WO2017099591A1 - Traitement de cancers à mutation braf résistants aux inhibiteurs - Google Patents

Traitement de cancers à mutation braf résistants aux inhibiteurs Download PDF

Info

Publication number
WO2017099591A1
WO2017099591A1 PCT/NL2016/050851 NL2016050851W WO2017099591A1 WO 2017099591 A1 WO2017099591 A1 WO 2017099591A1 NL 2016050851 W NL2016050851 W NL 2016050851W WO 2017099591 A1 WO2017099591 A1 WO 2017099591A1
Authority
WO
WIPO (PCT)
Prior art keywords
treatment
protein kinase
cancer
inhibitor
subject
Prior art date
Application number
PCT/NL2016/050851
Other languages
English (en)
Inventor
Jos Bernard Poell
Rodrigo Leonardus BEIJERSBERGEN
Original Assignee
Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis
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 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis filed Critical Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis
Publication of WO2017099591A1 publication Critical patent/WO2017099591A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Cancer is one of the leading causes of death in developed countries. Cancer is a heterologous class of diseases characterized by uncontrolled cell division and the ability of these cells to invade other tissues, either by direct growth into adjacent tissue or by migration of cells to distant sites. The proliferative properties of the cells initially give rise to a tumor or neoplasm. A tumor is considered a cancer when its cells acquire the ability to invade other tissues e.g. by forming secondary tumors at other sites in the body.
  • the unregulated growth is caused by mutations in DNA of, for example, genes that control cell division or cell growth.
  • the presence of one or more of these mutations, which can be inherited or acquired, can lead to uncontrolled cell division and cancer.
  • Cancer can cause many different symptoms, depending on the site and character of the malignancy and whether there is metastasis.
  • a definitive diagnosis usually includes biological and biochemical determinations on tissue obtained by biopsy. Once diagnosed, cancer is usually treated with surgery, chemotherapy and/or radiation.
  • chemotherapy chemotherapy and/or radiation.
  • the available drug therapies for metastatic disease are unfortunately often only palliative in nature and seldom offer a long-term cure.
  • Cancer cells often have an addiction to the signals generated by the cancer-causing genes (oncogenes). Cancer drugs that selectively inhibit the products of activated oncogenes can therefore have a dramatic effect on cancer cell viability.
  • Treatment with such targeted drugs provides significant clinical results for patients with, for example, Non-Small Cell Lung Cancer (NSCLC) characterized by activating mutations in EGFR or by translocations of the ALK kinase.
  • NSCLC Non-Small Cell Lung Cancer
  • Anti-cancer therapies using targeted drugs are frequently ineffective due to resistance of the tumor cells to therapy. Resistance may be acquired during therapy.
  • resistance may be intrinsic, i.e. not acquired of induced by the anti-cancer therapy.
  • the resistance is intrinsic the tumor cells already originally lack sensitivity to one or more anti-cancer drugs.
  • cancers in subjects have acquired resistance to a targeted drug inhibitor which initially had successfully been used in these subjects to treat the cancer.
  • a cancer in which acquired resistance is often observed is melanoma.
  • мелаnoma has an incidence and mortality in Europe of about 100.000 and 22.000 persons in Europe alone. Treatment of melanoma typically includes surgical removal of the melanoma, adjuvant treatment, chemo- and immunotherapy, and/or radiation therapy. The chance of a cure is greatest when the melanoma is discovered while it is still small and thin, and can be removed by surgery. More than 50% of (cutaneous) melanomas carry a mutation in the protein kinase referred to as BRAF (RAF is an acronym for Rapidly Accelerated Fibrosarcoma). In approximately 90% the mutation in the gene results in the substitution of glutamic acid for valine at codon 600 (BRAF V600E). Other known mutations include BRAF V600K and BRAF V600R.
  • BRAF protein kinase
  • the MAPK pathway plays a significant role in modulating cellular responses to extracellular stimuli, particularly in response to growth factors, and the pathway controls cellular events including cell proliferation, cell-cycle arrest, terminal differentiation and apoptosis (Peyssonnaux et al., Biol Cell. 93(l-2):53-62 (2001)).
  • FIG. 1 Limited sensitivity of resistant BRAF-mutant melanoma cells to drug withdrawal.
  • A Colony formation assay (8 days) of PLX4032-resistant BRAF-mutant melanoma cell lines on continued drug exposure (2 ⁇ PLX4032) versus drug withdrawal.
  • B Colony formation assay (8 days) of double-resistant (DR) lines derived from single-resistant lines (R). Cells were cultured under continued drug exposure (2 ⁇ PLX4032 and 200 nM PD0325901) or drug withdrawal.
  • PMA stimulates MAPK signaling in BRAF-mutant melanoma.
  • A Western blot of markers for MAPK pathway activation in two BRAF-mutant melanoma cell lines.
  • FIG. 3 Resistant BRAF-mutant melanoma cells are sensitive to PMA after drug withdrawal.
  • A Colony formation assays of parental (P) and resistant (R) BRAF-mutant melanoma treated for 8 days with no drug, 2 ⁇ PLX4032 or 10 nM PMA. BRAF-mutant melanoma cells were cultured in the presence of PLX4032 before the start of the 8 day colony formation
  • B Western blot analysis of phospho-ERK in response to 1 hour treatment with 10 nM PMA or 2 ⁇ PLX4032 in parental cells and resistant cells after PLX4032 withdrawal. GAPDH is used as a loading control.
  • Figure 4 Effect of Prostratin on BRAF inhibitor resistant melanoma cells after drug withdrawal.
  • 20,000 cells were seeded per well in a 6-wells plate and grown with the indicated drugs for 7 days.
  • Resistant A375R were grown in the presence of 2 ⁇ PLX4032 before the start of the experiment.
  • FIG. 5 PMA sensitivity of resistant BRAF-mutant melanoma after prolonged withdrawal.
  • A Colony formation assay (8 days) of A375R and mel888R cells comparing sensitivity to no drug, PMA (10 nM) and PLX4032 (2 ⁇ ) between resistant cells cultured on 2 ⁇ PLX4032 or after 6 or 12 day drug withdrawal.
  • B Colony formation assay of A375 cells overexpressing EGFR with no drug, 2 ⁇ PLX4032 or 10 nM PMA in the presence or absence of 20 ng/mL EGF.
  • A Colony formation assay of A375DR and mel888DR treated with different combination of 2 ⁇ PLX4032 and 100 or 200 nM PD0325901 in the presence or absence of 10 nM PMA.
  • FIG. 7 Colony formation assay of mel888 (parental), mel888R (PLX4032-resistant) untreated or treated with 2 ⁇ Prostratin. 200 nM PD0325901 or the combination. Resistant mel888R cells and mel888DR were grown in the presence of 2 ⁇ PLX4032 or 2 ⁇ PLX4032 and 200 nM PD0325901 , respectively, before the start of the experiment. 20,000 cells were seeded in a 6-wells plate and grown with the indicated drugs for 7 days. Colonies were visualized with crystal violet staining.
  • Figure 8 Colony formation assay of BRAF inhibitor resistant mel888R cells after drug withdrawal and treatment with either 10nM PMA or 2 ⁇ Prostratin fro the indicated times. After the indicated times, medium containing PMA and Prostratin were removed and replaced with medium alone. After 10 days the colonies were visualized with crystal violet staining.
  • FIG. 9 Colony formation assay (8 days) of A375R cells treated with 0.5 ⁇ GF109203X (PKCi) or 100 nM PD0325901 (MEKi) in the presence or absence of 10 nM PMA.
  • Acquired resistance indicates that a cancer has acquired reduced sensitivity or has become resistant to the effects of a drug after being exposed to it, or a drug targeting the same mechanism or pathway, for a certain period of time. Acquired resistance to the therapy with a drug often manifests either a diminished amount of tumor regression for the same dose of a drug or the need for an increased dose for an equal amount of tumor regression.
  • the term also indicates that a cancer may also become resistant to a first drug after being exposed to a second drug targeting the same mechanism of pathway in the cancer cell. For example, resistance may be acquired to a first ERK-inhibitor due to exposure to a second ERK-inhibitor (and to which the cancer will also have developed resistance).
  • resistance may be intrinsic, i.e. not acquired of induced by the anti-cancer therapy.
  • the resistance is intrinsic the tumor cells already originally lack sensitivity to one or more anticancer drugs. Since the resistance can be intrinsic or acquired the observed reduction in sensitivity is either compared to fully sensitive "normal" cancer cells, which are responsive to the therapeutically effective dosage of the applied anticancer drug and/or compared to the original sensitivity upon therapy onset.
  • Drug this relates to a chemical entity or biological product, or combination of chemical entities or biological products, administered to a person (for example, intravenous, intramuscular, subcutaneous, intradermal, intraperitoneal, intrathecal, intrapleural, intrauterine, rectal, oral, vaginal, topical, intratumor) to treat or prevent or control a disease or condition.
  • a person for example, intravenous, intramuscular, subcutaneous, intradermal, intraperitoneal, intrathecal, intrapleural, intrauterine, rectal, oral, vaginal, topical, intratumor
  • the chemical entity or biological product is preferably, but not necessarily a low molecular weight compound, but may also be a larger compound, for example, an oligomer of nucleic acids, amino acids, or carbohydrates including without limitation proteins, oligonucleotides, ribozymes, DNAzymes, glycoproteins, siRNAs, lipoproteins, aptamers, and modifications and combinations thereof.
  • Drug may have many modes of actions.
  • the drugs referred to herein are generally inhibitors (of the activity) of enzyme(s) or activators (of the activity) of enzymes.
  • Effective amount this means the amount of a drug which is effective for at least a statistically significant fraction of subjects to treat any symptom or aspect of the cancer. Effective amounts can be determined routinely.
