[go: up one dir, main page]

WO2025039073A1 - Inhibiteur d'hélicase à arn - Google Patents

Inhibiteur d'hélicase à arn Download PDF

Info

Publication number
WO2025039073A1
WO2025039073A1 PCT/CA2024/051074 CA2024051074W WO2025039073A1 WO 2025039073 A1 WO2025039073 A1 WO 2025039073A1 CA 2024051074 W CA2024051074 W CA 2024051074W WO 2025039073 A1 WO2025039073 A1 WO 2025039073A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
cells
myc positive
elf4a1
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.)
Pending
Application number
PCT/CA2024/051074
Other languages
English (en)
Inventor
Gerard Pelletier
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.)
Royal Institution for the Advancement of Learning
Original Assignee
Royal Institution for the Advancement of Learning
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 Royal Institution for the Advancement of Learning filed Critical Royal Institution for the Advancement of Learning
Publication of WO2025039073A1 publication Critical patent/WO2025039073A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4355Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having oxygen as a ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered

Definitions

  • This disclosure relates to the field of inhibitors of RNA helicases, methods of making same and uses thereof in the treatment of cancer.
  • Breast cancer is a heterogeneous disease that is classified into three main histological subtypes.
  • the histological subtypes inform on patient outcome and treatment options for clinical management of the disease.
  • the treatments include estrogen/progesterone receptor (ER/PR)- positive (65% incidence), human epidermal growth factor receptor 2 (HER2)-positive ( ⁇ 20% incidence) and triple negative breast cancers (TNBC: ER-/PR-/HER2-) (15% incidence).
  • ER/PR estrogen/progesterone receptor
  • HER2 human epidermal growth factor receptor 2
  • TNBC triple negative breast cancers
  • TNBC tumors tend to be more aggressive and are more likely to be diagnosed in younger (premenopausal) women.
  • Standard of care therapy for women with ER+ disease includes endocrine therapies that disrupt ER signaling and/or reduce ER levels within breast cancer cells (tamoxifen, fulvestrant) or alternatively impair estrogen production (aromatase inhibitors such as letrozole).
  • HER2+ patients are treated with trastuzumab, a HER2+ monoclonal antibody, which is highly effective for women diagnosed with early stage disease. In contrast, for women with TNBC, effective targeted therapies remain elusive, and chemotherapy is the standard of care.
  • chemotherapeutic regimens minimally impact disease-free and overall survival rates in non-responders or in women who experience relapse.
  • a high degree of intra- tumoral heterogeneity was thought to, at least in part, underpin intrinsic or acquired chemoresistance for many TNBC tumors. Indeed, following neo-adjuvant chemotherapy, patients diagnosed with TNBC tumors showed decreased 3-year survival rates, and increased rate of recurrence and progression to metastatic disease following treatment compared to women with non-TNBC disease.
  • the high degree of genomic instability for many TNBCs has informed pre- clinical studies to explore whether poly ADP ribose polymerase (PARP) inhibitors and/or immune checkpoint inhibitors represent promising classes of compounds for this subtype.
  • PARP poly ADP ribose polymerase
  • an enantiomeric mixture comprising the compounds of formula la and lb:
  • compositions comprising the compound of formula, the enantiomeric mixture or the full racemate as defined herein.
  • a composition comprising a full racemate of the compound of formula I and a pharmaceutically acceptable excipient.
  • a composition comprising a racemate of the compounds of formula la and lb and a pharmaceutically acceptable excipient.
  • MYC positive cancers including lung cancer, leukemia, breast cancer, myeloproliferative disorders, colorectal cancer, medulloblastoma, renal, hepatocellular cancer, melanoma, ovarian cancer, prostate cancer, esophageal adenocarcinoma, liposarcoma, esophageal squamous cancer, gastrointestinal stromal tumor, glioma, myxofibrosarcoma, leiomyosarcoma, neuroblastoma, synovial sarcoma, mesothelioma, gastric cancer, thyroid cancer, lymphoma, osteosarcoma, rhabdomyosarcoma, fibrosarcoma, epithelial cancer, and neural cancer.
  • the MYC positive cancer can be breast cancer.
  • FIG. 1 A is a graph assessing the compound-induced clamping of elF4A1 to fluorescein amidities (FAM) - labelled RNA of the indicated nucleotide composition.
  • the AmP obtained with elF4A1 «ATP «poly (NN)s RNA was measured for the indicated compounds at 10 pM.
  • FIG. 1 B is a graph assessing compound-induced clamping of DDX3X to FAM-labelled poly(AG)8 RNA.
  • the AmP was measured for 2 pM protein and 2 pM compound concentration.
  • the AmP obtained relative to DMSO is shown.
  • n 3 ⁇ SD
  • FIG. 1C is a graph showing the relative dissociation of pre-formed elF4A1 «ATP «Cmpd*FAM-poly (AG)a complexes measured as a function of time in the presence of 1000-fold molar excess poly (AG)s RNA.
  • DMSO ti/2 ⁇ 4.1 ⁇ 1 min
  • CR-1-31 B 10 pM
  • MG-002 (10 pM), ti/2 ⁇ 68 ⁇ 2.8 min
  • eFT226 (10 pM), t-1/2 ⁇ 27 ⁇ 5.8 min. Error values were calculated from the 95% confidence intervals.
  • FIG. 1 D is a graph of the differential scanning fluorimetry (DSF) analysis of elF4A1 (8 pM) in the presence of the indicated compounds (15 pM), 15 pM poly(AG)s, and 1 mM AMPPNP.
  • the transition midpoint temperature shifts (AT50) are: CR-1-31 B, 7.3°C; eFT226, 7°C; MG-002, 7.2°C; PatA, 10°C.
  • n 3 ⁇ SD.
  • FIG. 1 F is a schematic of bicistronic mRNA reporters with (AG)- or (UC)- enriched 5’ leader regions.
  • FIG. 11 is a bar graph showing that MG-002 is a potent inhibitor of cellular translation.
  • FIG. 1J is a graph showing the sucrose density gradient for polysomes isolated from eHAP1 cells exposed to vehicle or 15 nM MG-002 for 1 h.
  • FIG. 2D is a bar graph showing the activity of CR-1 -31 B, eFT226, and MG-002 towards the TNBC cell line MDA_MB-31 .
  • FIG. 2H is a Western blot analysis of the indicated proteins in cells exposed to MG- 002 (100 nM), CR-1-31 B (100 nM), or doxorubicin (Dox, 0.5 pg/ml) for 24 h.
  • FIG. 3A is a graph showing the activity of MG-002 cells towards parental eHAP1 or elF4A1 F163L /elF4A2- cells.
  • FIG. 3B is a schematic showing that in eHAP1 cells, rocaglate-induced clamping of elF4A1 to mRNAs leads to inhibition of translation and cell death.
  • FIG. 3C is a schematic showing that elF4A1 F163L /elF4A2 _ eHAP1 cells are significantly resistant to rocaglates as clamping of elF4A1 F163L to RNA is impaired.
  • FIG. 3D is a schematic showing an overexpression of wild type (wt) elF4A1 in elF4A1 F163L /elF4A2 _ cells is expected to re-sensitize these to rocaglate-induced translation inhibition and cell death.
  • FIG. 3E is a Western blot of proteins from stable cell lines ectopically expression elF4A1 , elF4A2, or DDX3X. Blots were probed with antibodies targeting the V5 tag or proteins indicated to the right.
