatory properties mediated by MyD88, TLR2, and TLR9; (iv) C. Ferraro et al. (2000), “Anthracyclines Trigger Apoptosis of Both G0-G1 and Cycling Peripheral Blood Lymphocytes and Induce Massive Deletion of Mature T and B Cells”, Cancer Res.60: 1901-1907, which stated that anthracyclines induce apoptosis and ceramide production, as well as activate caspase-3 in resting and cycling cells; the apoptosis induced is independent from CD95-L/CD95 and TNF/TNF-R; (v) K. Lee et al. (2009), “Anthracycline Chemotherapy Inhibits HIF-1 Transcriptional Activity and Tumor-Induced Mobilization of Circulating Angiogenic Cells”, Proc. Natl. Acad. Sci. USA 106: 2353- 2358, which provides another antineoplastic mechanism for anthracycline-based antibiotics, namely inhibition of HIF-1 mediated gene transcription, which, in turn, inhibits transcription of VEGF required for angiogenesis; HIF-1 also activates transcription of genes encoding glucose transporter GLUT1 and hexokinases HK1 and HK2, which are required for the high level of glucose uptake and phosphorylation that is observed in metastatic cancer cells, and pyruvate dehydrogenase kinase 1 (PDK1), which shunts pyruvate away from the mitochondria, thereby increasing lactate production; patients with HiF-1a overexpression based on immunohistochemical resuits were suggested to be good candidates for treatment with anthracyciine-based antibiotics.
[0050] Several clinical trials have investigated the pharmacokinetics of bisantrene in humans, to one trial of patients given a 90 min infusion at 260 mg/m2 a biphasic elimination with an initial half-life of 65 ± 15 min, a terminal half -life of 1142 ± 226 min, and a steady state volume of distribution (Vdss) of 1845 L/m2. Plasma clearance in this trial was 735 mL/min/m2, with 11.3% of the administered dose excreted unchanged in the urine in 24 h. In another trial , doses of 80-250 mg/m2 were assessed, and the initial and terminal half-lives were 0.6 h and 24.7 h, respectively, with a clearance of 1045.5 ± 51.0 mL/kg/h and a calculated volume of distribution of 42.1 ± 5.9 L/kg. In this study only 3.4 ± 1.1% of the administered dose was found in the urine over 96 h. to three other single dose studies triphasic elimination was reported, one with
Figure imgf000013_0001
«, p, and y of 3.44 min, 1.33 h and 26.13 h, respectively, another was 3 min, 1 h, and 8 h respectively, and the last revealed clearances of 0.1 h, 1.9 h and 43.9 h, respectively. In one report a large volume of distribution (687 L/m2) was interpreted as tissue sequestration of the drug with a subsequent depot effect, in a 72 h infusion study, a plasma concentration of 12 ± 6 ng/mL was observed at a dose of 56 mg/m2, while a dose of 260 mg/m2 resulted in a plasma concentration of 55 ± 8 ng/mL. in this trial plasma clearance was 1306 ± 179 mL/min/m2 with urinary excretion of 4.6% of the dose in 24 h. Finally, in another study, a 5-day schedule of 60 min infusions revealed a tl4 a and p of 0.9 and 9.7 h, respectively with 7.1% of the dose excreted in the urine.
[0051] The structure of bisantrene dihydrochloride is shown in Formula (I)
Figure imgf000013_0002
[0052] Bisantrene dihydrochloride is a tricyclic aromatic compound with the chemical name, 9,10-anthracenedicarboxaldehyde bis[(4,5-dihydro-1H-imidazol-2- yl)hydrazine] dihydrochloride. The molecular formula of bisantrene dihydrochloride is C22H22N8 ^ 2HCl and the molecular weight, 471.4 g/mol. The alkylimidazole side chains are very basic and, at physiologic pH, are positively charged. This is believed to facilitate electrostatic attractions to negatively charged ribose phosphate groups in DNA. [0053] Bisantrene is typically administered intravenously, either centrally or peripherally. [0054] Various formulations suitable for use in the administration of bisantrene or derivatives or analogs thereof are known in the art. United States Patent No. 4,784,845 to Desai et al. discloses a composition of matter for delivery of a hydrophobic drug (i.e., bisantrene or a derivative or analog thereof) comprising: (i) the hydrophobic drug; (ii) an oleaginous vehicle or oil phase that is substantially free of butylated hydroxyanisole (BHA) or butylated hydroxytoluene (BHT); (iii) a co- surfactant or emulsifier; (iv) a co-surfactant or auxiliary emulsifier; and (v) benzyl alcohol as a co-solvent. United States Patent No.4,816,247 by Desai et al. discloses a composition of matter for delivery by intravenous, intramuscular, or intraarticular routes of hydrophobic drugs (such as bisantrene or a derivative or analog thereof) comprising: (i) the hydrophobic drug; (ii) a pharmaceutically acceptable oleaginous vehicle or oil selected from the group consisting of: (a) naturally occurring vegetable oils and (b) semisynthetic mono-, di-, and triglycerides, wherein the oleaginous vehicle or oil is free of BHT or BHA; (iii) a surfactant or emulsifier; (iv) a co-surfactant or emulsifier; (v) an ion-pair former selected from C6-C20 saturated or unsaturated aliphatic acids when the hydrophobic drug is basic and a pharmaceutically acceptable aromatic amine when the hydrophobic drug is acidic; and (vi) water. United States Patent No.5,000,886 to Lawter et al. and United States Patent No. 5,143,661 to Lawter et al. disclose compositions for delivery of pharmaceutical agents such as bisantrene or a derivative or analog thereof comprising a microcapsule, wherein the microcapsule includes a hardening agent that is a volatile silicone fluid. United States Patent No.5,070,082 to Murdock et al., United States Patent No.5,077,282 to Murdock et al., and United States Patent No.5,077,283 to Murdock et al. disclose prodrug forms of poorly soluble hydrophobic drugs, including bisantrene and derivatives and analogs, that are salts of a phosphoramidic acid. United States Patent No.5,116,827 to Murdock et al. and United States Patent No. 5,212,291 to Murdock et al. disclose prodrug forms of poorly soluble hydrophobic drugs, including bisantrene and derivatives and analogs, that are quinolinecarboxylic acid derivatives. United States Patent No.5,378,456 to Tsou includes compositions containing an anthracene antitumor agent, such as bisantrene or a derivative or analog thereof, in which the bisantrene or derivative or analog thereof is conjugated to or admixed with a divinyl ether-maleic acid (MVE) copolymer. United States Patent No.5,609,867 to Tsou discloses polymeric 1,4-bis derivatives of bisantrene and copolymers of bisantrene and another monomer, such as a dianhydride. [0055] Methods and compositions described herein can use a derivative or analog of bisantrene in place of bisantrene itself. Derivatives and analogs of bisantrene are described in in US Patents 10,500,19 and 10,548,876, by Garner et al. Methods for treating melanoma [0056] Surprisingly, the present studies have shown that immune checkpoint inhibitors and bisantrene (or pharmaceutically acceptable salts thereof) show beneficial antitumor effects in a mouse model of melanoma. Furthermore, bisantrene was able to increase the number of antitumorigenic M1 macrophages when combined with a murine PD-1 inhibitor relative to mice treated with the PD-1 inhibitor alone. In addition, bisantrene/Anti-PD-1 combination produced a significant decrease in protumorigenic M2b macrophages in tumours relative to PD-1 inhibition alone, indicating that bisantrene is able to elicit an antitumor immunophenotype through selective effects on myeloid cell populations. Bisantrene/anti-PD-1 combinations also potently downregulated the expression of CTLA4, a validated immunosuppressive checkpoint target, in CD8+ T-cells, priming these key mediators of antitumor immunity for effective destruction of tumor cells. These effects offer an exciting novel approach for the enhancement of immune checkpoint therapy through the modulation of myeloid cell disposition and immunosuppressive lymphocyte signatures in melanoma patients. It