WO2017200492A1 - Method of treating cancer - Google Patents

Method of treating cancer Download PDF

Info

Publication number
WO2017200492A1
WO2017200492A1 PCT/SG2017/050263 SG2017050263W WO2017200492A1 WO 2017200492 A1 WO2017200492 A1 WO 2017200492A1 SG 2017050263 W SG2017050263 W SG 2017050263W WO 2017200492 A1 WO2017200492 A1 WO 2017200492A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
virus
cells
cell
therapy
Prior art date
Application number
PCT/SG2017/050263
Other languages
French (fr)
Inventor
Bertil Lindmark
Original Assignee
Aslan Pharmaceuticals Pte Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aslan Pharmaceuticals Pte Ltd filed Critical Aslan Pharmaceuticals Pte Ltd
Publication of WO2017200492A1 publication Critical patent/WO2017200492A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2086IL-13 to IL-16
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present disclosure relates to a method of treating cancer, use of medicaments in a staged combination therapy for the treatment of cancer (also referred to herein as multimodal cancer therapy), and use of a medicament or medicaments in the manufacture of a multimodal therapy for the treatment of cancer, for example linked with dynamic input of tumor status, to optimise intervention and timing.
  • the treatment of cancer is still a huge challenge and burden to society.
  • the present inventor believes that the effectiveness of cancer therapy can be greatly increased by employing a multimodal method of treatment. That is conscious choice of sequence and the timing of multiple treatments to achieve improved outcomes, for patients.
  • Cancer cells are highly adapted to corrupting and hijacking many natural biological processes that exist in the body. Cancers are sometimes based on mutations and modifications of cells and thus can readily change and adapt. Given this the present inventor believes that is unlikely that one or two modes of treatment will be effective for the treatment of the majority of cancers.
  • the inventor has designed a new method to treat cancer, which involves a radical resetting of the immune system, which will be much more efficacious than just using simple steps, such as immune check point inhibitors or combinations thereof.
  • the method of present disclosure can be considered a "reboot" of the immune system, which after treatment is then able to fight the cancer.
  • the present disclosure provides a method of treating cancer comprising the steps;
  • the treatment schedule of the present disclosure is a paradigm shift in cancer treatment, because at the present time standard of care cancer treatment generally only involves one or two of the above steps.
  • cancer therapy regimes are a combination or cocktail of the therapeutic agents from the outset, and cancer adjuvant therapy is known (i.e. chemotherapy or radiation therapy after surgery to excise a tumor) subsequent cancer therapies are not employed until the first line or second therapy has failed.
  • cancer adjuvant therapy i.e. chemotherapy or radiation therapy after surgery to excise a tumor
  • the multimodal therapy of the present disclosure is not the same as simply trying different treatments after the failure of a treatment. Instead it involves pro- actively employing multiples modes of treatment, in particular in the order specified herein.
  • the method comprises the further step of tumour surveillance.
  • the therapeutic steps are taken in fast succession, in particular while surveying and reading "efficacy", for example by studying effects on immune parameters and/or variables showing circulating tumor components.
  • Efficacy here is not employed in the strict technical sense but simply to effects more generically.
  • the present multimodal treatment comprises multiple treatment steps, which are employed at specific times, for example over a period of 1 to 12 months, such as 1 to 2 months, 1 to 3 months, 1 to 4 months, 1 to 5 months, 1 to 6 months, 1 to 7 months, 1 to 8 months, 1 to 9 months, 1 to 10 months or 1 to 11 months.
  • the "subsequent" therapies are not employed because a therapy has failed i.e. the further therapy is not started because there is resistance to "first line or second line” therapy.
  • step b) follows step a) or step al).
  • step c) follows step b).
  • step d) follows step c).
  • step e) follows step d).
  • one or more treatment steps may overlap or be at the same time.
  • step a) and al) overlap or are at the same time. In one embodiment step a) and b) overlap or are at the same time. In one embodiment step al) and b) overlap or are at the same time. In one embodiment step b) and c) overlap or are at the same time. In one embodiment step c) and d) overlap or are at the same time. In one embodiment step d) and e) overlap or are at the same time.
  • step a), al) and b) overlap or are at the same time. In one embodiment step a), b) and c) overlap or are at the same time. In one embodiment step al), b) and c) overlap or are at the same time. In one embodiment step b), c) and d) overlap or are at the same time. In one embodiment step c), d) and e) overlap or are at the same time.
  • step a), al), b) and c) overlap or are at the same time. In one embodiment step a), b), c) and d) overlap or are at the same time. In one embodiment step al), b), c) and d) overlap or are at the same time. In one embodiment step b), c), d) and e) overlap or are at the same time.
  • step a), b), c), d) and e) overlap or are at the same time.
  • step al), b), c), d) and e) overlap or are at the same time
  • one or more of the steps do not overlap, for example 2, 3, 4 or all of the step do not overlap.
  • step b) and d) do not overlap and are not at the same time.
  • step e) does not overlap with step c).
  • the method comprises a pre-step of surgery, for example to remove tumour mass and/or lymph nodes and the like.
  • the method comprises irradiation.
  • step a) is a treatment selected from the group comprising irradiation, chemotherapy, anti-proliferation pathway therapy (for example anti- oestrogen, anti-androgen, anti-HER, anti-BCR/ABL, anti-VEGF, anti-PDGF, anti-BRAF antibody molecule [including a bispecific antibody, full-length or an antibody binding fragment], with or without combining with the appropriate chemotherapy) and a combination of two or more of the same.
  • anti-proliferation pathway therapy for example anti- oestrogen, anti-androgen, anti-HER, anti-BCR/ABL, anti-VEGF, anti-PDGF, anti-BRAF antibody molecule [including a bispecific antibody, full-length or an antibody binding fragment], with or without combining with the appropriate chemotherapy
  • At least one agent employed in step a) is an antiproliferative agent, for example selected from the group 25-hydroxcholesterol, 3-hydroxtyrosol, 7 ⁇ - hydroxycholestrol, aloe-emodin, apigenin, artesunate, berberine, caffeine power (C0750), dichloromethylenediphosphonic acid (for example as a salt, such as a disodium salt), emodin, HA 14-1, hyperforin, N-acetyl-D-sphingosine, N-hexanoyl-D-sphingosine, oleuropein, pathenolide, tangeretin, tanshinone IIA, ⁇ -cryptoxanthine, ⁇ -cryptoxanthin, ⁇ -lonone, trans-cinnamaldehyde and combinations thereof.
  • an antiproliferative agent for example selected from the group 25-hydroxcholesterol, 3-hydroxtyrosol
  • agents that can be employed alone or in a combination in step a) include high doses of vitamin Bl, parp inhibitors, a gene product of NME2, myrtillin (delphinidin-3-O-glucoside), tulipanin (delphinidin-3-O-rutinoside), violdelphin (delphinidin 3-rutinoside-7-0-(6-0-(4-(6-0-(4-hydroxybenzoyl) -p-D-glucosyl)oxy benzoyiJ-p-D-glucoside), nasunin (delphinidin-3-(p-coumaroylrutinoside)-5-glucoside) and combinations of two or more of the same.
  • therapy employed in step a) is Varlitinib [(i?)-N4-[3-Chloro- 4-(thiazol-2-ylmethoxy)-phenyl]-N6-(4-methyl-4, 5,-dihydro-oxazol-2-yl)-quinazoline- 4,6-diamine] or a pharmaceutically acceptable salt thereof, which may be employed alone or in a combination therapy.
  • the Varlitinib is administered at a dose in the range lOOmg to 900mg on each occasion, in particular 200 mg, 300mg, 400mg or 500mg each dose, such as 400mg, for example administered once or twice daily, such as twice daily.
  • the therapy of step a) comprises a DHODH inhibitor, such as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
  • a DHODH inhibitor such as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
  • CEP-9722 a PARP-1 and PARP-2 inhibitor
  • INO-1001 (3-aminobenzamide) a potent inhibitor of PARP (with IC50 of ⁇ 50 nM in CHO cells and a mediator of oxidant- induced myocyte dysfunction during reperfusion)
  • BGB-290 a PARP-1 and PARP-2 inhibitor BGP-15 a PARP inhibitor (which has been (structure not shown); shown to protect against ischemia- reperfusion injury);
  • E7016 (previously known as GPI-21016) a PARP inhibitor (undergoing Phase I trials in combination with temozolomide for advanced solid tumours and gliomas);
  • ABT-737 a PARP-1 and PARP-2 inhibitor
  • the therapy in step al), i.e. the decluttering of immune cells is performed by administering a compound, such as a CXCR2 antagonist, an anti-GCF, cyclophosphamide and/or irradiation, and a combination of two or more of the same.
  • a compound such as a CXCR2 antagonist, an anti-GCF, cyclophosphamide and/or irradiation, and a combination of two or more of the same.
  • decluttering refers to neutralisation of immune cells. This neutralisation may, for example include reduction in number of immune cells. Generally speaking, decluttering results in a reduction in overall number of immune cells, but may also encompass total depletion, including local and systemic depletion.
  • decluttering of immune cells is performed by removing specific immune cell populations, for example using medication and/or techniques such as a dialysis.
  • the immune cells that are "decluttered" are selected from a lymphocyte (such as a T lymphocyte, such as a T cell or a NKT cell), a B cell, a natural killer cell and a combination of two or more of the same.
  • a lymphocyte such as a T lymphocyte, such as a T cell or a NKT cell
  • B cell such as a B cell
  • natural killer cell such as a B cell
  • a combination of two or more of the same are selected from a lymphocyte (such as a T lymphocyte, such as a T cell or a NKT cell), a B cell, a natural killer cell and a combination of two or more of the same.
  • the immune cells which are decluttered are tumor associated macrophages.
  • Tumor associated macrophages can be decluttered by NKT cells engineered to encode a cytokine, for example as disclosed in WO2013/040371, incorporated herein by reference.
  • the immune cell which is decluttered is a lymphocyte, for example a NK cell or a T lymphocyte, such as a T cell or a NKT cell.
  • a lymphocyte for example a NK cell or a T lymphocyte, such as a T cell or a NKT cell.
  • the immune cells that are decluttered are B cells.
  • the immune cell that is decluttered is a natural killer cell.
  • NK cells can be targeted by monoclonal antibodies or binding fragments specific to CD56.
  • Mature T lymphocytes can be removed by medicaments such as alemtuzumab.
  • Immune cells with surface expression of CD52 can be removed by Campath.
  • the CXCR2 antagonist is selected from an antibody or binding fragment thereof, MK-7123, SB265610, SB 225002:
  • GSK1325756 GSK1325756, AZD5069, and combinations thereof.
  • the CXCR2 antagonist is SCH-527123.
  • the decluttering step al) is performed by employing anti- neutrophil antibodies.
  • the step al) is performed using a G-CSF antibody, for example CSL324, which is a humanized G-CSFR antibody.
  • the debulking in step b) is surgery to reduce a tumor mass.
  • the surgery is followed by chemotherapy and/or (in particular or) irradiation therapy.
  • Debulking refers to a reduction in the total mass of a tumor. This includes breaking a tumor mass into smaller fragments as well as shrinking the tumor mass. Methods of reducing a tumor mass include cutting (excising), ablating (including cryoablation, liquid nitrogen treatment of topical cancer cells and combinations of two or more of the same.
  • the debulking in step b) is performed by administering chemotherapy selected from alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents, such as doxorubicin, 5-fluorouracil (5-FU), paclitaxel, gemcitabine, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
  • chemotherapy selected from alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents, such as doxorubicin, 5-fluorouracil (5-FU), paclitaxel, gemcitabine, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
  • the debulking in step b) is selected from treatment with an oncolytic virus, an antibody drug conjugate and a combination thereof.
  • the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus.
  • the oncolytic virus is selected from Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep-hIL12, CavatakTM, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, Toca 511.
  • the antibody drug conjugate is selected from the group comprising AGS-15E, AGS-16C3F, AGS-22M6E, AMG-172, AMG-595, ASG-5ME, brentuximab vedotin, DEDN-6526A, DMUC-5754A, gemtuzumab ozogamicin, glembatumumab vedotin, GSK2857916, hRS7-SN-38, IMGN-242, IMGN-289, IMGN-388, IMGN-529, IMGN-633, IMGN-853, Indatuximab, indatuximab ravtansine, Inotuzumab ozogamicin, lorvotuzumab mertansine, MEDI-547, milatuzumab doxorubicin, MLN- 0264, MLN-2704, PF-06263507, pinatuzumab vedo
  • step b) comprises administering varlitinib or a salt thereof, as a monotherapy or as part of a combination therapy, for example as disclosed elsewhere herein.
  • step b) comprises administering a DHODH inhibitor, such as as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
  • a DHODH inhibitor such as as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
  • activating the immune cells in step c) comprises administering a RON inhibitor, an AXL kinase inhibitor, a MER kinase agonist, a cytokine (such as IL-13, IL-15, IL-4 or IL-2), a tumour vaccine for example comprising a tumour antigen and adjuvant, a dendritic cell vaccine and a combination of two or more of the same.
  • a RON inhibitor an AXL kinase inhibitor
  • a MER kinase agonist such as IL-13, IL-15, IL-4 or IL-2
  • a cytokine such as IL-13, IL-15, IL-4 or IL-2
  • a tumour vaccine for example comprising a tumour antigen and adjuvant, a dendritic cell vaccine and a combination of two or more of the same.
  • activating immune cells refers to the functional activation of the immune cells, for example the activation of T cells by stimilulation with antigens (including where the antigen is provided on antigen presenting cells), and the activation of B cells to produce tumor specific antibodies. Activating may also include the recruitment of immune cells to the tumor site. Use of appropriate cytokines can stimulate and/or activate immune cells.
  • the RON inhibitor is N-(4-(2-Amino-3-chloropyridin-4- yloxy)-3-fluorophenyl)-4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3- carboxamide disclosed in WO 2008/058229.
  • activating the immune cells in step c) comprises administering one or more check-point inhibitors.
  • V155991 and V158411 are CHKl and CHK2 inhibitors,:
  • MK-8776 is a selective CHKl inhibitor (with CHIR124 is CHKl inhibitor (is a novel an IC50 of 3 nM in a cell-free assay.
  • the and potent inhibitor with IC50 of 0.3 molecule shows 500-fold selectivity against nM in a cell-free assay. It shows 2,000- CHK2); fold selectivity against CHK2 and 500 to
  • GDC-0425 is a selective CHKl a natural product derived from sponges that inhibitor;
  • KU55933 is a potent and selective KU59403 is an ATM inhibitor; ATM inhibitor vis-a-vie DNA-PK
  • VE-822 is a selective ATR inhibitor
  • NU6027 is a potent ATR inhibitor; with a Ki of 0.2 nM and an about 150
  • PD-321852 is a CHKl and WEEl PD-407824 is a CHKl and WEEl inhibitor;
  • PV1019 is a CHK2 inhibitor
  • CCT241533 is a CHK2 inhibitor with about 63
  • the check-point inhibitor is a PD-1 or PD-L1 inhibitor.
  • the checkpoint inhibitor is a combination of 2 or several immune checkpoint inhibitors such as PD-1 plus CTL4, or PD-1 plus LAG3 (or TIM3 or GITR).
  • step d) i.e. increasing antigen presentation and/or enhancing T cell response, comprises administering a therapy selected from the group comprising a T cell therapy (for example where T cells are activated in vitro to generate CTLs specific to an antigen), a cell therapy comprising engineered chimeric receptors, a PD1 inhibitor, a PD-L1 inhibitors, a CTLA4 inhibitor, a LAG3 inhibitor and a combination of two or more of the same.
  • a therapy selected from the group comprising a T cell therapy (for example where T cells are activated in vitro to generate CTLs specific to an antigen), a cell therapy comprising engineered chimeric receptors, a PD1 inhibitor, a PD-L1 inhibitors, a CTLA4 inhibitor, a LAG3 inhibitor and a combination of two or more of the same.
  • Increasing antigen presentation refers to the enhancement of the presentation of antigens to antigen presenting cells, for example by using a combination of treatments to make tumors necrotic, thereby providing a lot of potential antigenic material for presentation by antigen presenting cells.
  • Other examples of increasing antigen presentation include enhancing the ability of dendritic cells and macrophages to phagocytose tumor and present tumor antigen; and blocking or inhibiting do-not-eat- me signals (CD47) on tumour cells that prevent phagocytosis of tumour cells.
  • onclotytic viruses carrying a transgene can be used to infect the cancer cell and express the "protein or peptide" encoded on the cancer cells surface to increase antigen expression.
  • Antigen in this context simply means a target for therapy or immune cells, in particular a target for immunotherapy.
  • step d) comprises administering a therapy selected from the group comprising an oncolytic virus, a DLL inhibitor, ADH-1 molecular targeting therapy and combinations of two or more of the same
  • the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus, such as Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep-hIL12, CavatakTM, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, or Toca 511.
  • the oncolytic virus is a virus selected from an a
  • Residual tumor cells refers to tumor cells that remain after the previous therapy, for exampleat the primary tumor site or in the vicinity, including circulating tumor cells, dormant cancer cells, cancer stems cells and combinations of the two or more of the sameThe goal of this step is to eradicate any remaining traces of cancer cells and minimise recurrence of the cancer.
  • step e), i.e. clearing residual tumor cells comprises targeting cancer stem cells.
  • the cancer stem cell therapy comprises targeting one or surface markers, for example with one or more monoclonal antibodies.
  • the marker is selected from CD133, CD44, CD24, ESA, CD34 + CD38, and CD47.
  • step e) comprises an agent, such as an antibody or binding fragment thereof, specific to TRAIL or NF-aB.
  • step e) comprises an agent, such as an antibody or binding fragment thereof, specific to a cell marker Epcam, CD 133, CD44, CD24, ESA, CD34 + /CD38- and CD47.
  • an agent such as an antibody or binding fragment thereof, specific to a cell marker Epcam, CD 133, CD44, CD24, ESA, CD34 + /CD38- and CD47.
  • the cell markers are targeted by a chimeric antigen receptor.
  • antibody or antibody binding fragment is in the format of an antibody drug conjugate.
  • Multimodal or stage cancer therapy refers to discrete phases of therapy, for example each phase lasting in the region of 1 week to 6 months, such as 2 weeks to 1 month, 2 weeks to 2 months, 2 weeks to 3 months, 2 weeks to 4 months or 2 weeks to 5 months.