  • FIG. 3F is a graph showing the cytotoxicity of elF4A1 F163L /elF4A2 _ eHAP1 cells ectopically expressing GFP, elF4A1 , elF4A2, or DDX3X following a two-day exposure to MG-002.
  • n 3 ⁇ SD.
  • FIG. 3H is a Western blot of elF4A1 F163L /elF4A2 _ eHAP1 cells ectopically expressing GFP, elF4A1 , or elF4A3. Blots were probed with antibodies targeting the V5 tag or elF4A3.
  • FIG. 4A is a graph showing the average plasma concentration of MG-002 at the indicated time points following delivery of 0.5 mg/kg MG-002 by oral gavage (PO).
  • FIG. 4B is a graph showing polysomes isolated from livers of mice treated with vehicle (control).
  • FIG. 4C is a graph showing polysomes isolated from livers of mice treated at the indicated time points following delivery of 0.5 mg/kg MG-002 PO (time point 7 h).
  • FIG. 4D is a graph showing polysomes isolated from livers of mice treated at the indicated time points following delivery of 0.5 mg/kg MG-002 PO (time point 24 h).
  • FIG. 4M here should we add FIG. 4C of the paper
  • FIG. 5A shows the gel of the puromycin assays assessing de novo protein synthesis in 4T1-526 cells treated with increasing concentration of MG-002 (1-50 nM) compared to vehicle control. Puromycin incorporation was visualized by immunoblot analysis using puromycin-specific antibodies and Ponceau S staining verified similar protein content between the various conditions.
  • FIG. 5B is a quantification of the data of Fig. 5A to determine the IC50 for the ability of MG-002 to inhibit protein synthesis in 4T1-526 cells.
  • FIG. 5C is a comparative to Fig. 5B with eFT226 instead of MG-002 in 4T1-526 cells.
  • FIG. 5D is a graph of the live cell counts using the IC50 concentrations calculated from
  • MG-002 was tested for its ability to inhibit the growth of 4T1-526 cells after 72 hours in culture, compared to the vehicle control.
  • Technical duplicate mean ⁇ SD was plotted, and statistical analysis performed with a One-way ANOVA (Dunnett’s multiple comparisons test). *, p ⁇ 0.05; **, p ⁇ 0.01 .
  • FIG. 6A is a graph showing the tumour volume of primary tumor growth of lung metastatic 4T1-526 TNBC over time up to reaching ⁇ 100 mm 3 in mouse.
  • FIG. 6B is a bar graph showing the change in mouse weight of the mice of Fig. 6A.
  • FIG. 6C is a bar graph showing the change in liver weight of the mice of Fig. 6A.
  • FIG. 6D is a bar graph showing the change in % Ki67 positive cells in the mice of Fig. 6A.
  • FIG. 6E is a bar graph showing the change in % cleaved Casp3 positive cells in the mice of Fig. 6A.
  • FIG. 6F is a bar graph showing the change in % Myc positive pixels for the mice of Fig. 6A.
  • FIG. 6G is a graph showing the Pearson correlation examining the relationship between Myc positivity and the percentage Ki67 positive cells (individual vehicle-treated tumors are represented by circles; individual MG-002 treated tumors are represented by squares).
  • FIG. 6H is a graph showing the Pearson correlation examining the relationship between Myc positivity and the percentage of cleaved Casp3 positive cells (individual vehicle- treated tumors are represented by circles; individual MG-002 treated tumors are represented by square.
  • FIG. 7A is a graph showing the tumor volume overtime for a 4T1-526 mammary tumor that was allowed to develop in the mammary fat pads of BALB/c mice and when the tumors reached ⁇ 100 mm 3 (arrow), the animals were randomized into three groups: 0.5 mg/kg MG-002, 0.5 mg/kg eFT226, or vehicle control with intraperitoneal administration.
  • FIG. 7B is a bar graph showing the change in weight for the mice of Fig. 7A.
  • FIG. 7C is a bar graph showing the change in liver weight at the end point of the mice of Fig. 7A.
  • FIG. 7D is a graph showing the tumor volume overtime for a 4T1-526 mammary tumor that was allowed to develop in the mammary fat pads of BALB/c mice and when the tumors reached ⁇ 100 mm 3 (arrow), the animals were randomized into three groups: 0.5 mg/kg MG-002, 0.5 mg/kg eFT226, or vehicle control with intravenous administration.
  • FIG. 7E is a bar graph showing the change in weight for the mice of Fig. 7D.
  • FIG. 7F is a bar graph showing the change in liver weight at the end point of the mice of Fig. 7D.
  • FIG. 7G is a graph showing the tumor volume overtime for a MDA-MB-231 mammary tumor that was allowed to develop in the mammary fat pads of BALB/c mice and when the tumors reached ⁇ 100 mm 3 (arrow), the animals were randomized into two groups: 0.5 mg/kg MG-002 or vehicle control with intraperitoneal administration.
  • FIG. 7H is a bar graph showing the change in weight for the mice of Fig. 7G.
  • FIG. 7I is a bar graph showing the change in liver weight at the end point of the mice of Fig. 7G.
  • FIG. 8A is a schematic showing diagram outlining the experimental schedule to assess the ability of MG-002 to prevent the formation of spontaneous lung metastases from the primary tumor (MFP, mammary fat pad).
  • MFP primary tumor
  • FIG. 8B is a bar graph showing the experimental results of 4T1- 526 following the schedule of Fig. 8A where mice were either treated by oral gavage with MG-002 (0.5 mg/kg), or vehicle control every three days until surgical resection. Lung (4T1-526) metastatic area was quantified by histology (15-16 mice/group) and the average metastatic burden is shown ⁇ SD.
  • FIG. 8C is a bar graph showing the experimental results of MDA-MB-231 following the schedule of Fig. 8A where mice were either treated by oral gavage with MG-002 (0.5 mg/kg), eFT226 (0.5 mg/kg), or vehicle control every three days until surgical resection.
  • Liver (MDA-MB- 231) metastatic area was quantified by histology (15-16 mice/group) and the average metastatic burden is shown ⁇ SD.
  • FIG. 8D is a schematic diagram outlining the experimental schedule to assess the ability of MG-002 to treat established lung metastases following lung colonization.
  • FIG. 8E is a bar graph showing the experimental results of the schedule of Fig. 8D where mice were either treated by oral gavage with MG-002 (0.5 mg/kg) or vehicle control every three days until the experimental endpoint.
  • the metastatic area in the lungs was quantified by histology and the average metastatic burden is presented ⁇ SD.
  • FIG. 9B is a bar graph showing the change in mouse weight of the mice of Fig. 9A.
  • FIG. 9C is a graph of tumour volume over time for the mice of Fig. 9A sacrificed four days after the start of drug treatment.
  • FIG. 9D is a bar graph showing the histology analysis results with Ki67 specific antibodies on the mice harvested in Fig. 9C.
  • FIG. 9E is a bar graph showing the histology analysis results with cleaved caspase-3 specific antibodies on the mice harvested in Fig. 9C.
  • FIG. 9F is a bar graph showing mice with established metastases that were randomized into four groups and treated as outlined in Fig. 9A.
  • Statistical analysis was performed with a Two-way ANOVA (Tukey’s multiple comparisons test).
  • PDX patient-derived xenograft
  • FIG. 11 is an image showing the three dimensional structure determined for the compound of formula lb.
  • FIG. 12 is a graph showing the activity of MG-002 cells towards wild type eHAP1 or elF4A1 F163L /elF4A2- cells.