is contemplated that derivatives of bisantrene possessing substantially the same activity will also act enhance with antitumor immune stimulation when combined with checkpoint inhibitors in methods and compositions according to the present invention. [0057] As of 2022 the FDA has approved three PD-1 inhibitors (pembrolizumab [Keytruda], nivolumab [Opdivo], ostarlimab [Jemperli], and cemiplimab [Libtayo]), one PD-L1 inhibitor/LAG3 inhibitor combination (nivolumab-relatlimab), and one CTLA-4 inhibitor (ipilimumab) for use. Other PD-1/ PD-L1 immune checkpoint inhibitors of potential consideration include atezolizumab, bevacizumab, dostarlimab, and durvalumab. [0058] Thus, one aspect of the invention provides a method of treating a patient with melanoma, especially immune checkpoint inhibitor resistant melanoma, said method comprising administering to said patient a therapeutically effective amount of an immune checkpoint inhibitor, such as a PD-1 inhibitor, and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. [0059] The immune checkpoint inhibitor may inhibit one or more potential immune checkpoint targets associated with melanoma. According to certain embodiments, the immune checkpoint inhibitor or inhibitor combination inhibits PD-1, PD-L1, CTLA- 4, LAG3 or a combination of at least any two of these targets. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1, PD-L1, LAG3. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1 or PD-L1. According to certain embodiments, the immune checkpoint inhibitor inhibits PD-1. [0060] According to certain embodiments the at least one immune checkpoint inhibitor or inhibitor combination is selected from the group consisting of: ipilimumab; nivolumab; cemiplimab; nivolumab-relatlimab; and pembrolizumab. [0061] According to specific embodiments, the at least one immune checkpoint inhibitor is selected from the group consisting of: nivolumab; cemiplimab; nivolumab- relatlimab; and pembrolizumab. [0062] According to specific embodiments of the present invention, the at least one immune checkpoint inhibitor is selected from the group consisting of nivolumab, cemiplimab or pembrolizumab. [0063] According to specific embodiments of the present invention, the second agent is bisantrene or a pharmaceutically acceptable salt thereof. [0064] According to certain embodiments, the bisantrene can be administered as a drug compound or as a component of a pharmaceutical composition, as discussed further below. [0065] According to certain specific embodiments, the method comprises administering to said patient a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof. [0066] The pharmaceutical compositions and medicaments of the present invention may be administered to a subject by standard enteral or parenteral routes, including, but not limited to, injection (such as intravenous, subcutaneous, intramuscular, bolus, etc.), or by, for example, topical, oral, sublingual, nasal, pulmonary, otic, rectal or vaginal administration routes. In some embodiments, pharmaceutical compositions according to the invention may be administered to a subject by themselves or in combination with other pharmaceutical composition(s). In the latter case, the administration may be simultaneous or sequential, or administration of the pharmaceutical composition(s) may be independent of one another. [0067] In certain embodiments bisantrene (or derivative, or pharmaceutically acceptable salt of either) is administered intravenously, either centrally or peripherally, including by intramuscular, subcutaneous and/or intradermal injection. [0068] In general, the pharmaceutical compositions and medicaments of the present invention can be administered in a manner compatible with the route of administration and physical characteristics of the subject (including health status) and in such a way that the desired effect(s) are induced (i.e. therapeutically effective and/or preventative). For example, the appropriate dosage may depend on a variety of factors including, but not limited to, a subject’s physical characteristics (e.g. age, weight, sex), whether the composition or medicament is being used as single agent, the progression (i.e. pathological state) of the disease or disorder being treated, and other factors readily apparent to those of ordinary skill in the art. [0069] Suitable dosages, dosage frequencies, dosage durations, and routes of administration for chemotherapeutic agents are known in the art. As suggested by Figure 5, bisantrene, derivatives of bisantrene, or pharmaceutically acceptable salts of either can be administered in the same pharmaceutical composition as an immune checkpoint inhibitor, in a separate composition as, but simultaneously with the immune checkpoint inhibitor, or at a different time. If the bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of bisantrene or derivative of bisantrene, is administered at a different time than the immune checkpoint inhibitor, it can either be administered before or after the immune checkpoint inhibitor and/or at different timings, being administered according to different timing and/or frequency regimes. One of ordinary skill in the art can determine a suitable schedule for administration based on variables such as the age, weight, and sex of the subject, the susceptibility of the subject to side effects of the active agents, genetic markers, the dose of active agent(s), the subjects history with prior active agent(s), and other pharmacokinetic parameters such as heart, liver or kidney function. [0070] Figure 5 shows a prophetic example of a bisantrene/immune checkpoint inhibitor trial to overcome treatment resistance in a model of melanoma. [0071] The methods and compositions provided herein enable a subject to receive a therapy more frequently without having the dosage regimen significantly altered by the risk of side-effects, such as cardiotoxicity. The dose(s) of a immune checkpoint inhibitor and bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of bisantrene or derivative of bisantrene, may be administered to a subject in one or more doses per day. In some cases, the daily dose of the chemotherapeutic agent may be administered together with bisantrene, derivative of bisantrene, or pharmaceutically acceptable salt of either, in a single dose. [0072] The pharmaceutical compositions described herein may be administered to a patient one or more times per day. In some cases, the pharmaceutical composition may be administered to a patient once per day. In some cases, the pharmaceutical composition may be administered to a patient at least 2 times, 3 times, 4 times 5 times, or 6 times per day. For example, a pharmaceutical composition may be administered to a patient 3 times per day. [0073] In methods described herein, suitable dosages of bisantrene (or a derivative of bisantrene, or a pharmaceutically acceptable salt of either) can be determined by one of ordinary skill in the art. The selected dosage level depends upon a variety of pharmacokinetic factors including the amount of active agent(s) being administered, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the severity of the condition, other health considerations affecting the subject, and the status of liver and kidney function of the subject. It also depends on the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular therapeutic agent employed, as well as the age, weight, condition, general health and prior medical history of the subject being treated, and like factors. Methods for determining optimal dosages are described in the art, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Co., 20th ed., 2000, and Gilman et al., (Eds), (1990), “Goodman And Gilman's: The Pharmacological Bases of Therapeutics”, Pergamon Press. Optimal dosages for a given set of conditions can be ascertained by those skilled in the art using conventional dosage-determination tests in view of