  • the therapies may overlap but generally will not simply be a combination therapy.
  • Combination therapy as employed herein is two or more active agents both administered over the same period of treatment.
  • Neutrophils are key for angiogenesis, which turns cancers to fast growth via the so called angiogenic shift (7)
  • TAMs Tumor associated Macrophages
  • Macrophage type 2 is the most prevalent TAM and is not effective in antigen presentation (11)
  • Dendritic cells capture antigens from tumour and migrate into the draining lymph nodes for presentation.
  • the antigens can be presented with MHC class I and class II molecules.
  • Tumours interfere with DC maturation through IL-10 and TSLP.
  • the DCs are skewed towards a Th2 lineage which promotes tumour development.
  • pDCs are also inhibited due to ligation of ILT7 with BST2 on tumour cells.
  • the inhibited pDCs induce CD4+ T cells to differentiate to become immunosuppressive.
  • GM-CSF in combination with other chemokines, can induce differentiation of inflammatory DCs that in turn will activate T cell functions.
  • chemokines can induce differentiation of inflammatory DCs that in turn will activate T cell functions.
  • Antibody therapy for engagement of surface molecules on DCs such as dectinl, DCSIGN/CD209 and CD40 putatively polarize DCs towards the proinflammatory phenotype. (19, 20, 21)
  • a specific example of cancer metabolism inhibitor would be DHODH inhibitors that inhibit de novo synthesis of pyrimidines. This inhibition will specifically inhibit rapidly dividing cells. DHODH inhibition will also lead to induction of p53 and resulting apoptosis once a threshold of p53 levels is breached. (39)
  • Neo-antigen presentation can also be greatly enhanced by blocking or inhibiting do-not-eat-me signals (CD47) on tumour cells that prevent phagocytosis of tumour cells.
  • ICI immune checkpoint inhibitor
  • ICIs Single or combinations or sequences of ICIs can be used and have shown additive effects in the clinic (25) b.
  • These ICI therapies are not limited to T cell mediators and may also affect DCs, NK, NKT cells. They are a combination of antagonists to inhibitory signals and agonists to activation signals on the immune cells. (25)
  • CTC Circulating tumour cells
  • CSC Cancer Stem Cells
  • DCC dormant cancer cells
  • CSCs The phenotype of CSCs will differ depending on the cellular context.
  • therapies against CSC have also shown efficacy, in reducing tumour recurrence and demonstrating long-term efficacy. (29, 30, 31, 32)
  • Tumors signal via exosomes in the circulation to seeding areas about status and to steer grafting probabilities (34)
  • Thrombocytes carry RNA and other tumor markers that tell status in tumor (35)
  • Tumors recruit a number of cells and they are either supporting cells, or just bystanders, which are likely to physically block access to the cancer. It may also be that these supporting or bystander cells, feed of fuel the cancer cell growth. Therefore we hypothesise therapy should focus on removing these before shifting to changing the macrophage and dendritic cell infiltration.
  • the immunostimulation by the means of making antigens available en masse, to the macrophages and the dendritic cells, should occur after the macrophages and the dendritic cells have been activated, and should precede application of the immune checkpoint inhibitors.
  • Killing circulating tumor cells and cancer stem cells and dormant cancer cells is described as the last step. However this step can be applied at initiation and/or during the subsequent steps, and may be repeated together with immune reactivation at subsequent sessions.
  • the timely feedback of the status of the tumor is important, since this multi-stage immuno, pathway and tumor escape/survival directed therapy needs to be steered and directed.
  • daily measures of anti-tumour activity may enable one to be able to direct the therapy better (i.e. understanding when the neutrophils are down and depleted, and when the myeloid derived suppressor cells have been turned off, and when the macrophages have turned to Ml phenotype and how the immune checkpoint inhibition is successful in attacking the tumor).
  • the detection of circulating tumor cells and RNA and DNA products from the tumor provides an indication whether the strategies to kill cancer stem cells have been successful.
  • the method is different from other methods in that it foresees usage of key therapies for a limited period of time, for a therapy period of less than 2 months is envisioned for one or more stages. This therapy in parts or in full may need to be repeated at a later stage, potentially with changes that may take instruction from the ongoing immune and cancer diagnostics.
  • a 2 week period of proliferation and nutrient/metabolic inhibition may be sufficient before taking down neutrophils and debulking of tumor.
  • This step may take a week, followed by macrophage/dendritic cell type change (3-7 days), and then turning on the antigen production and presentation (1 to 2 weeks) before turning on the immune defense with T-cells and NK cells via the immune checkpoint inhibitors for at least 2 weeks.
  • the killing of circulating and dormant cancer cells should be started, along with attempts to eradicate cancer stem cells.
  • This new method represents a way to declare total war against the cancer and is believed to cause maximal effect in the shortest period of time, while keeping side effects to a minimum. It also is a new way to try to learn as much possible about the specific cancer and to be able to adapt therapy to achieve best effect and avoid side effects.
  • the method should be meaningful both in early cancer (e.g. neo-adjuvant setting pre surgery) as well as a treatment alternative later in the cancer development.
  • Treatment of cancer refers to administration of the combination therapy of the present disclosure for the purpose of reducing symptoms, ameliorating symptoms, slowing progressing (including spreading) of the cancer or a form thereof (such as metastatic cancer), stopping progression (including spreading) of the cancer or a form thereof (such as metastatic cancer), shrinking tumors, reducing metastases, reducing or eliminating certain populations of cancer cells, for example cancer stem cell populations, sending the cancer into remission, maintaining the cancer in remission, prophylactic treatment of cancer, curing cancer or a combination of one or more the same, as appropriate.
  • Treatment of cancer as employed herein also refers to administration of the combination therapy of the present disclosure for the purpose of enhancing concomitant anti-cancer therapy, and for the purpose (at least in part) to reduce side effects and/or reducing the dose required of concomitant anti-cancer therapy.
  • Lithium compound as employed herein refers to an active agent employed in therapy comprising the alkali metal lithium, for example lithium salts comprising a counter-ion such as selected from the group comprising aspartate, bromide, chloride, citrate, lithium carbonate, lithium sulfate and orotate.
  • a counter-ion such as selected from the group comprising aspartate, bromide, chloride, citrate, lithium carbonate, lithium sulfate and orotate.
  • Lithium salts are well established treatments for mood disorders.
  • the main side effects are taste derangement, tremor and kidney side effects.
  • these side effects are much less toxic than those encountered by patients receiving normal traditional cancer therapy, and the doses employed for cancer therapy may be lower than those employed for the treatment of mood disorders, especially when the lithium is employed in combination of disulfiram.
  • the patient receiving the combination therapy of the present disclosure is monitored for plasma levels of lithium, for example when the treatment is started and/or on a monthly, three monthly, six monthly or annual basis.
  • the dose of lithium employed is a dose that does not elicit side effects in the majority of patients.
  • the majority of patients as employed herein refers to wherein at 51% of patients who receive that dose do not encounter therapy limiting side effects or only encounter minor side effects associated with the lithium such as 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100% of patients do not encounter therapy limiting side effects.
  • the lithium acts in cancer cells (for example cancer cells which are differentiated, i.e. non-cancer-stem-cells) at least via inhibiting the enzyme glycogen synthase kinase-3 (GSK-3), which is linked to the insulin and the Wnt pathways.
  • GSK-3 glycogen synthase kinase-3
  • the inhibition of GSK-3 may be associated with antiproliferative effects.
  • lithium may also be linked to hormone receptors such as estrogen and androgen.
  • other effects of lithium may be exerted on bone marrow and on adaptive and innate immune defenses which are not necessarily associated to the GSK3 mediated effects.
  • lithium seems to increase the proliferation and self-renewal of stem cells, including cancer stem cells. That could be seen as a negative in terms of cancer therapy. However, in this instance it may be positive because it prevents the cancer cells evading treatment.
  • lithium driving proliferation and self-renewal prevents the cancer cells becoming dormant.
  • the latter is thought to be a mechanism for cancer cell resistance.
  • preventing the option for the cancer cells to become dormant may enhance immediate efficacy of cancer therapy and may allow more or all of the cancer cell populations present to be treated. This in turn may improve the effectiveness of the cancer therapy and/or reduce the risk for cancer relapse.
  • self-renewal may also mean that the effectiveness of concomitant therapies (such as traditional cancer therapies or therapies directed to cancerous at renewal mechanisms) may have an increased therapeutic window or have a therapeutic window which is shifted towards more "selectively" effecting cancer cells (or certain types of cancer cells, such as cancer stem cells).
  • concomitant therapies such as traditional cancer therapies or therapies directed to cancerous at renewal mechanisms
  • Another mechanism of lithium efficacy may be on the bone marrow and the T- cells, which through the inhibition of GSK3 may become more effective, for example providing prolonged T cell proliferation and increased IL-2 production.
  • immune-stimulatory effects provided by employing the combination therapy of the present disclosure which renders the patient more receptive to therapy, for example in the form of immune-oncology compounds.
  • Immune-oncology compounds also referred to as cancer immune therapy herein refers to therapies that target tumor associated antigens (TAAs) and stimulate immune responses to the cancer.
  • TAAs tumor associated antigens
  • lithium and disulfiram may work additively or in synergy, to decrease the survival of cancer cells, such as cancer stem cells.
  • Disulfiram inhibits the aldehyde dehydrogenase, which is a key marker for cancer stem cells, and is a key enzyme for defense against reactive oxygen species (ROS), which may otherwise harm the cancer stem cell.
  • ROS reactive oxygen species
  • disulfiram may cause cancer stem cell reduction, for example via accumulation of reactive oxygen species leading to necrosis, apoptosis or autophagy thereby increasing death of cancer stem cells.
  • Necrosis as employed herein refers to cell death through injury or damage to the cell.
  • Apoptosis as employed herein refers to hastening programmed cell death, which is sometimes referred to as cell assisted suicide.
  • Autophagy as employed herein refers to a complex process of intracellular degradation, which may deliver nutrients and/or energy to other cells.
  • the combination therapy of the present disclosure makes it difficult for cancer cells to stay in quiescent dormant mode, for example by provoking self-renewal stimulated by the lithium part of the combined therapy. This renders the cancer stem cells vulnerable to cancer treatments, either via the disulfiram part or by other anti-cancer therapeutics.
  • ROS reactive oxygen species
  • Reactive oxygen species refers to peroxides, superoxides, hydroxyl radicals, singlet oxygen and the like.
  • ROS reactive oxygen species
  • DSF Disulfiram
  • aldehyde dehydrogenase aldehyde dehydrogenase
  • its' inhibition causes upstream accumulation of ethanol aldehyde which is linked to physical discomfort.
  • Aldehyde dehydrogenase is also a marker of cancer stem cells and the present inventors believe that disulfiram via inhibition of aldehyde dehydrogenase increases the concentration of reactive oxygen species. This increased concentration of reactive oxygen species in turn damages the cancer stem cells. Thus is hypothesized that DSF suppresses self-renewal capability mainly through the activation of reactive oxygen species (ROS)-p38 MAPK pathway.
  • ROS reactive oxygen species
  • TNF-alpha TNF-alpha
  • vascular endothelial growth factor TNF-alpha
  • MMP-2 and MMP-9 from cultured macrophages, which may contribute to the overall effectiveness of the combination therapy.
  • effects of lithium on the bone marrow and on the immune system may add to the breadth of potential positives of the therapy of the present disclosure.
  • the combination therapy of the present disclosure has beneficial effects in protecting non-cancerous tissue from the toxic effect of cancer therapy that may be employed in combination with the therapy comprising disulfiram and a lithium compound.
  • Combination treatment refers to a treatment wherein the duration of time when the patient is on or taking the medicament comprising a lithium compound is also the time when the patient is taking or is administered a medicament comprising the disulfiram (i.e. treatment regimens for lithium and disulfiram at least overlap).
  • a lithium compound and a disulfiram are administered or taken in the same 7 days period. In one embodiment the disulfiram is administered or taken on the same day as the lithium compound.
  • the combination therapy of the present disclosure is employed as adjuvant therapy, for example post-surgery or post-chemotherapy.
  • the combination therapy of the present disclosure is employed as neo-adjuvant therapy.
  • the combination treatment of the present disclosure may be employed in combination with one or more further cancer treatments, for example
  • the therapy may reduce the side effects of such and may allow usage of higher and more effective doses of other cancer therapies to be employed and tolerated.
  • a suitable dose may be chosen by the practitioner based on the nature of the cancer being treated.
  • Chemotherapeutic agent as employed herein is intended to refer to specific antineoplastic chemical agents or drugs that are selectively destructive to malignant cells and tissues. Chemotherapeutic agent and chemotherapy or cytotoxic agent are employed interchangeably herein unless the context indicates otherwise. Examples inlcude alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents. Other examples of chemotherapy include doxorubicin, 5-fluorouracil (5-FU), paclitaxel, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
  • 5-fluorouracil 5-FU
  • paclitaxel paclitaxel
  • capecitabine paclitaxel
  • irinotecan irinotecan
  • platins such as cisplatin and oxaliplatin.
  • alkylating agents which may be employed in the method of the present disclosure include an nitrogen mustards alkylating agent, nitrosoureas, tetrazines, aziridines, platins and derivatives, and non-classical alkylating agents.
  • platinum containing chemotherapeutic agent also referred to as platins
  • chemotherapeutic agent such as cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin and lipoplatin (a liposomal version of cisplatin), in particular cisplatin, carboplatin and oxaliplatin.
  • the dose for cisplatin ranges from about 20 to about 270 mg/m 2 depending on the exact cancer. Often the dose is in the range about 70 to about 100mg/m 2 .
  • the dose for cisplatin ranges from about 20 to about 270 mg/m 2 depending on the exact cancer. Often the dose is in the range about 70 to about 100mg/m 2 .
  • hemotherapeutic agent is a topoisomerase inhibitor.
  • Nitrogen mustards include mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide and busulfan.
  • Nitrosoureas include N-Nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU) and semustine (MeCCNU), fotemustine and streptozotocin.
  • Tetrazines include dacarbazine, mitozolomide and temozolomide.
  • Aziridines include thiotepa, mytomycin and diaziquone (AZQ).
  • antimetabolites examples include anti-folates (for example methotrexate and pemetrexed), purine analogues (for example thiopurines, such as azathiopurine, mercaptopurine, thiopurine, fludarabine (including the phosphate form), pentostatin and cladribine), pyrimidine analogues (for example fluoropyrimidines, such as 5-fluorouracil and prodrugs thereof such as capecitabine [Xeloda®]), floxuridine, gemcitabine, cytarabine, decitabine, raltitrexed(tomudex) hydrochloride, cladribine and 6-azauracil.
  • anti-folates for example methotrexate and pemetrexed
  • purine analogues for example thiopurines, such as azathiopurine, mercaptopurine, thiopurine, fludarabine (including the phosphate form),
  • anthracyclines examples include daunorubicin (Daunomycin), daunorubicin (liposomal), doxorubicin (Adriamycin), doxorubicin (liposomal), epirubicin, idarubicin, valrubicin currenlty used only to treat bladder cancer and mitoxantrone an anthracycline analog, in particular doxorubicin.
  • anti-microtubule agents examples include vinca alkaloids and taxanes.
  • Vinca alkaloids include completely natural chemicals for example vincristine and vinblastine and also semi-synthetic vinca alkaloids, for example vinorelbine, vindesine, and vinflunine.
  • Taxanes include paclitaxel, docetaxel, abraxane, carbazitaxel and derivatives of thereof.
  • Derivatives of taxanes as employed herein includes reformulations of taxanes like taxol, for example in a micelluar formulaitons, derivatives also include chemical derivatives wherein synthetic chemistry is employed to modify a starting material which is a taxane.
  • Topoisomerase inhibitors which may be employed in a method of the present disclosure include type I topoisomerase inhibitors, type II topoisomerase inhibitors and type II topoisomerase poisons.
  • Type I inhibitors include topotecan, irinotecan, indotecan and indimitecan.
  • Type II inhibitors include genistein and ICRF 193 which has the following structure:
  • Type II poisons include amsacrine, etoposide, etoposide phosphate, teniposide and doxorubicin and fluoroquinolones.
  • he chemotherapeutic agent is a parp inhibitor, in particular as disclosed herein.
  • the combination therapy is administered with or following a treatment regime comprising a platinum containing chemotherapeutic agent, for example cisplatin, carboplatin or oxaliplatin.
  • a combination of chemotherapeutic agents employed is, for example a platin and 5-FU or a prodrug thereof, for example cisplatin or oxaplatin and capecitabine or gemcitabine, such as FOLFOX.
  • the chemotherapy comprises a combination of chemotherapy agents, in particular cytotoxic chemotherapeutic agents.
  • the chemotherapy combination comprises a platin, such as cisplatin and fluorouracil or capecitabine.
  • the chemotherapy combination in capecitabine and oxaliplatin in capecitabine and oxaliplatin (Xelox).
  • the chemotherapy is a combination of folinic acid and 5-FU, optionally in combination with oxaliplatin.
