US20140073645A1 - Treatment of Solid Tumours - Google Patents

Treatment of Solid Tumours Download PDF

Info

Publication number
US20140073645A1
US20140073645A1 US14/006,277 US201214006277A US2014073645A1 US 20140073645 A1 US20140073645 A1 US 20140073645A1 US 201214006277 A US201214006277 A US 201214006277A US 2014073645 A1 US2014073645 A1 US 2014073645A1
Authority
US
United States
Prior art keywords
methyl
cells
inhibiting agent
autophagy
iron chelator
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/006,277
Other languages
English (en)
Inventor
Stig Linder
Märten Fryknas
Rolf Larsson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vivolux AB
Original Assignee
Vivolux AB
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 Vivolux AB filed Critical Vivolux AB
Assigned to VIVOLUX AB reassignment VIVOLUX AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LINDER, STIG, FRYKNAS, MARTEN, LARSSON, ROLF
Publication of US20140073645A1 publication Critical patent/US20140073645A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/44221,4-Dihydropyridines, e.g. nifedipine, nicardipine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to a treatment of a solid tumour, in particular a disseminated solid tumour, in a person affected by cancer and to a means for such treatment.
  • New and effective anticancer drugs need to be developed for patients that suffer from disseminated cancer.
  • Developing drugs for solid tumours is associated with specific problems due to complex biophysical and metabolic conditions in 3-D tumour tissue which may be difficult to mimic in experimental in vitro systems. Hypoxia and limited diffusion of nutrients is known to lead to quiescence and resistance to conventional anticancer agents and radiation therapy.
  • anticancer drugs must be able to penetrate into tumour parenchyme to reach cancer cells at toxic concentrations.
  • Multicellular spheroids mimic human solid tumours better than 2-D monolayer cultures (2-4), and many clinically used drugs show limited potency on cancer cells grown as MCS (5, 6). Therefore, MCS are better suited than monolayer cultures for screening drugs active on solid tumours.
  • Apoptosis is mediated by the activation of caspases.
  • Autophagy is an evolutionarily conserved mechanism for degradation of long-lived cellular proteins and damaged cell organelles. The formation of autophagosomes is a main characteristic of autophagy. Autophagosome formation requires activation of class III phosphatidylinositol-3-kinase and is also dependent of two ubiquitin-like conjugation systems (Atg-Atg12 and Atg8) (7). Autophagy protects cells during conditions of nutrient deprivation, and cells undergo apoptosis when autophagy is inhibited (8-10). Morphological features of autophagy have also been observed during cell death under conditions of caspase inhibition (11).
  • a cell permeable iron chelator optionally in combination with an autophagy inhibiting agent, for treating a solid cancer tumour in a person.
  • the compound of general formula I is intended to include any pharmaceutically suitable salt or complex or prodrug thereof.
  • R is particularly preferred for R to be methyl. It is preferred for both of R, R 1 to be methyl.
  • R 1 is methyl, in particular 6-methyl or 8-methyl.
  • N-(1-pyridine-2-yl-methylidene)-N-(9H-1,3,4,9-tetraaza-fluoren-2-yl)-hydrazine is substituted as follows:
  • the compound of the general formula I in which R 1 is C 1 -C 4 alkyl may be additionally substituted by C 1 -C 4 alkyl at one of positions 6, 7 or 8 of the tetraazaflourenyl moiety not substituted by R 1 .
  • a pharmaceutical composition comprising a compound of the invention and a pharmaceutical carrier.
  • the pharmaceutical composition of the invention can be administered by any suitable route, such perorally or parenterally.
  • Suitable carriers comprise dimethyl sulfoxide and aqueous media, such as mixtures comprising dimethyl sulfoxide and water.
  • Preferred fluid carriers are those capable of dissolving the compound of the invention.
  • Other preferred fluid carriers, in particular aqueous carriers are those comprising the compound of the invention in finely dispersed form, such as in form of microparticles of a size of 10 ⁇ m or smaller.
  • a method of treating a solid cancer in a person comprising administering to the person a pharmacologically effective dose of a compound of the invention or a pharmaceutically acceptable salt thereof.
  • the pharmacologically effective dose is preferably administered comprised by the pharmaceutical composition of the invention.
  • the compound of the invention is a cell permeable iron chelator. While not wishing to bound by theory, the inventors believe the anti-cancer effect of the compound of the invention to be based on its iron-chelating properties.
  • an iron chelator such the compound of the invention
  • an autophagy inhibiting agent is chloroquine.
  • an autophagy inhibiting agent and a cell permeable iron chelator in combination in the treatment of a solid tumour.
  • the autophagy inhibiting agent and the cell permeable iron chelator can be administered in form of a pharmaceutical composition comprising them or in form of separate pharmaceutical compositions. If administered in form of a pharmaceutical composition, the combination comprises a pharmaceutically acceptable carrier.
  • the cell permeable iron chelator is preferably selected from N-(1-pyridine-2-yl-methylidene)-N-(9H-1,3,4,9-tetraaza-fluoren-2-yl)-hydrazine or a pharmaceutically acceptable salt thereof of the general formula I, wherein R is H or methyl and R 1 is H or C 1 -C 4 alkyl, for treating a solid tumour in a person affected by cancer. It is particularly preferred for R to be methyl. It is preferred for both of R, R 1 to be methyl. It is preferred for R 1 to be in particular 6-methyl or 8-methyl.