the experimental data for an agent. [0074] According to certain embodiments, administration of bisantrene is performed at a dosage of from about 0.1 mg/m2/day to about 400 mg/m2/day, such as from about 0.2 mg/m2/day to about 300 mg/m2/day, from about 0.5 mg/m2/day to about 200 mg/m2/day, from about 0.5 mg/m2/day to about 100 mg/m2/day, from about 0.5 mg/m2/day to about 50 mg/m2/day, from about 0.5 mg/m2/day to about 30 mg/m2/day, from about 0.5 mg/m2/day to about 20 mg/m2/day, from about 1.0 mg/m2/day to about 10 mg/m2/day, from about 1.0 mg/m2/day to about 8 mg/m2/day, about 1 mg/m2/day, about 2 mg/m2/day, about 3 mg/m2/day, about 4 mg/m2/day, about 5 mg/m2/day, about 6 mg/m2/day, about 7 mg/m2/day, or about 10 mg/m2/day. In some embodiments, bisantrene is administered daily or weekly, once every two weeks, once every three weeks, once every four weeks, over a period of, for example, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 21 days, 28 days, 35 days, 42 days, 49 days, 56 days or 63 days. In certain embodiments, bisantrene is administered once or multiple times over a period of 28 days, optionally once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, at a dosage of about 20-50 mg/m2/28 days. Administration of pharmaceutically acceptable salts of bisantrene or derivatives or pharmaceutically acceptable salts thereof may be performed at similar dosage rates, adjusted for molar equivalence. [0075] Dosing regimens for immune checkpoint inhibitors will depend on the particular immune checkpoint inhibitor, and have been published for known immune checkpoint inhibitors, including for treatment of melanoma. In addition, suitable dosages of a given immune checkpoint inhibitor can be readily determined by one of ordinary skill in the art using known approaches and techniques as discussed above in relation to bisantrene. [0076] According to certain embodiments, and as a result of the beneficial biologic activity observed when bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either) is administered with a immune checkpoint inhibitor, a therapeutic outcome may be achieved more effectively for a given dose rate for that immune checkpoint inhibitor. This biologic activity of the bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either) may also allow for use, or expanded use of immune checkpoint inhibitor which at normal dosages may have deleterious side-effects, such as immune-related adverse events. [0077] Alternatively, when administered in combination with bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either), the immune checkpoint inhibitor may be administered to a patient at a lower dosage than it would normally be administered, over a longer period while maintaining a comparable ongoing therapeutic outcome. This is particularly important for active agents with deleterious side-effects, such as immune-related adverse events. Thus, in certain embodiments of methods according to the present invention, when administered in combination with bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either), the immune checkpoint inhibitor may be administered at a rate which is significantly lower than the recommended dosage rate for that immune checkpoint inhibitor when administered alone, such as at a rate which is at least 10%, 20%, 30%, 40%, 50%, 60% or 70% lower than the dosage rate for that immune checkpoint inhibitor when administered alone. Additional active agents [0078] Programmed cell death 1 ligand 1 (PD-L1) and its receptor programmed cell death 1 (PD-1) regulate the activation of immune cells. Checkpoint inhibitors can assist in suppressing this regulation, allowing immune cells to destroy tumors. Known checkpoint inhibitors include antibodies that target PD-1, PD-L1, CTLA4 and LAG3. [0079] In addition to immune checkpoint inhibitors, some studies have shown that immunomodulators such as cytokines that regulate immune cell maturation, growth and activation, including interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and Granulocyte-macrophage colony-stimulating factor (GM-CSF) have also been approved for treatment of various cancers via immunostimulation. Interleukin-2 (or the recombinant human version Aldesleukin/ Proleukin) has been approved for the treatment of melanoma. [0080] Also in addition to immune checkpoint inhibitors, targeted therapeutic agents that have been FDA approved for treating melanoma include: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib. [0081] Thus, methods according to the present invention may further comprise administration of at least one additional therapeutic agent, such as immunomodulators, protein kinase inhibitors, or combinations of immunomodulators, protein kinase inhibitors for the treatment of melanoma. [0082] The protein kinase inhibitor may inhibit one or more potential protein kinase targets associated with melanoma. According to certain embodiments, the protein kinase inhibitor inhibits at least one or more of RAS, MEK and BRAF. According to certain embodiments, the protein kinase inhibitor inhibits BRAF. [0083] According to certain embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib. [0084] According to specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and trametinib. [0085] According to more specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: dabrafenib, encorafenib and vemurafenib. [0086] According to still more specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and vemurafenib. [0087] According to even more specific embodiments the protein kinase inhibitor is vemurafenib. [0088] According to certain embodiments the additional therapeutic agent is an immunomodulator selected from interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and GM-CSF. According to certain embodiments the at least one additional therapeutic agent comprises interleukin-2 (Aldesleukin/ Proleukin). [0089] Suitable dosages, dosage frequencies, dosage durations, and routes of administration for these additional agents are known in the art. As suggested by Figure 5, these additional agents can either be administered simultaneously with the bisantrene or the derivative or analog of bisantrene, or at a different time than the bisantrene or the derivative or analog of bisantrene. If the additional agent is administered at a different time than the bisantrene or the derivative or analog of bisantrene, it can either be administered before or after the bisantrene or the derivative or analog of bisantrene and/or at different timings, being administered according to different timing and/or frequency regimes. Similarly, and independently of bisantrene considerations, these additional agents can either be administered simultaneously with the immune checkpoint inhibitor, or at a different time than the immune checkpoint inhibitor. If the additional agent is administered at a different time than the immune checkpoint inhibitor, it can either be administered before or after the immune checkpoint inhibitor and/or at different timings, being administered according to different timing and/or frequency regimes. One of ordinary skill in the art can determine a suitable schedule for administration based on variables such as the age, weight, and sex of the patient, the severity of the cancer, genetic markers such as further described below, and pharmacokinetic parameters such as liver and kidney function. Pharmaceutical compositions for the treatment of melanoma [0090] The present invention also provides pharmaceutical compositions for the treatment of melanoma comprising at least one immune checkpoint inhibitor, as described above, and a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. As described above, the compositions of the invention may comprise an additional agent selected from immunomodulators, protein kinase inhibitors or combinations thereof. [0091] According to certain embodiments, the immune checkpoint inhibitor or inhibitor combination inhibits PD-1, PD-L1, CTLA-4, LAG3 or a combination of at least any two of these targets. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1, PD-L1 or CTLA-4. According to certain embodiments, the immune checkpoint inhibitor inhibits at least one or more of PD-1 or PD-L1. According to certain embodiments, the immune checkpoint inhibitor inhibits PD-1. [0092] According to certain embodiments the at least one immune checkpoint inhibitor or inhibitor combination is selected from the group consisting of: ipilimumab; nivolumab; nivolumab-relatlimab; cemiplimab; dostarlimab; and pembrolizumab. [0093] According to certain embodiments, the at least one immune checkpoint inhibitor is selected from the group consisting of: nivolumab; nivolumab-relatlimab; and pembrolizumab. [0094] According to specific embodiments of the present invention, the at least one immune checkpoint inhibitor is selected from the group consisting of nivolumab or pembrolizumab. [0095] According to specific embodiments of the present invention, the second agent is bisantrene or a pharmaceutically acceptable salt thereof. [0096] According to specific embodiments of the present invention, the composition may comprise a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof. [0097] Compositions according to the present invention may further comprise administration of at least one additional therapeutic agent for the treatment melanoma. [0098] According to certain embodiments the additional therapeutic agent is a checkpoint inhibitor or an immunomodulator such as those discussed above. According to certain embodiments the at least one additional therapeutic agent comprises a an immunomodulator selected from interleukin-2 (Aldesleukin/ Proleukin), interferon alpha-2a, interferon alpha-2b (and Peginterferon alpha-2b), and GM-CSF. According to certain embodiments the at least one additional therapeutic agent comprises interleukin-2 (Aldesleukin/ Proleukin). [0099] According to certain embodiments the additional therapeutic agent is at least one protein kinase inhibitor selected from the group consisting of: binimetinib, cobimetinib, dabrafenib, encorafenib, trametinib, and vemurafenib. According to specific embodiments the at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and trametinib. According to other specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: dabrafenib, encorafenib and vemurafenib. According to still more specific embodiments at least one protein kinase inhibitor is selected from the group consisting of: binimetinib, cobimetinib and vemurafenib. According to even more specific embodiments the protein kinase inhibitor is vemurafenib. [0100] Compositions according to the present invention may be administered by any route, and in a form suitable for that route, as known in the art. Thus, compositions according to the present invention may be adapted for administration by enteral or parenteral routes, including by injection (such as intravenous, subcutaneous, intramuscular, bolus, etc.), or by, for example, topical, oral, sublingual, nasal, pulmonary, otic, rectal or vaginal administration routes. [0101] Typically, the pharmaceutical compositions described herein include at least one pharmaceutically acceptable carrier or excipient and/or diluents. For preparing the pharmaceutical compositions and medicaments, inert, pharmaceutically acceptable carriers can be either solid or liquid. Liquid form preparations include solutions, suspensions and emulsions, for example water or water-propylene glycol solutions for parenteral injection. Also included are solid form preparations, such as tablets, or amorphous or crystalline powders, including lyophilized preparations, that are intended to be converted, shortly before use, to liquid form preparations for either oral or injection administration. Such liquid forms include solutions, suspensions and emulsions. Examples of pharmaceutically acceptable carriers and methods of manufacture for various compositions may be found in, for example, “Remington: The Science and Practice of Pharmacy”, Mack Publishing Co., 20th ed., 2000, and Gilman et al., (Eds), (1990), “Goodman And Gilman's: The Pharmacological Bases of Therapeutics”, Pergamon Press. [0102] Pharmaceutically acceptable carriers and excipients include: (i) a liquid carrier; (ii) an isotonic agent; (iii) a wetting or emulsifying agent; (iv) a preservative; (v) a buffer; (vi) an acidifying agent; (vii) an antioxidant; (viii) an alkalinizing agent; (ix) a carrying agent; (x) a chelating agent; (xi) a coloring agent; (xii) a complexing agent; (xiii) a solvent; (xiv) a suspending and/or viscosity-increasing agent; (xv) an oil; (xvi) a penetration enhancer; (xvii) a polymer; (xviii) a stiffening agent; (xix) a protein; (xx) a carbohydrate; (xxi) a bulking agent; and (xxii) a lubricating agent. [0103] Other pharmaceutically acceptable carriers and excipients known in the art may be used. [0104] The carriers, diluents, excipients and adjuvants must be “acceptable” in terms of being compatible with the other ingredients of the composition or medicament, and are generally not deleterious to the subject thereof. Non-limiting examples of pharmaceutically acceptable carriers or diluents are demineralised or distilled water; saline solution; vegetable-based oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil; sesame oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oil, arachis oil or coconut oil; silicone oils, including polysiloxanes, such as methyl polysiloxane, phenyl polysiloxane and methylphenyl polysolpoxane; volatile silicones; mineral oils such as liquid paraffin, soft paraffin or squalane; cellulose derivatives such as methyl cellulose, ethyl cellulose, carboxymethylcellulose, sodium carboxymethylcellulose or hydroxylpropyl- methylcellulose; lower alkanols, for example ethanol or isopropanol; lower aralkanols; lower polyalkylene glycols or lower alkylene glycols, for example polyethylene glycol, polypropylene glycol, ethylene glycol, propylene glycol, 1,3- butylene glycol or glycerin; fatty acid esters such as isopropyl palmitate, isopropyl myristate or ethyl oleate; polyvinylpyrolidone; agar; gum tragacanth or gum acacia, and petroleum jelly. Typically, the carrier or carriers will form from about 10% to about 99.9% by weight of the composition, vaccine or medicament. [0105] For administration as an injectable solution or suspension, non-toxic parenterally acceptable diluents or carriers can include, Ringer’s solution, isotonic saline, phosphate buffered saline, ethanol and 1,2 propylene glycol. Methods for preparing parenterally administrable pharmaceutical compositions and medicaments are apparent to those of ordinary skill in the art, and are described in more detail in, for example, “Remington: The Science and Practice of Pharmacy”, Mack Publishing Co., 20th ed., 2000, and Gilman et al., (Eds), (1990), “Goodman And Gilman's: The Pharmacological Bases of Therapeutics”, Pergamon Press. [0106] For oral administration, some examples of suitable carriers, diluents, excipients and adjuvants include peanut oil, liquid paraffin, sodium carboxymethylcellulose, methylcellulose, sodium alginate, gum acacia, gum tragacanth, dextrose, sucrose, sorbitol, mannitol, gelatine and lecithin. In addition these oral formulations may contain suitable flavouring and colourings agents. When used in capsule form the capsules may be coated with compounds such as glyceryl monostearate or glyceryl stearate which delay disintegration. [0107] Solid forms for oral administration may contain binders acceptable in human and veterinary pharmaceutical practice, sweeteners, disintegrating agents, diluents, flavourings, coating agents, preservatives, lubricants and/or time delay agents. Suitable binders include gum acacia, gelatine, corn starch, gum tragacanth, sodium alginate, carboxymethylcellulose or polyethylene glycol. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, guar gum, xanthan gum, bentonite, alginic acid or agar. Suitable diluents include lactose, sorbitol, mannitol, dextrose, kaolin, cellulose, calcium carbonate, calcium silicate or dicalcium phosphate. Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate. [0108] Liquid forms for oral administration may contain, in addition to the above agents, a liquid carrier. Suitable liquid carriers include water, oils such as olive oil, peanut oil, sesame oil, sunflower oil, safflower oil, arachis oil, coconut oil, liquid paraffin, ethylene glycol, propylene glycol, polyethylene glycol, ethanol, propanol, isopropanol, glycerol, fatty alcohols, triglycerides or mixtures thereof. [0109] Suspensions for oral administration may further comprise dispersing agents and/or suspending agents. Suitable suspending agents include sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, poly-vinyl- pyrrolidone, sodium alginate or acetyl alcohol. Suitable dispersing agents include lecithin, polyoxyethylene esters of fatty acids such as stearic acid, polyoxyethylene sorbitol mono- or di-oleate, -stearate or- laurate, polyoxyethylene sorbitan mono- or di-oleate, -stearate or -laurate and the like. [0110] In certain embodiments, the pharmaceutical composition may comprise a liposome. A liposomal formulation suitable for bisantrene or a cardioprotective derivative thereof comprises small unilamellar or multilamellar liposomes of size range between 0.01 and 100 ^M, and between about 50-95% liposome-entrapped bisantrene, composed of hydrogenated soy phosphatidylcholine, distearoyl phosphatidylglycerol, and cholesterol of natural or synthetic origin lipids, in aqueous solution which can be reconstituted from a lyophilized form to an injectable liposome suspension. The composition is prepared by reconstituting a lyophilized bisantrene/liposome composition to a liposome concentrate, then diluting the concentrate for parenteral administration for the treatment of melanoma. [0111] In yet another embodiment, the pharmaceutical composition may comprise a complex with a beta-cyclodextrin. A liposomal formulation suitable for bisantrene, a therapeutically active derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or derivative thereof, comprises a complex formed in aqueous solution which may be reconstituted from a lyophilized form to an injectable suspension. One such composition is prepared by reconstituting a lyophilized bisantrene/beta- cyclodextrin composition to a concentrate, then diluting the concentrate for parenteral administration. Beta-cyclodextrin complexes and methods for preparing such complexes are known in the art and are described in., for example, WO 2019/073296 by Rothman. [0112] Various formulations suitable for use in the administration of bisantrene, a therapeutically active derivative of bisantrene, or a pharmaceutically acceptable salt of bisantrene or derivative thereof, are known in the art. United States Patent No. 4,784,845 to Desai et al. discloses a composition for delivery of a hydrophobic drug (i.e., bisantrene or a cardioprotective derivative thereof) comprising: (i) the hydrophobic drug; (ii) an oleaginous vehicle or oil phase that is substantially free of butylated hydroxyanisole (BHA) or butylated hydroxytoluene (BHT); (iii) a co- surfactant or emulsifier; (iv) a co-surfactant or auxiliary emulsifier; and (v) benzyl alcohol as a co-solvent. United States Patent No.4,816,247 by Desai et al. discloses a composition for delivery by intravenous, intramuscular, or intraarticular routes of hydrophobic drugs (such as bisantrene or a cardioprotective derivative or analog thereof) comprising: (i) the hydrophobic drug; (ii) a pharmaceutically acceptable oleaginous vehicle or oil selected from the group consisting of: (a) naturally occurring vegetable oils and (b) semisynthetic mono-, di-, and triglycerides, wherein the oleaginous vehicle or oil is free of BHT or BHA; (iii) a surfactant or emulsifier; (iv) a co-surfactant or emulsifier; (v) an ion-pair former selected from C6-C20 saturated or unsaturated aliphatic acids when the hydrophobic drug is basic and a pharmaceutically acceptable aromatic amine when the hydrophobic drug is acidic; and (vi) water. [0113] According to certain embodiments, compositions according to the present invention may be adapted for administration of bisantrene at a dosage of from about 0.1 mg/m2/day to about 100 mg/m2/day, such as from about 0.2 mg/m2/day to about 50 mg/m2/day, from about 0.5 mg/m2/day to about 20 mg/m2/day, from about 1.0 mg/m2/day to about 10 mg/m2/day, from about 1.0 mg/m2/day to about 8 mg/m2/day, about 1 mg/m2/day, about 2 mg/m2/day, about 3 mg/m2/day, about 4 mg/m2/day, about 5 mg/m2/day, about 6 mg/m2/day, about 7 mg/m2/day, or about 10 mg/m2/day. In some embodiments, compositions according to the present invention may be adapted for administration of bisantrene once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, over a period of, for example, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 21 days, 28 days, 35 days, 42 days, 49 days, 56 days or 63 days. In certain embodiments, compositions according to the present invention may be adapted for administration of bisantrene over a period of 28 days, optionally once or multiple times daily or weekly, once every two weeks, once every three weeks, once every four weeks, at a dosage of about 20-50 mg/m2/28 days. Compositions according to the present invention comprising pharmaceutically acceptable salts of bisantrene or therapeutically active derivatives or pharmaceutically acceptable salts thereof may be adapted to deliver similar dosage rates, adjusted for molar equivalence. [0114] Dosing regimens for immune checkpoint inhibitors will depend on the particular immune checkpoint inhibitor, and have been published for known immune checkpoint inhibitors, including for treatment of melanoma. In addition, suitable dosages of a given immune checkpoint inhibitor can be readily determined by one of ordinary skill in the art using known approaches and techniques as discussed above in relation to bisantrene. Compositions according to the invention may be formulated to include the immune checkpoint inhibitor(s) at their recommended dosage rates, or different dosage rates as discussed below. [0115] According to certain embodiments, and as a result of the beneficial biologic activity observed when bisantrene (or derivative of bisantrene, or a pharmaceutically acceptable salt of either) is administered with a immune checkpoint inhibitor, the dosage of immune checkpoint inhibitor in a composition according to the present invention may be lower than it would be when administered to a patient by itself. This is particularly important for active agents with deleterious side-effects, such as immune related adverse events. Thus, in certain embodiments of compositions according to the present invention, the immune checkpoint inhibitor may be included in the composition at a dosage which is significantly lower than the recommended dosage rate for that immune checkpoint inhibitor when administered alone, such as at a rate which is at least 10%, 20%, 30%, 40%, 50%, 60% or 70% lower than the dosage rate for that immune checkpoint inhibitor when administered alone. [0116] Similarly, where compositions according to the present invention include an additional therapeutic agent, being an immunomodulatory or a protein kinase inhibitor, that additional therapeutic agent may be included in the composition at its known recommended rate(s) for the treatment of melanoma, or even lower rates if synergy amongst the components is determined. Uses of compositions for the manufacture of medicaments for the treatment of melanoma [0117] The present invention also provides uses of compositions according to the invention for the manufacture of a medicament for the treatment of