  • the chemotherapy is a combination of folinic acid, 5-FU and irinotecan (FOLFIRI), optionally in combination with oxaliplatin (FOLFIRINOX).
  • the regimen consists of: irinotecan (180 mg/m 2 IV over 90 minutes) concurrently with folinic acid (400 mg/m 2 [or 2 x 250 mg/m 2 ] IV over 120 minutes); followed by fluorouracil (400-500 mg/m 2 IV bolus) then fluorouracil (2400-3000 mg/m 2 intravenous infusion over 46 hours). This cycle is typically repeated every two weeks.
  • the dosages shown above may vary from cycle to cycle.
  • the combination employs a microtubule inhibitor, for example vincristine sulphate, epothilone A, N-[2-[(4-Hydroxyphenyl)amino]-3- pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), ataxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • a microtubule inhibitor for example vincristine sulphate, epothilone A, N-[2-[(4-Hydroxyphenyl)amino]-3- pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), ataxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • the combination employs an mTor inhibitor.
  • mTor inhibitors include: everolimus (RADOOl), WYE-354, KU-0063794, papamycin (Sirolimus), Temsirolimus, Deforolimus(MK-8669), AZD8055 and BEZ235(NVP-
  • the chemotherapy combination employs a microtubule inhibitor, for example vincristine sulphate, epothilone A, N-[2-[(4- Hydroxyphenyl)amino]-3-pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), a taxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • a microtubule inhibitor for example vincristine sulphate, epothilone A, N-[2-[(4- Hydroxyphenyl)amino]-3-pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), a taxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • a microtubule inhibitor for example vincristine sulphate, epothilone A,
  • the chemotherapy combination employs a MEK inhibitor.
  • MEK inhibitors include: AS703026, CI-1040 (PD184352), AZD6244 (Selumetinib), PD318088, PD0325901, AZD8330, PD98059, U0126-EtOH, BIX 02189 or BIX 02188.
  • the chemotherapy combination employs an AKT inhibitor.
  • AKT inhibitors include: MK-2206 and AT7867.
  • the combination employs an aurora kinase inhibitor.
  • aurora kinase inhibitors include: Aurora A Inhibitor I, VX-680, AZD1152- HQPA (Barasertib), SNS-314 Mesylate, PHA-680632, ZM-447439, CCT129202 and Hesperadin.
  • the chemotherapy combination employs a p38 inhibitor, for example as disclosed in WO2010/038086, such as N-[4-( ⁇ 4-[3-(3-tert-Butyl-l-p-tolyl- lH-pyrazol-5-yl)ureido]naphthalen-l-yloxy ⁇ methyl)pyridin-2-yl]-2-methoxyacetamide.
  • a p38 inhibitor for example as disclosed in WO2010/038086, such as N-[4-( ⁇ 4-[3-(3-tert-Butyl-l-p-tolyl- lH-pyrazol-5-yl)ureido]naphthalen-l-yloxy ⁇ methyl)pyridin-2-yl]-2-methoxyacetamide.
  • the combination employs a Bcl-2 inhibitor.
  • Bcl- 2 inhibitors include: obatoclax mesylate, ABT-737, ABT-263(navitoclax) and TW-37.
  • the chemotherapy combination comprises an antimetabolite such as capecitabine (xeloda), fludarabine phosphate, fludarabine (fludara), decitabine, raltitrexed (tomudex), gemcitabine hydrochloride and cladribine.
  • an antimetabolite such as capecitabine (xeloda), fludarabine phosphate, fludarabine (fludara), decitabine, raltitrexed (tomudex), gemcitabine hydrochloride and cladribine.
  • the chemotherapy combination comprises ganciclovir, which may assist in controlling immune responses and/or tumour vasculation.
  • the gastric cancer is selected from the group comprising adenocarcinoma of the stomach, squamous cell carcinomas, lymphoma of the stomach, gastric stromal tumour, and neuroendocrine tumours.
  • the liver cancer is, for example selected from the group hepatocellular carcinoma, cholangiocarcinoma, angiosarcoma, and hepatoblastoma, in particular hepatocellular carcinoma.
  • the primary liver cancer is stage 1, 2, 3 or 4.
  • the liver cancer is secondary or metastasized liver cancer.
  • Prostate cancer refers to cancer of the prostate, for example ductal adenocarcinoma, transitional cell (urothelial cancer), squamous cell cancer, carcinoid of the prostate, small cell cancer or sarcoma and sarcomatoid cancer.
  • Pancreatic cancer as employed herein includes exocrine cancers (including rare forms thereof such as cystitic tumours, and cancer of the acinar cells), endocrine pancreatic tumours (including gastrinomas, insulinomas, somatostatinomas, VIPomas, glucagonomas), pancreatoblastoma, sarcomas of the pancreas and lymphoma.
  • Biliary tract cancer refers to cholangiocarcinoma (intrahepatic, extrahepatic), gall bladder cancer and ampullary carcinoma.
  • Colorectal cancer refers to cancer or the colon and/or rectum and includes squamous cell cancers, carcinoid tumours, sarcomas and lymphomas.
  • Breast cancer refers to cancer of the breast and includes ductal carcinoma in situ, lobular carcinoma in situ, invasive ductal breast cancer, invasive lobular breast cancer, invasive breast cancer, Paget's disease, angiosarcoma of the breast and rare types of breast cancer such as medullary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast metaplastic breast cancer, basal type breast cancer and papillary breast cancer.
  • Lung cancers are classified according to histological type and are categorized by the size and appearance of the malignant cells seen by a histopathologist under a microscope.
  • two broad classes are distinguished: non-small cell lung carcinoma and small cell lung carcinoma.
  • the epithelial cancer is lung cancer, for example small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC).
  • SCLC small-cell lung cancer
  • NSCLC non-small-cell lung cancer
  • Non-small-cell lung carcinoma The three main subtypes of NSCLC are adenocarcinoma, squamous-cell carcinoma and large-cell carcinoma.
  • adenocarcinoma Nearly 40% of lung cancers are adenocarcinoma, which usually originates in peripheral lung tissue.
  • a subtype of adenocarcinoma, the bronchioloalveolar carcinoma, is more common in female never-smokers, and may be associated with a better long term survival.
  • Squamous-cell carcinoma accounts for about 30% of lung cancers. They typically occur close to large airways. A hollow cavity and associated cell death are commonly found at the center of the tumor. About 9% of lung cancers are large-cell carcinoma. These are so named because the cancer cells are large, with excess cytoplasm, large nuclei and conspicuous nucleoli.
  • SCLC Small-cell lung carcinoma-In small-cell lung carcinoma
  • the cells contain dense neurosecretory granules (vesicles containing neuroendocrine hormones), which give this tumor an endocrine/paraneoplastic syndrome association.
  • These cancers grow quickly and spread early in the course of the disease. Sixty to seventy percent have metastatic disease at presentation.
  • the cancer is non-small lung carcinoma.
  • the cancer is liver cancer, for example a liver metastasis from a primary cancer, for example colon cancer, which has spread to the liver.
  • the liver cancer is HCC hepatocellular carcinoma.
  • renal cancer for example renal cell carcinoma and/or urothelial cell carcinoma.
  • Other examples of renal cancer include squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma, Bellini duct carcinoma, clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma, and carcinoid tumor of the renal pelvis.
  • reninoma juxtaglomerular cell tumor
  • angiomyolipoma renal oncocytoma
  • Bellini duct carcinoma clear-cell sarcoma of the kidney
  • mesoblastic nephroma Wilms' tumor
  • mixed epithelial stromal tumor mixed epithelial stromal tumor
  • the cancer is bladder cancer, for example is any of several types of malignancy arising from the epithelial lining (i.e. the urothelium) of the urinary bladder.
  • the epithelial lining i.e. the urothelium
  • the bladder cancers are transitional cell carcinoma.
  • the other 10% are squamous cell carcinoma, adenocarcinoma, sarcoma, small cell carcinoma, and secondary deposits from cancers elsewhere in the body.
  • the staging of this cancer is given below.
  • the current disclosure extends to any stage of bladder cancer.
  • Cancerous ovarian tumours can start from three common cell types:
  • Germ Cells - cells that are destined to form eggs • Stromal Cells - Cells that release hormones and connect the different structures of the ovaries
  • the present disclosure relates to treatment of ovarian cancer from any source, for example as described herein, in particular epithelium cells.
  • Epithelial ovarian carcinomas account for 85 to 90 percent of all cancers of the ovaries.
  • Epithelial ovarian tumors develop from the cells that cover the outer surface of the ovary. Most epithelial ovarian tumors are benign (noncancerous). There are several types of benign epithelial tumors, including serous adenomas, mucinous adenomas, and Brenner tumors. Cancerous epithelial tumors are carcinomas - meaning they begin in the tissue that lines the ovaries. These are the most common and most dangerous of all types of ovarian cancers. Unfortunately, almost 70 percent of women with the common epithelial ovarian cancer are not diagnosed until the disease is advanced in stage.
  • LMP tumors tumors of low malignant potential
  • Germ Cell Tumors Ovarian germ cell tumors develop from the cells that produce the ova or eggs. Most germ cell tumors are benign (non-cancerous), although some are cancerous and may be life threatening. The most common germ cell malignancies are maturing teratomas, dysgerminomas, and endodermal sinus tumors. Germ cell malignancies occur most often in teenagers and women in their twenties. Today, 90 percent of patients with ovarian germ cell malignancies can be cured and their fertility preserved.
  • Stromal Tumors - Ovarian stromal tumors are a rare class of tumors that develop from connective tissue cells that hold the ovary together and those that produce the female hormones, estrogen and progesterone. The most common types are granulosa- theca tumors and Sertoli-Leydig cell tumors. These tumors are quite rare and are usually considered low-grade cancers, with approximately 70 percent presenting as Stage I disease (cancer is limited to one or both ovaries).
  • Primary Peritoneal Carcinoma The removal of one's ovaries eliminates the risk for ovarian cancer, but not the risk for a less common cancer called Primary Peritoneal Carcinoma.
  • Primary Peritoneal Carcinoma is closely rated to epithelial ovarian cancer (most common type). It develops in cells from the peritoneum (abdominal lining) and looks the same under a microscope. It is similar in symptoms, spread and treatment. Stages of Ovarian Cancer
  • stage of a tumor can be determined during surgery, when the doctor can tell if the cancer has spread outside the ovaries.
  • the treatment plan and prognosis (the probable course and outcome) will be determined by the stage of cancer.
  • Stage I - Growth of the cancer is limited to the ovary or ovaries.
  • Stage IA - Growth is limited to one ovary and the tumor is confined to the inside of the ovary. There is no cancer on the outer surface of the ovary. There are no ascites present containing malignant cells. The capsule is intact.
  • Stage IB - Growth is limited to both ovaries without any tumor on their outer surfaces. There are no ascites present containing malignant cells. The capsule is intact.
  • Stage IC The tumor is classified as either Stage IA or IB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries;
  • Stage II - Growth of the cancer involves one or both ovaries with pelvic extension.
  • Stage IIA - The cancer has extended to and/or involves the uterus or the fallopian tubes, or both.
  • Stage IIB - The cancer has extended to other pelvic organs.
  • Stage IIC The tumor is classified as either Stage IIA or IIB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries; (2) the capsule has ruptured; and (3) there are ascites containing malignant cells or with positive peritoneal washings.
  • Stage III - Growth of the cancer involves one or both ovaries, and one or both of the following are present: (1) the cancer has spread beyond the pelvis to the lining of the abdomen; and (2) the cancer has spread to lymph nodes.
  • the tumor is limited to the true pelvis but with histologically proven malignant extension to the small bowel or omentum.
  • Stage IIIA - During the staging operation, the practitioner can see cancer involving one or both of the ovaries, but no cancer is grossly visible in the abdomen and it has not spread to lymph nodes. However, when biopsies are checked under a microscope, very small deposits of cancer are found in the abdominal peritoneal surfaces.
  • Stage IIIB The tumor is in one or both ovaries, and deposits of cancer are present in the abdomen that are large enough for the surgeon to see but not exceeding 2 cm in diameter. The cancer has not spread to the lymph nodes.
  • Stage IIIC The tumor is in one or both ovaries, and one or both of the following is present: (1) the cancer has spread to lymph nodes; and/or (2) the deposits of cancer exceed 2 cm in diameter and are found in the abdomen.
  • Stage IV This is the most advanced stage of ovarian cancer. Growth of the cancer involves one or both ovaries and distant metastases (spread of the cancer to organs located outside of the peritoneal cavity) have occurred. Finding ovarian cancer cells in pleural fluid (from the cavity which surrounds the lungs) is also evidence of stage IV disease.
  • the ovarian cancer is: type I, for example IA, IB or IC; type II, for example IIA, IIB or IIC; type III, for example IIIA, IIIB or III C; or type IV.
  • the breast cancer is one selected from the group comprising ductal carcinoma in situ, lobular carcinoma in situ, invasive breast cancer, invasive lobular breast cancer, Paget's disease, angiosarcoma of the breast, medulllary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast, metaplastic breast cancer, lymphoma of the breast, basal type breast cancer, phyllodes or cystosarcoma phyllodes and papillary breast cancer.
  • the prostate cancer is selected from the group comprising ductal adenocarcinoma, transitional cell (urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers.
  • the prostate cancer is selected from the group comprising ductal adenocarcinoma, transitional cell (urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers and a combination of two or more of the same.
  • Thyroid cancer refers to cancer of the thyroid originating from follicular or parafollicular thyroid cells and includes papillary thyroid cancer (75% to 85% of cases); follicular thyroid cancer (10% to 20% of cases); medullary thyroid cancer (5% to 8% of cases)- cancer of the parafollicular cells, often part of multiple endocrine neoplasia type 2; poorly differentiated thyroid cancer; anaplastic thyroid cancer (less than 5% of cases) is not responsive to treatment and can cause pressure symptoms, thyroid lymphoma, squamous cell thyroid carcinoma, sarcoma of thyroid.
  • Renal cancer refers to cancer of the kidney, for example renal cell carcinoma and transitional cell carcinoma of the renal pelvis, such as squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma, bellini duct carcinoma, clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma; carcinoid tumor of the renal pelvis.
  • reninoma juxtaglomerular cell tumor
  • angiomyolipoma angiomyolipoma
  • renal oncocytoma bellini duct carcinoma
  • clear-cell sarcoma of the kidney mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor
  • Bladder cancer as employed herein refers to cancer of the bladder including transitional cell bladder cancer, carcinoma in situ, papillary cancer and rarer types of bladder cancer such as squamous cell cancer, adenocarcinoma and a combination of two or more of the same.
  • Esophageal cancer as employed herein refers to cancer of the oesphagus including esophageal squamous-cell carcinomas, esophageal adenocarcinomas, and variants of squamous-cell carcinoma, and non-epithelial tumors, such as leiomyosarcoma, malignant melanoma, rhabdomyosarcoma, lymphoma, among others and a combination of two or more of the same.
  • Head and neck cancer refers to cancer of the neck and/or head, including mouth cancer, nasopharyngeal cancer, oropharyngeal cancer, paranasal sinus cancer, salivary gland cancer and a combination of two or more of the same.
  • the chemotherapy combination comprises ganciclovir, which may assist in controlling immune responses and/or tumour vasculation.
  • Embodiments of the invention may be combined. Embodiments are described herein as comprising certain features/elements. The disclosure also extends to separate embodiments consisting or consisting essentially of said features/elements.
  • the IL-6-STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression Q Zhu,l,4 X Zhang,l,4,* L Zhang,l W Li,2 H Wu,2 X Yuan,l F Mao,l M Wang.1 W Zhu,l H Qian,l and W Xul,3,*
  • Macrophage plasticity and polarization in vivo Veritas. Sica Al, Mantovani A.
  • TNF skews monocyte differentiation from macrophages to dendritic cells. Chomarat P 1 , Dantin C, Bennett L, Banchereau J, Palucka AK.
  • T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions.
  • Lithium an anti-psychotic drug, greatly enhances the generation of induced pluripotent stem cells; Quan Wang,l Xinxiu Xu,2 Jun Li,3 Jing Liu,4 Haifeng Gu,l Ru Zhang,2 Jiekai Chen,4 Yin Kuang,5 Jian Fei,5 Cong Jiang,6 Ping Wang,6 Duanqing Pei,4 Sheng Ding,3 and Xin Xiel,2,*
  • FIG. 2 Schematic diagram showing multimodal cancer treatment according to the present method
  • Example 1 Protocol for treating a cholangiocarcinoma patient Example 1 Protocol for treating a cholangiocarcinoma patient.
  • a pre-step as much cancerous tissue as possible is removed by surgery.
  • the patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent (eg capecitabine or gemcitabine) for 12-28 days.
  • a chemotoxic agent eg capecitabine or gemcitabine
  • AZD5069 is given as a monotherapy or concomitantly with a T-cell stimulator for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of chemotherapy comprising gemcitabine and cisplatin are given.
  • treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
  • the final stage of the therapy is one cycle of antibody drug conjugate, wherein the antibody is specific to Epcam, potentially in combination with an immune-oncology (IO) agent such as pemrolizumab.
  • IO immune-oncology
  • a pre-step as much cancerous tissue as possible is removed by surgery.
  • the patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent (eg capecitabine or gemcitabine) for 12-28 days.
  • a chemotoxic agent eg capecitabine or gemcitabine
  • AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of chemotherapy comprising 5-FU and leucovorin are given.
  • treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
  • the final stage of the therapy is one cycle of antibody drug conjugate, wherein the antibody is specific to Epcam, potentially in combination with an 10 agent such as pemrolizumab.
  • Example 3 Protocol for treating a cholangiocarcinoma patient
  • AZD5069 eg capecitabine or gemcitabine
  • AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of
  • an IO agent such as pemrolizumab.
  • treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
  • the final stage of the therapy targeted radiation therapy in combination with a PARP inhibitor potentially in combination with an 10 agent such as pemrolizumab.
  • Example 4 Protocol for treating a cholangiocarcinoma patient
  • AZD5069 e.g. capecitabine or gemcitabine
  • AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of
  • treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
  • the final stage of the therapy targeted radiation therapy in combination with a PARP inhibitor potentially in combination with an 10 agent such as pemrolizumab.