  • FIG. 14 is a titration curve for MG-002 synthesized by Wuxi or Nuchem and for compound lb.
  • the elF4F complex contains the elF4E cap binding protein, the DEAD-box RNA helicase elF4A, and a large scaffolding protein, elF4G.
  • elF4F catalyzes the recruitment of 40S ribosomes (and associated initiation factors) to mRNA 5’ cap structures.
  • Mammalian cells encode three related elF4A proteins: (i) elF4A1 [DDX2A], (ii) elF4A2 [DDX2B], and (iii) elF4A3 [DDX48], elF4A1 and elF4A2 share 90% amino acid identity, can recycle through the elF4F complex, and are implicated in translation.
  • DDX48 is a component of the exon junction complex (EJC) and shares 66% amino acid identity with elF4A1.
  • elF4A1 is the more abundant and best studied family member.
  • elF4A in translation is directly proportional to cap-proximal secondary structure, which imparts unequal responses to different mRNAs following changes in elF4F levels/activity. For example, increased elF4F activity preferentially leads to the production of proteins regulating cell proliferation, survival, migration, and differentiation, as well as chemoresistance. This, in combination with high reliance of TNBC cells on elF4F activity, positions elF4F complex as a target to overcome intra-tumor heterogeneity.
  • the compound of formula I has been identified and characterized as an inhibitor targeting the elF4A helicase subunit of elF4F.
  • the compound for formula I has a plurality of possible stereoisomers.
  • the compound of formula I refers to a stereoisomeric mixture that contains two or more stereoisomers, where at least one of the stereoisomers is the compound of formula lb.
  • the enantiomer mixture has four stereoisomers and includes both the compounds of formula la and lb. In other embodiments, there is provided an enantiomeric mixture containing both formula la and lb.
  • a full racemate of the compound of formula I (which comprises formulas la and lb) as well as other possible stereoisomers.
  • the full racemate can refer to a mixture containing all possible stereoisomers.
  • a racemic mixture of compounds la and lb This racemic mixture is referred to herein as MG-002.
  • the present compounds can be formulated as salts or solvates thereof and can be prepared as crystals suitable for pharmaceutical use
  • the compound of formula I is derived from rocaglates.
  • Rocaglates are cyclopenta[b]benzofuran natural products produced by plants of the Aglaia genus that inhibit eukaryotic protein synthesis.
  • Rocaglates target the translation process by acting as molecular staples to induce clamping of elF4A (and elF4F) to polypurine-rich RNA sequences.
  • CR-1-31 B exhibits clear selectivity for blocking translation of mRNAs with polypurine-rich 5’ leader regions (over those with polypyrimidine rich 5’ leaders); whereas, the same does not hold true for silvestrol which inhibits both class of reporters similarly.
  • RocA has been shown to also induce clamping of DDX3X to poly (AG) RNA, albeit with significantly lower affinity compared to elF4A1 .
  • Silvestrol and RocA are well studied rocaglates. They have been very useful in unraveling the mechanism of action for this class of compounds, they demonstrate cytotoxic selectivity towards transformed cells, and this class of compounds have anti-tumor activity.
  • Introduction of a hydroxamate group at the C-2 position yielded a series of bioactive molecules with improved activity, among which CR-1-31 B is the best characterized.
  • the improved potency of CR-1 -31 B compared to RocA is likely related to the ability of the hydroxamate moiety to act as a hydrogen bond acceptor and/or as a bidentate chelating group, possibly stabilizing interactions with elF4A1 Q195 and D194.
  • a second modification introduced to the rocaglate core that improved activity was addition of a nitrile group on one of the ring, which can form hydrogen bonds with N167 of elF4A1.
  • the substitution of one of the aryl ring for a pyridine ring was found to lower lipophilicity of the core while maintaining n-stacking with RNA, leading to the development of a clinical candidate, eFT226 (Zotatifin).
  • the compound I of the present disclosure incorporates the advantages including having the pyridine ring, the hydroxamate moiety and the nitrile group to arrive at an improved inhibitory activity.
  • the cancer is characterized by cell proliferation dependent, driven or promoted by elF4A1 or elF4F.
  • the compound of formula lb is an inhibitor of elF4A1 and elF4F, and therefore limits cell proliferation and tumor growth.
  • the compound of formula lb or stereoisomeric mixtures comprising the same cause unscheduled and non-productive RNA clamping by the RNA helicase eukaryotic initiation factor (elF) 4A.
  • Compound lb or stereoisomeric mixtures comprising the same potently inhibit mRNA translation initiation by preventing ribosome recruitment to mRNAs. Moreover, compound lb or stereoisomeric mixtures comprising the same have a potent efficacy in limiting primary TNBC tumor growth. Even more importantly, considering that metastatic spread is a major cause of mortality in TNBC, compound lb or stereoisomeric mixtures comprising the same attenuate metastatic spread. Compound lb or stereoisomeric mixtures comprising the same showed a superior translation inhibition activity compared to the compounds of the prior art, is orally bioavailable, and shows single agent efficacy toward primary and metastatic TNBC pre-clinical models.
  • MYC positive cancers include lung cancer (small cell or non-small cell), leukemia (acute or chronic), breast cancer, myeloproliferative disorders (e.g.
  • myelodysplasia and multiple myeloma colorectal cancer, medulloblastoma, renal, hepatocellular cancer, melanoma, ovarian cancer, prostate cancer, esophageal adenocarcinoma, liposarcoma (pleomorphic or dedifferentiated), esophageal squamous cancer, gastrointestinal stromal tumor, glioma, myxofibrosarcoma, leiomyosarcoma, neuroblastoma, synovial sarcoma, mesothelioma, gastric cancer, thyroid cancer, lymphoma (e.g.
  • non-Hodgkin’s lymphoma lymphoma
  • osteosarcoma rhabdomyosarcoma
  • fibrosarcoma epithelial cancer (e.g. bladder, pancreatic, cervical, esophageal, endometrial, and head and neck), and neural cancer (e.g. meningioma, schwannoma, and retinoblastoma)
  • neural cancer e.g. meningioma, schwannoma, and retinoblastoma
  • the cancer is a breast cancer such as a triple negative breast cancer or is a metastatic cancer for example a cancer that metastasized into the lungs.
  • the present disclosure provides a method for the prevention, treatment and/or alleviation of a cancer comprising administering to a subject in need thereof a therapeutically effective amount of the compound of formula lb or an stereoisomeric mixture comprising the same.
  • the prevention of a metastatic cancer is particularly advantageous as metastasis is linked with poor prognosis.
  • the prevention is only contemplated for breast cancer types having a known genetic factor that increases the likelihood of developing cancer.