  • the term includes both pharmacological effectiveness and physiological safety.
  • Pharmacological effectiveness refers to the ability of the treatment to result in a desired biological effect in the subject such as improvement of symptoms, a cure, a reduction in disease load, reduction in tumor mass or cell numbers, extension of life, improvement in quality of life, or other effect generally recognized as positive by medical doctors familiar with treating the particular type of disease or condition.
  • Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (often referred to as side-effects) resulting from administration of the treatment.
  • Subject this is to indicate the organism to be treated.
  • the subject may be any subject in accordance with the present invention, including, e.g., mammals, such as dogs, cats, horses, rats, mice, monkeys, and humans.
  • mammals such as dogs, cats, horses, rats, mice, monkeys, and humans.
  • the subject is a human patient.
  • any method, use or composition described herein can be implemented with respect to any other method, use or composition described herein.
  • Embodiments discussed in the context of methods, use and/or compositions of the invention may be employed with respect to any other method, use or composition described herein.
  • an embodiment pertaining to one method, use or composition may be applied to other methods, uses and compositions of the invention as well.
  • the inventors of the present invention have surprisingly found that substances that activate protein kinase C (PKC) are effective in the treatment of a mitogen-activated protein kinase (MAPK) pathway inhibitor resistant cancer.
  • PKC activators are effective in the treatment of cancer in subjects who are not or no longer responding to MAPK-pathway inhibitor therapies.
  • the present invention provides for the use of a protein kinase C activator in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer is a subject.
  • PKC Protein kinase C
  • DAG diacylglycerol
  • Ca2+ calcium ions
  • the PKC family consists of fifteen isozymes in humans and is divided into three subfamilies, based on their second messenger requirements: conventional (or classical), novel, and atypical. ⁇ is sometime referred to as a fourth subfamily; it resembles the "novel" PKC isoforms but differs by having a putative transmembrane domain.
  • PKC are thought to have a role in signal transduction in response to physiological stimuli (Nishizuka (1989) Cancer 10: 1892), as well as in neoplastic transformation and differentiation (Glazer (1994) Protein Kinase C. J.F. Kuo, ed., Oxford U. Press (1994): 171-198).
  • Dowling et al. reviewed the role of PKC as a target in anti-cancer treatment. They conclude that research into the role of PKC in cancer was primarily based on the assumption that increased PKC activation and expression may promote carcinogen induced tumorigenesis, but that increasing evidence suggests that PKC can act as both tumor suppressors and oncogenes.
  • This invention relates to contacting a PKC activator with cancer cells of a subject in a manner sufficient to increase the amount and/or activity of a PKC in the cancer cell.
  • the contacting with the PKC activator is sufficient to stimulate the activity of PKC in the cancer cells of the subject.
  • Activators of PKC are known to the skilled person.
  • PMA as described in the examples.
  • the PKC activator that may be used in the invention may, for example, be a macrocyclic lactone or a benzolactam.
  • the PKC activator may also be a pyrrolidinone.
  • the macrocyclic lactone may, for example, be bryostatin, e.g. bryostatin-1 , -2, -3, -4, -5, -6, -7, -8, -9, -10, -1 1 , -12, -13, -14, -15, -16, -17, or - 18.
  • the macrocyclic lactone may also be neristatin, for example neristatin- 1.
  • Another example is ingenol mebutate.
  • a preferred example is Prostratin (12-Deoxyphorbol-13-acetate;
  • Prostratin is a protein kinase C activator found in the bark of the mala tree of Samoa, Homalanthus nutans (Euphorbiaceae). Synthesis of Prostrating and related compounds have been described (Science (2008) 320(5876): 649-652). For an overview on Prostratin, reference is made to Miana (2015) Mini Rev Med Chem. 5(13):1 122-30.
  • the PKC activators are amenable to combinatorial synthetic techniques and thus libraries of the compounds can be generated to optimize pharmacological parameters, including, but not limited to efficacy and safety of the compositions.
  • PKC modulators see for example W01997043268, US5652232; US6043270; US6080784 and others.
  • the cancer in the subject is a cancer that is resistant to mitogen-activated protein kinase pathway inhibitors (MAPK inhibitors).
  • the cancer in the patient is a cancer that has reduced sensitivity or is insensitive to the effect of drugs that are inhibitors of the mitogen-activated protein kinase pathway.
  • the resistance maybe acquired resistance of may be intrinsic resistance.
  • the resistance of the cancer is a resistance that has been acquired as the consequence of prior treatment of the subject with mitogen-activated protein kinase pathway inhibitors.
  • the progression of cancer may be monitored by methods well known to the skilled person.
  • the progression may be monitored by way of visual inspection of the cancer, such as, by means of X-ray, CT scan or MRI.
  • the progression may be monitored by way of tumor biomarker detection.
  • the subject is monitored at various time points throughout the treatment of the cancer.
  • the progression of a cancer may be monitored by analyzing the progression of cancer at a second time point and comparing this analysis to an analysis at a first time point. An increased growth of the cancer indicates progression of the cancer and resistance to the treatment.
  • Acquired resistance to mitogen-activated protein kinase pathway inhibitors is a common phenomenon observed in subjects treated with such inhibitors. Resistance of a cancer to treatment with mitogen-activated protein kinase pathway inhibitors is readily recognized by the skilled person, for example by observing a diminished amount of tumor regression for the same dose of a drug or the need for an increased dose for an equal amount of tumor regression. Resistance may be acquired in response to treatment with single mitogen- activated protein kinase pathway inhibitors and/or in response to treatments involving more than one mitogen-activated protein kinase pathway inhibitor or treatments with combinations including at least one mitogen-activated protein kinase pathway inhibitor (e.g.
  • vemurafenib an inhibitor targeting mutated B-raf; see below
  • resistance to develops on average within 7 months of initial use, as is witnessed by a decrease in initial favorable response to the drug (i.e. less tumor regression, or return of tumor growth after initial stabilization, or as determined by measuring response or expression/activity of marker genes or proteins (e.g. of the MAPK pathway) of cell obtained from the subject by biopsies; see for example Trunzer et al. J Clin Oncol. 2013 May 10;31 (14):1767-74.
  • Attempts to overcome or prevent resistance of a cancer to treatment with mitogen-activated protein kinase pathway inhibitors include treatment with combinations of mitogen-activated protein kinase pathway inhibitors and PI3K/mTOR inhibitors.
  • a number of combinations of MEK and PI3K/mTOR pathway inhibitors combinations have entered early phase clinical trials, however their benefit in the setting of BRAF/MEK inhibitor resistance remains untested (see. e.g. Gowrishankar in "Melanoma - From Early Detection to Treatment” (2013), edited by Guy Huynh Thien Due, ISBN 978-953-51-0961-7, DOI: 10.5772/53629).
  • the current inventors have found that cancers that show resistance to mitogen-activated protein kinase pathway inhibitors are discontinued for the treatment with the mitogen- activated protein kinase pathway inhibitor and are at the same time treated with an activator of PKC respond very well to such treatment, as shown in the examples. It was surprisingly found that such cells have become very sensitive to activation of PKC by the PKC activator.
  • the PKC activator is effective in the treatment of cancers that do not, or to a limited extent, respond to inhibitors of enzymes that form part of the mitogen-activated protein kinase pathway.
  • the mitogen-activated protein kinase pathway (MAPK pathway) is one of the most studied pathways in cancer biology, well known to the skilled person and sometimes also referred to as the MAPK/ERK pathway, the RAS-RAF-MEK-ERK pathway, or the RAS-RAF- MEK-ERK-RSK pathway.
  • the MAPK pathway is a chain or pathway of proteins in the cell that communicates a signal from a receptor on the surface of the cell to the DNA in the nucleus of the cell.
  • activated RAS activates the protein kinase activity of RAF kinase
  • RAF kinase phosphorylates and activates MEK (MEK1 and MEK2)
  • MEK phosphorylates and activates a mitogen-activated protein kinase
  • MAPK phosphorylates ribosomal protein S6 kinase (RSK).
  • a MAPK pathway inhibitor is a compound that inhibits signaling through the MAPK pathway, preferably by inhibiting the activity of one of the proteins forming the chain or pathway.
  • the MAPK pathway inhibitor may do so by, for example, reducing the biological activity of a protein of the pathway, or my reducing expression of an mRNA encoding a protein of the pathway, or my reducing the expression of a protein of the pathway.
  • RAF inhibitor for example a BRAF inhibitor
  • a compound that may reduce the biological activity of RAF for example BRAF; or that may reduce the expression of an mRNA encoding a RAF polypeptide, for example BRAF; or that may reduce the expression of a RAF polypeptide, for example BRAF.
  • the above is likewise applicable with respect to inhibitors of the other proteins of the MAPK pathway.
  • the MAPK pathway inhibitor is an inhibitor of a protein of the MAPK pathway, preferably an inhibitor of RAS protein, an inhibitor of RAF protein, an inhibitor of MEK protein, an inhibitor of ERK protein and/or an inhibitor of RSK protein.