Abstract

Disclosed herein is a method of treating cancer, comprising: a) reducing or stopping cancer proliferation, b) debulking tumour mass, c) activating immune cells, d) increasing antigen presentation and/or enhancing T cell response, and e) clearing residual tumour cells.

Description

METHOD OF TREATING CANCER
The present disclosure relates to a method of treating cancer, use of medicaments in a staged combination therapy for the treatment of cancer (also referred to herein as multimodal cancer therapy), and use of a medicament or medicaments in the manufacture of a multimodal therapy for the treatment of cancer, for example linked with dynamic input of tumor status, to optimise intervention and timing.
BACKGROUND
The treatment of cancer is still a huge challenge and burden to society. The present inventor believes that the effectiveness of cancer therapy can be greatly increased by employing a multimodal method of treatment. That is conscious choice of sequence and the timing of multiple treatments to achieve improved outcomes, for patients.
The involvement of the immune system in defense against cancer growth has been evident for a long time but with the emergence of the immune check-point inhibitor therapies (for example, PD-1, PDL-1, CTLA4) the understanding of the field has rapidly expanded. It is clear that tumors attract immune cells. However, it is also clear that many of these cells that are effective against viral and bacterial infections, offer little effect against cancer, and may even be counterproductive and cancer supporting.
It is now thought that tumor associated macrophages actually feed and protect the cancer cells. Stroma, containing immune cells, builds up around cancer cells and creates a protected environment where the cancer flourishes.
Cancer cells are highly adapted to corrupting and hijacking many natural biological processes that exist in the body. Cancers are sometimes based on mutations and modifications of cells and thus can readily change and adapt. Given this the present inventor believes that is unlikely that one or two modes of treatment will be effective for the treatment of the majority of cancers.
Therefore, the inventor has designed a new method to treat cancer, which involves a radical resetting of the immune system, which will be much more efficacious than just using simple steps, such as immune check point inhibitors or combinations thereof.
In some ways, the method of present disclosure can be considered a "reboot" of the immune system, which after treatment is then able to fight the cancer.
SUMMARY OF THE DISCLOSURE
Thus, the present disclosure provides a method of treating cancer comprising the steps;
a) reducing or stopping cancer proliferation,
al) declutter of tumor infiltrating immune cells,
b) debulking tumour mass, and/or increasing antigen presentation, c) activating immune cells,
d) increasing antigen presentation and/or enhancing T cell response, and e) clearing residual tumor cells.
The treatment schedule of the present disclosure is a paradigm shift in cancer treatment, because at the present time standard of care cancer treatment generally only involves one or two of the above steps.
Whilst some cancer therapy regimes (like chemotherapy regimens) are a combination or cocktail of the therapeutic agents from the outset, and cancer adjuvant therapy is known (i.e. chemotherapy or radiation therapy after surgery to excise a tumor) subsequent cancer therapies are not employed until the first line or second therapy has failed.
The multimodal therapy of the present disclosure is not the same as simply trying different treatments after the failure of a treatment. Instead it involves pro- actively employing multiples modes of treatment, in particular in the order specified herein.
In one embodiment the method comprises the further step of tumour surveillance.
In one embodiment the therapeutic steps are taken in fast succession, in particular while surveying and reading "efficacy", for example by studying effects on immune parameters and/or variables showing circulating tumor components. Efficacy here is not employed in the strict technical sense but simply to effects more generically.
The fast delivery of a sequence of therapies and the elected timing of the next treatment step may be important to reset the immune system while stopping further cancer growth. Thus in one embodiment the present multimodal treatment comprises multiple treatment steps, which are employed at specific times, for example over a period of 1 to 12 months, such as 1 to 2 months, 1 to 3 months, 1 to 4 months, 1 to 5 months, 1 to 6 months, 1 to 7 months, 1 to 8 months, 1 to 9 months, 1 to 10 months or 1 to 11 months.
This fast succession of different modes of treatment is highly advantageous because it minimises the time available for the cancer cells to "regroup" and develop strategies to make them resistant to treatment.
In one embodiment the "subsequent" therapies are not employed because a therapy has failed i.e. the further therapy is not started because there is resistance to "first line or second line" therapy.
In one embodiment step b) follows step a) or step al). In one embodiment step c) follows step b). In one embodiment step d) follows step c). In one embodiment step e) follows step d). In one embodiment one or more treatment steps may overlap or be at the same time.
In one embodiment step a) and al) overlap or are at the same time. In one embodiment step a) and b) overlap or are at the same time. In one embodiment step al) and b) overlap or are at the same time. In one embodiment step b) and c) overlap or are at the same time. In one embodiment step c) and d) overlap or are at the same time. In one embodiment step d) and e) overlap or are at the same time.
In one embodiment step a), al) and b) overlap or are at the same time. In one embodiment step a), b) and c) overlap or are at the same time. In one embodiment step al), b) and c) overlap or are at the same time. In one embodiment step b), c) and d) overlap or are at the same time. In one embodiment step c), d) and e) overlap or are at the same time.
In one embodiment step a), al), b) and c) overlap or are at the same time. In one embodiment step a), b), c) and d) overlap or are at the same time. In one embodiment step al), b), c) and d) overlap or are at the same time. In one embodiment step b), c), d) and e) overlap or are at the same time.
In one embodiment step a), b), c), d) and e) overlap or are at the same time. In one embodiment step al), b), c), d) and e) overlap or are at the same time
In one embodiment one or more of the steps do not overlap, for example 2, 3, 4 or all of the step do not overlap.
In one embodiment step b) and d) do not overlap and are not at the same time.
In one embodiment step e) does not overlap with step c).
In one embodiment the method comprises a pre-step of surgery, for example to remove tumour mass and/or lymph nodes and the like.
In one embodiment the method comprises irradiation.
In one embodiment step a) is a treatment selected from the group comprising irradiation, chemotherapy, anti-proliferation pathway therapy (for example anti- oestrogen, anti-androgen, anti-HER, anti-BCR/ABL, anti-VEGF, anti-PDGF, anti-BRAF antibody molecule [including a bispecific antibody, full-length or an antibody binding fragment], with or without combining with the appropriate chemotherapy) and a combination of two or more of the same.
In one embodiment at least one agent employed in step a) is an antiproliferative agent, for example selected from the group 25-hydroxcholesterol, 3-hydroxtyrosol, 7β- hydroxycholestrol, aloe-emodin, apigenin, artesunate, berberine, caffeine power (C0750), dichloromethylenediphosphonic acid (for example as a salt, such as a disodium salt), emodin, HA 14-1, hyperforin, N-acetyl-D-sphingosine, N-hexanoyl-D-sphingosine, oleuropein, pathenolide, tangeretin, tanshinone IIA, β-cryptoxanthine, β-cryptoxanthin, β-lonone, trans-cinnamaldehyde and combinations thereof. Other agents that can be employed alone or in a combination in step a) include high doses of vitamin Bl, parp inhibitors, a gene product of NME2, myrtillin (delphinidin-3-O-glucoside), tulipanin (delphinidin-3-O-rutinoside), violdelphin (delphinidin 3-rutinoside-7-0-(6-0-(4-(6-0-(4-hydroxybenzoyl) -p-D-glucosyl)oxy benzoyiJ-p-D-glucoside), nasunin (delphinidin-3-(p-coumaroylrutinoside)-5-glucoside) and combinations of two or more of the same.
In one embodiment therapy employed in step a) is Varlitinib [(i?)-N4-[3-Chloro- 4-(thiazol-2-ylmethoxy)-phenyl]-N6-(4-methyl-4, 5,-dihydro-oxazol-2-yl)-quinazoline- 4,6-diamine] or a pharmaceutically acceptable salt thereof, which may be employed alone or in a combination therapy.
The Varlitinib, is administered at a dose in the range lOOmg to 900mg on each occasion, in particular 200 mg, 300mg, 400mg or 500mg each dose, such as 400mg, for example administered once or twice daily, such as twice daily.
In one embodiment the therapy of step a) comprises a DHODH inhibitor, such as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
In one embodiment the PARP inhibitor one or more compounds independently selected from of the followin :
Figure imgf000005_0001
Phase III trials for ovarian, breast and evaluated in Phase III studies for breast, other solid cancers); pancreatic, non-small cell lung cancer, lymphoma and multiple myeloma);
Figure imgf000006_0001
CEP-9722 a PARP-1 and PARP-2 inhibitor; INO-1001 (3-aminobenzamide) a potent inhibitor of PARP (with IC50 of <50 nM in CHO cells and a mediator of oxidant- induced myocyte dysfunction during reperfusion)
BGB-290 a PARP-1 and PARP-2 inhibitor BGP-15 a PARP inhibitor (which has been (structure not shown); shown to protect against ischemia- reperfusion injury);
H
E7016 (previously known as GPI-21016) a PARP inhibitor (undergoing Phase I trials in combination with temozolomide for advanced solid tumours and gliomas);
Figure imgf000006_0002
MP-124 a PARP-1 inhibitor (structure not shown);
Figure imgf000007_0001
ABT-737 a PARP-1 and PARP-2 inhibitor,
or a pharmaceutically acceptable salt or solvate of any one of the same.
In one embodiment the therapy in step al), i.e. the decluttering of immune cells, is performed by administering a compound, such as a CXCR2 antagonist, an anti-GCF, cyclophosphamide and/or irradiation, and a combination of two or more of the same.
As used herein, decluttering refers to neutralisation of immune cells. This neutralisation may, for example include reduction in number of immune cells. Generally speaking, decluttering results in a reduction in overall number of immune cells, but may also encompass total depletion, including local and systemic depletion.
In one embodiment decluttering of immune cells is performed by removing specific immune cell populations, for example using medication and/or techniques such as a dialysis.
In one embodiment the immune cells that are "decluttered" are selected from a lymphocyte (such as a T lymphocyte, such as a T cell or a NKT cell), a B cell, a natural killer cell and a combination of two or more of the same.
In one embodiment the immune cells which are decluttered are tumor associated macrophages. Tumor associated macrophages can be decluttered by NKT cells engineered to encode a cytokine, for example as disclosed in WO2013/040371, incorporated herein by reference.
In one embodiment the immune cell, which is decluttered is a lymphocyte, for example a NK cell or a T lymphocyte, such as a T cell or a NKT cell.
In one embodiment the immune cells that are decluttered are B cells.
In one embodiment the immune cell that is decluttered is a natural killer cell. NK cells can be targeted by monoclonal antibodies or binding fragments specific to CD56.
Mature T lymphocytes can be removed by medicaments such as alemtuzumab. Immune cells with surface expression of CD52 can be removed by Campath. In one embodiment the CXCR2 antagonist is selected from an antibody or binding fragment thereof, MK-7123, SB265610, SB 225002:
Figure imgf000008_0001
GSK1325756, AZD5069, and combinations thereof.
In one embodiment the CXCR2 antagonist is SCH-527123.
In one embodiment the decluttering step al) is performed by employing anti- neutrophil antibodies.
In one embodiment the step al) is performed using a G-CSF antibody, for example CSL324, which is a humanized G-CSFR antibody.
In one embodiment the debulking in step b) is surgery to reduce a tumor mass. In one embodiment the surgery is followed by chemotherapy and/or (in particular or) irradiation therapy.
Debulking as employed herein refers to a reduction in the total mass of a tumor. This includes breaking a tumor mass into smaller fragments as well as shrinking the tumor mass. Methods of reducing a tumor mass include cutting (excising), ablating (including cryoablation, liquid nitrogen treatment of topical cancer cells and combinations of two or more of the same.
In one embodiment the debulking in step b) is performed by administering chemotherapy selected from alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents, such as doxorubicin, 5-fluorouracil (5-FU), paclitaxel, gemcitabine, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
In one embodiment the debulking in step b) is selected from treatment with an oncolytic virus, an antibody drug conjugate and a combination thereof.
In one embodiment the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus.
In one embodiment the oncolytic virus is selected from Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep-hIL12, Cavatak™, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, Toca 511.
In one embodiment the antibody drug conjugate (ADCC) is selected from the group comprising AGS-15E, AGS-16C3F, AGS-22M6E, AMG-172, AMG-595, ASG-5ME, brentuximab vedotin, DEDN-6526A, DMUC-5754A, gemtuzumab ozogamicin, glembatumumab vedotin, GSK2857916, hRS7-SN-38, IMGN-242, IMGN-289, IMGN-388, IMGN-529, IMGN-633, IMGN-853, Indatuximab, indatuximab ravtansine, Inotuzumab ozogamicin, lorvotuzumab mertansine, MEDI-547, milatuzumab doxorubicin, MLN- 0264, MLN-2704, PF-06263507, pinatuzumab vedotin, polatuzumab, PSMA-ADC, RG- 7450, RG-7599, SAR-3419, SAR-566658, SC16LD6.6, SGN-15, SGN-19A, SGN-CD33A, SGN-LIV1A, trastuzumab emtansine and combinations thereof.
In one embodiment step b) comprises administering varlitinib or a salt thereof, as a monotherapy or as part of a combination therapy, for example as disclosed elsewhere herein.
In one embodiment step b) comprises administering a DHODH inhibitor, such as as 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid or a pharmaceutically acceptable salt thereof.
In one embodiment activating the immune cells in step c) comprises administering a RON inhibitor, an AXL kinase inhibitor, a MER kinase agonist, a cytokine (such as IL-13, IL-15, IL-4 or IL-2), a tumour vaccine for example comprising a tumour antigen and adjuvant, a dendritic cell vaccine and a combination of two or more of the same.
As used herein activating immune cells refers to the functional activation of the immune cells, for example the activation of T cells by stimilulation with antigens (including where the antigen is provided on antigen presenting cells), and the activation of B cells to produce tumor specific antibodies. Activating may also include the recruitment of immune cells to the tumor site. Use of appropriate cytokines can stimulate and/or activate immune cells.
In one embodiment the RON inhibitor is N-(4-(2-Amino-3-chloropyridin-4- yloxy)-3-fluorophenyl)-4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3- carboxamide disclosed in WO 2008/058229.
In one embodiment activating the immune cells in step c) comprises administering one or more check-point inhibitors.
In one embodiment the checkpoint inhibitor is one or more compounds
Figure imgf000009_0001
Figure imgf000010_0001
422, V155991 and V158411 are CHKl and CHK2 inhibitors,:
Figure imgf000010_0002
VEfe00£5$992, Γ':
Figure imgf000011_0001
Gero pHHS (610) f 79
VsSuas sre tse average of si tsasi St !tii«¾itsfis ± SO
potent CHKl
Figure imgf000011_0002
MK-8776 is a selective CHKl inhibitor (with CHIR124 is CHKl inhibitor (is a novel an IC50 of 3 nM in a cell-free assay. The and potent inhibitor with IC50 of 0.3 molecule shows 500-fold selectivity against nM in a cell-free assay. It shows 2,000- CHK2); fold selectivity against CHK2 and 500 to
K2/4
Figure imgf000011_0003
Debromohymenialdisine (SKF 108753) is GDC-0425 is a selective CHKl a natural product derived from sponges that inhibitor;
in inhibits CHKl and CHK2;
Figure imgf000012_0001
Figure imgf000013_0001