  • the BRCA1 and BRCA2 genes are known to have an impact on an individual’s chances of developing breast cancer.
  • the compound of the present disclosure can be used to prevent the cancer.
  • the prevention of cancer as contemplated herein refers to patients in remission that are receiving treatments to maintain remission, in the context of a relapse following a remission or a partial remission.
  • Administration is by any of the routes normally used for introducing an agent into ultimate contact with blood.
  • the agent described herein can be administered in any suitable manner, preferably with pharmaceutically acceptable carriers or excipients.
  • An advantage of the compound of formula lb or stereoisomeric mixtures comprising the same is that they have an excellent oral bioavailability which allows the non-invasive and most convenient form of administration.
  • pharmaceutically acceptable carrier “excipients”, “physiologically acceptable vehicle” and the like are to be understood as referring to an acceptable carrier that may be administered to a subject, together with the compound, and which does not impair the pharmacological activity thereof.
  • pharmaceutically acceptable carrier or “pharmaceutical carrier” are known in the art and include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or 0.9% saline.
  • pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, collating agents, inert gases and the like.
  • a pharmaceutical composition comprising a compound of formula lb and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be used in the prevention, alleviation or treatment methods described herein.
  • pharmaceutical composition means therapeutically effective amounts (dose) of the compound together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, and/or carriers.
  • a “therapeutically effective amount” as used herein in the context of the pharmaceutical composition refers to that amount which provides a therapeutic effect for a given condition and administration regimen.
  • compositions are liquids or lyophilized or otherwise dried formulations and include diluents of various buffer content (e.g., Tris-HCI, acetate, phosphate), pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, and detergents (e.g., Tween 20TM, Tween 80TM, Pluronic F68TM, bile acid salts).
  • buffer content e.g., Tris-HCI, acetate, phosphate
  • pH and ionic strength e.g., Tris-HCI, acetate, phosphate
  • additives such as albumin or gelatin to prevent absorption to surfaces
  • detergents e.g., Tween 20TM, Tween 80TM, Pluronic F68TM, bile acid salts.
  • the pharmaceutical composition can comprise pharmaceutically acceptable solubilizing agents (e.g., glycerol, polyethylene glycerol), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., thimerosal, benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol, complexation with metal ions, or incorporation of the material into or onto particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, hydrogels, etc, or onto liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts, or spheroplasts.
  • solubilizing agents e.g., glycerol, polyethylene glycerol
  • anti-oxidants e.g., as
  • Controlled or sustained release compositions include formulation in lipophilic depots (e.g., fatty acids, waxes, oils). Also contemplated by the present disclosure are particulate compositions coated with polymers (e.g., poloxamers or poloxamines).
  • the pharmaceutical composition further comprises or be administered in conjunction with one or more other therapeutic agents particularly in the case of metastatic cancer.
  • other therapeutic agents include but are not limited to doxorubicin, dexamethasone, and cisplatinum.
  • Suitable methods of administering the agent are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.
  • the preventive or therapeutic compounds of the present invention may be administered, either orally or parenterally, systemically or locally.
  • the compound of formula lb is administered orally for example in the form of a tablet ordroplets.
  • Other examples include, intravenous injection such as drip infusion, intramuscular injection, intraperitoneal injection, subcutaneous injection, suppositories, intestinal lavage, and the like.
  • excipient(s) or carrier(s) must be “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of the formulation and not being deleterious to the recipient thereof.
  • Standard accepted excipient(s) or carrier(s) are well known to skilled practitioners and described in numerous textbooks.
  • the amount of the compound described herein and used in accordance with the disclosure can be determined by the attending physician or pharmacist. It will be appreciated that the amount of a compound required will vary not only with the particular compound selected but also with the route of administration, the extent of the condition for which treatment is required (e.g. stage I, II, III or IV of cancer) and the age and condition of the patient. It will be understood that the scope of the method of treatment or uses described herein is not particularly limited, but includes in principle any therapeutically useful outcome including preventing, treating or slowing the progression of conditions defined herein.
  • Unlabelled and 5’-end fluorescein amidities (FAM) labelled poly (AG)s and (UC)s RNAs were obtained from Integrated DNA Technologies (IDT). MG-002 was custom synthesized by WuXi Chemistry Services (China) and NuChem (Sygnature Discovery - Canada). eFT226 was also purchased from GLPBIO (Montclair, CA) or was synthesized by WuXi. CR-1-31 B was purchased from MedChem Express.
  • eHAP1 cells were obtained from Horizon Discovery and maintained in Iscove’s Modified Dulbecco’s Medium (IMDM) supplemented with 10% fetal bovine serum, 1 % penicillin- streptomycin antibiotics (Pen-Strep), and 2 mM L-glutamine at 37 °C and 5% CO2. 293T/17 cells were grown in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% bovine growth serum, 1 % penicillin-streptomycin (Pen-Strep), and 2 mM L-glutamine. Tests for mycoplasma contamination were routinely performed.
  • DMEM Modified Eagle Medium
  • MDA-MB-231 cells were obtained from the American Type Culture Collection (ATCC) and were grown in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% bovine growth serum, 1 % Pen-Strep, and 2 mM L-glutamine.
  • IMR-90 cells were obtained from ATCC and were grown in Eagles Minimum Essential Medium (EMEM) supplemented with 10% bovine growth serum, 1 % Pen-Strep, and 2 mM L-glutamine.
  • MRC-5 were obtained from ATCC and were grown in Eagles Minimum Essential Medium (EMEM) supplemented with 10% bovine growth serum, 1 % Pen-Strep, and 2 mM L-glutamine.
  • HUVEC cells were obtained from Lonza and propagated in Endothelial Basal Medium (EBM) supplemented with Endothelial Supplemental Mix.
  • the compounds were synthesized by WuXi or NuChem. Many synthesis pathways are possible and the scheme below presents a non-limitative exemplary synthesis path. Specifically, the scheme below details the synthesis steps by WuXi to obtain a stereoisomeric mixture from which a racemate of the compounds of formula la and lb was extracted.