  • a RAS protein is a polypeptide belonging to the RAS family, more in particular to polypeptides as encoded by H-ras, K-ras, and N-ras in humans.
  • the RAS protein is a GTP-binding protein having the function to transduce signals to e.g. RAF protein in the MAPK signaling pathway.
  • RAS inhibitors are known to the skilled person.
  • Non-limitative examples include farnesyltransferase inhibitors including SCH66336 (Lonafarnib), R1 15777 (Zarnesta), BMS- 214662 and FTI-277, the geranylgeranyltransferase I inhibitor (GGTI)-2166 and trans- farnesylthiosalicylic acid (FTS, Salirasib).
  • a RAF protein is a polypeptide belonging to the RAF kinase family.
  • RAF kinases are a family of three serine/threonine-specific protein kinases that are related to retroviral oncogenes.
  • the three RAF kinase family members are ARAF (A-RAF; for example Genbank Accession NO: NP001243125), BRAF (B-RAF; (for example, Genbank Accession NO: NP004324)) and CRAF (C-RAF; (e.g. Gene accession number 5894; Refseq RNA Accessions NM_002880.3 ; protein NP_002871.1), and are well-known to the skilled person.
  • RAF kinase inhibitors are known to the skilled person.
  • Non-limitative examples include the compounds GW5074, BAY 43-9006, CHIR-265 (Novartis), Vemurafenib, PLX4720 (Tsai et al. 2008 PNAS 105(8):3041), PLX4032 (RG7204), GDC-0879 (Klaus P. Hoeflich et al. Cancer Res.2009 April 1 ;69:3042-3051), sorafenib tosylate (e.g. from Bayer and Onyx Pharmaceuticals as Nexavar), dasatinib (also known as BMS-354825, e.g.
  • RAF inhibitors include Vemurafenib (Roche/Plexxikon), Dabrafenib (GSK), LGX818 (Novartis), TAK-632 (Takeda), MLN2480 (Takeda/Millennium), PLX4032- 4720 (Plexxikon).
  • a MEK polypeptide e.g. Gene accession numbers 5604 or 5605; Refseq RNA Accessions NM_002755.3 or NM_030662.3; protein NP_002746.1 or NP_109587.1
  • MEK1 e.g.
  • MEK comprises both MEK1 and MEK2: MAP/ERK kinase 1 , MEK1 , PRKMK1 , MAPKK1 , MAP2K1 , MKK1 are the same enzyme, known as MEK1 , MAP/ERK kinase 2, MEK2, PRKMK2, MAPKK2, MAP2K2, MKK2 are the same enzyme, known as MEK2.
  • MEK1 and MEK2, together MEK can phosphorylate serine, threonine and tyrosine residues in protein or peptide substrates.
  • MEK inhibitors include but are not limited to the MEK inhibitors PD184352 and PD98059, inhibitors of MEKI and MEK2 U0126 (see Favata, M., et al., J. Biol. Chem. 273, 18623, 1998) and SL327 (Carr et al Psychopharmacology (Berl). 2009 Jan;201 (4):495-506), and those MEK inhibitors discussed in Davies et al (2000) (Davies et al Biochem J. 351 , 95- 105).
  • Another example is PDI 84352 (Allen, Lee et al Seminars in Oncology, Oct. 2003, pp. 105-106, vol.
  • MEK162 (Novartis) is another example.
  • Other known MEK inhibitors may be selected from PD-325901 (Pfizer), XL518 (Genentech), PD-184352 (Allen and Meyer Semin Oncol.
  • MEK inhibitors include Trametinib (GSK), Cobimetinib (GDC-0973) (Genentech/Exelixis), MEK162 (Novartis/Array BioPharma), AZD6244 (AstraZeneca/Array BioPharma), R05126766 (Roche/Chugai), GDC-0623 (Genentech/Chugai), PD0325901 (Pfizer), and Selumetinib.
  • An ERK protein is a polypeptide having serine/threonine protein kinase activity, e.g.
  • ERK phosphorylates and activates MAP (microtubule-associated proteins), and having at least 85% amino acid identity to the amino acid sequence of a human ERK, e.g. to ERK1 (e.g. Gene accession number 5595; Refseq RNA Accessions NM_001040056.2; protein NP_001035145.1) or ERK2 (e.g. Gene accession number 5594; Refseq RNA Accessions NM_002745.4 ; protein NP_002736.3).
  • ERK inhibitors are known to the skilled person, and includes such inhibitors as disclosed in WO2002058687, for example SL-327 (Carr et al Psychopharmacology (Berl).
  • ERK inhibitors may be found in WO2002058687, AU2002248381 , US20050159385, US2004102506, US2005090536, US2004048861 , US20100004234, HR20110892, WO201 1163330, TW200934775, EP2332922, WO2011041152, US2011038876, WO2009146034, HK11 17159, WO2009026487, WO2008115890, US2009186379, WO2008055236, US2007232610, WO2007025090, and US2007049591.
  • Further non-limiting examples or ERK-inhibitors include BVD-523, FR 180204 (CAS No.
  • ERK inhibitors include SCH772984 (Merck/Schering- Plough), VTX11e (Vertex) and GDC-0994 (Roche/Genentech).
  • An RSK protein e.g. EC 2.7.11.1 ; e.g.
  • rsk ribosomal s6 kinase
  • RSK protein is a MAP kinase activated protein kinase (MAPKAP kinase) and described in, e.g., Leukemia, 17: 1263-1293 (2003).
  • RSK is phosphorylated and activated by Erk1 and ERK-2 in response to many growth factors, polypeptide hormones and neurotransmitters.
  • RSK inhibitors are known to the skilled person and include, for example, Kaempherol-3-0-(4'- O-acetyl-a-L-rhamnopyranoside), or such inhibitors as disclosed in EP1845778.
  • the used inhibitors may also be inhibitors that inhibit (gene) expression of a protein of the MAPK pathway, for example by interfering with mRNA stability or translation.
  • the MAPK pathway inhibitor is selected from small interfering RNA (siRNA), which is sometimes referred to as short interfering RNA or silencing RNA, or short hairpin RNA (shRNA), which is sometimes referred to as small hairpin RNA.
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • the skilled person knows how to design such small interfering nucleotide sequence, for example as described in handbooks such as Doran and Helliwell RNA interference: methods for plants and animals Volume 10 CABI 2009. /pet
  • the inventors have found that when a cancer displays resistance to inhibitors of the MAPK pathway, for example inhibitors of the different proteins of the MAPK pathway, such as those described above, such cancer is in particular sensitive to treatment with a PKC activator.
  • PKC activator treatment of the cancer that displays resistance to the MAPK pathway inhibitor should take place in the absence of any MAPK pathway inhibitor, for example after treatment with the MAPK pathway inhibitor is discontinued.
  • the amount of the PKC activator, preferably prostratin, that may be administered to the subject is an effective amount.
  • more than one PKC activator may be used.
  • the PKC activator may also be combined with other drugs useful in the treatment of the cancer and/or in alleviating symptoms related to the cancer or the treatment thereof. The skilled person understand how to determine the effective amount, for example, by performing dose-finding studies while monitoring tumor pro- or regression.
  • the present invention relates to using a PKC activator to treat a cancer which has acquired resistance to a MAPK pathway inhibitor, irrespective of the molecular mechanism responsible for it.
  • the present invention also provides methods of treating a cancer in a subject, comprising administering an effective amount of a PKC activator to said subject having a cancer, wherein said cancer is refractory to a MAPK pathway inhibitor.
  • refractory means that the cancer (including a tumor and/or any metastasis thereof), upon treatment with at least one MAPK pathway inhibitor, shows no or only weak anti-cancer response (e.g., anti-proliferative response; such as, no or only weak inhibition of tumor growth) after the treatment.
  • the cancer that is resistant to a mitogen-activated protein kinase pathway inhibitor is a BRAF-mutation harboring cancer.
  • the cancer of the subject is a BRAF-mutated cancer, i.e. a cancer harboring mutation in BRAF.
  • BRAF-mutated cancer or "BRAF-mutation harboring cancer” is known to the skilled person.
  • BRAF e.g.
  • BRAF is a serine/threonine protein kinase and participates in the RAS/RAF/MEK/ERK/RSK mitogen activated protein kinase pathway (MAPK pathway, see Williams & Roberts, Cancer Metastasis Rev. 13(1):105- 16 (1994); Fecher et al 2008 Curr Opin Oncol 20, 183-189 or Cargnello M, Roux PP. Microbiol Mol Biol Rev.
  • BRAF mutations are found in different types of cancer. For example, approximately 40-60% of (cutaneous) melanomas carry a mutation in the BRAF protein. Approximately 90% of these mutations result in the substitution of glutamic acid for valine at codon 600 (BRAF V600E, although other mutations are also known (e.g. BRAF V600K and BRAF V600R). Such mutation in BRAF typically leads to proliferation and survival of the cancer cells (Davies et al Nature 2002; 417:949-54; Curtin et al N Engl J Med 2005;353:2135-47).
  • BRAF-mutation harboring cancers includes the use of trametinib and/or vemurafenib. Also the combination of the BRAF inhibitor vemurafenib and the MEK inhibitor cobimetinib demonstrated a statistically significant improvement in overall survival (Larkin (2015) J. Clin Oncol. 33 (suppl; abstr 9006). Another combination used is dabrafenib (a BRAF inhibitor), a BRAF inhibitor, plus trametinib (Mekinist®), a MEK inhibitor.
  • BRAF-mutation harboring cancers that are resistant to treatment with MAPK pathway inhibitors are, after discontinuation of the treatment with such MAPK pathway inhibitors, treated with an effective amount of a PKC activator.
  • a PKC activator By treatment of the cancers with the PKC activator, in accordance with the invention disclosed herein, progression of a cancer as a consequence of the resistance to the MAPK pathway targeting drugs, may be halted and/or regression of the cancer may be obtained.
  • the protein kinase C activator that is used in the treatment of the mitogen- activated protein kinase pathway resistant cancer is administered to the subject to be treated after any treatment with a mitogen-activated protein kinase pathway inhibitor is discontinued.
  • a mitogen-activated protein kinase pathway inhibitor is discontinued.
  • cancer- treating effects were obtained when the PKC activator was provided after treatment with drugs targeting protein of the MAPK pathway was stopped or discontinued.
  • This embodiment is in particular relevant for cancer that has acquired resistance to a drug targeting the MAPK pathway, e.g. an inhibitor of RAF, MER and/or ERK protein.
  • the treatment with the PKC activator is in the absence of any effective amount of a drug that act as an inhibitor on the MAPK pathway, for example drugs that inhibit enzymes of the MAPK pathway, irrespective of the fact whether the subject was prior treated with said MAPK pathway targeting drug or with any other MAPK pathway targeting drug, or not.