melanoma, especially immune checkpoint inhibitor resistant melanoma in a patient. [0118] The parameters for such manufacture, including immune checkpoint inhibitors, bisantrene, bisantrene derivatives and salts thereof, as well as immunomodulators, protein kinase inhibitors, or both as optional additional therapeutic agent(s), and dosages/dosing for all actives, dosage forms and other components are described above. Kits for the treatment of melanoma [0119] The present invention also provides kits for the treatment of melanoma, especially immune checkpoint resistant melanoma, the kit comprising at least one immune checkpoint inhibitor and at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof. [0120] The parameters for such kits, including immune checkpoint inhibitors, bisantrene, bisantrene derivatives and salts thereof, as well as immunomodulators, protein kinase inhibitors, or both as optional additional therapeutic agent(s), and dosages/dosing for all actives, dosage forms and other components are described above. [0121] In some cases, the kit may also comprise vials, tubes, needles, packaging, or other materials. [0122] Kits with unit doses of one or more of the active agents described herein, usually in injectable doses, are provided. Such kits may include containers containing unit doses, informational package inserts describing the use and attendant benefits of the drugs in treating the disease, and optionally appliance(s) or device(s) for delivery of the composition. [0123] The kit may further comprise any device suitable for administration of the composition. For example, a kit may comprise a needle suitable for intravenous administration. [0124] In some cases, kits may be provided with instructions. The instructions may be provided in the kit or they may be accessed electronically. The instructions may provide information on how to use the compositions of the present disclosure. The instructions may further provide information on how to use the devices of the present disclosure. The instructions may provide information on how to perform the methods of the disclosure. In some cases, the instructions may provide dosing information. The instructions may provide drug information such as the mechanism of action, the formulation of the drug, adverse risks, contraindications, and the like. In some cases, the kit is purchased by a physician or health care provider for administration at a clinic or hospital. In some cases, the kit is purchased by a laboratory and used for screening candidate compounds. [0125] Preferred forms of the present invention will now be described, by way of example only, with reference to the following examples, including comparative data, and which are not to be taken to be limiting to the scope or spirit of the invention in any way. EXAMPLES Example 1: Evaluation of Efficacy of Zantrene and RMP1-14 in the Subcutaneous B16-F10 Mouse Cancer Tumor Model in Female C57BL/6 Mice [0126] Two hundred and seventy female C57BL/6 mice were subcutaneously inoculated with B16-F10 mouse skin cancer cells. Six days post-inoculation, animals were randomised into six groups for efficacy assessment (20 per group) and six groups for flow cytometry analysis (10 per group). [0127] Antibody control rat IgG2a and RMP1-14 (each 10 mg/kg) were administered via intraperitoneal injection (i.p.) every three days. Vehicle (8% w/v Sulfobutyl-ether-β-Cyclodextrin (SBECD) and Zantrene (5 or 15 mg/kg) were administered via intravenous injection (i.v.) every second day. The high dose of Zantrene was reduced from 15 mg/kg to 10 mg/kg from Study Day 8 onwards due to adverse events at the higher dose. Combinations of rat IgG2a / Vehicle, RMP1- 14/Vehicle, rat IgG2a / 5 mg/kg Zantrene, rat IgG2a / 15 mg/kg Zantrene, RMP1-14 / 5 mg/kg Zantrene and RMP1-14 / 15 mg/kg Zantrene were administered. Ten doses of Vehicle (8% w/v SBECD) and Zantrene, and seven doses of rat IgG2a and RMP1- 14 were administered to efficacy groups. Seven doses of Vehicle (8% w/v SBECD) and Zantrene, and five doses of rat IgG2a and RMP1-14 were administered to flow groups. [0128] Animals were observed daily. Body weights, tumor volumes and clinical observations were recorded three times per week. Floor food was given to animals throughout the study. Flow study animals were euthanised on Study Day 7 or 13 (five per group at each time-point), unless earlier due to ethical limits or were found dead. Tumors and spleens were excised and weighed for flow cytometry analysis. Efficacy study animals were euthanised as ethical limits were reached or were found dead. Remaining animals were euthanised and the study was terminated on Study Day 20. [0129] The majority of animals treated with 10 mg/kg rat IgG2a / Vehicle (8% w/v SBECD) and 10 mg/kg RMP1-14 / Vehicle (8% w/v SBECD) were euthanised from Study Day 9 onwards as tumor burden reached the ethical limit. [0130] Assessment of tumor growth in efficacy study animals on Study Day 12 reveled smaller tumors in animals receiving treatments that included Zantrene as a component of the combination compared with 10 mg/kg rat IgG2a / Vehicle (8% w/v SBECD) control treatment and 10 mg/kg RMP1-14 / Vehicle (8% w/v SBECD). [0131] Enhanced inhibition of tumor growth was observed for the combination of Zantrene 5 mg/kg/ RMP1-1410 mg/kg compared to Zantrene 5 mg/kg/IgG2a isotype control 10 mg/kg at Day 14 in well-established tumor with a volume >170 mm3 by Day 5 following implantation. [0132] Spleen levels of CD4+ PD-1+ cells (PD-1+ as % of CD4+) on Study Day 7 were substantially higher in animals treated with RMP1-14 monotherapy compared with all other treatments, significantly (p≤0.05) [0133] Combination of Bisantrene 5 mg/kg with/RMP1-1410 mg/kg produced a 56% increase in the number of antitumor M1 macrophages relative to animals treated with RMP1-1410 mg/kg alone (p = 0.0189). [0134] Additionally, a 56% decrease in the number of protumorigenic M2b macrophages was found in tumors from mice treated Bisantrene 5 mg/kg with/RMP1-1410 mg/kg combination relative to RMP1-1410 mg/kg alone at Day 13 (p = 0.0264) [0135] Combination of Bisantrene 5 mg/kg with/RMP1-1410 mg/kg produced a 83% decrease in the number of CD8+ T-cells co-expressing immunosuppressive checkpoint protein CTLA4 relative to animals treated with RMP1-1410 mg/kg alone (p = 0.0279). ADVANTAGES OF THE INVENTION [0136] The present invention provides a new paradigm for treating of melanoma by the administration of bisantrene, an antineoplastic agent that has multiple mechanisms of action, including DNA intercalation, inhibition of topoisomerase, inhibition of the fat and obesity associated protein (FTO), and activation of the immune system. Bisantrene is well tolerated and, in particular, lacks the cardiotoxicity that is characteristic of some other anthracene derivatives. Bisantrene can be used together with other therapeutic agents that are used for the treatment of melanoma. [0137] The inventions illustratively described herein can suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” “containing,” etc. shall be read expansively and without limitation. As used herein the use of the term “comprising” as a transitional phrase in claims is intended to include therein the use of the transitional phrases “consisting essentially of” or “consisting of” if the narrower transitional phrases are not expressly excluded. Although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the inventions herein disclosed can be resorted by those skilled in the art, and that such modifications and variations are considered to be within the scope of the inventions disclosed herein. The inventions have been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the scope of the generic disclosure also form part of these inventions. This includes the generic description of each invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised materials specifically resided therein.