  • Preferred iron chelating compounds of the invention for use in the combination comprise N-(1-pyridine-2-yl-methylidene)-N-(9H-1,3,4,9-tetraaza-fluoren-2-yl)-hydrazine of the general formula I, wherein is
  • R R 1 R 2 also by methyl 8-methyl H particularly preferred CB21 methyl H H preferred methyl 6-methyl H preferred H 6-methyl H preferred H 8-methyl methyl preferred
  • iron chelating compounds for use in combination with the autophagy inhibiting agent of the invention include deferoxamine, deferiprone, and deferasirox.
  • the autophagy inhibiting agent is preferably selected from chloroquine.
  • Other preferred autophagy inhibiting agents comprise hydroxychloroquine, 3-methyladenine, adenosine, bafilomycin A1, 5-amino-4-imidazole carboxamide riboside, wortmannin, and viniblastine.
  • a method of treating a solid tumour in a person affected by cancer comprising administering to said person a pharmacologically effective dose of the combination of autophagy inhibiting agent and cell permeable iron chelator of the invention in a close temporal relationship, such as at the same time or within one day or one week.
  • Administration may be by any suitable route, such as parenteral or per-oral in form of separate pharmaceutical combinations, one comprising the autophagy inhibitor and a pharmaceutically acceptable carrier, for instance dimethyl sulfoxide, or in a single pharmaceutical combination when administered at the same time, comprising a pharmaceutically acceptable carrier such as dimethyl sulfoxide.
  • a method of treating a solid cancer in a person comprising administering to the person the combination of autophagy inhibiting agent and cell permeable iron chelator in pharmacologically effective dose, either simultaneously or in a close timely relationship, such as within an hour or a day or a week.
  • Administration is preferably in form of the pharmaceutical composition(s) disclosed above, and by the parenteral or peroral or other suitable route.
  • FIG. 1 illustrates the in-vivo activity of CB21 on FaDu head-neck carcinoma xenografts. SCID mice carrying FaDu tumours were treated with mg/kg of CB21 and tumour volume was calculated.
  • FIGS. 2 a - 2 h illustrate the cytotoxicity of N-(1-pyridine-2-yl-ethylidene)-N-(9H-1,3,4,9-tetraaza-8-methyl-fluoren-2-yl)-hydrazine (in the following identified as “CB21”) to HCT116 MCS and the therapeutic window of the cytotoxic effect.
  • FIG. 2 a Morphology and caspase-3 induction in MCS treated with CB21. HCT116 MCS were treated for 6 hours with 6 ⁇ M CB21, followed by changing to drug-free medium and further incubation. MCS were sectioned and stained for active caspase-3.
  • FIG. 2 b Clonogenic outgrowth of cells treated with the indicated compounds.
  • FIG. 2 c Proliferation of monolayer HCT116 cells in the presence or absence of CB21.
  • Cells were seeded at 7,000 cells/well in 96 well plates and treated with 3, 6 and 12.5 ⁇ M CB21.
  • FIG. 2 d EdU (5-ethynyl-2′-deoxyuridine) incorporation into monolayer HCT116 cells 24 hours after addition of 6 ⁇ M CB21. Cells were incubated for 30 minutes with EdU, fixed and analyzed by ArrayScan.
  • FIG. 2 e Quantification of EdU signal in the experiment shown in FIG. 2 d .
  • FIG. 2 f Proliferation of monolayer hTERTRPEI cells in the presence or absence of CB21.
  • FIG. 2 g CB21 does not affect the viability of confluent hTERTRPE1 cells. Cells were seeded at 7,000 or 70,000 per well in 96 well plates in the presence or absence of CB21.
  • FIG. 2 h Morphology of HCT116 and hTERTRPEI cells after exposure to 6 ⁇ M CB21.
  • FIGS. 3 a - 3 e illustrate that CB21 is a potent iron chelator.
  • FIG. 3 a Scores according to the Connectivity Map (Cmap) database.
  • FIG. 3 b Viability of cells treated with 6 ⁇ M CB21 in the presence or absence of iron chloride. Left: HCT116 cells; right HCT116 p53 ⁇ / ⁇ cells.
  • FIG. 3 c Viability of HCT116 cells treated with different iron chelators.
  • FIG. 3 d Viability of HCT116 cells treated with 7 compounds with structures related to CB21.
  • FIG. 3 e CB21 is the most effective of a series of related compounds in reducing MCS viability.
  • FIGS. 4 a - 4 f illustrate the induction of autophagy by CB21 and other iron chelators.
  • FIG. 4 a Morphology of monolayer HCT116 cells exposed to 6 ⁇ M CB21 for 6 hours and an additional 42 hours or 90 hours in drug-free medium.
  • FIG. 4 b Staining of CB21-treated cells with an antibody to LC3.
  • FIG. 4 c Induction of LC3-I and LC3-II protein by CB21 in monolayer and MCS HCT116. Cells were treated for 6 hours (6 ⁇ M), and then incubated further. Protein were extracted and subjected to western blot analysis.
  • FIG. 4 a Morphology of monolayer HCT116 cells exposed to 6 ⁇ M CB21 for 6 hours and an additional 42 hours or 90 hours in drug-free medium.
  • FIG. 4 b Staining of CB21-treated cells with an antibody to LC3.
  • FIG. 4 c Induction of LC3-I and LC
  • FIG. 4 d Induction of LC3-I and LC3-II protein by CB21 in monolayer and MCS hTERTRPEI cells.
  • FIG. 4 e Induction of LC3-I and LC3-II by different iron chelators. Cells were treated with VLX50 (50 ⁇ M); Deferasirox (60 ⁇ M), deferoxamine (200 ⁇ M), ciclopiroxolamine (15 ⁇ M), CB21 (5 ⁇ M), rapamycin (0.1 ⁇ M), NVP-BEZ235 (0.2 ⁇ M).
  • FIG. 4 f Morphology of peripheral and core cells visualized by electron microscopy. MCS were treated for 6 hours with 6 ⁇ M C21 and incubated further in drug-free medium for the times indicated and sectioned. Note the appearance of swollen mitochondria at 24 hours of CB21 treatment.
  • FIGS. 5 a - 5 f Illustrate that inhibition of CB21-induced autophagy increases CB21-cytotoxicity.