Figure imgf000014_0001
Figure imgf000014_0002
KU55933 is a potent and selective KU59403 is an ATM inhibitor; ATM inhibitor vis-a-vie DNA-PK
Figure imgf000014_0003
VE-822 is a selective ATR inhibitor NU6027 is a potent ATR inhibitor; with a Ki of 0.2 nM and an about 150
selectivity over ATM;
Figure imgf000015_0001
PD-321852 is a CHKl and WEEl PD-407824 is a CHKl and WEEl inhibitor;
Figure imgf000015_0002
PV1019 is a CHK2 inhibitor; CCT241533 is a CHK2 inhibitor with about 63
Figure imgf000015_0003
Figure imgf000016_0001
In one embodiment the check-point inhibitor is a PD-1 or PD-L1 inhibitor.
In one embodiment the checkpoint inhibitor is a combination of 2 or several immune checkpoint inhibitors such as PD-1 plus CTL4, or PD-1 plus LAG3 (or TIM3 or GITR).
In one embodiment step d), i.e. increasing antigen presentation and/or enhancing T cell response, comprises administering a therapy selected from the group comprising a T cell therapy (for example where T cells are activated in vitro to generate CTLs specific to an antigen), a cell therapy comprising engineered chimeric receptors, a PD1 inhibitor, a PD-L1 inhibitors, a CTLA4 inhibitor, a LAG3 inhibitor and a combination of two or more of the same.
Increasing antigen presentation as used herein refers to the enhancement of the presentation of antigens to antigen presenting cells, for example by using a combination of treatments to make tumors necrotic, thereby providing a lot of potential antigenic material for presentation by antigen presenting cells. Other examples of increasing antigen presentation include enhancing the ability of dendritic cells and macrophages to phagocytose tumor and present tumor antigen; and blocking or inhibiting do-not-eat- me signals (CD47) on tumour cells that prevent phagocytosis of tumour cells.
Alternatively or in addition onclotytic viruses carrying a transgene can be used to infect the cancer cell and express the "protein or peptide" encoded on the cancer cells surface to increase antigen expression. Antigen in this context simply means a target for therapy or immune cells, in particular a target for immunotherapy.
In one embodiment step d) comprises administering a therapy selected from the group comprising an oncolytic virus, a DLL inhibitor, ADH-1 molecular targeting therapy and combinations of two or more of the same, for example the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus, such as Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep-hIL12, Cavatak™, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, or Toca 511.
Residual tumor cells as used herein refers to tumor cells that remain after the previous therapy, for exampleat the primary tumor site or in the vicinity, including circulating tumor cells, dormant cancer cells, cancer stems cells and combinations of the two or more of the sameThe goal of this step is to eradicate any remaining traces of cancer cells and minimise recurrence of the cancer.
In one embodiment step e), i.e. clearing residual tumor cells comprises targeting cancer stem cells.
In one embodiment the cancer stem cell therapy comprises targeting one or surface markers, for example with one or more monoclonal antibodies. In one embodiment the marker is selected from CD133, CD44, CD24, ESA, CD34 + CD38, and CD47.
In one embodiment step e) comprises an agent, such as an antibody or binding fragment thereof, specific to TRAIL or NF-aB.
In one embodiment step e) comprises an agent, such as an antibody or binding fragment thereof, specific to a cell marker Epcam, CD 133, CD44, CD24, ESA, CD34+/CD38- and CD47.
In one embodiment the cell markers are targeted by a chimeric antigen receptor.
In one antibody or antibody binding fragment is in the format of an antibody drug conjugate.
DETAILED DISCLOSURE
Multimodal or stage cancer therapy as employed herein refers to discrete phases of therapy, for example each phase lasting in the region of 1 week to 6 months, such as 2 weeks to 1 month, 2 weeks to 2 months, 2 weeks to 3 months, 2 weeks to 4 months or 2 weeks to 5 months. The therapies may overlap but generally will not simply be a combination therapy.
Combination therapy as employed herein is two or more active agents both administered over the same period of treatment.
The theory behind the method of the present disclosure is as follows:
1. Stop cancer continuing growing via use of cell cycle inhibitors and proliferation pathway inhibitors, and nutrient pump/metabolic inhibitors, to decrease growth of tumor burden:
a. Cancers use de-breaking of cell cycle to go on fast growth (1)
b. Proliferation pathway inhibitors and metabolic inhibitors can slow cancer growth (2)
2. Remove Neutrophils and Tregs and de-clog cancer (remove non-functional immune cells and other cancer supportive elements) to make it accessible to immune therapy and to stop building of cancer tissue
a. Mesenchymal stem cells and Neutrophils are steered by cancer G-CSF (2) b. Mesenchymal stem cell (MSC) are attracted to early stage tumor (2) c. MCS steer Neutrophils via G-CSF to become tumor supportive (2)
d. Inhibitors of G-CSF slow cancer (3)
e. Tumors with higher blood Neutrophil to Lymphocyte ratio have worse prognosis (4)
f. Patients who become neutropenic during chemotherapy have better prognosis (5)
g. Neutrophils steer dendritic cells and Tregs (6)
h. Neutrophils are key for angiogenesis, which turns cancers to fast growth via the so called angiogenic shift (7)
i. Tumor associated Macrophages (TAMs) support tumor growth (8)
j. Removing Neutrophils using an antibody, increases survival in PDX model (9) k. In order for allogenic T-cell transplantation to work, best results are achieved by combination of cyclophosphamide and radiation to remove Tregs and other immune cells (10)
3. Then shift dendritic cells and macrophages to macrophage type 1, the inflammatory kind
a. Macrophage type 2 is the most prevalent TAM and is not effective in antigen presentation (11)
b. Intervention with inhibitor of RON kinase shifts macrophages to Type 1 and increase efficacy of PD-1 therapy (12) c. Potential steering of M2 macrophage to Ml subtype with inhibition of IL-4 and 1L-13 since these gluococorticoid hormones have been shown to induce M2 Macrophage phenotype. (12)
d. Dendritic cells capture antigens from tumour and migrate into the draining lymph nodes for presentation. The antigens can be presented with MHC class I and class II molecules. (13)
e. Tumours interfere with DC maturation through IL-10 and TSLP. The DCs are skewed towards a Th2 lineage which promotes tumour development. (14) f. pDCs are also inhibited due to ligation of ILT7 with BST2 on tumour cells.
The inhibited pDCs induce CD4+ T cells to differentiate to become immunosuppressive. (15)
g. GM-CSF, in combination with other chemokines, can induce differentiation of inflammatory DCs that in turn will activate T cell functions. (16, 17, 18) h. Antibody therapy for engagement of surface molecules on DCs such as dectinl, DCSIGN/CD209 and CD40 putatively polarize DCs towards the proinflammatory phenotype. (19, 20, 21)
Secure major antigen presentation to make dendritic cells and macrophages phagocytose tumor and present tumor antigen
a. Use either of a series of technologies (radiation, chemo, oncolytic virus, cancer vaccine, toxin loaded antibodies, cancer metabolism inhibitors or nutrient pump inhibitors or other) to make tumor necrotic and allow APCs lots of material to present. (40)
b. A specific example of cancer metabolism inhibitor would be DHODH inhibitors that inhibit de novo synthesis of pyrimidines. This inhibition will specifically inhibit rapidly dividing cells. DHODH inhibition will also lead to induction of p53 and resulting apoptosis once a threshold of p53 levels is breached. (39)
c. With findings that the neoantigen repertoire for tumours vary greatly across tumours, exome sequencing for tumours can yield personalized vaccine targets based on predicted MHC binding capacity. (22, 23)
d. Neo-antigen presentation can also be greatly enhanced by blocking or inhibiting do-not-eat-me signals (CD47) on tumour cells that prevent phagocytosis of tumour cells. (24)
Use immune checkpoint inhibitor (ICI) to enhance T-cell response (or use highly targeted CAR-T construct to seek out and destroy tumor tissue
a. Single or combinations or sequences of ICIs can be used and have shown additive effects in the clinic (25) b. These ICI therapies are not limited to T cell mediators and may also affect DCs, NK, NKT cells. They are a combination of antagonists to inhibitory signals and agonists to activation signals on the immune cells. (25)
6. Eradicate remnant tumor by using either of the following technologies to specifically kill Circulating tumour cells (CTC) and Cancer Stem Cells (CSC) dormant cancer cells (DCC)
a. Oncolytic virus kills CTC (26)
b. Inhibition of ALDH by eg. disulfiram increases reactive oxygen species (ROS) and kills CSC (27)
c. Inhibition of NANOG indirectly through HIFla inhibition by eg. disulfiram may inhibit CSCs (28)
d. The phenotype of CSCs will differ depending on the cellular context. In well- validated systems, such as breast and hemaotopoietic cancers, therapies against CSC have also shown efficacy, in reducing tumour recurrence and demonstrating long-term efficacy. (29, 30, 31, 32)
e. Starting proliferation of dormant cells by eg. Lithium turns DCCs non- dormant and accessible for killing via eg. chemo, irradiation or other means. Chemotherapy and irradiation methods may only impact cycling cells. By driving cells out of GO into the cell cycle - you would be able to trigger apoptosis at cell cycle checkpoint. (33)
7. Use fast dynamic diagnostics to follow immune status in the tumor and direct therapy
a. Tumors signal via exosomes in the circulation to seeding areas about status and to steer grafting probabilities (34)
b. Thrombocytes carry RNA and other tumor markers that tell status in tumor (35)
c. Survey both CTC and circulating immune cell subsets to gain an idea about heightened nature of immune system in response to tumour, as well as, resolution of immune response. (36)
d. This is particularly important as several immune checkpoint therapies have demonstrated pseudo-progression, increase in tumour size via RECIST due to immune cell infiltration and inflammation. The anti-tumour effect is only visible after several weeks. Hence it would be critical to have a dynamic marker of immune activity that can be read out at earlier timepoints. (37) e. Use combination of RNA, DNA, cell and immune diagnostics to determine response to therapy and best next steps (38)
Tumors recruit a number of cells and they are either supporting cells, or just bystanders, which are likely to physically block access to the cancer. It may also be that these supporting or bystander cells, feed of fuel the cancer cell growth. Therefore we hypothesise therapy should focus on removing these before shifting to changing the macrophage and dendritic cell infiltration.
In turn changing the macrophage and dendritic cell set up is important to perform before applying ICIs since these activated Ml and active dendritic cells can provide immunogenic material to the T-cells.
Generally it is useful in the present method that the immunostimulation, by the means of making antigens available en masse, to the macrophages and the dendritic cells, should occur after the macrophages and the dendritic cells have been activated, and should precede application of the immune checkpoint inhibitors.
Killing circulating tumor cells and cancer stem cells and dormant cancer cells is described as the last step. However this step can be applied at initiation and/or during the subsequent steps, and may be repeated together with immune reactivation at subsequent sessions.
The timely feedback of the status of the tumor is important, since this multi-stage immuno, pathway and tumor escape/survival directed therapy needs to be steered and directed. For example daily measures of anti-tumour activity may enable one to be able to direct the therapy better (i.e. understanding when the neutrophils are down and depleted, and when the myeloid derived suppressor cells have been turned off, and when the macrophages have turned to Ml phenotype and how the immune checkpoint inhibition is successful in attacking the tumor).
Additionally, the detection of circulating tumor cells and RNA and DNA products from the tumor provides an indication whether the strategies to kill cancer stem cells have been successful.
Furthermore, the diagnosis of autoimmune reactions early allowing early intervention will be key, since several of the immunostimulation combos and efforts will elicit autoimmune reactions that need to be staved off quickly. The learning from this monitoring is likely to be important since individuals' response to these combined therapies may be highly specific both in kind, quality, quantity and timing.
The method is different from other methods in that it foresees usage of key therapies for a limited period of time, for a therapy period of less than 2 months is envisioned for one or more stages. This therapy in parts or in full may need to be repeated at a later stage, potentially with changes that may take instruction from the ongoing immune and cancer diagnostics.
In one embodiment we expect that a 2 week period of proliferation and nutrient/metabolic inhibition may be sufficient before taking down neutrophils and debulking of tumor. This step may take a week, followed by macrophage/dendritic cell type change (3-7 days), and then turning on the antigen production and presentation (1 to 2 weeks) before turning on the immune defense with T-cells and NK cells via the immune checkpoint inhibitors for at least 2 weeks. During the latter period, if not before, the killing of circulating and dormant cancer cells should be started, along with attempts to eradicate cancer stem cells.
This new method represents a way to declare total war against the cancer and is believed to cause maximal effect in the shortest period of time, while keeping side effects to a minimum. It also is a new way to try to learn as much possible about the specific cancer and to be able to adapt therapy to achieve best effect and avoid side effects.
The method should be meaningful both in early cancer (e.g. neo-adjuvant setting pre surgery) as well as a treatment alternative later in the cancer development.
The tumor mass and whether or not mass metastasis has occurred will be key factors whether or not the therapy will be effective. However, given the reported cases of durable responses in melanoma with immune checkpoint inhibitors, even after many lines of therapy, make it possible to believe in efficacy even at protracted and far progressed disease.
Treatment of cancer as employed herein refers to administration of the combination therapy of the present disclosure for the purpose of reducing symptoms, ameliorating symptoms, slowing progressing (including spreading) of the cancer or a form thereof (such as metastatic cancer), stopping progression (including spreading) of the cancer or a form thereof (such as metastatic cancer), shrinking tumors, reducing metastases, reducing or eliminating certain populations of cancer cells, for example cancer stem cell populations, sending the cancer into remission, maintaining the cancer in remission, prophylactic treatment of cancer, curing cancer or a combination of one or more the same, as appropriate.
Treatment of cancer as employed herein also refers to administration of the combination therapy of the present disclosure for the purpose of enhancing concomitant anti-cancer therapy, and for the purpose (at least in part) to reduce side effects and/or reducing the dose required of concomitant anti-cancer therapy.
Lithium compound as employed herein refers to an active agent employed in therapy comprising the alkali metal lithium, for example lithium salts comprising a counter-ion such as selected from the group comprising aspartate, bromide, chloride, citrate, lithium carbonate, lithium sulfate and orotate.
Lithium salts are well established treatments for mood disorders. The main side effects are taste derangement, tremor and kidney side effects. However, these side effects are much less toxic than those encountered by patients receiving normal traditional cancer therapy, and the doses employed for cancer therapy may be lower than those employed for the treatment of mood disorders, especially when the lithium is employed in combination of disulfiram.
It is common clinical practice to monitor plasma levels of lithium, so that the likelihood of toxicity is reduced. Therefore, in one embodiment the patient receiving the combination therapy of the present disclosure is monitored for plasma levels of lithium, for example when the treatment is started and/or on a monthly, three monthly, six monthly or annual basis.
Thus in one embodiment the dose of lithium employed is a dose that does not elicit side effects in the majority of patients.
The majority of patients as employed herein refers to wherein at 51% of patients who receive that dose do not encounter therapy limiting side effects or only encounter minor side effects associated with the lithium such as 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100% of patients do not encounter therapy limiting side effects.
Minor side effects as employed herein are effects that a negligible i.e. that do not significantly impact on the patients' quality of life and thus at most are slightly inconvenient.
It is believed by the present inventors that the lithium acts in cancer cells (for example cancer cells which are differentiated, i.e. non-cancer-stem-cells) at least via inhibiting the enzyme glycogen synthase kinase-3 (GSK-3), which is linked to the insulin and the Wnt pathways. The inhibition of GSK-3 may be associated with antiproliferative effects. However, lithium may also be linked to hormone receptors such as estrogen and androgen. Thus other effects of lithium may be exerted on bone marrow and on adaptive and innate immune defenses which are not necessarily associated to the GSK3 mediated effects.
Paradoxically, lithium seems to increase the proliferation and self-renewal of stem cells, including cancer stem cells. That could be seen as a negative in terms of cancer therapy. However, in this instance it may be positive because it prevents the cancer cells evading treatment.
Thus lithium driving proliferation and self-renewal prevents the cancer cells becoming dormant. The latter is thought to be a mechanism for cancer cell resistance. Thus preventing the option for the cancer cells to become dormant may enhance immediate efficacy of cancer therapy and may allow more or all of the cancer cell populations present to be treated. This in turn may improve the effectiveness of the cancer therapy and/or reduce the risk for cancer relapse.
Furthermore, self-renewal may also mean that the effectiveness of concomitant therapies (such as traditional cancer therapies or therapies directed to cancerous at renewal mechanisms) may have an increased therapeutic window or have a therapeutic window which is shifted towards more "selectively" effecting cancer cells (or certain types of cancer cells, such as cancer stem cells).
Another mechanism of lithium efficacy may be on the bone marrow and the T- cells, which through the inhibition of GSK3 may become more effective, for example providing prolonged T cell proliferation and increased IL-2 production. Thus, there may be immune-stimulatory effects provided by employing the combination therapy of the present disclosure which renders the patient more receptive to therapy, for example in the form of immune-oncology compounds.
Immune-oncology compounds also referred to as cancer immune therapy herein refers to therapies that target tumor associated antigens (TAAs) and stimulate immune responses to the cancer.