  • the term MG-002 refers to the racemate of the compounds of formula la and lb. MG-002 racemate was used in the experiments of the present example unless stated otherwise.
  • BL21 (DE3) codon + E. coli cells were transformed with pET15b-HiS6-elF4A1 , cultured at 37°C until the optical density (ODeoo) reached 0.6, at which point induction was undertaken by the addition of 1 mM (isopropyl-p-D-thiogalactopyranoside) IPTG at 37°C for 3 h.
  • Recombinant HiS6-elF4A1 was purified on a Ni(2 + )-NTA agarose column and the eluent applied to a Q- Sepharose fast flow matrix.
  • the protein was eluted with a linear salt gradient (100-500 mM KCI), dialyzed against Buffer A (20 mM Tris-CI [pH 7.5], 10% glycerol, 0.1 mM ethylenediaminetetraacetic acid (EDTA)) overnight at 4°C, and stored in aliquots at -80°C.
  • Buffer A 20 mM Tris-CI [pH 7.5], 10% glycerol, 0.1 mM ethylenediaminetetraacetic acid (EDTA)
  • elF4A1 protein was added to 10 nM FAM-labeled RNA in binding buffer (14.4 mM 4-(2-hydroxyethyl)-1 -piperazineethanesulfonic acid (HEPES) - NaOH [pH 8], 108 mM NaCI, 1 mM MgCI 2 , 14.4% glycerol, 0.1 % dimethyl sulfoxide (DMSO), 2 mM dithiothreitol (DTT)) and 1 mM adenosine triphosphate (ATP) in the presence or absence of indicated compound in black, low volume 384 well plates (Corning 3820).
  • binding buffer (14.4 mM 4-(2-hydroxyethyl)-1 -piperazineethanesulfonic acid (HEPES) - NaOH [pH 8], 108 mM NaCI, 1 mM MgCI 2 , 14.4% glycerol, 0.1 % dimethyl sulfoxide (DMSO),
  • DFS Differential scanning fluorimetry
  • elF4A1 8 pM of recombinant elF4A1 was incubated with compound (15 pM) or DMSO in DSF buffer (20 mM HEPES [pH 7.5], 70 mM KCI, 2 mM DTT, 1 mM Mg(OAc) 2 , 1 mM AMPPNP, 7.5X Sypro Orange (S-6650, Thermo Fisher), and 15 pM poly (AG)s RNA.
  • DSF buffer 20 mM HEPES [pH 7.5], 70 mM KCI, 2 mM DTT, 1 mM Mg(OAc) 2 , 1 mM AMPPNP, 7.5X Sypro Orange (S-6650, Thermo Fisher), and 15 pM poly (AG)s RNA.
  • the samples were heated and read from 37°C to 75°C using 1 °C/min ramp rate in the CFX96 TouchTM Real-Time PCR Detection System (BioRad).
  • Cell viability assays were performed using sulforhodamine B reagent (SRB). Cells (5000/well) were seeded into 96 well plates and treated with compound at the indicated concentrations. Cells were then washed with phosphate buffered saline (PBS) and fixed with 50% trichloroacetic acid (TCA) for 1 h at 4°C, rinsed with water and air-dried. Fixed cells were stained with 0.5% SRB/1 % acetic acid for 1 h, plates were washed with 1% acetic acid four times, and dried.
  • SRB sulforhodamine B reagent
  • the bound SRB was resuspended in 100 pl 20mM Tris base (pH adjusted to 9) and the OD550nm was measured using a SpectraMaxTM M5 microplate reader (Molecular Devices). Relative viability was calculated by normalizing to the DMSO control.
  • Radioimmunoprecipitation assay buffer (RIPA) buffer (20 mM Tris-HCI [pH 7.6], 100 mM NaCI, 1 mM EDTA,1 mM ethylene glycol- bis(p-aminoethyl ether (EGTA), 1% Thermo Scientific Surfact-Amps NP40, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate (SDS), 20 mM B-glycerophosphate, 10 mM NaF, 1 mM phenylmethylsulfonyl fluoride (PMSF), 4 pg/mL aprotinin, 2 pg/mL leupeptin, 2 pg/mL pepstatin).
  • RIPA radioimmunoprecipitation assay buffer
  • MDA-MB231 cells were seeded in 12-well plates in DMEM supplemented with 10% FBS and antibiotics. Twenty-four hours later, cells were treated with 2.5 mM thymidine dissolved in media to synchronize them in S phase, as described in Whitfield ML, Sherlock G, Saldanha AJ, Murray JI, Ball CA, Alexander KE, et al. Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol Biol Cell. 2002;13(6):1977-2000. Twenty hours later, cells were washed twice with warm PBS and released into fresh media that contained either DMSO or 10 nM of CR-1-31-B, MG-002, or eFT226.
  • cells were placed in an incubation chamber of a Zeiss Axiovert 200M microscope to maintain temperature and CO2 levels. Bright-field imaging was performed. Images were taken every 10 minutes at 10x total magnification for a total of 40 hours. For each condition, 50 cells were followed from the time of starting the experiment to the time they entered mitosis (minutes to mitotic entry), which roughly represent the time they spent in late S and G2 phase of the cell cycle. A one-way ANOVA with Tukey multiple comparisons test was used to compare the average minutes to mitotic entry for each treatment to the vehicle control.
  • Lentivirus harboring cDNAs to elF4A1 (TRCN0000491404) and elF4A3 (TRCN0000471580) were obtained from the Genetic Perturbation Service (GPS) of the Goodman Cancer Research Center and McGill Biochemistry Dept.
  • the elF4A1 cDNA was cloned into pLX-TRC317 and DDX3X was cloned into pPRIME-CMV-GFP-PGK-Puro-recipient.
  • Virus was produced in 293T cells and used to infect parental or elF4A1 F163L /elF4A2- eHAP1 cells. Following puromycin selection (2 pg/pL for 2 days), cells were expanded for experiments.
  • Triton X-100 and sodium deoxycholate were added to the supernatant to a final concentration of 0.5%.
  • Samples were loaded on 10-50% sucrose gradients and centrifuged in an SW40 rotor at 35,000 rpm for 2 h. Gradients were collected using a Brandel tube piercer and delivering 60% sucrose through the bottom of the centrifuge tube. Recording of the data was performed using InstaCalTM Version 5.70 and TracerDaqTM Version 1 .9.0.0 (Measurement Computing Corporation, Norton, MA).
  • mice Female mice (8-10 weeks old) were purchased from Charles River Laboratories (Quebec, Canada). 4T1-526 (50,000 cells) or MDA-MB-231 (1x10 6 cells) were injected into the mammary fat pads of Balb/c or SCID-Beige mice, respectively as described. For the experimental metastasis assays, 1x10 5 4T1-526 cells were injected into the tail vein of animals. All mice had ad libitum access to food and water and were housed within the animal facilities of the Lady Davis Institute, on a 12h light day cycle, mean temperature 22.5 C ⁇ 1 .5 °C.
  • MG-002 and eFT226 were injected either intraperitoneally, intravenously or by oral gavage (0.5 mg/kg every three days).
  • Urothelial carcinoma patient-derived xenograft (PDX) JP- PDX01 were established and viably preserved at McGill University, were cut into pieces and then inserted into a pocket in the subcutaneous space of NSG mice. Mice were randomized to treatment arms once the average tumor volume reached at least 100 mm 3 .
  • MG-002 was resuspended in carrier (PBS) and administered by PO at 0.0625 mg/kg (every three days). Tumor volume and mouse weight were measured twice to thrice weekly. Tumor volume was calculated as length*(width) 2 *0.52.
  • Animal studies were approved by the Animal Resource Centre at McGill University in accordance with guidelines from the Canadian Council of Animal Care.
  • MG-002 induces elF4A1'RNA clamping to potently inhibit translation
  • RNA clamping activity of rocaglates can be conveniently measured using the fluorescence polarization (FP) assay. It was found that like CR-1-31 B, both eFT226 and MG-002 discriminated between poly (AG)s and poly (UC)8 RNA templates (Fig. 1A). As previously noted, the unrelated natural product, pateamine A (PatA), stabilized binding of elF4A1 to both polypurine and polypyrimidine RNA templates. Relative to elF4A, the tested rocaglates only minimally influenced DDX3X:RNA clamping (Fig. 1 B).