  • FIG. 5 a HCT116 monolayer cells were treated with 6 ⁇ M CB21 and/or 10 ⁇ M 3-MA and cell viability was determined after 48 hours.
  • FIG. 5 b HCT116 monolayer cells were transfected with siRNA to Beclin/Atg6 or control siRNA. After 24 hours cells were treated with 6 ⁇ M CB21 as indicated.
  • FIG. 5 c Bafilomycin A (10 ⁇ M) inhibits formation of LC3 positive vesicles in CB21-treated cells. Cells were fixed and stained for LC3 after drug treatment at the times indicated.
  • FIG. 5 a HCT116 monolayer cells were treated with 6 ⁇ M CB21 and/or 10 ⁇ M 3-MA and cell viability was determined after 48 hours.
  • FIG. 5 b HCT116 monolayer cells were transfected with siRNA to Beclin/Atg6 or control si
  • FIG. 5 d Bafilomycin A increases the cytotoxicity of CB21.
  • Cells were treated with 6 ⁇ M CB21 and/or 10 ⁇ M bafilomycin A and cells were photographed at the times Indicated.
  • FIG. 5 e Chloroquine increases the cytotoxicity of CB21 in monolayer culture.
  • HCT116 cells were treated with 6 ⁇ M CB21 and/or 10 ⁇ M chloroquine.
  • Cell proliferation was monitored by calculation of culture confluency.
  • FIG. 5 f Chloroquine increases the cytotoxicity of CB21 in MCS culture. Cell viability was determined using the acid phosphatase test. Note that background levels of acid phosphatase activity was observed in HCT116 MCS cultures containing no viable cells (generally ⁇ 30%), probably due to enzyme trapping.
  • FIGS. 6 a - 6 e Illustrate that CB21 induces a p53 and hypoxia response.
  • FIG. 6 a Gene expression profile induced by CB21 were analyzed by Affymetrix microarrays and the representation of genes associated with hypoxia, p53 networks and mitosis is shown.
  • FIG. 6 b Analysis of p53 and HIF-1a by western blotting. HCT116 cells were treated with 6 ⁇ M CB21 for the times indicated.
  • FIG. 6 c Induction of a HIF-1a-promoter driver GFP reporter by CB21.
  • FIG. 6 d Induction of BNIP3 by CB21 in HCT116 and hTERT-RPE1 cells.
  • HCT116 cells were transfected with siRNA to BNIP3 or control siRNA and treated with 6.25 ⁇ M CB21 after 24 hours. Viability was determined after 48 hours using the acid phosphatase assay.
  • FIGS. 7 a - 7 d illustrate that CB21 reduces respiration and inhibits mTOR.
  • FIG. 7 a Effect on glucose transport.
  • FIG. 7 b CB21 reduces oxygen consumption.
  • FIG. 7 c CB21 reduces hypoxia in HCT116 MCS. HCT116 MCS were treated as indicated and processed for pimonidazole immunohistochemistry. Note that CB21 reduces the area staining positive for pimonidazole adducts ( ⁇ 10 mm Hg O2).
  • FIG. 7 d CB21 inhibits phosphorylation of 4EBP1.
  • HCT116 cells were treated with CB21 for the times indicated and protein extracts were processed for western blotting. Reduction in 4EBP1 phosphorylation and induction of AKT phosphorylation should be noted.
  • FIGS. 8 a - 8 c illustrate that CB21 cytotoxicity is enhanced by glucose starvation.
  • FIG. 8 a Morphology of HCT116 MCS after incubation in the presence or absence of glucose for 24 hours.
  • FIG. 8 b HCT116 monolayer cells were treated with different concentrations of CB21 in glucose-containing or glucose-free medium. The levels of caspase-cleaved K18 was determined using M30 CytoDeath ELISA.
  • FIG. 8 c HCT116 monolayer cells were treated as in FIG. 8 b . Viability was determined using the acid phosphatase assay.
  • Compounds of the invention were obtained from compound libraries. They can be prepared according to methods described in the literature, such as in WO 02/089809, or by their non-inventive modifications. The compounds were dissolved in DMSO. A final concentration of 0.5% DMSO was reached in cell cultures.
  • HCT116 colon carcinoma cells were maintained in McCoy's 5A modified medium/10% fetal calf serum at 37° C. in 5% CO 2 .
  • MCS were prepared using a modification of our previously described method (12).
  • a cell suspension containing 10,000 cells (200 ⁇ l) was added to each well of poly-HEMA coated 96 well plates. The wells were then overfilled by adding an additional 170 ⁇ l media to acquire a convex surface curvature.
  • Plasticine spacers (3 mm) were placed in the corners of each plate to prevent the lids from touching the media. The plates were then inverted in order to allow the cells to sediment to the liquid/air interface and incubated in gentle shaking. After 24 hrs incubation the plates were returned to normal.
  • NP40 was added to the culture medium to a concentration of 0.1% to extract caspase-cleaved K18 from MCS and to include material released to the medium from dead cells.
  • Caspase cleaved keratin-18 (K18-Asp396) was determined using 25 mL medium/extract using the M30 CytoDeath ELISA assay (a variant of the M30-Apoptosense® ELISA (13) developed for in-vitro use (Peviva AB, Bromma, Sweden)).
  • hTERT-RPE1 cells were obtained from Clontech Laboratories, Mountain View, Calif.
  • hTERT-RPE1 is an immortalized human retinal epithelial cell line that stably expressed human telomerase reverse transcriptase (hTERT).
  • MCS Immunological assays.
  • MCS produced by the hanging drop method in 96 well plates were fixed in paraformaldehyde, dehydrated, embedded in paraffin and sectioned. Each sample contained 32 MCS (MCS from each 96 well plate were pooled into 3 groups). The sections were deparaffinized with xylene, rehydrated and microwaved, and then incubated overnight with the monoclonal primary antibodies diluted in 1% (weight/volume) bovine serum albumin and visualized by standard avidin-biotin-peroxidase complex technique (Vector Laboratories, Burlingame, Calif., USA). Counterstaining was performed with Mayer's haematoxylin.
  • Antibody MIB-1 (against the nuclear proliferation-associated antigen Ki67) was obtained from Immunotech SA, Marseille, France and used at a dilution of 1:150; antibody against active caspase-3 was obtained from Pharmingen and used at a dilution of 1:50.