Thus lithium and disulfiram may work additively or in synergy, to decrease the survival of cancer cells, such as cancer stem cells.
Disulfiram inhibits the aldehyde dehydrogenase, which is a key marker for cancer stem cells, and is a key enzyme for defense against reactive oxygen species (ROS), which may otherwise harm the cancer stem cell. Thus disulfiram may cause cancer stem cell reduction, for example via accumulation of reactive oxygen species leading to necrosis, apoptosis or autophagy thereby increasing death of cancer stem cells.
Necrosis as employed herein refers to cell death through injury or damage to the cell.
Apoptosis as employed herein refers to hastening programmed cell death, which is sometimes referred to as cell assisted suicide.
Autophagy as employed herein refers to a complex process of intracellular degradation, which may deliver nutrients and/or energy to other cells.
We hypothesize that the combination therapy of the present disclosure makes it difficult for cancer cells to stay in quiescent dormant mode, for example by provoking self-renewal stimulated by the lithium part of the combined therapy. This renders the cancer stem cells vulnerable to cancer treatments, either via the disulfiram part or by other anti-cancer therapeutics.
However, if the cancer cells manage to stay in quiescent dormant mode the disulfiram part of the therapy may be deleterious to them through, for example the accumulation of reactive oxygen species (ROS).
Reactive oxygen species (ROS) as employed herein refers to peroxides, superoxides, hydroxyl radicals, singlet oxygen and the like. The accumulation of reactive oxygen specifies causes significant damage to cell structure, which ultimately is likely to cause cell death. Disulfiram (DSF) is a widely used anti addiction medicine (ethanol addiction). Its main target is aldehyde dehydrogenase, and its' inhibition causes upstream accumulation of ethanol aldehyde which is linked to physical discomfort.
Aldehyde dehydrogenase is also a marker of cancer stem cells and the present inventors believe that disulfiram via inhibition of aldehyde dehydrogenase increases the concentration of reactive oxygen species. This increased concentration of reactive oxygen species in turn damages the cancer stem cells. Thus is hypothesized that DSF suppresses self-renewal capability mainly through the activation of reactive oxygen species (ROS)-p38 MAPK pathway.
In addition disulfiram, via inhibition of SOD-1, is thought to inhibit the release of
TNF-alpha, vascular endothelial growth factor, and MMP-2 and MMP-9 from cultured macrophages, which may contribute to the overall effectiveness of the combination therapy.
It is believed that by attacking cancers cell via two independent pathways, using well tolerated drugs, and particularly the ability to target cancer stems cells will lead to significant improvements of cancer treatment.
In addition, effects of lithium on the bone marrow and on the immune system may add to the breadth of potential positives of the therapy of the present disclosure.
Furthermore it may be that the combination therapy of the present disclosure has beneficial effects in protecting non-cancerous tissue from the toxic effect of cancer therapy that may be employed in combination with the therapy comprising disulfiram and a lithium compound.
Combination treatment as employed herein refers to a treatment wherein the duration of time when the patient is on or taking the medicament comprising a lithium compound is also the time when the patient is taking or is administered a medicament comprising the disulfiram (i.e. treatment regimens for lithium and disulfiram at least overlap).
In one embodiment a lithium compound and a disulfiram are administered or taken in the same 7 days period. In one embodiment the disulfiram is administered or taken on the same day as the lithium compound.
In one embodiment the combination therapy of the present disclosure is employed as adjuvant therapy, for example post-surgery or post-chemotherapy.
In one embodiment the combination therapy of the present disclosure is employed as neo-adjuvant therapy.
The combination treatment of the present disclosure may be employed in combination with one or more further cancer treatments, for example
• concomitant with chemotherapy (including where chemotherapy follows radiation therapy), or • concomitantly with radiation therapy (including where radiation therapy follows chemotherapy)
• concomitant with chemotherapy and radiation therapy
• concomitant with immune-modulating agents such as PD-1 inhibitors and IDOl- inhibitors and agents active on the macrophage type shift (type 2 to type 1 shift)
In one embodiment the therapy may reduce the side effects of such and may allow usage of higher and more effective doses of other cancer therapies to be employed and tolerated.
A suitable dose may be chosen by the practitioner based on the nature of the cancer being treated.
Chemotherapeutic agent as employed herein is intended to refer to specific antineoplastic chemical agents or drugs that are selectively destructive to malignant cells and tissues. Chemotherapeutic agent and chemotherapy or cytotoxic agent are employed interchangeably herein unless the context indicates otherwise. Examples inlcude alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents. Other examples of chemotherapy include doxorubicin, 5-fluorouracil (5-FU), paclitaxel, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
Examples of alkylating agents, which may be employed in the method of the present disclosure include an nitrogen mustards alkylating agent, nitrosoureas, tetrazines, aziridines, platins and derivatives, and non-classical alkylating agents.
Examples of a platinum containing chemotherapeutic agent (also referred to as platins), such as cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin and lipoplatin (a liposomal version of cisplatin), in particular cisplatin, carboplatin and oxaliplatin.
The dose for cisplatin ranges from about 20 to about 270 mg/m2 depending on the exact cancer. Often the dose is in the range about 70 to about 100mg/m2.
The dose for cisplatin ranges from about 20 to about 270 mg/m2 depending on the exact cancer. Often the dose is in the range about 70 to about 100mg/m2.hemotherapeutic agent is a topoisomerase inhibitor.
Nitrogen mustards include mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide and busulfan. Nitrosoureas include N-Nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU) and semustine (MeCCNU), fotemustine and streptozotocin. Tetrazines include dacarbazine, mitozolomide and temozolomide.
Aziridines include thiotepa, mytomycin and diaziquone (AZQ).
Examples of antimetabolites, which may be employed in the method of the present disclosure, include anti-folates (for example methotrexate and pemetrexed), purine analogues (for example thiopurines, such as azathiopurine, mercaptopurine, thiopurine, fludarabine (including the phosphate form), pentostatin and cladribine), pyrimidine analogues (for example fluoropyrimidines, such as 5-fluorouracil and prodrugs thereof such as capecitabine [Xeloda®]), floxuridine, gemcitabine, cytarabine, decitabine, raltitrexed(tomudex) hydrochloride, cladribine and 6-azauracil.
Examples of anthracyclines, which may be employed in the method of the present disclosure, include daunorubicin (Daunomycin), daunorubicin (liposomal), doxorubicin (Adriamycin), doxorubicin (liposomal), epirubicin, idarubicin, valrubicin currenlty used only to treat bladder cancer and mitoxantrone an anthracycline analog, in particular doxorubicin.
Examples of anti-microtubule agents, which may be employed in the method of the present disclosure, include include vinca alkaloids and taxanes.
Vinca alkaloids include completely natural chemicals for example vincristine and vinblastine and also semi-synthetic vinca alkaloids, for example vinorelbine, vindesine, and vinflunine.
Taxanes include paclitaxel, docetaxel, abraxane, carbazitaxel and derivatives of thereof. Derivatives of taxanes as employed herein includes reformulations of taxanes like taxol, for example in a micelluar formulaitons, derivatives also include chemical derivatives wherein synthetic chemistry is employed to modify a starting material which is a taxane.
Topoisomerase inhibitors, which may be employed in a method of the present disclosure include type I topoisomerase inhibitors, type II topoisomerase inhibitors and type II topoisomerase poisons. Type I inhibitors include topotecan, irinotecan, indotecan and indimitecan. Type II inhibitors include genistein and ICRF 193 which has the following structure:
Figure imgf000027_0001
Type II poisons include amsacrine, etoposide, etoposide phosphate, teniposide and doxorubicin and fluoroquinolones.
In one embodiment he chemotherapeutic agent is a parp inhibitor, in particular as disclosed herein.
In one embodiment the combination therapy is administered with or following a treatment regime comprising a platinum containing chemotherapeutic agent, for example cisplatin, carboplatin or oxaliplatin. In one embodiment a combination of chemotherapeutic agents employed is, for example a platin and 5-FU or a prodrug thereof, for example cisplatin or oxaplatin and capecitabine or gemcitabine, such as FOLFOX.
In one embodiment the chemotherapy comprises a combination of chemotherapy agents, in particular cytotoxic chemotherapeutic agents.
In one embodiment the chemotherapy combination comprises a platin, such as cisplatin and fluorouracil or capecitabine.
In one embodiment the chemotherapy combination in capecitabine and oxaliplatin (Xelox).
In one embodiment the chemotherapy is a combination of folinic acid and 5-FU, optionally in combination with oxaliplatin.
In one embodiment the chemotherapy is a combination of folinic acid, 5-FU and irinotecan (FOLFIRI), optionally in combination with oxaliplatin (FOLFIRINOX). The regimen consists of: irinotecan (180 mg/m2 IV over 90 minutes) concurrently with folinic acid (400 mg/m2 [or 2 x 250 mg/m2] IV over 120 minutes); followed by fluorouracil (400-500 mg/m2 IV bolus) then fluorouracil (2400-3000 mg/m2 intravenous infusion over 46 hours). This cycle is typically repeated every two weeks.
The dosages shown above may vary from cycle to cycle.
In one embodiment the combination employs a microtubule inhibitor, for example vincristine sulphate, epothilone A, N-[2-[(4-Hydroxyphenyl)amino]-3- pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), ataxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
In one embodiment the combination employs an mTor inhibitor. Examples of mTor inhibitors include: everolimus (RADOOl), WYE-354, KU-0063794, papamycin (Sirolimus), Temsirolimus, Deforolimus(MK-8669), AZD8055 and BEZ235(NVP-
BEZ235).
In one embodiment the chemotherapy combination employs a microtubule inhibitor, for example vincristine sulphate, epothilone A, N-[2-[(4- Hydroxyphenyl)amino]-3-pyridinyl]-4-methoxybenzenesulfonamide (ABT-751), a taxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
In one embodiment the chemotherapy combination employs a MEK inhibitor. Examples of MEK inhibitors include: AS703026, CI-1040 (PD184352), AZD6244 (Selumetinib), PD318088, PD0325901, AZD8330, PD98059, U0126-EtOH, BIX 02189 or BIX 02188.
In one embodiment the chemotherapy combination employs an AKT inhibitor. Examples of AKT inhibitors include: MK-2206 and AT7867. In one embodiment the combination employs an aurora kinase inhibitor. Examples of aurora kinase inhibitors include: Aurora A Inhibitor I, VX-680, AZD1152- HQPA (Barasertib), SNS-314 Mesylate, PHA-680632, ZM-447439, CCT129202 and Hesperadin.
In one embodiment the chemotherapy combination employs a p38 inhibitor, for example as disclosed in WO2010/038086, such as N-[4-({4-[3-(3-tert-Butyl-l-p-tolyl- lH-pyrazol-5-yl)ureido]naphthalen-l-yloxy}methyl)pyridin-2-yl]-2-methoxyacetamide.
In one embodiment the combination employs a Bcl-2 inhibitor. Examples of Bcl- 2 inhibitors include: obatoclax mesylate, ABT-737, ABT-263(navitoclax) and TW-37.
In one embodiment the chemotherapy combination comprises an antimetabolite such as capecitabine (xeloda), fludarabine phosphate, fludarabine (fludara), decitabine, raltitrexed (tomudex), gemcitabine hydrochloride and cladribine.
In one embodiment the chemotherapy combination comprises ganciclovir, which may assist in controlling immune responses and/or tumour vasculation.
Cancer Types In more Detail
In one embodiment the gastric cancer is selected from the group comprising adenocarcinoma of the stomach, squamous cell carcinomas, lymphoma of the stomach, gastric stromal tumour, and neuroendocrine tumours.
In one embodiment the liver cancer is, for example selected from the group hepatocellular carcinoma, cholangiocarcinoma, angiosarcoma, and hepatoblastoma, in particular hepatocellular carcinoma. In one embodiment the primary liver cancer is stage 1, 2, 3 or 4. In one embodiment the liver cancer is secondary or metastasized liver cancer.
Prostate cancer as employed herein refers to cancer of the prostate, for example ductal adenocarcinoma, transitional cell (urothelial cancer), squamous cell cancer, carcinoid of the prostate, small cell cancer or sarcoma and sarcomatoid cancer.
Pancreatic cancer as employed herein includes exocrine cancers (including rare forms thereof such as cystitic tumours, and cancer of the acinar cells), endocrine pancreatic tumours (including gastrinomas, insulinomas, somatostatinomas, VIPomas, glucagonomas), pancreatoblastoma, sarcomas of the pancreas and lymphoma.
Biliary tract cancer as employed herein refers to cholangiocarcinoma (intrahepatic, extrahepatic), gall bladder cancer and ampullary carcinoma.
Colorectal cancer as employed herein refers to cancer or the colon and/or rectum and includes squamous cell cancers, carcinoid tumours, sarcomas and lymphomas.
Breast cancer as employed herein refers to cancer of the breast and includes ductal carcinoma in situ, lobular carcinoma in situ, invasive ductal breast cancer, invasive lobular breast cancer, invasive breast cancer, Paget's disease, angiosarcoma of the breast and rare types of breast cancer such as medullary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast metaplastic breast cancer, basal type breast cancer and papillary breast cancer.
Lung cancers are classified according to histological type and are categorized by the size and appearance of the malignant cells seen by a histopathologist under a microscope. For therapeutic purpose, two broad classes are distinguished: non-small cell lung carcinoma and small cell lung carcinoma.
In one embodiment the epithelial cancer is lung cancer, for example small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC).
Non-small-cell lung carcinoma-The three main subtypes of NSCLC are adenocarcinoma, squamous-cell carcinoma and large-cell carcinoma.
Nearly 40% of lung cancers are adenocarcinoma, which usually originates in peripheral lung tissue. A subtype of adenocarcinoma, the bronchioloalveolar carcinoma, is more common in female never-smokers, and may be associated with a better long term survival.
Squamous-cell carcinoma accounts for about 30% of lung cancers. They typically occur close to large airways. A hollow cavity and associated cell death are commonly found at the center of the tumor. About 9% of lung cancers are large-cell carcinoma. These are so named because the cancer cells are large, with excess cytoplasm, large nuclei and conspicuous nucleoli.
Small-cell lung carcinoma-In small-cell lung carcinoma (SCLC), the cells contain dense neurosecretory granules (vesicles containing neuroendocrine hormones), which give this tumor an endocrine/paraneoplastic syndrome association. Most cases arise in the larger airways (primary and secondary bronchi). These cancers grow quickly and spread early in the course of the disease. Sixty to seventy percent have metastatic disease at presentation.
In one embodiment the cancer is non-small lung carcinoma.
In one embodiment the cancer is liver cancer, for example a liver metastasis from a primary cancer, for example colon cancer, which has spread to the liver. In one embodiment the liver cancer is HCC hepatocellular carcinoma.
In one embodiment there is provided treatment of renal cancer, for example renal cell carcinoma and/or urothelial cell carcinoma. Other examples of renal cancer include squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma, Bellini duct carcinoma, clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma, and carcinoid tumor of the renal pelvis. In one embodiment the cancer is bladder cancer, for example is any of several types of malignancy arising from the epithelial lining (i.e. the urothelium) of the urinary bladder. About 90% of bladder cancers are transitional cell carcinoma. The other 10% are squamous cell carcinoma, adenocarcinoma, sarcoma, small cell carcinoma, and secondary deposits from cancers elsewhere in the body. The staging of this cancer is given below.
T (Primary tumour)
• TX Primary tumour cannot be assessed
• TO No evidence of primary tumour
· Ta Non-invasive papillary carcinoma
• Tis Carcinoma in situ ('flat tumour')
• Tl Tumour invades subepithelial connective tissue
• T2a Tumour invades superficial muscle (inner half)
• T2b Tumour invades deep muscle (outer half)
· T3 Tumour invades perivesical tissue:
• T3a Microscopically
• T3b Macroscopically (extravesical mass)
• T4a Tumour invades prostate, uterus or vagina
• T4b Tumour invades pelvic wall or abdominal wall
N (Lymph nodes)
• NX Regional lymph nodes cannot be assessed
• NO No regional lymph node metastasis
• Nl Metastasis in a single lymph node 2 cm or less in greatest dimension
• N2 Metastasis in a single lymph node more than 2 cm but not more than 5 cm in greatest dimension, or multiple lymph nodes, none more than 5 cm in greatest dimension
• N3 Metastasis in a lymph node more than 5 cm in greatest dimension
M (Distant metastasis)
• MX Distant metastasis cannot be assessed
· M0 No distant metastasis
• Ml Distant metastasis.
The current disclosure extends to any stage of bladder cancer.
There are more than 30 different types of ovarian cancer which are classified according to the type of cell from which they start. Cancerous ovarian tumours can start from three common cell types:
• Surface Epithelium - cells covering the lining of the ovaries
• Germ Cells - cells that are destined to form eggs • Stromal Cells - Cells that release hormones and connect the different structures of the ovaries
The present disclosure relates to treatment of ovarian cancer from any source, for example as described herein, in particular epithelium cells. Epithelial ovarian carcinomas (EOCs) account for 85 to 90 percent of all cancers of the ovaries.
Common Epithelial Tumors - Epithelial ovarian tumors develop from the cells that cover the outer surface of the ovary. Most epithelial ovarian tumors are benign (noncancerous). There are several types of benign epithelial tumors, including serous adenomas, mucinous adenomas, and Brenner tumors. Cancerous epithelial tumors are carcinomas - meaning they begin in the tissue that lines the ovaries. These are the most common and most dangerous of all types of ovarian cancers. Unfortunately, almost 70 percent of women with the common epithelial ovarian cancer are not diagnosed until the disease is advanced in stage.
There are some ovarian epithelial tumors whose appearance under the microscope does not clearly identify them as cancerous. These are called borderline tumors or tumors of low malignant potential (LMP tumors). The method of the present disclosure includes treatment of the latter.
Germ Cell Tumors - Ovarian germ cell tumors develop from the cells that produce the ova or eggs. Most germ cell tumors are benign (non-cancerous), although some are cancerous and may be life threatening. The most common germ cell malignancies are maturing teratomas, dysgerminomas, and endodermal sinus tumors. Germ cell malignancies occur most often in teenagers and women in their twenties. Today, 90 percent of patients with ovarian germ cell malignancies can be cured and their fertility preserved.