  • a drug that act as an inhibitor on the MAPK pathway for example drugs that inhibit enzymes of the MAPK pathway
  • the PKC activator may be administered to the subject.
  • the PKC activator may be administered to the subject certain time after the last dose of the MAPK pathway targeting drug was administered.
  • the length of the time between the last dose of the MAPK pathway targeting drug and the first dose of the PKC activator will depend on the pharmacodynamics characteristics of the drug compounds used in the treatment regime and can readily be established by the skilled person practicing the invention.
  • the protein kinase C activator for use in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject is used in a treatment that comprises
  • step b) treatment of the subject with a protein kinase C activator after the treatment of step a) is discontinued in step b).
  • a subject in particular a subject having a cancer characterized by the presence of a mutated BRAF, is initially treated in line with the accepted treatment in the art, for example with a combination of a BRAF inhibitor and a MEK inhibitor, or with only a BRAF inhibitor or MEK inhibitor.
  • the accepted treatment in the art for example with a combination of a BRAF inhibitor and a MEK inhibitor, or with only a BRAF inhibitor or MEK inhibitor.
  • During treatment normally regression/progression of the cancer is monitored. Once it is observed that the subject does not respond anymore, or once response to the treatment is reduced, treatment with the MAPK pathway targeting drugs is discontinued. After the treatment is discontinued, the patient is provided with a PKC activator as according to the invention.
  • the treatment with the MAPK pathway inhibitors (step a)), prior to the treatment with the PKC activator, comprises a treatment with a BRAF inhibitor, a MEK inhibitor or a combination of at least a BRAF inhibitor and a MEK inhibitor.
  • cancers that acquired resistance to in particular the above- indicated drugs are very sensitive to treatment with a PKC activator after treatment with MAPK pathway inhibitor(s) is discontinued.
  • the invention is not in particular limited to any PKC activator. Indeed, the inventors have shown that the effect of the PKC activators can be completely rescued by addition of a PKC inhibitor, proving that the observed effect is via PKC activation per se.
  • the PKC activator is phorbol-12-myristate-13-acetated (PMA), Bryostatin or Prostratin. In particular the latter two are preferred.
  • the PKC activator may be provided in the form of a pharmaceutical composition comprising the PKC activator and one or more pharmaceutical excipients.
  • the amount of the PKC activator will depend on the particular PKC activator employed in the invention and may be established using routine experimentation available in the art, including for example ascending dose studies and the like.
  • the use of the protein kinase C activator in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject is not in particular limited to any specific type of cancer, it is preferred the cancer is melanoma, colon cancer, papillary thyroid carcinoma, ovarian carcinoma, astrocytoma, ganglioglioma, craniopharyngioma, Langerhans cell histiocytosis, hairy cell leukemia, or ameloblastoma.
  • cancers are BRAF-mutation harboring cancers, as discussed in detail herein.
  • the cancer is melanoma, preferably BRAF-mutated melanoma.
  • the protein kinase C activator for use in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject as disclosed herein wherein the mitogen-activated protein kinase pathway inhibitor is a RAS inhibitor, RAF inhibitor, MEK inhibitor, ERK inhibitor and/or RSK inhibitor and/or wherein the treatment further comprises the use of an Receptor Tyrosine Kinases inhibitor, preferably a EGFR inhibitor, HER2 inhibitor, HER3 inhibitor, Met inhibitor, Axl inhibitor or PDGF inhibitor.
  • the cancer is resistant to, for example has acquired resistance, to a RAS inhibitor, a RAF inhibitor, a MEK inhibitor, an ERK inhibitor and/or a RSK inhibitor.
  • the inhibitor to which the cancer is resistant for example to which the cancer acquired resistance may be any of the known inhibitors of RAS, RAF, MEK, ERK and/or RSK inhibitors mentioned herein.
  • the mitogen-activated protein kinase pathway inhibitor treatment may further comprise the use of a Receptor Tyrosine Kinases inhibitor, preferably an EGFR inhibitor, a HER family inhibitor, Met inhibitor, Axl inhibitor or PDGF inhibitor.
  • a Receptor Tyrosine Kinases inhibitor preferably an EGFR inhibitor, a HER family inhibitor, Met inhibitor, Axl inhibitor or PDGF inhibitor.
  • the cancer is prior treated with a combination of at least one mitogen-activated protein kinase pathway inhibitor and a further inhibitor of a Receptor Tyrosine Kinases inhibitor, preferably an EGFR inhibitor (Gefitinib, Erlotinib), HER family inhibitor (Trastuzumab, Lapatinib, Pertuzumab, Afatinib, Dacometinib), Met inhibitor (XL184 (Cabozantinib), Tivantinib) , Axl inhibitor (BGB324, TP-0903) or PDGF inhibitor (Imatinib, Sunitinib, Sorafinib).
  • EGFR inhibitor Gafitinib, Erlotinib
  • HER family inhibitor Trastuzumab, Lapatinib, Pertuzumab, Afatinib, Dacometinib
  • Met inhibitor XL184 (Cabozantinib)
  • Tivantinib axl inhibitor
  • the protein kinase C activator is, when the cancer is first treated with a mitogen-activated protein kinase pathway inhibitor, to preferably to be administered to a subject after the treatment with the mitogen-activated protein kinase pathway inhibitor is discontinued.
  • the time between the last treatment with the mitogen-activated protein kinase pathway inhibitor and the first treatment with the PKC activator will depend on, for example, the kind of mitogen-activated protein kinase pathway inhibitor used in the prior treatment and/or the PKC activator to be administered to the subject.
  • the protein kinase C activator for use in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject as disclosed herein is administered within a period of 1 - 28 days after treatment of the subject with a mitogen-activated protein kinase pathway inhibitor is discontinued.
  • a period of 1 day (24 hours) between the final administration of the mitogen- activated protein kinase pathway inhibitor and the first administration of the PKC activator.
  • the period is within 1 - 14 days after treatment, even more preferably within 1 - 7 days after treatment with the mitogen-activated protein kinase pathway inhibitor is discontinued.
  • the PKC activator may be administered to the subject 1 , 2, 3, 4, 5, 6... , 1 1 , 12, 13...20, 21 , 22, 23...28 days after the last treatment with the mitogen-activated protein kinase pathway inhibitor.
  • PKC activator administration is repeated more than once; in some embodiments the PKC activator administration is repeated at regular intervals. In other embodiments the interval is between 6 hours and two weeks, between 12 hours and one week, or between one day and three days. In one embodiment, the administration of the PKC activator is continued for a fixed period of time. In one embodiment, the administration of the PKC activator is repeated for a period greater than one day. In another embodiment, the administration of the PKC activator is repeated for a period of between one day and one month, or is repeated for a period greater than one month.
  • the treatment with the PKC activator may be a combined treatment with other drugs useful in the treatment of the cancer in the subject.
  • progression of cancer in a subject may be monitored at a time point after the subject has initiated the treatment, for example the treatment with the mitogen-activated protein kinase pathway inhibitor.
  • progression of the cancer while being treated with mitogen-activated protein kinase pathway inhibitor, is indicative of cancer that is resistant to such mitogen-activated protein kinase pathway inhibitor and, according to the invention, the treatment with the mitogen-activated protein kinase pathway inhibitor should be discontinued and be replaced by treatment with the PKC activator.
  • Administering effective amounts of the PKC activator according to the invention can treat one or more aspects of the cancer disease, including, but not limited to, causing tumor regression; causing cell death; causing apoptosis; causing necrosis; inhibiting cell proliferation; inhibiting tumor growth; inhibiting tumor metastasis; inhibiting tumor migration; inhibiting tumor invasion; reducing disease progression; stabilizing the disease; reducing or inhibiting angiogenesis; prolonging subject survival; enhancing subject's quality of life; reducing adverse symptoms associated with cancer; and reducing the frequency, severity, intensity, and/or duration of any of the aforementioned aspects.
  • cancer in particular mitogen-activated protein kinase pathway inhibitor resistant cancer, in particular BRAF-mutation harboring mitogen-activated protein kinase pathway inhibitor resistant cancers can be effectively treated with PKC activators.
  • the PKC activator is provided to a subject (contacted with the cancer) under conditions that the cancer is not at the same time treated with a MAPK pathway inhibitor.
  • small populations of cells remain even after the treatment with the PKC activator.
  • these cells, after the treatment with the PKC activator are sensitive to further treatment with a MAPK pathway inhibitor.
  • the protein kinase C activator for use in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject as disclosed herein wherein further mitogen-activated protein kinase pathway inhibitors are administered after treatment of the subject with the protein kinase C activator is discontinued.
  • a subject having a mitogen-activated protein kinase pathway inhibitor resistant cancer is first treated with a PKC activator for a time sufficiently to treat the cancer, and that may cause the cancer to regress. After a certain period of time, the treatment with the PKC activator is discontinued, after which the remaining cancer is further treated with a MAPK pathway inhibitor, for example as disclosed herein.