  • Connectivity Map The Connectivity Map (CMAP) (www.broad.mit.edu/cmap) build 02 contains genome-wide expression data for 1300 compounds (6100 instances, including replicates, different doses and cell lines).
  • CMAP Connectivity Map
  • RNA expression analysis was performed using Genome U133 Plus 2.0 Arrays according to the GeneChip Expression Analysis Technical Manual (Rev. 5, Affymetrix Inc., Santa Clara, Calif.).
  • Raw data was normalized with MAS5 (Affymetrix) and gene expression ratios for drug treated vs. vehicle control cells were calculated to generate lists of regulated genes. Filter criteria were present call for all genes in the treated cell line and an expression cut-off of at least 100 arbitrary expression units. For CMAP compatibility reasons only probes present on HG U133A were used. To retrieve a ranked compound list the 40 most up and down regulated genes (i.e. probes) for each compound were uploaded into the CMAP and compared with the 6100 instances in the CMAP database.
  • Oxygen consumption Measurement of respiration was performed as described (16). Succinate (5 mM) in the presence of rotenone (2 mM), malate+pyruvate (5 mM each) and TMPD (0.5 mM)+ascorbate (1 mM) were used as mitochondrial substrates.
  • mice Treatment of mouse xenografts. When HCT116 tumours in SCID mice had grown to a size of 200 mm 3 the mice were injected with drugs i.p., and tumour size measured daily.
  • the Compound of the Invention Induces Apoptosis and Reduces Viability of MCS
  • the Compound of the Invention is a Cell Permeable Iron Chelator
  • CMap Connectivity Map
  • CB21 The anti-proliferative activity of CB21 was compared with that of other known iron chelators. CB21 was found to be more potent than VLX50, deferasirox, ciclopiroxolamine, deferoxamine ( FIG. 3 c ). Structure-activity relationships were examined by use of a number of structurally related compounds ( FIGS. 3 d , 3 e ). These studies showed that CB21 was the most effective compound in both monolayer and MCS cultures.
  • the Compound of the Invention Induces a Widespread Autophagic Response
  • the anti-tumourigenic activity of iron chelators is generally attributed to inhibition of ribonucleotide reductase, leading to inhibition of cell proliferation (18).
  • MCSs contain mostly non-proliferating cells.
  • the finding of induction of cytotoxic effects on MCSs by the iron chelator CB21 thus was unexpected.
  • the mechanism(s) of action was studied in more detail. Visual inspection of CB21-treated cells revealed that cells contained multiple large cytoplasmic vesicles ( FIG. 4 a ). These vesicles stained positively with an antibody to microtubule associated protein 1 light chain 3 (LC3), suggesting that they were associated with autophagy. LC3 staining was observed at 24 h and was stronger at 42 h ( FIG. 4 b ).
  • HCT116 cells were treated with cytotoxic concentrations of different iron chelators for 24 h.
  • Induction of LC3-I and LC3-II was observed in all instances, showing that LC3 induction was a general effect of iron chelators ( FIG. 4 e ).
  • Induction of LC3-I and -II by iron chelators was much stronger compared to that observed after treatment with rapamycin or NVP-BEZ235 (no induction observed at 24 h, weak induction at 6 h).
  • HCT116 MCS were treated with CB21 for 6 h, washed and incubated for different time periods, fixed, sectioned, and examined by electron microscopy. Large vesicles were observed in cells starting at 24 h after treatment ( FIG. 4 f ). Notably, a common feature of CB21-treated MCS was the early appearance of enlarged and swollen mitochondria ( FIG. 4 f ). Most importantly, massive vacuolization occurred in a time-dependent manner not only in cells in the MCS periphery but also in the center of the MCS ( FIG. 4 f ). It was concluded that CB21 induces the formation of vesicles in the cells of the central cores of MCS, found to be resistant to apoptosis, and that this response was associated with loss of viability of these cells.
  • Autophagy is generally considered to be a survival response to stress conditions but may also be a mechanism of programmed cell death (20, 21).
  • 3-MA 3-MA
  • a PI3K inhibitor commonly used as an inhibitor of autophagy was potentiated by 3-MA
  • a knock-down of Beclin/Atg6 using siRNA was performed. This resulted in almost complete knock-down of the expression of this protein. Beclin/Atg6 knock-down reduced the viability of HCT116 cells by ⁇ 50%; viability was further reduced by CB21 toxicity ( FIG. 5 b ).
  • Chloroquine is a lysosomotropic agent widely used to inhibit the maturation of autophagosomes into degradative autolysosomes (22).
  • CQ has no effect on its own on the proliferation of HCT116 cells.
  • the combination of CQ and CB21 resulted in a strong potentiation of cell death on monolayer HCT116 cells ( FIG. 5 e ).
  • Examination of cytotoxicity to MCS of the combination of CQ and CB21 revealed an effect potentiated in comparison with the effect of either CQ or CB21 on MCS ( FIG. 5 f ).
  • CB21 induced a number of hypoxia responsive genes and also a number of genes known to be regulated by p53 ( FIG. 6 a ).
  • a large induction was also observed using a reporter cell line where GFP is regulated by the HIF-la promoter ( FIG. 6 c ).
  • BNIP3 is a known target of HIF-1 a (23). BNIP3 expression has been reported to induce extensive cytoplasmic vacuolization and autophagy (24). CB21 was found to induce the expression of BNIP3 protein in HCT116 cells ( FIG. 6 d ). BNIP3 expression was, however, also strongly induced by CB21 in hTERT-RPE1 cells ( FIG. 6 d ). This finding is not consistent with BNIP3 being a presumed mechanism of CB21-induced autophagy. Knock-down of BNIP3 using siRNA did not decrease induction of LC3-ll and cell death by CB21 ( FIG. 6 e ).