Stromal Tumors - Ovarian stromal tumors are a rare class of tumors that develop from connective tissue cells that hold the ovary together and those that produce the female hormones, estrogen and progesterone. The most common types are granulosa- theca tumors and Sertoli-Leydig cell tumors. These tumors are quite rare and are usually considered low-grade cancers, with approximately 70 percent presenting as Stage I disease (cancer is limited to one or both ovaries).
Primary Peritoneal Carcinoma-The removal of one's ovaries eliminates the risk for ovarian cancer, but not the risk for a less common cancer called Primary Peritoneal Carcinoma. Primary Peritoneal Carcinoma is closely rated to epithelial ovarian cancer (most common type). It develops in cells from the peritoneum (abdominal lining) and looks the same under a microscope. It is similar in symptoms, spread and treatment. Stages of Ovarian Cancer
Once diagnosed with ovarian cancer, the stage of a tumor can be determined during surgery, when the doctor can tell if the cancer has spread outside the ovaries. There are four stages of ovarian cancer - Stage I (early disease) to Stage IV (advanced disease). The treatment plan and prognosis (the probable course and outcome) will be determined by the stage of cancer.
Following is a description of the various stages of ovarian cancer:
Stage I - Growth of the cancer is limited to the ovary or ovaries.
Stage IA - Growth is limited to one ovary and the tumor is confined to the inside of the ovary. There is no cancer on the outer surface of the ovary. There are no ascites present containing malignant cells. The capsule is intact.
Stage IB - Growth is limited to both ovaries without any tumor on their outer surfaces. There are no ascites present containing malignant cells. The capsule is intact.
Stage IC - The tumor is classified as either Stage IA or IB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries;
(2) the capsule has ruptured; and (3) there are ascites containing malignant cells or with positive peritoneal washings.
Stage II - Growth of the cancer involves one or both ovaries with pelvic extension.
Stage IIA - The cancer has extended to and/or involves the uterus or the fallopian tubes, or both.
Stage IIB - The cancer has extended to other pelvic organs.
Stage IIC - The tumor is classified as either Stage IIA or IIB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries; (2) the capsule has ruptured; and (3) there are ascites containing malignant cells or with positive peritoneal washings.
Stage III - Growth of the cancer involves one or both ovaries, and one or both of the following are present: (1) the cancer has spread beyond the pelvis to the lining of the abdomen; and (2) the cancer has spread to lymph nodes. The tumor is limited to the true pelvis but with histologically proven malignant extension to the small bowel or omentum.
Stage IIIA - During the staging operation, the practitioner can see cancer involving one or both of the ovaries, but no cancer is grossly visible in the abdomen and it has not spread to lymph nodes. However, when biopsies are checked under a microscope, very small deposits of cancer are found in the abdominal peritoneal surfaces.
Stage IIIB - The tumor is in one or both ovaries, and deposits of cancer are present in the abdomen that are large enough for the surgeon to see but not exceeding 2 cm in diameter. The cancer has not spread to the lymph nodes.
Stage IIIC - The tumor is in one or both ovaries, and one or both of the following is present: (1) the cancer has spread to lymph nodes; and/or (2) the deposits of cancer exceed 2 cm in diameter and are found in the abdomen. Stage IV - This is the most advanced stage of ovarian cancer. Growth of the cancer involves one or both ovaries and distant metastases (spread of the cancer to organs located outside of the peritoneal cavity) have occurred. Finding ovarian cancer cells in pleural fluid (from the cavity which surrounds the lungs) is also evidence of stage IV disease.
In one embodiment the ovarian cancer is: type I, for example IA, IB or IC; type II, for example IIA, IIB or IIC; type III, for example IIIA, IIIB or III C; or type IV.
In one embodiment the breast cancer is one selected from the group comprising ductal carcinoma in situ, lobular carcinoma in situ, invasive breast cancer, invasive lobular breast cancer, Paget's disease, angiosarcoma of the breast, medulllary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast, metaplastic breast cancer, lymphoma of the breast, basal type breast cancer, phyllodes or cystosarcoma phyllodes and papillary breast cancer.
In one embodiment the prostate cancer is selected from the group comprising ductal adenocarcinoma, transitional cell (urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers.
In one embodiment the prostate cancer is selected from the group comprising ductal adenocarcinoma, transitional cell (urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers and a combination of two or more of the same.
Thyroid cancer as employed herein refers to cancer of the thyroid originating from follicular or parafollicular thyroid cells and includes papillary thyroid cancer (75% to 85% of cases); follicular thyroid cancer (10% to 20% of cases); medullary thyroid cancer (5% to 8% of cases)- cancer of the parafollicular cells, often part of multiple endocrine neoplasia type 2; poorly differentiated thyroid cancer; anaplastic thyroid cancer (less than 5% of cases) is not responsive to treatment and can cause pressure symptoms, thyroid lymphoma, squamous cell thyroid carcinoma, sarcoma of thyroid.
Renal cancer as employed herein refers to cancer of the kidney, for example renal cell carcinoma and transitional cell carcinoma of the renal pelvis, such as squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma, bellini duct carcinoma, clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma; carcinoid tumor of the renal pelvis.
Bladder cancer as employed herein refers to cancer of the bladder including transitional cell bladder cancer, carcinoma in situ, papillary cancer and rarer types of bladder cancer such as squamous cell cancer, adenocarcinoma and a combination of two or more of the same. Esophageal cancer as employed herein refers to cancer of the oesphagus including esophageal squamous-cell carcinomas, esophageal adenocarcinomas, and variants of squamous-cell carcinoma, and non-epithelial tumors, such as leiomyosarcoma, malignant melanoma, rhabdomyosarcoma, lymphoma, among others and a combination of two or more of the same.
Head and neck cancer as employed herein refers to cancer of the neck and/or head, including mouth cancer, nasopharyngeal cancer, oropharyngeal cancer, paranasal sinus cancer, salivary gland cancer and a combination of two or more of the same.
In one embodiment the chemotherapy combination comprises ganciclovir, which may assist in controlling immune responses and/or tumour vasculation.
In the context of this specification "comprising" is to be interpreted as "including".
Aspects of the invention comprising certain elements are also intended to extend to alternative embodiments "consisting" or "consisting essentially" of the relevant elements.
Where technically appropriate, embodiments of the invention may be combined. Embodiments are described herein as comprising certain features/elements. The disclosure also extends to separate embodiments consisting or consisting essentially of said features/elements.
Technical references such as patents and applications are incorporated herein by reference.
Any embodiments specifically and explicitly recited herein may form the basis of a disclaimer either alone or in combination with one or more further embodiments. REFERENCES (all incorporated herein by reference)
1. Clin Lab. 2016;62(3) :409-16. SNP rsl059234 in CDKN1A Gene Correlates with
Prognosis in Resected Gastric Adenocarcinoma. Nie X, Wang X, Lin Y, Yu Y, Liu W, Xie F, Wang X, Tan J, Huang Q.
2. Proc Natl Acad Sci U S A. 2015 Feb 10; 112(6) : E566-E575. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils Amy-Jo Casbon,a Damien Reynaud,b Chanhyuk Park,a Emily Khuc,a Dennis D. Gan,a Koen Schepers,b Emmanuelle Passegue,b and Zena Werba,l
3. Oncoimmunology. 2015 Nov 24;5(3) :elll5177. eCollection 2016. G-CSF regulates macrophage phenotype and associates with poor overall survival in human triple- negative breast cancer. Hollmen Ml, Karaman SI, Schwager SI, Lisibach Al, Christiansen AJ1, Maksimow M2, Varga Z3, Jalkanen S2, Detmar Ml. 4 Semin Immunol. 2016 Apr 7. pii: S1044-5323(16)30004-5. Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Moses Kl, Brandau S2.
5. Med Oncol. 2014 Jun;31(6):969. Neutrophil-lymphocyte ratio as a predictor of outcomes for patients with hepatocellular carcinoma undergoing TAE combined with Sorafenib. Wei Kl, Wang M, Zhang W, Mu H, Song TQ.
6. Cell Death Dis. 2014 Jun; 5(6): el295. The IL-6-STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression Q Zhu,l,4 X Zhang,l,4,* L Zhang,l W Li,2 H Wu,2 X Yuan,l F Mao,l M Wang.1 W Zhu,l H Qian,l and W Xul,3,*
7. Proc Natl Acad Sci U S A. 2006 Aug l5; 103(33) : 12493-12498; Published online 2006 Aug 4. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis Hiroaki Nozawa,*† Christopher Chiu,* and Douglas Hanahand:
8. Immunology, 2013 Oct; 5(1): 1075-87. Targeting tumor-infiltrating macrophages to combat cancer Roheena Z Panni,l David C Linehan,l,2 and David G
DeNardo*,2,3,4
9. J Exp Med. 1995;181:435-440. Inhibition of tumor growth by elimination of
granulocytes. Pekarek LA, Starr BA, Toledano AY, Schreiber H.
10. Curr Opin Immunol. 2009 Apr;21(2):233-40. Adoptive cell therapy for the
treatment of patients with metastatic melanoma. Rosenberg SA1, Dudley ME.
11. Gut. 2016 Mar 24. pii: gutjnl-2015-310049. Pharmacological macrophage inhibition decreases metastasis formation in a genetic model of pancreatic cancer. Griesmann HI, Drexel C2, Milosevic N2, Sipos B3, Rosendahl J 4, Gress TM2, Michl PI.
12. J Clin Invest. 2012 Mar;122(3):787-95. doi: 10.1172/JCI59643. Epub 2012 Mar 1.
Macrophage plasticity and polarization: in vivo Veritas. Sica Al, Mantovani A.
13. Annu Rev Immunol. 2000;18:767-811. Immunobiology of dendritic cells.
Banchereau Jl, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K.
14. J Exp Med. 2007 May 14;204(5):1037-47. Epub 2007 Apr 16. Breast cancer
instructs dendritic cells to prime interleukin 13-secreting CD4+ T cells that facilitate tumor development. Aspord CI, Pedroza-Gonzalez A, Gallegos M, Tindle S, Burton EC, Su D, Marches F, Banchereau J, Palucka AK.
15. J Exp Med. 2009 Jul 6;206(7):1603-14. Regulation of TLR7/9 responses in
plasmacytoid dendritic cells by BST2 and ILT7 receptor interaction. Cao Wl, Bover L, Cho M, Wen X, Hanabuchi S, Bao M, DB, Wang YH, Shaw JL, Du Q, Li C, Arai N, Yao
Z, Lanier LL, Liu YJ. 16. J Leukoc Biol. 1998 Sep;64(3):358-67. Interferon-alpha and granulocyte- macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells. Paquette RL1, Hsu NC, Kiertscher SM, Park AN, Tran L, Roth MD, Glaspy J A.
17. J Immunol.2003 Sep l;171(5):2262-9. TNF skews monocyte differentiation from macrophages to dendritic cells. Chomarat P1, Dantin C, Bennett L, Banchereau J, Palucka AK.
18. J Exp Med. 2001 Oct l;194(7):1013-20. Interleukin 15 skews monocyte
differentiation into dendritic cells with features of Langerhans cells.
Mohamadzadeh M1, Berard F, Essert G, Chalouni C, Pulendran B, Davoust J, Bridges
G, Palucka AK, Banchereau J.
19. PLoS One. 2010 Oct 18;5(10): el3418. Human dendritic cells activated via dectin-1 are efficient at priming Thl7, cytotoxic CD8 T and B cell responses. Agrawal S1, Gupta S, Agrawal A.
20. Blood. 2005 Aug 15;106(4):1278-85. Epub 2005 May 5. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via ahumanized anti-DC-SIGN antibody. Tacken PJ1, de Vries IJ, Gijzen K, Joosten B, Wu D, Rother RP, Faas SJ, Punt CJ, Torensma R, Adema GJ, Figdor CG.
21. Nature. 1998 Jun 4;393(6684):480-3. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Schoenberger SP1, Toes RE, van der Voort
EI, Offringa R, Melief CJ.
22. Oncoimmunology. 2014 Jun 25;3:e29311. eCollection 2014; Personal neoantigen cancer vaccines: The momentum builds. Fritsch EF1, Hacohen N2, Wu CJ3.
23. J Clin Invest. 2015 Sep;125(9):3413-21. Tumor neoantigens: building a framework for personalized cancer immunotherapy. Gubin MM, Artyomov MN Mardis ER,
Schreiber RD.
24. Proc Natl Acad Sci U S A. 2013 Jul 2;110(27):11103-8. Anti-CD47 antibody- mediated phagocytosis of cancer by macrophages primes an effective antitumor T- cellresponse. Tseng D1, Volkmer JP, Willingham SB, Contreras-Trujillo H, Fathman JW, Fernhoff NB, Seita J, Inlay MA, Weiskopf K, Miyanishi M, Weissman IL.
25. Nature Reviews Cancer, 12, 252-264 (April 2012). Doi: 10.1038/nrc3239; The
blockade of immune checkpoints in cancer immunotherapy. Drew M Pardoll
26. Nature review Drug Discovery, September 2015 Vol 14(9): 642-662. Cancer
immunotherapy Oncolytic viruses: a new class of immunotherapy drugs, Howard L. Kaufman, Frederick J Kohlhapp & Andrew Zloza 27. Br J Cancer. 2013 Oct l;109(7): 1876-85. Disulfiram targets cancer stem-like cells and reverses resistance and cross-resistance in acquired paclitaxel-resistant triple- negative breast cancer cells. Liu PI, Kumar IS, Brown S, Kannappan V, Tawari PE, Tang JZ, Jiang W, Armesilla AL, Darling JL, Wang W.
28. Acta Pharmacol Sin. 2013 Sep;34(9):1208-16. Disulfiram deregulates HIF-a
subunits and blunts tumor adaptation to hypoxia in hepatoma cells. Park HJ1, Kim MS, Cho K, Yun JH, Choi YJ, Cho CH.
29. J Cell Mol Med. 2013 Jan; 17(1): 30-54. Hypoxia-inducing factors as master
regulators of sternness properties and altered metabolism of cancer- and
metastasis-initiating cells Murielle Mimeault* and Surinder K Batra*
30. Cancer Res. 2009 Oct 1;69(19):7507-11. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Hirsch HA1, Iliopoulos D, Tsichlis PN, Struhl K.
31. Nat Med. 2006 Oct;12(10):1167-74. Epub 2006 Sep 24. Targeting of CD44
eradicates human acute myeloid leukemic stem cells. Jin L1, Hope KJ, Zhai
Q, Smadja-Joffe F, Dick JE.
32. Cell Stem Cell. 2009 Jul 2;5(l):31-42. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Jin L1, Lee EM, Ramshaw HS, Busfield SJ, Peoppl AG, Wilkinson L, Guthridge MA, Thomas D, Barry EF, Boyd A, Gearing DP, Vairo G, Lopez AF, DickJE, Lock RB.
33. Cell Res. 2011 Oct; 21(10): 1424-1435. Lithium, an anti-psychotic drug, greatly enhances the generation of induced pluripotent stem cells; Quan Wang,l Xinxiu Xu,2 Jun Li,3 Jing Liu,4 Haifeng Gu,l Ru Zhang,2 Jiekai Chen,4 Yin Kuang,5 Jian Fei,5 Cong Jiang,6 Ping Wang,6 Duanqing Pei,4 Sheng Ding,3 and Xin Xiel,2,*
34. Cancer Metastasis Rev. 2013 Dec; 32(0): 10.1007/sl0555-013-9441-9. Exosomes in Cancer Development, Metastasis and Drug Resistance: A Comprehensive Review Asfar S. Azmi,l,* Bin Bao,l and Fazlul H. Sarkarl,2,*
35. Cancer Cell 2015 Nov 9; 28(5): 666-676. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics, Myron G. Best, Nik Sol, Irsan Kooi, Jihane Tannous, Bart A. Westerman,
Francois Rustenburg, Pepijn Schellen, Heleen Verschueren, Edward Post, Jan Koster, Bauke Ylstra, Najim Ameziane, Josephine Dorsman, Egbert F. Smit, Henk M. Verheul, David P. Noske, Jaap C. Reijneveld, R. Jonas A. Nilsson, Bakhos A. Tannousl2, Pieter Wesselingl2, Thomas Wurdingerl2,
36. J Mol Diagn. 2015 May; 17(3): 209-224; Do Circulating Tumor Cells, Exosomes, and Circulating Tumor Nucleic Acids Have Clinical Utility? A Report of the Association for Molecular Pathology Bert Gold,*†* Milena Cankovic,*:}: Larissa V. Furtado,*§ Frederick Meier,*:}: and Christopher D. Gocke*
37. J Clin Oncol. 2015 Nov 1; 33(31): 3541-3543; Pseudoprogression and Immune- Related Response in Solid Tumors. Chiou VL1, Burotto M2.
38. Oncoimmunology. 2015 Oct 29;5(3):el093277. Small cell lung cancer: Recruitment of macrophages by circulating tumor cells. Hamilton Gl, Rath B2, Klameth L2, Hochmair MJ3.
39. Oncogene, 2013 Mar 21: 32(12): 1570-1579; Inhibition of autophagy and tumor growth in colon cancer by miR-502, Haiyan Zhai, BO Song, Xiao Hu, Wei Zhu and Jingfang Ju
40. Semin Oncol. 2012, June; 39(3): 323-339; The tipping point for combination
therapy: cancer vaccines with radiation, chemotherapy or targeted small molecule inhibitors James W. Hodge, Andressa Ardiani, Benedetto Fasaci, Anna R. Kwilas and Sofia Gameiro
BRIEF DESCRIPTION OF FIGURES
Figure 1 Flow diagram showing process of the CIM-MIMEC method
Figure 2 Schematic diagram showing multimodal cancer treatment according to the present method
EXAMPLES
Example 1 Protocol for treating a cholangiocarcinoma patient.
As a pre-step as much cancerous tissue as possible is removed by surgery. The patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent (eg capecitabine or gemcitabine) for 12-28 days. Following this, AZD5069 is given as a monotherapy or concomitantly with a T-cell stimulator for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of chemotherapy comprising gemcitabine and cisplatin are given.
After chemotherapy is complete or concomitantly, treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
The final stage of the therapy is one cycle of antibody drug conjugate, wherein the antibody is specific to Epcam, potentially in combination with an immune-oncology (IO) agent such as pemrolizumab.
Example 2 Protocol for treating a cholangiocarcinoma patient.
As a pre-step as much cancerous tissue as possible is removed by surgery. The patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent (eg capecitabine or gemcitabine) for 12-28 days. Following this AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of chemotherapy comprising 5-FU and leucovorin are given.
After or concomitantly chemotherapy is complete, treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
The final stage of the therapy is one cycle of antibody drug conjugate, wherein the antibody is specific to Epcam, potentially in combination with an 10 agent such as pemrolizumab.
Example 3 Protocol for treating a cholangiocarcinoma patient.
As a pre-step as much cancerous tissue as possible is removed by surgery. The patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent
(eg capecitabine or gemcitabine) for 12-28 days. Following this AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of
chemotherapy comprising gemcitabine and cisplatin are given potentially in
combination with an IO agent such as pemrolizumab.
After chemotherapy is complete or concomitantly, treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
The final stage of the therapy targeted radiation therapy in combination with a PARP inhibitor, potentially in combination with an 10 agent such as pemrolizumab.
Example 4 Protocol for treating a cholangiocarcinoma patient.
As a pre-step as much cancerous tissue as possible is removed by surgery. The patient is then given a dose of 300-400mg bidaily of varlitinib together with a chemotoxic agent
(e.g. capecitabine or gemcitabine) for 12-28 days. Following this AZD5069 is given as a monotherapy for 4 to 6 weeks. After completing the latter 4, 5 or 6 cycles of
chemotherapy comprising 5-FU and leucovorin are given.
After chemotherapy is complete or concomitantly, treatment with permbrolizumab is given at 2 mg/Kg or 200mg every 3 weeks for 3, 6, 9, 12, 15 or 18 weeks.
The final stage of the therapy targeted radiation therapy in combination with a PARP inhibitor potentially in combination with an 10 agent such as pemrolizumab.
In all of these examples, a careful assessment of the immune status of the patient and the presence or absence of circulating tumor material like circulating tumor cells and circulating tumor DNA, is monitored on a weekly basis, and changes in the treatment regimen may be considered based on these findings.