  • timing and period of treatment of each individual step can be established by the skilled person using routine experimentation.
  • a subject having a cancer is first treated with a treatment comprising the use of MAPK pathway inhibitors (e.g. RAS, MEK and/or ERR inhibitors).
  • MAPK pathway inhibitors e.g. RAS, MEK and/or ERR inhibitors
  • the subject once it observed the cancer has acquired resistance to the MAPK pathway inhibitor (the subject now has, within the context of the current invention, a mitogen-activated protein kinase pathway inhibitor resistant cancer) treatment with the MAPK pathway inhibitor is discontinued.
  • the subject is provided with treatment comprising a PKC activator. Also during this stage cancer progression/regression may be monitored. After a certain period of time, for example, once a reduction in the therapeutic effect of the treatment comprising the PKC activator is observed, and in this embodiment of the invention, treatment with the PKC activator is discontinued.
  • a further treatment may be provided to the patient, comprising the further use of MAPK pathway inhibitors.
  • the skilled person understands such treatment may be repeated several times if so desired in view of the treatment of the subject (MAPK pathway inhibitor - PKC activator - MAPK pathway inhibitor - PKC activator ).
  • a protein kinase C activator for use in the treatment of cancer in a subject, wherein the treatment comprises administering the protein kinase C activator to cancer after the cancer has acquired resistance to treatment with a mitogen-activated protein kinase pathway inhibitor, preferably wherein the cancer is a BRAF-mutation harboring cancer.
  • the subject is first treated with a MAPK pathway inhibitor until the cancer acquires resistance to the MAPK pathway inhibitor, for example, as determined as disclosed herein.
  • treatment with the MAPK pathway inhibitor is discontinued.
  • the subject, now having a mitogen-activated protein kinase (MAPK) pathway inhibitor resistant cancer is treated with a PKC activator, as disclosed herein.
  • MAPK mitogen-activated protein kinase
  • the protein kinase C activator for use in the treatment of cancer in a subject as disclosed herein, wherein the treatment comprises a) treatment of the subject with one or more mitogen-activated protein kinase pathway inhibitor(s), preferably a BRAF inhibitor, a MEK inhibitor, or a combination of at least a BRAF inhibitor and a MEK inhibitor;
  • mitogen-activated protein kinase pathway inhibitor(s) preferably a BRAF inhibitor, a MEK inhibitor, or a combination of at least a BRAF inhibitor and a MEK inhibitor
  • step b) treatment of the subject with a protein kinase C activator after the treatment of step a) is discontinued in step b), essentially as already disclosed herein.
  • a protein kinase C activator in the manufacture of a medicament a) for the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject, preferably a BRAF-mutation harboring cancer; and/or
  • a cancer in a subject preferably a BRAF-mutation harboring cancer
  • the treatment comprises administering the protein kinase C activator to cancer after the cancer has acquired resistance to treatment with a mitogen-activated protein kinase pathway inhibitor.
  • a mitogen-activated protein kinase pathway inhibitor for use in the treatment of a cancer in a subject, preferably a BRAF-mutation harboring cancer, wherein the treatment comprises
  • step b) treatment of the subject with a protein kinase C activator after the treatment of step a) is discontinued in step b).
  • a mitogen-activated protein kinase pathway inhibitor in the manufacture of a medicament for the treatment of a cancer in a subject, preferably a BRAF-mutation harboring cancer, wherein the treatment comprises administering a protein kinase C activator to cancer after the cancer has acquired resistance to treatment with the mitogen-activated protein kinase pathway inhibitor.
  • a method of treatment of a mitogen- activated protein kinase pathway inhibitor resistant cancer in a subject preferably a BRAF- mutation harboring cancer
  • the method comprising treatment of the subject with a protein kinase C activator.
  • method of treatment of cancer in a subject preferably a BRAF-mutation harboring cancer, the method comprising:
  • mitogen-activated protein kinase pathway inhibitor(s) preferably a BRAF inhibitor, a MEK inhibitor, or a combination of at least a BRAF inhibitor and a MEK inhibitor;
  • step c) treatment of the subject with a protein kinase C activator after the treatment of step a) is discontinued in step b).
  • step c) starts within a period of 1 - 28 days after step b).
  • Prostratin for use in the treatment of cancer in a subject, preferably for use in the treatment of a mitogen-activated protein kinase pathway inhibitor resistant cancer in a subject.
  • Prostratin for use in the treatment of cancer in a subject as disclosed herein wherein the cancer is a BRAF-mutation harboring cancer.
  • the cancer is a melanoma.
  • the treatment may further comprise the discontinuation of the treatment with the PKC activator, followed by further treatment with a MAPK pathway inhibitor once the treatment with the PKC activator is discontinued, as detailed herein, including any further repeat steps ((MAPK pathway inhibitor - PKC activator - MAPK pathway inhibitor - PKC activator ).
  • the step of providing the further MAPK pathway inhibitors to the subject, after the treatment with the PKC activator is discontinued starts within a period of 1 - 28 days after the treatment with the PKC activator is discontinued.
  • the treatment with the PKC activator as disclosed herein does not comprise the simultaneous treatment with a MAPK pathway inhibitor.
  • BRAF inhibitors A common mechanism to BRAF inhibitors in the clinic is the acquisition of an activating mutation in the RAS oncogene.
  • BRAF-mutant melanoma cells that have become resistant to the combination of BRAF and MEK inhibitors also show increased vulnerability to exogenous activation of PKC, for example by PMA, characterized by an induction of senescence and cell cycle defects.
  • MAPK-pathway inhibitor resistant cancers in particular BRAF-mutant melanoma as well as colorectal cancer, gains a specific vulnerability to PKC activation causing MAPK (hyper-)activation.
  • A375, mel888, D10, WM266-4 and AO cell lines were obtained from in-house stocks and authenticated by STR profiling (BaseClear, Leiden). Resistant cell lines were previously described (Muller et al. 2014. Nat Commun.15;5:5712) and authenticated by STR profiling. Patient-derived naive and resistant melanoma lines were provided by the Peeper lab (Dutch Cancer Institute, The Netherlands).
  • pCDH_ER-RasV12_puro was created by cloning ER-RasV12 from Addgene #21 19 into pCDH_puro (System Biosciences #CD510B-1).
  • PLX4032_EGFR_blast was picked from the Broad ORF library.
  • pLKO_shNF1_puro was picked from the Broad TRC collection (TRCN_39717).
  • Constructs were cotransfected with lentiviral packaging vectors (3: 1 : 1 : 1) into 293T producer cells using 4 ⁇ g of PEI (Sigma-Aldrich) per ⁇ g of plasmid DNA. 12 ⁇ g plasmid DNA was used for one 10 cm dish. Medium was refreshed after overnight incubation.
  • virus supernatant was collected and filtered. Infections were achieved by applying virus supernatant in 1 :3 - 1 :50 dilution (optimal dilution was determined per construct and cell line) in the presence of 8 ⁇ g/mL hexadimethrine bromide (Sigma-Aldrich). Cells were put on puromycin (2 ⁇ g/mL) or blasticidin (10 ⁇ g/mL) selection 24 hours after infection for 3-5 days.
  • ELISA on phospho-ERK was executed according to manufacturer's instructions (Cell Signaling, 7246), except the readout, which was achieved using ECL substrate and measurement of luminescence on a Perkin-Elmer Envision. Luminescence signals were normalized to protein input as determined by BCA assay and standardized to the appropriate control sample.
  • BRAF and MEK inhibitor double-resistant (DR) lines through exposure to 2 ⁇ PLX4032 and 0.2 ⁇ PD0325901 for ⁇ 2 months.
  • A375DR and mel888DR were sensitive to drug withdrawal (Fig. 1 B). This effect was accompanied by an increase in the levels of phospho-c- Raf and phospho-ERK compared to untreated and drug treated cells.
  • drug withdrawal in single resistant cell lines did not lead to a remarkable increase in phospho-ERK compared to the parental cells or drug continuation.
  • the growth inhibitory effect of drug withdrawal in A375DR and mel888DR cells was limited, with cells surviving and continue to proliferate in the absence of drug.
  • a PKC activator activates the MAPK pathway in BRAF-mutant melanoma
  • the MAPK kinase pathway can be activated by exogenous stimulation of PKC.
  • PMA phorbol 12-myristate 13-acetate, also known as TPA.
  • PMA is a diacylglycerol mimic that activates several protein kinase C (PKC) isoforms.
  • PKC protein kinase C
  • Resistant BRAF-mutant melanomas are sensitive to MAPK hyper-activation by PKC activators
  • PMA had little effect on growth of parental lines, but intermediate to strong effect on growth of resistant lines after withdrawal of the BRAF inhibitor PLX4032 (Fig. 3A). Although PMA activates MAPK signaling through activation of PKC, we only observed highly elevated phospho-ERK levels in resistant mel888 in contrast to a very modest increase in resistant A375 (Fig. 3B). To ascertain that the detrimental effects of PMA were indeed caused by MAPK activation, we titrated in a MEK inhibitor with the PMA treatment. In all resistant cell lines, the detrimental effect of PMA could be rescued by MEK inhibition in a dose-dependent fashion.
  • Oncogenic Ras activation is synthetic lethal with PMA in BRAF-mutant melanoma
  • Double-resistant BRAF-mutant melanoma are hypersensitive to PMA
  • Figure 7 shows the effect of Prostratin on BRAF inhibitor or BRAF/MEK inhibitor resistant mel888 cells compared to the parental cells.
  • Mell888R resistant to BRAF inhibitor
  • mel888DR cells resistant to the combination of BRAF and MEK inhibitors
  • PD MEK inhibitor