  • the Compound of the Invention Inhibits Oxygen Consumption and Decreases mTOR Activity
  • HCT116 monolayer cultures were treated with the mitochondrial uncoupling agent carbonylcyanide-3-chlorophenylhydrazone, CCCP, known to increase oxygen consumption.
  • CCCP mitochondrial uncoupling agent carbonylcyanide-3-chlorophenylhydrazone
  • the mammalian target of rapamycin is a serine/threonine kinase regulating cell growth in response to nutrient status. It is well established that metabolic stress affects the activity of the mTOR pathway (27). The mTOR pathway regulates mitochondrial oxygen consumption and oxidative capacity (28, 29).
  • phosphorylation of the mTOR substrate 4EBP1 was examined. As shown in FIG. 7D , 4EBP1 phosphorylation is inhibited by CB21. The decrease in phosphorylation is associated with an increased AKT-phosphorylation.
  • Inhibition of mTORC1 is known to release a negative feedback loop involving, resulting in strong Akt activation (30).
  • HCT116 MCS was treated with rapamycin (a specific pharmacological inhibitor of mTOR-raptor complex formation) and stained sections with pimonidazole. A reduction of pimonidazole staining was observed, although not as strong as with CB21 ( FIG. 7 c ; Table 1).
  • NVP-BEZ235 a compound in clinical trials.
  • NVP-BEZ235 was found to decrease 4EBP1 phosphorylation in HCT116 cells grown both under monolayer or MCS conditions ( FIG. 7 d ).
  • NBPBEZ235 did not affect the viability of the cells in the core of the MCS.
  • Oxygen consumption has been reported to decrease in the interior regions of tumour MCS, possibly as a consequence of decreased proliferative activity (32, 33).
  • Other investigators have found that oxygen consumption is rather uniform in viable regions of MCS (34); it has been reported that fibroblast clones at the same stage of transformation may have quite distinct metabolic activity in MCS culture (33).
  • a further decrease induced by CB21 is expected to lead to an increased dependence of glucose.
  • monolayer cells may compensate increased glucose dependence by increased uptake (as shown in FIG. 8 a ), glucose will be limiting in MCS. As shown in FIG.
  • HCT116 MCS core cells are dependent on glucose for survival: glucose depletion leads necrosis of central areas, an effect is reminiscent of that of CB21 ( FIG. 2 ). Based on these considerations it was tested whether glucose starvation increases the sensitivity of HCT116 monolayer cells to CB21. This was indeed the case: glucose starvation decreased cell viability and increased apoptosis by CB21. These findings are likely to at least partly explain the sensitivity of central core cells to CB21.
  • the in-vivo anti-tumour activity by CB21 was examined in the HCT116 model. Tumours were allowed to grow to a size of ⁇ 0.2 mL and then treated with CB21. A clear anti-tumour effect of the compound CB21 was observed ( FIG. 1 ).
  • a number of iron chelators have been developed that exhibit anti-tumour activity, including Triapine (35), Tachpyr (36) and Trensox (37). Iron is important for many metabolic reactions, including the formation of deoxyribonucleotides from ribonucleotides by ribonucleotide reductase (38).
  • CB21 Another effect of CB21, shared by other iron chelators (41), is the induction of LC3 positive cytoplasmic vesicles and LC3-II protein.
  • LC3 induction was found to be much less pronounced in hTERT-RPE1 cells.
  • the induction of LC3-II by iron chelators was significantly stronger than that observed with mTOR inhibitors, suggesting that LC3-II induction was not mediated exclusively by mTOR inhibition.
  • Glucose starvation of MCS induced cell death of the core cells consistent with the concept that survival of this cell population is dependent on glucose.
  • the increased dependence of glucose observed after treatment with CB21 is very likely contributing to cell death of the population of core cells.
  • Apoptosis did not appear to be the main mechanism of cell death by CB21, as evidenced by weak caspase-3 induction compared to the strong induction of caspase-3 in peripheral cells (not shown).
  • Conditions of poor cellular energy status may lead to resistance to apoptosis (also explaining the resistance of core cells to NSC647889-induced apoptosis (not shown). It seems that CB21 induces increased glucose dependence of HCT116 cells, and that this leads to decreased viability of hypoxic cells in MCS cores.
  • Autophagy is a catabolic degradation response to metabolic stress, which strives to maintain homeostasis through degradation of proteins and organelles.
  • PI3K-Akt-mTOR, LKB1-AMPK-mTOR and p53 are the main regulators of the autophagic pathway.
  • Autophagy is believed to be involved in mediating resistance of cancer cells to anticancer therapy and to be an attractive therapeutic target in anticancer drug resistance (20, 43).
  • CB21 induced a remarkable autophagic response, characterized be strong LC3-I and -II induction.
  • the present invention reveals inhibition of autophagy to potentiate the cytotoxicity of CB21.
  • Lum J J et al Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 005;120:237-48.
  • Triapinel (3- aminopyridine -2- carboxaldehyde - thiosemicarbazone ): A potent inhibitor of ribonucleotide reductase activity with broad spectrum antitumour activity. Biochem Pharmacol 2000; 59:983-91.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US14/006,277 2011-03-21 2012-03-14 Treatment of Solid Tumours Abandoned US20140073645A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
SE1100201-1 2011-03-21
SE1100201 2011-03-21
PCT/SE2012/000034 WO2012128689A1 (en) 2011-03-21 2012-03-14 Treatment of solid tumours