Claims

Claims
1. A method of treating cancer comprising the steps;
a) reducing or stopping cancer proliferation,
b) debulking tumour mass,
c) activating immune cells,
d) increasing antigen presentation and/or enhancing T cell response, and
e) clear residual tumor cells.
2. A method according to claim 1, which comprises the further step of tumour
surveillance.
3. A method according to claim 1 or 2, wherein method comprises a pre-step of
surgery.
4. A method according to any one of claims 1 to 3, wherein step a) is selected from irradiation, chemotherapy, a CXCR2 antagonist and a combination of two or more the same.
5. A method according to claim 4, wherein the chemotherapy is selected from
alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents, such as doxorubicin, 5-fluorouracil (5-FU), paclitaxel, gemcitabine, capecitabine, irinotecan, and platins such as cisplatin and oxaliplatin.
6. A method according to claim 4 or 5, wherein the CXCR2 antagonist is selected from an antibody or binding fragment thereof, MK-7123, SB265610, SB 225002,
GSK1325756, AZD5069, and combinations thereof.
7. A method according to any one of claims 1 to 6, further comprising prior to step al) a step of decluttering of immune cells.
8. A method according to claim 7, wherein the decluttering is selected from treatment with a CXCR2 antagonist, an anti-GCF, cyclophosphamide, irradiation and a combination of two or more of the same.
9. A method according to claim 7 or 8, wherein the decluttering is performed by
removing specific immune cell populations, for example using medication and/or techniques such as dialysis.
10. A method according to claim 9, wherein the immune cell is selected from a
lymphocyte (such as a T cell or NKT cell), a B cell, a natural killer cell, a tumor associated macrophage, and a combination of two or more of the same, in particular a tumor associated macrophage.
11. A method according to any one of claims 4 to 8, wherein the CXCR2 antagonist is selected from an antibody or binding fragment thereof, MK-7123, SB265610, SB 225002:
Figure imgf000042_0001
N ½, GSK1325756, AZD5069, SCH-527123 and combinations thereof, in particular SCH-527123.
12. A method according to any one of claims 1 to 11, wherein the debulking in step b) is selected from treatment with an oncolytic virus, an antibody drug conjugate and a combination thereof.
13. A method according to claim 12, wherein the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus.
14. A method according to claim 13, wherein the oncolytic virus is selected from
Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep- hIL12, Cavatak™, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, Toca 511.
15. A method according to claim 13 or 14, where the antibody drug conjugate (ADCC) is selected from the group comprising AGS-15E, AGS-16C3F, AGS-22M6E, AMG-172, AMG-595, ASG-5ME, brentuximab vedotin, DEDN-6526A, DMUC-5754A,
gemtuzumab ozogamicin, glembatumumab vedotin, GSK2857916, hRS7-SN-38, IMGN-242, IMGN-289, IMGN-388, IMGN-529, IMGN-633, IMGN-853, Indatuximab, indatuximab ravtansine, Inotuzumab ozogamicin, lorvotuzumab mertansine, MEDI- 547, milatuzumab doxorubicin, MLN-0264, MLN-2704, PF-06263507, pinatuzumab vedotin, polatuzumab, PSMA-ADC, RG-7450, RG-7599, SAR-3419, SAR-566658, SC16LD6.6, SGN-15, SGN-19A, SGN-CD33A, SGN-LIV1A, trastuzumab emtansine and combinations thereof.
16. A method according to any one of claims 1 to 15, wherein activating the immune cells in step c) comprises administering a RON inhibitor, an AXL kinase inhibitor, a MER kinase agonist, a cytokine (such as IL-13), a tumour vaccine for example comprising a tumour antigen and adjuvant, a dendritic cell vaccine and a
combination of two or more of the same.
17. A method according to claim 16, wherein the RON inhibitor is N-(4-(2-Amino-3- chloropyridin-4-yloxy]-3-fluorophenyl]-4-ethoxy-l-(4-fluorophenyl]-2-oxo-l,2- dihydropyridine-3-carboxamide disclosed in WO 2008/058229.
18. A method according to any one of claims 1 to 17, wherein activating the immune cells in step c) comprises administering a check-point inhibitor.
19. A method according to claim 18, wherein the check-point inhibitor is a PD-1 or PD- Ll inhibitor.
20. A method according to any one of claims 1 to 19, wherein step d) comprises
administering a therapy from the group comprising a T cell therapy (for example where T cells are activated in vitro to generate CTLs specific to an antigen), a cell therapy comprising engineered chimeric receptors, a PD1 inhibitor, a PD-L1 inhibitors, a CTLA4 inhibitor, a LAG3 inhibitor and a combination of two or more of the same.
21. A method according to any one of claims 1 to 20, wherein step e) comprises
administering a therapy selected from the group comprising an oncolytic virus, a DLL inhibitor, ADH-1 molecular targeting therapy and combinations of two or more of the same.
22. A method according to claim 21, wherein the oncolytic virus is a virus selected from an adenovirus, herpes simplex virus, reovirus, measles virus, Newcastle disease virus, Seneca Valley virus, Vesicular stomatitis virus, polio virus, ECHO enterovirus, Coxsackie virus, and vaccinia virus.
23. A method according to claim 22, wherein the oncolytic virus is selected from
Enadenotucirev, talimogene laherparepvec, RIGVIR, Ad5-yCD/mutTKSR39rep- hIL12, Cavatak™, CG0070, DNX-2401, G207, HF10, Imlygic®, JX-594, MG1-MA3, MV-NIS, OBP-301, Reolysin®, Toca 511.
24. A method according to any one of claims 1 to 23, wherein step e) comprises
targeting cancer stem cells.
25. A method according to claim 24, wherein the cancer stem cells are treated with a combination therapy comprising lithium and disulfiram.
26. A method according to any one of claims 1 to 25, wherein the cancer is an epithelial cancer.
27. A method according to claim 26, wherein the epithelial cancer is selected from liver cancer (such as hepatocellular carcinoma), biliary tract cancer, breast cancer (such as non-ER+ breast cancer, in particular double negative breast cancer ), prostate cancer, colorectal cancer, ovarian cancer, endometrial cancer, cervical cancer, lung cancer, gastric cancer, pancreatic, bone cancer, bladder cancer, head and neck cancer, thyroid cancer, skin cancer, renal cancer, and oesophagus cancer, for example gastric cancer.
28. A method according to claim 27, wherein the cancer is selected from the group comprising liver cancer (such as hepatocellular carcinoma), breast cancer, biliary duct cancer prostate cancer, colorectal cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, gastric cancer, oesophageal cancer, kidney cancer, head and neck cancers and combinations thereof.
PCT/SG2017/050263 2016-05-20 2017-05-19 Method of treating cancer WO2017200492A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1608918.7A GB201608918D0 (en) 2016-05-20 2016-05-20 Method
GB1608918.7 2016-05-20

Publications (1)

Publication Number Publication Date
WO2017200492A1 true WO2017200492A1 (en) 2017-11-23

Family

ID=56369705

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2017/050263 WO2017200492A1 (en) 2016-05-20 2017-05-19 Method of treating cancer

Country Status (2)

Country Link
GB (1) GB201608918D0 (en)
WO (1) WO2017200492A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108543077A (en) * 2018-04-10 2018-09-18 武汉迈德森医药科技股份有限公司 A kind of mesoporous silica nano-particle and its preparation method and application of disulfiram monomer
WO2019183455A1 (en) * 2018-03-22 2019-09-26 Windmil Therapeutics, Inc. Prostate cancer specific marrow infiltrating lymphocytes and uses thereof
CN110947004A (en) * 2018-09-26 2020-04-03 江苏康缘药业股份有限公司 Pharmaceutical composition, application thereof, sterile container and kit

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190275092A1 (en) * 2016-11-01 2019-09-12 Dnatrix, Inc. Combination therapy for treatment of brain cancers

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009083232A1 (en) * 2007-12-28 2009-07-09 Deutsches Krebsforschungszentrum Parvovirus cancer therapy and combination with chemotherapy
WO2010135242A1 (en) * 2009-05-19 2010-11-25 Eden Pharmaceuticals, Inc. Combinations of immunostimulatory agents, oncolytic virus, and additional anticancer therapy
WO2013112942A1 (en) * 2012-01-25 2013-08-01 Dna Trix, Inc. Biomarkers and combination therapies using oncolytic virus and immunomodulation
WO2014022138A2 (en) * 2012-07-30 2014-02-06 Alex Wah Hin Yeung Live and in-vivo tumor specific cancer vaccine system developed by co-administration of either at least two or all three of the following components such as tumor cells, an oncolytic virus vector with transgenic expression of gm-csf and an immune checkpoint modulator
WO2015027915A1 (en) * 2013-09-02 2015-03-05 北京中康万达医药科技有限公司 In-vivo individualized system immunological therapeutic method and device
US20150086541A1 (en) * 2013-09-21 2015-03-26 Carlos Estuardo Aguilar-Cordova Methods of Cytotoxic Gene Therapy To Treat Tumors
WO2015061752A1 (en) * 2013-10-25 2015-04-30 Pharmacyclics, Inc. Treatment using bruton's tyrosine kinase inhibitors and immunotherapy
WO2015153514A1 (en) * 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
WO2016065330A1 (en) * 2014-10-24 2016-04-28 Aladar Szalay Combination immunotherapy approach for treatment of cancer

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009083232A1 (en) * 2007-12-28 2009-07-09 Deutsches Krebsforschungszentrum Parvovirus cancer therapy and combination with chemotherapy
WO2010135242A1 (en) * 2009-05-19 2010-11-25 Eden Pharmaceuticals, Inc. Combinations of immunostimulatory agents, oncolytic virus, and additional anticancer therapy
WO2013112942A1 (en) * 2012-01-25 2013-08-01 Dna Trix, Inc. Biomarkers and combination therapies using oncolytic virus and immunomodulation
WO2014022138A2 (en) * 2012-07-30 2014-02-06 Alex Wah Hin Yeung Live and in-vivo tumor specific cancer vaccine system developed by co-administration of either at least two or all three of the following components such as tumor cells, an oncolytic virus vector with transgenic expression of gm-csf and an immune checkpoint modulator
WO2015027915A1 (en) * 2013-09-02 2015-03-05 北京中康万达医药科技有限公司 In-vivo individualized system immunological therapeutic method and device
US20150086541A1 (en) * 2013-09-21 2015-03-26 Carlos Estuardo Aguilar-Cordova Methods of Cytotoxic Gene Therapy To Treat Tumors
WO2015061752A1 (en) * 2013-10-25 2015-04-30 Pharmacyclics, Inc. Treatment using bruton's tyrosine kinase inhibitors and immunotherapy
WO2015153514A1 (en) * 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
WO2016065330A1 (en) * 2014-10-24 2016-04-28 Aladar Szalay Combination immunotherapy approach for treatment of cancer

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019183455A1 (en) * 2018-03-22 2019-09-26 Windmil Therapeutics, Inc. Prostate cancer specific marrow infiltrating lymphocytes and uses thereof
CN112074280A (en) * 2018-03-22 2020-12-11 温德弥尔治疗公司 Prostate cancer specific marrow infiltrating lymphocytes and uses thereof
CN108543077A (en) * 2018-04-10 2018-09-18 武汉迈德森医药科技股份有限公司 A kind of mesoporous silica nano-particle and its preparation method and application of disulfiram monomer
CN108543077B (en) * 2018-04-10 2021-04-30 武汉迈德森医药科技股份有限公司 Mesoporous silica nanoparticle of disulfiram monomer and preparation method and application thereof
CN110947004A (en) * 2018-09-26 2020-04-03 江苏康缘药业股份有限公司 Pharmaceutical composition, application thereof, sterile container and kit
CN110947004B (en) * 2018-09-26 2021-05-18 江苏康缘药业股份有限公司 Pharmaceutical composition, application thereof, sterile container and kit

Also Published As

Publication number Publication date
GB201608918D0 (en) 2016-07-06

Similar Documents

Publication Publication Date Title
US10849899B2 (en) Combination therapy comprising Varlitinib and an anticancer agent
AU2018233032B2 (en) TEC family kinase inhibitor adjuvant therapy
Wuerth et al. Drug-repositioning opportunities for cancer therapy: novel molecular targets for known compounds
Romiti et al. Metronomic chemotherapy for cancer treatment: a decade of clinical studies
Pasini et al. The role of chemotherapy in metastatic gastric cancer
WO2017200492A1 (en) Method of treating cancer
WO2018136010A1 (en) Combination therapy
WO2018136009A1 (en) Combination therapy
US20190134034A1 (en) Method of Treating Liver Cancer
Guo et al. Advances and challenges in immunotherapy of small cell lung cancer
Bhoopathi et al. The quest to develop an effective therapy for neuroblastoma
Hou et al. Autophagy and tumour chemotherapy
Jackson et al. A review of the anti-tumor potential of current therapeutics targeting the mitochondrial protease ClpP in H3K27-altered, diffuse midline glioma
Choudhary et al. Different chemosensitization approaches in gastric cancer
Zhao et al. Trends of rapamycin in survival benefits of liver transplantation for hepatocellular carcinoma
Erika et al. Old tyrosine kinase inhibitors and newcomers in gastrointestinal cancer treatment
Ge et al. Review and prospect of immune checkpoint blockade therapy represented by PD-1/PD-L1 in the treatment of clear cell renal cell carcinoma
Sailo et al. Molecular alterations involved in pancreatic cancer chemoresistance and chemosensitization strategies
WO2021178449A1 (en) Compositions and methods for the treatment of pancreatic cancer
Sookprasert et al. Systemic therapy for locally advanced and metastatic cholangiocarcinoma
Hamza et al. Recent Concepts in the Pathogenesis and Management of Colorectal Cancer
Eckel et al. Emerging drugs for biliary cancer
Balar et al. A new era of immune therapeutics for pancreatic cancer: Monoclonal antibodies paving the way
Thakre et al. A LITERARY EXPLORATION OF CANCER TREATMENT TECHNOLOGIES
JP2023549698A (en) Compositions of nanoparticles for the treatment of cancer

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17799790

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17799790

Country of ref document: EP

Kind code of ref document: A1