Abstract

L'invention concerne des méthodes de traitement du cancer, notamment du cancer qui est résistant à un traitement par des inhibiteurs de la voie MAPK, par exemple ayant acquis une résistance aux dits inhibiteurs de la voie MAPK. La présente invention concerne des compositions pharmaceutiques destinées à être utilisées dans un tel traitement, des programmes thérapeutiques utilisant de telles compositions et un procédé de traitement. L'invention est basée sur l'utilisation d'activateurs de PKC.
PCT/NL2016/050851 2015-12-07 2016-12-06 Traitement de cancers à mutation braf résistants aux inhibiteurs WO2017099591A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NL2015916 2015-12-07
NL2015916 2015-12-07

Publications (1)

Publication Number Publication Date
WO2017099591A1 true WO2017099591A1 (fr) 2017-06-15

Family

ID=55858853

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2016/050851 WO2017099591A1 (fr) 2015-12-07 2016-12-06 Traitement de cancers à mutation braf résistants aux inhibiteurs

Country Status (1)

Country Link
WO (1) WO2017099591A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
WO2022173241A1 (fr) * 2021-02-15 2022-08-18 연세대학교 산학협력단 Composition pour réguler le sucre, contenant un activateur de pkc atypique
US11680297B2 (en) 2013-09-16 2023-06-20 The Johns Hopkins University Activities of multiple cancer-related pathways are associated with BRAF mutation and predict the resistance to BRAF/MEK inhibitors in melanoma cells
WO2023125788A1 (fr) * 2021-12-31 2023-07-06 Edigene Therapeutics (Beijing) Inc. Biomarqueurs pour le traitement du cancer colorectal