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2012/000034 A-371-Of-International WO2012128689A1 (en) 2011-03-21 2012-03-14 Treatment of solid tumours

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/625,337 Division US10022380B2 (en) 2011-03-21 2017-06-16 Treatment of solid tumours

Publications (1)

Publication Number Publication Date
US20140073645A1 true US20140073645A1 (en) 2014-03-13

Family

ID=46879602

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/006,277 Abandoned US20140073645A1 (en) 2011-03-21 2012-03-14 Treatment of Solid Tumours
US15/625,337 Active US10022380B2 (en) 2011-03-21 2017-06-16 Treatment of solid tumours

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/625,337 Active US10022380B2 (en) 2011-03-21 2017-06-16 Treatment of solid tumours

Country Status (20)

Country Link
US (2) US20140073645A1 (xx)
EP (1) EP2688569B1 (xx)
JP (1) JP6064215B2 (xx)
KR (1) KR101937279B1 (xx)
CN (1) CN103547268B (xx)
AU (1) AU2012231814B2 (xx)
BR (1) BR112013024211B1 (xx)
CA (1) CA2830081C (xx)
DK (1) DK2688569T3 (xx)
EA (1) EA025180B1 (xx)
ES (1) ES2681703T3 (xx)
HU (1) HUE039021T2 (xx)
IL (1) IL228411A (xx)
MX (1) MX2013010770A (xx)
PL (1) PL2688569T3 (xx)
PT (1) PT2688569T (xx)
SG (2) SG193494A1 (xx)
TR (1) TR201809040T4 (xx)
WO (1) WO2012128689A1 (xx)
ZA (1) ZA201307036B (xx)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150259349A1 (en) * 2012-09-21 2015-09-17 Vivolux Ab Means and Method for Treating Solid Tumors
WO2018119207A1 (en) * 2016-12-21 2018-06-28 The Medical College Of Wisconsin, Inc. Synergistic inihibition of tumor cell proliferation induced by combined treatment of metformin compounds and iron chelators
CN110585194A (zh) * 2019-10-12 2019-12-20 广州医科大学附属第五医院 倍半萜内酯类化合物在制备溶酶体自噬抑制剂以及抗癌药物中的应用
CN111849873A (zh) * 2020-07-30 2020-10-30 扬州大学 一种诱导鸡的胚胎干细胞自噬的方法
US20210369692A1 (en) * 2017-11-23 2021-12-02 Deutsches Krebsforschungszentrum Iron chelators in tumor therapy
US11504346B2 (en) 2013-11-03 2022-11-22 Arizona Board Of Regents On Behalf Of The University Of Arizona Redox-activated pro-chelators