  • FP fluorescence polarization
  • CR-1 -31 B and MG-002 were similarly effective at inhibiting cap-dependent translation from a reporter harboring a 37% (AG)-rich 5’ leader region (Fig. 1 E).
  • eFT226 was the least active of the three tested compounds (Fig. 1 E).
  • MG-002 like CR- 1 -31 B, also preferentially inhibited translation of an mRNA reporter enriched for (AG)-repeats, relative to a reporter with a (UC)-rich 5’ leader (Figs. 1 F-1 H).
  • HCV hepatits C virus
  • IRS internal ribosome entry site
  • MG-002 stabilizes elF4A1-RNA complexes, preferentially inhibits mRNAs with polypurine leader regions, and is a potent inhibitor of translation in vitro and in cellula.
  • MG-002 The anti-tumorigenic properties of MG-002 towards TNBCs were assessed. Exposure of MDA-MB-231 cells to MG-002 or CR-1 -31 B caused a profound delay in G2/M progression (Fig 2A), consistent with the previously reported activity of rocaglates against other cell lines. The consequences of prolonged exposure (2 days) to MG-002 was then assessed against two TNBC lines (4T1 , MDA-MB-231) and a model of ER+/HER2+ breast cancer (BT-474) to evaluate the generalizability of our findings (Figs. 2B-2D).
  • EC50 half maximal effective concentration
  • MG-002 cytotoxicity is elF4A1 and elF4A2 dependent
  • Rocaglates primarily exert their biological effects through elF4A1 engagement. However, RocA can induce RNA clamping of DDX3X, although the binding is at least 30-fold lower than for elF4A1.
  • HAP1 cells expressing either wild-type elF4A or an elF4A1 F163L mutant were tested to determine which is unable to bind the rocaglate compound. It was found that elF4A1 F163L /elF4A2 _ Hap1 cells were significantly more resistant to MG-002 when compared to those expressing the wild-type protein (Fig.
  • elF4A1 F163L /elF4A2 _ cells to ectopically express wild-type elF4A1 , elF4A2, elF4A3, or DDX3X; which should re-sensitize them to MG-002 induced cytotoxic activity due to compound- induced clamping of wild-type proteins (Figs. 3B-3D).
  • elF4A1 F163L /elF4A2 _ cells ectopically expressing elF4A1 or elF4A2 indeed showed increased sensitivity to MG-002 (Figs. 3E and 3F).
  • Sensitization to MG-002 was not observed with parental eHAP1 cells ectopically expressing elF4A1 or elF4A2 (Fig. 3G), suggesting that elF4A1 is not limiting for target engagement in these cells. Sensitization was also not observed in elF4A1 F163L /elF4A2 _ cells ectopically expressing DDX3X (Fig. 3F) or elF4A3 (Figs. 3H and 3I).
  • MG-002 shows similar mouse and human plasma protein binding as eFT226 (Table
  • MG-002 was also found to be stable in simulated gastric fluid (pH 1.2) (Table 1), prompting the investigation of its plasma concentration following drug delivery by oral gavage (PO). Surprisingly, it was found that PO delivery resulted in sustained presence of MG-002 in the plasma, with a terminal half-life of ⁇ 10 h (Fig. 4A, Table 1). The consequences of PO delivery on translation in vivo was assessed by analyzing liver polysomes following compound delivery by gavage (Figs. 4B-4D). The results indicate strong suppression of translation in the liver following PO delivery of MG-002 as evidenced by >3 fold decrease in polysome-to-monosome ratio 7h post-delivery, followed by partial translational recovery 24h post-treatment.
  • MG-002 distribution to plasma, spleen, lungs, and liver was also quite extensive 4h post-delivery PO (Fig. 4E).
  • MG- 002 administration was also well tolerated, with no observable impact on blood cells (Figs. 4F- 4K), organ weights (Fig. 4L), total body weights. Histopathological analysis of heart, lung, spleen, kidney, and bone marrow following six treatments with MG-002 did not uncover any overt tissue damage (Table 2). Based on these encouraging results, it was decided to undertake studies to assess the anti-cancer activity of MG-002 in vivo towards TNBC.
  • Table 1 Plasma protein binding, acid stability, and plasma half-life for MG-002 a Cmpd (50 pM) was incubated in Simulated Gastric Fluid (SGF) containing pepsin at pH 1 .2 for 1 h at 37°C.
  • SGF Simulated Gastric Fluid
  • n 3 ⁇ standard deviation (SD).
  • c Delivered IV 0.5 mg/kg. n 3 ⁇ SD.
  • MG-002 inhibits tumor cell growth in vivo following PO delivery
  • MG-002 The ability of MG-002 to accumulate in lung tissue following PO delivery (Fig. 4E), a frequent site of breast cancer metastasis, prompted us to assess the efficacy of MG-002 towards metastatic lesions. It was first assessed whether neo-adjuvant MG-002 treatment could prevent the formation of spontaneous lung and liver metastases (Figs. 8A-8C). 4T1-526 or MDA-MB-231 tumor bearing mice were treated with MG-002, eFT226 or vehicle control (0.5 mg/kg every 3 days) and primary tumors were subsequently surgically resected at a volume of 500 mm 3 .
  • MG-002 was effective as a monotherapy at reducing the formation of the murine spontaneous 4T1 -526 lung metastases (Fig. 8B). Furthermore, in the human metastatic MDA-MB-231 TNBC xenograft model, MG-002, but not eFT226, significantly decreased the formation of spontaneous liver metastases (Fig. 8C). Next, the ability of MG-002 to shrink breast cancer metastases that had already colonized the lungs was assessed (Fig. 8D). 4T1-526 breast cancer cells were injected into the tail vein to directly seed the lung.
  • MG-002 is associated with reduced lung metastatic burden, compared to vehicle control treated mice, although these differences were not statistically significant (Fig. 8E).
  • MG-002 is likely not sufficient as a monotherapy to treat individuals who present with distant metastases but metastatic cancer are considerably harder to treat and will require are known to require a multi-faced approach including more than one treatment agent.
  • the present data suggest that MG-002 monotherapy demonstrates anti-metastatic properties, primarily in preventing the likelihood that a patient will progress to metastatic disease.
  • MG-002 was also tested as a monotherapy to target urothelial carcinoma.
  • the consequences of prolonged exposure (2 days) to MG-002 was first assessed against one primary urothelial carcinoma cell line established from a metastatic tumor and cell viability was compromised with half maximal effective concentration (EC50) value for MG- 002 ⁇ 10 nM, indicating potent activity similar to other cancer cells tested (Fig. 2, 3).
  • EC50 half maximal effective concentration
  • MG-002/doxorubicin combination therapy reduces primary and metastatic disease burden
  • MG-002 Aggressive breast cancers are likely to require combination therapies to exploit the therapeutic benefit of MG-002. This was assessed by combining MG-002 with doxorubicin (Dox); a front-line chemotherapy that is already standard-of-care for TNBC. It was therefore investigated whether a combination of MG-002/Dox might be more effective than either single compound as monotherapy. Following mammary fat pad injection of 4T1-526, either the monotherapy or the combination treatment was started when tumors were ⁇ 100 mm 3 . Tumors were equally responsive to Dox or MG-002, but significantly more sensitive to combination treatment (Fig. 9A). Combination treatment did not lead to significant weight loss in treated animals, although animals failed to gain weight during treatment (Fig. 9B).