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5652232A (en) 1993-09-30 1997-07-29 Shionogi & Co., Ltd. Benzolactam derivatives
WO1997043268A1 (fr) 1996-05-10 1997-11-20 Georgetown University Derives de benzodizocine a substitution 8-hydrocarbyle, leur preparation et leur utilisation comme modulateurs de la proteine kinase c (pkc)
US6043270A (en) 1986-06-11 2000-03-28 Procyon Pharmaceuticals, Inc. Protein kinase C modulators V
US6080784A (en) 1986-06-11 2000-06-27 Procyon Pharmaceuticals, Inc. Protein kinase C modulators N
WO2002058687A2 (fr) 2001-01-25 2002-08-01 Board Of Regents, The University Of Texas System Inhibition de la erk reduisant ou prevenant a la fois la tolerance et la dependance aux analgesiques opioides et sensibilisation apres stimulation douloureuse
US20040048861A1 (en) 2000-02-05 2004-03-11 Guy Bemis Compositions useful as inhibitors of ERK
US20040102506A1 (en) 2000-02-05 2004-05-27 Michael Hale Pyrazole compositions useful as inhibitors of ERK
US20050090536A1 (en) 2000-09-15 2005-04-28 Hale Michael R. Isoxazole compositions useful as inhibitors of ERK
US20050159385A1 (en) 2003-12-19 2005-07-21 Mohapatra Shyam S. JAK/STAT inhibitors and MAPK/ERK inhibitors for RSV infection
US20070049591A1 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibitors of MAPK/Erk Kinase
WO2007076245A2 (fr) 2005-12-21 2007-07-05 Array Biopharma Inc. Nouveau sel hydrogenosulfate
US20070232610A1 (en) 2006-02-16 2007-10-04 Yongqi Deng Novel compounds that are ERK inhibitors
EP1845778A2 (fr) 2005-01-10 2007-10-24 University Of Virginia Patent Foundation Synthese d'inhibiteurs de p90rsk
WO2008055236A2 (fr) 2006-10-31 2008-05-08 Takeda Pharmaceutical Company Limited Inhibiteurs de kinase mapk/erk
WO2008115890A2 (fr) 2007-03-19 2008-09-25 Takeda Pharmaceutical Company Limited Inhibiteurs de mapk/erk kinase
WO2009026487A1 (fr) 2007-08-22 2009-02-26 The Ohio State University Research Foundation Procédés et compositions pour induire une dérégulation de la phosphorylation de epha7 et de erk dans des cas de leucémies humaines aiguës
US20090186379A1 (en) 2006-11-13 2009-07-23 Thomas David Reed ERK ligands and polynucleotides encoding ERK ligands
TW200934775A (en) 2007-11-12 2009-08-16 Takeda Pharmaceutical MAPK/ERK kinase inhibitors
WO2009146034A2 (fr) 2008-03-31 2009-12-03 Takeda Pharmaceutical Company Limited Inhibiteurs de kinases mapk/erk et procédés d’utilisation de ceux-ci
US20100004234A1 (en) 2004-09-27 2010-01-07 Kosan Biosciences Incorporated Specific kinase inhibitors
US20110038876A1 (en) 2007-06-18 2011-02-17 Robert Sun Heterocyclic compounds and use thereof as erk inhibitors
WO2011041152A1 (fr) 2009-09-30 2011-04-07 Schering Corporation Nouveaux composés inhibiteurs d'erk
EP2332922A1 (fr) 2001-10-23 2011-06-15 Merck Serono S.A. Azoles N-substitués et leur utilisation en tant que modulateurs de MEK-1 et/ou ERK-2
WO2011163330A1 (fr) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Nouveaux composés hétérocycliques utilisés comme inhibiteurs de erk
HRP20110892T1 (hr) 2005-12-13 2011-12-31 Schering Corporation Policiklični derivati indazola koji su inhibitori erk
US20140018329A1 (en) * 2007-01-31 2014-01-16 Biosuccess Biotech Co., Ltd. Compositions and methods of use of phorbol esters in the treatment of neoplasms

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6043270A (en) 1986-06-11 2000-03-28 Procyon Pharmaceuticals, Inc. Protein kinase C modulators V
US6080784A (en) 1986-06-11 2000-06-27 Procyon Pharmaceuticals, Inc. Protein kinase C modulators N
US5652232A (en) 1993-09-30 1997-07-29 Shionogi & Co., Ltd. Benzolactam derivatives
WO1997043268A1 (fr) 1996-05-10 1997-11-20 Georgetown University Derives de benzodizocine a substitution 8-hydrocarbyle, leur preparation et leur utilisation comme modulateurs de la proteine kinase c (pkc)
US20040048861A1 (en) 2000-02-05 2004-03-11 Guy Bemis Compositions useful as inhibitors of ERK
US20040102506A1 (en) 2000-02-05 2004-05-27 Michael Hale Pyrazole compositions useful as inhibitors of ERK
US20050090536A1 (en) 2000-09-15 2005-04-28 Hale Michael R. Isoxazole compositions useful as inhibitors of ERK
WO2002058687A2 (fr) 2001-01-25 2002-08-01 Board Of Regents, The University Of Texas System Inhibition de la erk reduisant ou prevenant a la fois la tolerance et la dependance aux analgesiques opioides et sensibilisation apres stimulation douloureuse
AU2002248381A1 (en) 2001-01-25 2002-08-06 Board Of Regents, The University Of Texas System Inhibition of erk to reduce or prevent tolerance to and dependence on opioid analgesics
EP2332922A1 (fr) 2001-10-23 2011-06-15 Merck Serono S.A. Azoles N-substitués et leur utilisation en tant que modulateurs de MEK-1 et/ou ERK-2
US20050159385A1 (en) 2003-12-19 2005-07-21 Mohapatra Shyam S. JAK/STAT inhibitors and MAPK/ERK inhibitors for RSV infection
US20100004234A1 (en) 2004-09-27 2010-01-07 Kosan Biosciences Incorporated Specific kinase inhibitors
EP1845778A2 (fr) 2005-01-10 2007-10-24 University Of Virginia Patent Foundation Synthese d'inhibiteurs de p90rsk
WO2007025090A2 (fr) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibiteurs de kinase mapk/erk
US20070049591A1 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibitors of MAPK/Erk Kinase
HRP20110892T1 (hr) 2005-12-13 2011-12-31 Schering Corporation Policiklični derivati indazola koji su inhibitori erk
WO2007076245A2 (fr) 2005-12-21 2007-07-05 Array Biopharma Inc. Nouveau sel hydrogenosulfate
US20070232610A1 (en) 2006-02-16 2007-10-04 Yongqi Deng Novel compounds that are ERK inhibitors
WO2008055236A2 (fr) 2006-10-31 2008-05-08 Takeda Pharmaceutical Company Limited Inhibiteurs de kinase mapk/erk
US20090186379A1 (en) 2006-11-13 2009-07-23 Thomas David Reed ERK ligands and polynucleotides encoding ERK ligands
US20140018329A1 (en) * 2007-01-31 2014-01-16 Biosuccess Biotech Co., Ltd. Compositions and methods of use of phorbol esters in the treatment of neoplasms
WO2008115890A2 (fr) 2007-03-19 2008-09-25 Takeda Pharmaceutical Company Limited Inhibiteurs de mapk/erk kinase
US20110038876A1 (en) 2007-06-18 2011-02-17 Robert Sun Heterocyclic compounds and use thereof as erk inhibitors
WO2009026487A1 (fr) 2007-08-22 2009-02-26 The Ohio State University Research Foundation Procédés et compositions pour induire une dérégulation de la phosphorylation de epha7 et de erk dans des cas de leucémies humaines aiguës
TW200934775A (en) 2007-11-12 2009-08-16 Takeda Pharmaceutical MAPK/ERK kinase inhibitors
WO2009146034A2 (fr) 2008-03-31 2009-12-03 Takeda Pharmaceutical Company Limited Inhibiteurs de kinases mapk/erk et procédés d’utilisation de ceux-ci
WO2011041152A1 (fr) 2009-09-30 2011-04-07 Schering Corporation Nouveaux composés inhibiteurs d'erk
WO2011163330A1 (fr) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Nouveaux composés hétérocycliques utilisés comme inhibiteurs de erk

Non-Patent Citations (43)

* Cited by examiner, † Cited by third party
Title
AHRONIAN, CANCER DISCOV., vol. 5, no. 4, 2015, pages 358 - 367
ALLEN, LEE ET AL., SEMINARS IN ONCOLOGY, vol. 30, October 2003 (2003-10-01), pages 105 - 106
ALLEN; MEYER, SEMIN ONCOL., vol. 30, no. 5, October 2003 (2003-10-01), pages 105 - 16
ASHENDEL ET AL., CANCER RES., vol. 43, 1983, pages 4333
BAST ET AL.: "Cancer Medicine, 5th ed.", B. C. DECKER INC.
CARGNELLO M; ROUX PP., MICROBIOL MOL BIOL REV., vol. 75, no. 1, March 2011 (2011-03-01), pages 50 - 83
CARR ET AL., PSYCHOPHARMACOLOGY (BERL, vol. 201, no. 4, January 2009 (2009-01-01), pages 495 - 506
CARR ET AL., PSYCHOPHARMACOLOGY (BERL, vol. 201, no. 4, January 2009 (2009-01-01), pages 495 - 5060
CURTIN ET AL., N ENGL J MED, vol. 353, 2005, pages 2135 - 47
DAVIES ET AL., BIOCHEM J., vol. 351, pages 95 - 105
DAVIES ET AL., NATURE, vol. 417, 2002, pages 949 - 54
DORAN; HELLIWELL: "RNA interference: methods for plants and animals", vol. 10, 2009, CABI
DOWLING ET AL., CANCERS, vol. 7, 2015, pages 1271 - 1291
FAVATA, M. ET AL., J. BIOL. CHEM., vol. 273, 1998, pages 18623
FECHER ET AL., CURR OPIN ONCOL, vol. 20, 2008, pages 183 - 189
FEDORENKO ET AL., BJC, vol. 112, 2015, pages 217 - 226
GLAZER: "Protein Kinase", 1994, OXFORD U. PRESS, pages: 171 - 198
GOWRISHANKAR: "Melanoma - From Early Detection to Treatment", 2013, ISBN: 978-953-51-09
IKAWA, MOL CELL BIOL., vol. 8, no. 6, 1988, pages 2651 - 4
KIKKAWA ET AL., J BIOL. CHEM., vol. 257, 1982, pages 13341
KLAUS P. HOEFLICH ET AL., CANCER RES., vol. 69, 1 April 2009 (2009-04-01), pages 3042 - 3051
LARKIN, J. CLIN ONCOL., vol. 33, 2015
LEUKEMIA, vol. 17, 2003, pages 1263 - 1293
LORUSSO ET AL., JOURNAL OF CLINICAL ONCOLOGY, vol. 23, no. 23, 2005, pages 5281 - 5293
MALGORZATA SZTILLER-SIKORSKA ET AL: "Natural Compounds' Activity against Cancer Stem-Like or Fast-Cycling Melanoma Cells", PLOS ONE, vol. 9, no. 3, 3 March 2014 (2014-03-03), pages e90783, XP055291942, DOI: 10.1371/journal.pone.0090783 *
MIANA, MINI REV MED CHEM., vol. 5, no. 13, 2015, pages 1122 - 30
MOHAMMAD ATEFI ET AL: "Reversing Melanoma Cross-Resistance to BRAF and MEK Inhibitors by Co-Targeting the AKT/mTOR Pathway", PLOS ONE, vol. 6, no. 12, 14 December 2011 (2011-12-14), pages e28973, XP055126888, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0028973 *
MULLER ET AL., NAT COMMUN., vol. 15, no. 5, 2014, pages 5712
NISHIZUKA, CANCER, vol. 10, 1989, pages 1892
PENG ET AL., CANCER CELL, vol. 28, no. 3, 2015, pages 384 - 398
PEYSSONNAUX ET AL., BIOL CELL, vol. 93, no. 1-2, 2001, pages 53 - 62
PRAHHALAD ET AL., NATURE, vol. 483, no. 7387, 2012, pages 100 - 103
RICHMAN, EXPERT OPIN. PHARMACOTHER, vol. 16, no. 9, 2015, pages 1285 - 97
SCIENCE, vol. 320, no. 5876, 2008, pages 649 - 652
SETON-ROGERS ET AL., NATURE REVIEWS CANCER, vol. 14, 2014, pages 7
SHARMA ET AL., CELL, vol. 141, no. 1, 2010, pages 69 - 80
SUN ET AL., CELL REP., vol. 10, no. 1, 2014, pages 86 - 93
TECLE ET AL., NIC & MEDICINAL CHEMISTRY LETTERS, vol. 19, no. 1, 1 January 2009 (2009-01-01), pages 226 - 229
TRUNZER ET AL., J CLIN ONCOL., vol. 31, no. 14, 10 May 2013 (2013-05-10), pages 1767 - 74
VAN ALLEN ET AL., CANCER DISCOV., 2013, Retrieved from the Internet <URL:http://dx.doi.org/10.1158/2159-8290.CD-13-0617>
WAGLE ET AL., J CLIN ONCOL., vol. 29, no. 22, 2011, pages 3085 - 96
WILLIAMS; ROBERTS, CANCER METASTASIS REV., vol. 13, no. 1, 1994, pages 105 - 16
YAO ET AL., CANCER CELL, vol. 28, no. 3, 2015, pages 370 - 383

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11680297B2 (en) 2013-09-16 2023-06-20 The Johns Hopkins University Activities of multiple cancer-related pathways are associated with BRAF mutation and predict the resistance to BRAF/MEK inhibitors in melanoma cells
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
WO2022173241A1 (fr) * 2021-02-15 2022-08-18 연세대학교 산학협력단 Composition pour réguler le sucre, contenant un activateur de pkc atypique
WO2023125788A1 (fr) * 2021-12-31 2023-07-06 Edigene Therapeutics (Beijing) Inc. Biomarqueurs pour le traitement du cancer colorectal

Similar Documents

Publication Publication Date Title
Huang et al. MST4 phosphorylation of ATG4B regulates autophagic activity, tumorigenicity, and radioresistance in glioblastoma
McCubrey et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascade inhibitors: how mutations can result in therapy resistance and how to overcome resistance
Li et al. Resveratrol inhibits Hexokinases II mediated glycolysis in non-small cell lung cancer via targeting Akt signaling pathway
Rattan et al. Metformin attenuates ovarian cancer cell growth in an AMP‐kinase dispensable manner
Ma et al. Novel agents on the horizon for cancer therapy
Heffernan-Stroud et al. Sphingosine kinase 1 in cancer
She et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors
Jacoby et al. Treatment with HIF-1α antagonist PX-478 inhibits progression and spread of orthotopic human small cell lung cancer and lung adenocarcinoma in mice
Tavallai et al. Rationally repurposing ruxolitinib (Jakafi®) as a solid tumor therapeutic
Ammoun et al. Nilotinib alone or in combination with selumetinib is a drug candidate for neurofibromatosis type 2
Ciuffreda et al. Signaling intermediates (MAPK and PI3K) as therapeutic targets in NSCLC
Botting et al. Mechanism of resistance and novel targets mediating resistance to EGFR and c-Met tyrosine kinase inhibitors in non-small cell lung cancer
Dong et al. Selective inhibition of PDGFR by imatinib elicits the sustained activation of ERK and downstream receptor signaling in malignant glioma cells
WO2017099591A1 (fr) Traitement de cancers à mutation braf résistants aux inhibiteurs
Howe et al. Focal adhesion kinase inhibitors in combination with erlotinib demonstrate enhanced anti-tumor activity in non-small cell lung cancer
Chow et al. Group I Paks as therapeutic targets in NF2-deficient meningioma
Nguyen et al. A selective inhibitor of ubiquitin-specific protease 4 suppresses colorectal cancer progression by regulating β-catenin signaling.
Belli et al. Regorafenib in combination with silybin as a novel potential strategy for the treatment of metastatic colorectal cancer
KR20190141164A (ko) 병용 요법
Fumarola et al. Impairing energy metabolism in solid tumors through agents targeting oncogenic signaling pathways
Wang et al. Identification of a novel PAK1 inhibitor to treat pancreatic cancer
Lin et al. Vorinostat combined with brigatinib overcomes acquired resistance in EGFR-C797S-mutated lung cancer
Hou et al. Integrin subunit beta 8 contributes to lenvatinib resistance in HCC
EP3355923B1 (fr) Inhibiteurs de l&#39;histone désacétylase utilisables dans le traitement d&#39;un mélanome résistant aux médicaments
Li et al. Degradation of AMPK‐α1 sensitizes BRAF inhibitor‐resistant melanoma cells to arginine deprivation

Legal Events

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

Ref document number: 16813156

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16813156

Country of ref document: EP

Kind code of ref document: A1