Claims

Claims:
1 . A method of treating a patient with melanoma, said method comprising administering to said patient a therapeutically effective amount of an immune checkpoint inhibitor and a therapeutically effective amount of at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
2. The method of claim 1 wherein said immune checkpoint inhibitor inhibits: PD-1 ; PD-L1 ; CTLA-4; LAG3 or a combination of at least any two of these targets.
3. The method of claim 1 , wherein the at least one immune checkpoint inhibitor is selected from the group consisting of:
(a) ipilimumab
(b) nivolumab
(c) nivolumab-relatlimab
(d) pembrolizumab
(e) cemiplimab
(f) dostarlimab
4. The method of claim 1 , wherein said at least one immune checkpoint inhibitor is selected from the group consisting of:
(a) nivolumab
(b) pembrolizumab
5. The method of any one of claims 1 to 4, wherein said second agent is bisantrene or a pharmaceutically acceptable salt thereof.
6. The method of any one of claims 1 to 5 which comprises administering to said patient a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof.
7. The method of any one of claims 1 to 6 which comprises treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma.
8. The method of any one of claims 1 to 7, further comprising administration of at least one additional therapeutic agent for the treatment of melanoma.
9. The method of claim 8, wherein the additional therapeutic agent comprises an immunomodulator, protein kinase inhibitor or a combination of immunomodulatory(s) and protein kinase inhibitor(s).
10. The method of claim 9, wherein the at least one additional therapeutic agent is selected from the group consisting of:
(a) binimetinib
(b) cobimetinib
(c) dabrafenib
(d) encorafenib
(e) trametinib
(f) vemurafenib
(g) interleukin-2; and
(h) proleukin.
11. The method of any one of claims 1 to 10 comprising administering said at least one immune checkpoint inhibitor to said patient prior to, simultaneously with, or after administration of said second agent to said patient.
12. The method of any one of claims 1 to 10, wherein said at least one immune checkpoint inhibitor and the second agent are administered to said patient at the same time, optionally in a single composition.
13. The method of any one of claims 1 to 12, wherein the dose of said at least one immune checkpoint inhibitor is at least 20% lower than the dose required of said at least one immune checkpoint inhibitor agent when administered without said second agent to achieve the same targeted outcome.
14. A pharmaceutical composition for the treatment of melanoma comprising at least one immune checkpoint inhibitor and a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
15. The composition of claim 14, wherein said immune checkpoint inhibitor inhibits one or more of: PD-1 ; PD-L1 ; CTLA-4; LAG3 or a combination of at least any two of these targets.
16. The composition of claim 14, wherein said at least one immune checkpoint inhibitor is selected from the group consisting of:
(a) ipilimumab
(b) nivolumab
(c) nivolumab-relatlimab
(d) pembrolizumab
(e) cemiplimab
(f) dostarlimab
17. The composition of claim 14, wherein said at least one immune checkpoint inhibitor is selected from the group consisting of:
(a) nivolumab
(b) pembrolizumab
18. The composition of any one of claims 14 to 17, further comprising at least one additional therapeutic agent for the treatment of melanoma.
19. The composition of claim 18, wherein the additional therapeutic agent comprises an immunomodulator, protein kinase inhibitor or a combination of immunomodulator(s) and protein kinase inhibitor(s).
20. The composition of claim 19, wherein the at least one additional therapeutic agent is selected from the group consisting of:
(a) binimetinib
(b) cobimetinib
(c) dabrafenib
(d) encorafenib
(e) trametinib
(f) vemurafenib
(g) interleukin-2; and
(h) proleukin.
21. The composition of any one of claims 14 to 20, wherein said second agent is bisantrene or a pharmaceutically acceptable salt thereof.
22. The composition of any one of claims 14 to 20 which comprises a therapeutically effective amount of one immune checkpoint inhibitor and a therapeutically effective amount of bisantrene or a pharmaceutically acceptable salt thereof.
23. The composition of any one of claims 14 to 22 for the treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma.
24. Use of a composition according to any one of claims 14 to 23 for the manufacture of a medicament for the treatment of melanoma in a patient.
25. The use of claim 24, wherein said medicament is for the treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma.
26. The use of claim 24 or claim 25, wherein said medicament comprises one immune checkpoint inhibitor and bisantrene or a pharmaceutically acceptable salt thereof.
27. A kit for the treatment of melanoma, said kit comprising at least one immune checkpoint inhibitor and at least a second agent comprising bisantrene or a derivative thereof, or a pharmaceutically acceptable salt of bisantrene or derivative thereof.
28. The kit of claim 27, wherein said immune checkpoint inhibitor inhibits one or more of: PD-1 ; PD-L1 ; CTLA-4; LAG3 or a combination of at least any two of these targets.
29. The kit of claim 27, wherein said at least one immune checkpoint inhibitor is selected from the group consisting of:
(a) ipilimumab
(b) nivolumab
(c) nivolumab-relatlimab
(d) pembrolizumab
(e) cemiplimab
(f) dostarlimab
30. The kit of any one of claims 27 to 29, wherein said second agent is bisantrene or a pharmaceutically acceptable salt thereof.
31 . The kit of any one of claims 27 to 29 which comprises one immune checkpoint inhibitor and bisantrene or a pharmaceutically acceptable salt thereof.
32. The kit of any one of claims 27 to 31 , further comprising at least one additional therapeutic agent for the treatment of melanoma, optionally wherein said additional therapeutic agent comprises an immunomodulator, protein kinase inhibitor or a combination of immunomodulator(s) and protein kinase inhibitor(s).
33. The kit of any one of claims 27 to 32 for the treatment of melanoma, optionally immune checkpoint inhibitor resistant melanoma.
PCT/AU2023/050566 2022-06-22 2023-06-22 Treatment of melanoma WO2023245248A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AU2022901702 2022-06-22
AU2022901702A AU2022901702A0 (en) 2022-06-22 Treatment of melanoma
AU2022901791A AU2022901791A0 (en) 2022-06-27 Treatment of melanoma
AU2022901791 2022-06-27