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105807976B (zh) 2014-12-31 2019-02-12 清华大学 静电传感器
WO2016168862A1 (en) * 2015-04-17 2016-10-20 Memorial Sloan-Kettering Cancer Center Use of mva or mvadeltae3l as immunotherapeutic agents against solid tumors
CN104997777A (zh) * 2015-07-24 2015-10-28 孔小乐 去铁酮的2比1锌络合物作为制备抗癌药物的应用
JP7103745B2 (ja) * 2015-10-05 2022-07-20 国立大学法人 岡山大学 癌幹細胞抑制剤、癌の転移又は再発の抑制剤並びに癌細胞の未分化マーカー発現抑制剤
JP6929299B2 (ja) 2015-12-18 2021-09-01 ヴィヴォルックス アーベー インドール誘導体を含む医薬組成物、その調製方法及びその使用
JP6900067B2 (ja) * 2017-02-07 2021-07-07 オンコクロス カンパニー,リミテッド 癌の転移抑制および治療用組成物
KR101859922B1 (ko) * 2017-02-07 2018-05-21 주식회사 온코크로스 클로페네신을 포함하는 대장암 증식 및 전이 억제용 조성물
CN106831776B (zh) * 2017-03-16 2018-12-07 河北科技大学 5,10-二氢吡啶并吡嗪并三嗪类化合物及其应用
CN109646679A (zh) * 2019-01-28 2019-04-19 中国科学院长春应用化学研究所 铁离子螯合剂及其可药用盐的用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030092716A1 (en) * 2001-05-04 2003-05-15 The Procter & Gamble Company Medicinal uses of hydrazones
WO2009035534A2 (en) * 2007-09-07 2009-03-19 The Cleveland Clinic Foundation Treatment of ischemic eye disease by the systematic pharmaceutical activation of hypoxia inducible factor (hif)

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0233101A1 (fr) * 1986-01-13 1987-08-19 Ire-Celltarg S.A. Dérivés de vinblastine et composition pharmaceutique les contenant
US5480906A (en) * 1994-07-01 1996-01-02 Eli Lilly And Company Stereochemical Wortmannin derivatives
IL121272A (en) * 1997-07-10 2000-06-01 Can Fite Technologies Ltd Pharmaceutical compositions comprising adenosine and their use for treating or preventing leukopenia
WO2004069801A1 (en) * 2003-02-05 2004-08-19 Unisearch Limited Metal ion chelators and therapeutic use thereof
US7767689B2 (en) 2004-03-15 2010-08-03 Ptc Therapeutics, Inc. Carboline derivatives useful in the treatment of cancer
WO2006055412A1 (en) * 2004-11-19 2006-05-26 Shiva Biomedical, Llc Methods of treating erythropoietin-resistance
JP2009536180A (ja) * 2006-05-09 2009-10-08 ノバルティス アクチエンゲゼルシャフト 鉄キレート剤と抗新生物剤を含む組合せ剤およびそれらの使用
KR101208587B1 (ko) * 2009-06-24 2012-12-06 여오영 하이드록시클로로퀸을 포함하는 항암 치료를 위한 국소 투여용 주사제 조성물
WO2014046589A1 (en) 2012-09-21 2014-03-27 Vivolux Ab Means and method for treating solid tumours

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030092716A1 (en) * 2001-05-04 2003-05-15 The Procter & Gamble Company Medicinal uses of hydrazones
WO2009035534A2 (en) * 2007-09-07 2009-03-19 The Cleveland Clinic Foundation Treatment of ischemic eye disease by the systematic pharmaceutical activation of hypoxia inducible factor (hif)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Sasaki et al. BMC Cancer, 2010, Vol. 10, No. 370, pgs. 1-11. *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150259349A1 (en) * 2012-09-21 2015-09-17 Vivolux Ab Means and Method for Treating Solid Tumors
US9562046B2 (en) * 2012-09-21 2017-02-07 Vivolux Ab Means and method for treating solid tumors
US11504346B2 (en) 2013-11-03 2022-11-22 Arizona Board Of Regents On Behalf Of The University Of Arizona Redox-activated pro-chelators
WO2018119207A1 (en) * 2016-12-21 2018-06-28 The Medical College Of Wisconsin, Inc. Synergistic inihibition of tumor cell proliferation induced by combined treatment of metformin compounds and iron chelators
EP3558282A4 (en) * 2016-12-21 2020-08-12 The Medical College of Wisconsin, Inc. SYNERGIC INHIBITION OF TUMOR CELL PROLIFERATION INDUCED BY A THERAPEUTIC ASSOCIATION OF METFORMIN COMPOUNDS AND IRON CHELATORS
US20210369692A1 (en) * 2017-11-23 2021-12-02 Deutsches Krebsforschungszentrum Iron chelators in tumor therapy
US11931349B2 (en) * 2017-11-23 2024-03-19 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Iron chelators in tumor therapy
CN110585194A (zh) * 2019-10-12 2019-12-20 广州医科大学附属第五医院 倍半萜内酯类化合物在制备溶酶体自噬抑制剂以及抗癌药物中的应用
CN111849873A (zh) * 2020-07-30 2020-10-30 扬州大学 一种诱导鸡的胚胎干细胞自噬的方法