  • Dox doxorubicin
  • breast tumors were harvested early during their treatment schedule, at a timepoint when reduced tumor volumes was first observed with MG-002 as a monotherapy, and an even further reduction with MG-002/Dox combination treatment (Fig. 9C).
  • Immunohistochemistry (IHC) analysis at these time points revealed no significant differences in the rates of cancer cell proliferation, with either MG-002 or doxorubicin single or combination treatments (Fig. 9D).
  • MG-002 treatment increased the rate of apoptosis in breast tumors, which was even more pronounced following MG-002/Dox treatment (Fig. 9E).
  • Cancer cells universally activate signal transduction pathways that upregulate the activity of protein synthesis machinery and/or perturb the expression of genes encoding the components of the translational apparatus to reprogram and augment translation. As a result, pharmacological inhibition of mRNA translation initiation has great therapeutic potential.
  • mTOR inhibitors which function upstream to prevent elF4F complex formation.
  • HR hormone receptor
  • This phase III randomized study that compared the effects of everolimus and exemestane (an aromatase inhibitor) with exemestane alone in women with advance HR+ disease.
  • MG-002 also significantly delayed the growth of primary TNBC tumors and impaired the formation of spontaneous lung metastases. Furthermore, MG-002 inhibited urothelial carcinoma cell proliferation in vitro and as a monotherapy, at a reduced dosage, strongly suppressed the growth of primary urothelial carcinoma in vivo. Moreover, MG-002 is likely to exert cytotoxic effects in other malignancies given promising pre-clinical studies showing that other elF4A inhibitors elicit strong anti-neoplastic effects in models of leukemia and pancreatic cancer.
  • MYC expression is quite sensitive to MG-002 treatment. Since MYC protein has a short half-life, levels are rapidly depleted from cells when exposed to MG-002. MYC is a well-known cancer driver, promoting tumorigenesis through a variety of mechanisms including stimulating cell proliferation, altering metabolism, inhibiting cell death, and impacting the tumor microenvironment. Genetic modelling of both the therapeutic impact and side effects of systemic MYC suppression in the mouse has shown profound anticancer activity, as well as the tolerability of such an approach. Indeed, exogenous administration of a MYC dominant-negative (Omomyc) is efficacious against TNBC and displays antimetastatic properties. However, targeting MYC for cancer treatment had proven quite challenging. The sensitivity of c-MYC translation to MG-002 offers a powerful approach by which to rapidly block MYC expression.
  • MG-002 functions as a molecular staple and clamps elF4A onto RNA with poly(AG)-rich tracts.
  • Cells encode two elF4A homologs, elF4A1 and elF4A2, both of which are targets for rocaglates.
  • elF4A1 is the more abundant isoform (58, 59) and overall levels of elF4A1 and elF4A2 in a tumor cell may contribute to MG-002 sensitivity.
  • eFT226 Clear differences in activity between MG-002 and eFT226 were noted.
  • the eFT226 compound is not as potent as MG-002 at inhibiting translation when compared to MG-002 (Figs. 1 B, and 1 E-1 G).
  • eFT226 is slightly better at clamping elF4A to poly(AG) RNA sequences (Fig. 1 A)
  • the data shows that the complexes formed are not as long lived and show a half-life that is approximately 2.5-fold less than those formed in the presence MG-002 (Fig. 1 C). This latter point may explain the lower potency at inhibiting protein synthesis on the part of eFT226.
  • M-002 was also more effective than eFT226 towards TNBCs in general in vitro (Figs. 2A-2H). Lastly, when delivered by gavage, eFT226 was less potent that MG-002. In particular as shown in Fig. 2A, MG-002 outperformed CR-1-31 B and eFT226 when evaluating the effect on cell division and time to mitotic entry. [0131] Identified in a yeast-based screen for rocaglate-resistant alleles, there are missense mutations in six amino acids that could confer resistance to two rocaglates. These are: T158, P159, F163, F192, Q195, and 1199 (human amino acid numbering used).
  • elF4A inhibition increases the radiosensitivity of tumors and also improves the response to targeted therapies, including MEK and CDK4/6 inhibitors.
  • elF4A inhibitors, and MG-002 specifically, increase responsiveness to standard-of-care chemotherapies, including doxorubicin.
  • MG- 002 is an orally available rocaglate capable of reducing primary tumor growth and blocking metastatic seeding.
  • Example 1 demonstrated the activity of MG-002 which is a racemic mixture of the compounds of formula la and lb.
  • Example 1 also demonstrated the activity of the compound of formula lb alone (Fig. 10A).
  • the specific structure of the compounds and enantiomeric mixtures were investigated to confirm chirality, particularly for the active compound lb.
  • the compound of formula lb crystallized from DMF/H2O, was collected from a shock- cooled single crystal at 150 K on a BrukerTM Venture Metaljet K-geometry diffractometer with a Metal Jet using a HeliosTM MX Mirror Optics as monochromator and a BrukerTM CMOS Photon III detector.
  • the structure was solved by intrinsic phasing methods with XT and refined by full-matrix least-squares methods against F 2 using XL within the graphical user interface of OLEX2. All non-hydrogen atoms were refined with anisotropic displacement parameters. The hydrogen atoms were refined isotropically on calculated positions using a riding model with their U ISO values constrained to 1 .5 times the L/ eq of their pivot atoms for terminal sp 3 carbon atoms and 1 .2 times for all other carbon atoms.
  • the Crystallographic Information Framework (CIF) file was generated using FinalCifTM. The three dimensional structure identified is shown in Fig. 11 . Ellipsoids were drawn at the 50% probability level and hydrogen atoms are shown as sphere of arbitrary size.
  • a FLuc and Rluc luciferase assay was performed as described in Example 1 a full racemate of the compound of formula I containing four stereoisomers, two of which were compounds la and lb and two had an unconfirmed structure (Fig. 13). Firefly (FF) and Renilla (Ren) luciferase reporter genes were used to determine expression. As can be seen from Fig. 13, the full racemate was active.
  • IC50 Half maximal inhibitory concentrations were determined using a non-linear regression model as explained in Example 1.
  • the titration graph is shown in Fig. 14 and was produced following the experimental protocol detailed in Example 1 .
  • the IC50 for the MG-002 racemate as obtained by Wuxi was 8.8 nM
  • for the MG-002 racemate as obtained by NuChem it was 11.8 nM

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne un composé de formule I, un sel ou un solvate de celui-ci, pour le traitement de cancers positifs pour MYC notamment le cancer du poumon, la leucémie, le cancer du sein, les troubles myéloprolifératifs, le cancer colorectal, le médulloblastome, le cancer du rein, le cancer hépatocellulaire, le mélanome, le cancer de l'ovaire, le cancer de la prostate, l'adénocarcinome œsophagien, le liposarcome, le cancer de l'œsophage squameux, la tumeur stromale gastro-intestinale, le gliome, le myxofibrosarcome, le léiomyosarcome, le neuroblastome, le sarcome synovial, le mésothéliome, le cancer gastrique, le cancer de la thyroïde, le lymphome, l'ostéosarcome, le rhabdomyosarcome, le fibrosarcome, le cancer épithélial et le cancer neuronal.
PCT/CA2024/051074 2023-08-18 2024-08-16 Inhibiteur d'hélicase à arn Pending WO2025039073A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363520384P 2023-08-18 2023-08-18
US63/520,384 2023-08-18

Publications (1)

Publication Number Publication Date
WO2025039073A1 true WO2025039073A1 (fr) 2025-02-27

Family

ID=94731164

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2024/051074 Pending WO2025039073A1 (fr) 2023-08-18 2024-08-16 Inhibiteur d'hélicase à arn

Country Status (1)

Country Link
WO (1) WO2025039073A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011140334A2 (fr) * 2010-05-06 2011-11-10 Trustees Of Boston University Composés, méthodes de fabrication ou d'identification de composés et leurs utilisations
WO2017091585A1 (fr) * 2015-11-25 2017-06-01 Effector Therapeutics, Inc. Composés inhibiteurs d'eif4-a et procédés associés
WO2019161345A1 (fr) * 2018-02-19 2019-08-22 Memorial Sloan-Kettering Cancer Center Agents et méthodes pour traiter des maladies dysprolifératives
WO2022089514A1 (fr) * 2020-10-28 2022-05-05 上海翰森生物医药科技有限公司 Biorégulateur polycyclique, procédé de préparation s'y rapportant et son application

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011140334A2 (fr) * 2010-05-06 2011-11-10 Trustees Of Boston University Composés, méthodes de fabrication ou d'identification de composés et leurs utilisations
WO2017091585A1 (fr) * 2015-11-25 2017-06-01 Effector Therapeutics, Inc. Composés inhibiteurs d'eif4-a et procédés associés
WO2019161345A1 (fr) * 2018-02-19 2019-08-22 Memorial Sloan-Kettering Cancer Center Agents et méthodes pour traiter des maladies dysprolifératives
WO2022089514A1 (fr) * 2020-10-28 2022-05-05 上海翰森生物医药科技有限公司 Biorégulateur polycyclique, procédé de préparation s'y rapportant et son application

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CENCIC REGINA, YOUNG K IM, SAI KIRAN NAINENI, MOHAMED MOUSTAFA-KAMAL, PREDRAG JOVANOVIC, VALERIE SABOURIN, MATTHEW G ANNIS, FRANCI: "A second-generation elF4A RNA helicase inhibitor exploits translational reprogramming as a vulnerability in triple-negative breast cancer", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 121, no. 4, 23 January 2024 (2024-01-23), pages e2318093121, XP093283505, DOI: 10.1073/pnas.2318093121 *
CHEN MINGMING; ASANUMA MIWAKO; TAKAHASHI MARI; SHICHINO YUICHI; MITO MARI; FUJIWARA KOICHI; SAITO HIRONORI; FLOOR STEPHEN N.; INGO: "Dual targeting of DDX3 and eIF4A by the translation inhibitor rocaglamide A", CELL CHEMICAL BIOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 28, no. 4, 8 December 2020 (2020-12-08), AMSTERDAM, NL , pages 475, XP086538909, ISSN: 2451-9456, DOI: 10.1016/j.chembiol.2020.11.008 *
DHANASEKARAN R ET AL.: "The MYC oncogene -the grand orchestrator of cancer growth and immune evasion", NATURE REVIEWS CLINICAL ONCOLOGY., vol. 19, no. 1, January 2022 (2022-01-01), pages 23 - 36, XP037645786, DOI: 10.1038/s41571-021-00549-2 *
ERNST JT ET AL.: "Design of development candidate eFT226, a first in class inhibitor of eukaryotic initiation factor 4A RNA helicase", JOURNAL OF MEDICINAL CHEMISTRY, vol. 63, no. 11, 29 May 2020 (2020-05-29), pages 5879 - 955, XP055890843, DOI: 10.1021/acs.jmedchem.0c00182 *
WOLFE AL ET AL.: "RNA G-quadruplexes cause eIF4A-dependent oncogene translation in cancer", NATURE, vol. 513, no. 7516, 4 September 2014 (2014-09-04), pages 65 - 70, XP055427834, DOI: 10.1038/ nature 13485 *

Similar Documents

Publication Publication Date Title
Hao et al. Lenvatinib in management of solid tumors
Yu et al. TRIB3-EGFR interaction promotes lung cancer progression and defines a therapeutic target
Zheng et al. A novel STAT3 inhibitor W2014-S regresses human non-small cell lung cancer xenografts and sensitizes EGFR-TKI acquired resistance
Xie et al. Preclinical characterization of anlotinib, a highly potent and selective vascular endothelial growth factor receptor‐2 inhibitor
US20240082218A1 (en) Pharmaceutical combinations comprising a kras g12c inhibitor and uses of a kras g12c inhibitor for the treatment of cancers
McInnes et al. PLK1 as an oncology target: current status and future potential
Zheng et al. Nutlin-3 overcomes arsenic trioxide resistance and tumor metastasis mediated by mutant p53 in Hepatocellular Carcinoma
Dei et al. Recent advances in the search of BCRP-and dual P-gp/BCRP-based multidrug resistance modulators
Singh et al. Single agent and synergistic activity of the “first-in-class” dual PI3K/BRD4 inhibitor SF1126 with sorafenib in hepatocellular carcinoma
Mahajan et al. Tumor-specific delivery of 5-fluorouracil–incorporated epidermal growth factor receptor–targeted aptamers as an efficient treatment in pancreatic ductal adenocarcinoma models
Zhang et al. Personalized clinical trials in hepatocellular carcinoma based on biomarker selection
Marcucci et al. Anti-cancer stem-like cell compounds in clinical development–an overview and critical appraisal
Yao et al. Discovery of a novel EGFR ligand DPBA that degrades EGFR and suppresses EGFR-positive NSCLC growth
JP2022506299A (ja) インテグリンリガンドを有する新規な細胞増殖抑制コンジュゲート
JP2021532159A (ja) (s)−5−アミノ−3−(4−((5−フルオロ−2−メトキシベンズアミド)メチル)フェニル)−1−(1,1,1−トリフルオロプロパン−2−イル)−1h−ピラゾール−4−カルボキサミドの噴霧乾燥分散体および製剤
Jing et al. Aurora kinase inhibitors: a patent review (2014-2020)
US20220280520A1 (en) Method of treating a methionine-dependent cancer
A Ucar et al. Inhibiting the interaction of cMET and IGF-1R with FAK effectively reduces growth of pancreatic cancer cells in vitro and in vivo
CN103298481A (zh) 用于治疗卵巢癌的催乳素受体拮抗剂和化疗药物的用途
JP7132848B2 (ja) 癌の治療のための合理的併用療法
Burns et al. CYT997: a novel orally active tubulin polymerization inhibitor with potent cytotoxic and vascular disrupting activity in vitro and in vivo
Geller et al. A phase 1 study of the c‐Met inhibitor, tivantinib (ARQ197) in children with relapsed or refractory solid tumors: A Children's Oncology Group study phase 1 and pilot consortium trial (ADVL1111)
WO2025039073A1 (fr) Inhibiteur d'hélicase à arn
BR112021004527A2 (pt) kits e métodos para tratamento contra câncer
WO2021208787A1 (fr) Polypeptide cible utilisé pour le criblage de médicaments et procédé de criblage associé

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: 24855171

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)