Publications (1)

Publication Number Publication Date
WO2023245248A1 true WO2023245248A1 (en) 2023-12-28

Family

ID=89378747

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/AU2023/050569 WO2023245249A1 (en) 2022-06-22 2023-06-22 Treatment of melanoma
PCT/AU2023/050566 WO2023245248A1 (en) 2022-06-22 2023-06-22 Treatment of melanoma

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/AU2023/050569 WO2023245249A1 (en) 2022-06-22 2023-06-22 Treatment of melanoma

Country Status (1)

Country Link
WO (2) WO2023245249A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015013579A1 (en) * 2013-07-26 2015-01-29 Update Pharma Inc. Compositions to improve the therapeutic benefit of bisantrene
WO2019073296A1 (en) * 2017-10-13 2019-04-18 Race Oncology Ltd. Liposomal formulations of bisantrene or derivatives or analogs thereof
WO2020072948A1 (en) * 2018-10-04 2020-04-09 Race Oncology Ltd. Method for preparing and delivering bisantrene formulations
WO2020208612A1 (en) * 2019-04-12 2020-10-15 Vascular Biogenics Ltd. Methods of anti-tumor therapy

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015013579A1 (en) * 2013-07-26 2015-01-29 Update Pharma Inc. Compositions to improve the therapeutic benefit of bisantrene
WO2019073296A1 (en) * 2017-10-13 2019-04-18 Race Oncology Ltd. Liposomal formulations of bisantrene or derivatives or analogs thereof
WO2020072948A1 (en) * 2018-10-04 2020-04-09 Race Oncology Ltd. Method for preparing and delivering bisantrene formulations
WO2020208612A1 (en) * 2019-04-12 2020-10-15 Vascular Biogenics Ltd. Methods of anti-tumor therapy

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Melanoma Treatment (PDQ®)-Patient Version", NATIONAL CANCER INSTITUTE, 1 March 2021 (2021-03-01), XP093123815, Retrieved from the Internet <URL:https://www.cancer.gov/types/skin/patient/melanoma-treatment-pdq> [retrieved on 20240125] *
TILLETT DANIEL: "Race Initiates FTO Melanoma Preclinical Study", RACE ONCOLOGY ASX ANNOUNCEMENT, 19 March 2021 (2021-03-19), XP093123813, Retrieved from the Internet <URL:https://raceoncology.com/race-initiates-fto-melanoma-preclinical-study/> [retrieved on 20240125] *
TILLETT DANIEL: "Zantrene Kills Melanoma Cancer Cells That Overproduce FTO", RACE ONCOLOGY ASXRELEASE, 30 September 2021 (2021-09-30), XP093123811, Retrieved from the Internet <URL:https://raceoncology.com/zantrene-kills-melanoma-cancer-cells-that-overproduce-fto/> [retrieved on 20240125] *

Also Published As

Publication number Publication date
WO2023245249A1 (en) 2023-12-28

Similar Documents

Publication Publication Date Title
EP2127652B1 (en) Method for treating cancer using anticancer agent in combination
JP6426706B2 (en) GLA monotherapy for use in cancer treatment
US20210077625A1 (en) Ray tracing technique for wireless channel measurements
KR101361445B1 (en) Composition comprising peptide, 5-fluorouracil, and mature dendritic cells for cancer treatment
Foletto et al. Cutaneous melanoma: new advances in treatment
EP2782994B1 (en) Small molecule enhancer for dendritic cell cancer vaccines
WO2021012886A1 (en) Anti-neoplastic combined pharmaceutical composition and application thereof
CN112587666A (en) Combination of a small molecule inhibitor of the PD-1/PD-L1 interaction and an anti-PD-1 antibody for the treatment of cancer
KR102358632B1 (en) Composition for preventing or treating colon cancer comprising streptonigrin and anticancer agent
JP2009539916A (en) Use of thymosin alpha 1 for the manufacture of a medicament for the treatment of stage IV malignant melanoma
WO2023245248A1 (en) Treatment of melanoma
Aschhoff Retrospective study of Ukrain treatment in 203 patients with advanced-stage tumors
CN112472803A (en) Application of combination of anti-PD-1 antibody and anti-CTLA-4 antibody in preparation of medicine for treating kidney cancer
US20210386736A1 (en) Combination therapy for treating cancer
US20230099637A1 (en) Palladium Hyaluronic Acid Particles and Methods of Managing Cancer or Angiogenic Conditions
US20210315898A1 (en) Combination therapy for treating cancer
WO2023168491A1 (en) Treatment of clear cell renal cell carcinoma
CN114732816B (en) Application of 2-aryl-1, 3-dihydrobenzimidazole derivative in preparation of medicines for treating kidney cancer
CN110740747A (en) NK-92 cell and IL-15 agonist combination therapy
US20230233474A1 (en) Use of mrnas encoding ox40l, il-23 and il-36gamma for treating cancer
JP2022551672A (en) breast cancer treatment
TW201513871A (en) Anti-CLUSTERIN monotherapy for cancer treatment
US20020123480A1 (en) Methods and compositions utilizing 2&#39;-5&#39; oligoadenylates in the treatment of disorders of cell proliferation
WO2004073719A1 (en) A combined therapy comprising an indolopyrrolocarbazole derivative and another antitumor agent

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: 23825696

Country of ref document: EP

Kind code of ref document: A1