Also Published As

Publication number Publication date
EA025180B1 (ru) 2016-11-30
CA2830081A1 (en) 2012-09-27
AU2012231814A1 (en) 2013-09-26
SG193494A1 (en) 2013-10-30
CA2830081C (en) 2020-09-22
US20170348317A1 (en) 2017-12-07
KR101937279B1 (ko) 2019-01-10
WO2012128689A1 (en) 2012-09-27
TR201809040T4 (tr) 2018-07-23
ES2681703T3 (es) 2018-09-14
PT2688569T (pt) 2018-08-06
ZA201307036B (en) 2014-05-28
HUE039021T2 (hu) 2018-12-28
BR112013024211A2 (pt) 2016-12-20
CN103547268A (zh) 2014-01-29
EP2688569B1 (en) 2018-05-30
PL2688569T3 (pl) 2018-09-28
AU2012231814B2 (en) 2016-06-09
EA201391286A1 (ru) 2014-03-31
IL228411A (en) 2016-12-29
MX2013010770A (es) 2014-03-27
US10022380B2 (en) 2018-07-17
SG10201605083SA (en) 2016-08-30
EP2688569A1 (en) 2014-01-29
JP6064215B2 (ja) 2017-02-01
EP2688569A4 (en) 2014-09-10
KR20140017619A (ko) 2014-02-11
CN103547268B (zh) 2017-02-22
JP2014508804A (ja) 2014-04-10
BR112013024211B1 (pt) 2020-12-15
IL228411A0 (en) 2013-12-31
DK2688569T3 (en) 2018-08-06

Similar Documents

Publication Publication Date Title
US10022380B2 (en) Treatment of solid tumours
Lu et al. Colistin-induced autophagy and apoptosis involves the JNK-Bcl2-Bax signaling pathway and JNK-p53-ROS positive feedback loop in PC-12 cells
Islam et al. Eugenol potentiates cisplatin anti‐cancer activity through inhibition of ALDH‐positive breast cancer stem cells and the NF‐κB signaling pathway
Reyes et al. Sorafenib and 2-deoxyglucose synergistically inhibit proliferation of both sorafenib-sensitive and-resistant HCC cells by inhibiting ATP production
Sun et al. Magnoflorine inhibits human gastric cancer progression by inducing autophagy, apoptosis and cell cycle arrest by JNK activation regulated by ROS
Li et al. Dichloroacetate and metformin synergistically suppress the growth of ovarian cancer cells
Dey et al. Synergistic therapy with tangeretin and 5-fluorouracil accelerates the ROS/JNK mediated apoptotic pathway in human colorectal cancer cell
Forkuo et al. In vitro anti-malarial interaction and gametocytocidal activity of cryptolepine
Pullarkat et al. Iron chelators induce autophagic cell death in multiple myeloma cells
WO2022062223A1 (zh) 金诺芬在制备用于治疗去势抵抗性前列腺癌药物中的应用
Shi et al. Enhancement of Anticancer Efficacy and Tumor Penetration of Sorafenib by Ionic Liquids
Su et al. Blocking Parkin/PINK1-mediated mitophagy sensitizes hepatocellular carcinoma cells to sanguinarine-induced mitochondrial apoptosis
Zhang et al. Toosendanin and isotoosendanin suppress triple-negative breast cancer growth via inducing necrosis, apoptosis and autophagy
KR102119150B1 (ko) N-1h-벤지미다졸-2-일-3-(1h-피롤-1-일) 벤자미드를 유효성분으로 함유하는 암 예방 또는 치료용 약학적 조성물
Han et al. Lidocaine enhances the efficacy of palbociclib in triple-negative breast cancer
Xia et al. Artesunate alleviates 5-fluorouracil-induced intestinal damage by suppressing cellular senescence and enhances its antitumor activity
Li et al. Differential effects of ginkgol C17: 1 on cisplatin‑induced cytotoxicity: Protecting human normal L02 hepatocytes versus sensitizing human hepatoma HepG2 cells
Goldberg et al. 3, 3′-Diindolylmethane (DIM) and its ring-substituted halogenated analogs (ring-DIMs) induce differential mechanisms of survival and death in androgen-dependent and–independent prostate cancer cells
US20220409611A1 (en) Methods of treating, ameliorating, and/or preventing cancer using pyrvinium compositions
Liu et al. New 1, 1-diarylethylene FOXM1 inhibitors potently reduce intracellular FOXM1 and suppress high-grade serous ovarian carcinoma cell viability
Carrero et al. Parasiticidal effect of 16α-bromoepiandrosterone (EpiBr) in amoebiasis and cysticercosis
KR20090058423A (ko) 소디움 메타아르세나이트를 함유하는 c형 바이러스성 간염치료용 조성물
OWADH et al. Forty Micromole Hydroxychloroquine Enhanced Cytotoxic Effect of Doxorubicin Against Laryngeal Cancer Cell Line HEp-2
Yin et al. Corynoxine and corynoxine B enhance the antitumor activity of gemcitabine in Panc-1 cells via ROS-p38 signaling pathway
WO2023104809A2 (en) 2-oxothiazole compositions for treatment of t-cell acute lymphoblastic leukaemia

Legal Events

Date Code Title Description
AS Assignment

Owner name: VIVOLUX AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LINDER, STIG;FRYKNAS, MARTEN;LARSSON, ROLF;SIGNING DATES FROM 20131120 TO 20131123;REEL/FRAME:031698/0068

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION