[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

WO2018005444A2 - Methods for treating cancer - Google Patents

Methods for treating cancer Download PDF

Info

Publication number
WO2018005444A2
WO2018005444A2 PCT/US2017/039430 US2017039430W WO2018005444A2 WO 2018005444 A2 WO2018005444 A2 WO 2018005444A2 US 2017039430 W US2017039430 W US 2017039430W WO 2018005444 A2 WO2018005444 A2 WO 2018005444A2
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
foregoing
compound
derivatives
pharmaceutically acceptable
Prior art date
Application number
PCT/US2017/039430
Other languages
French (fr)
Other versions
WO2018005444A3 (en
Inventor
Chiang Jia Li
Matthew HITRON
Youzhi Li
Wei Li
Waldo ORTUZAR
Original Assignee
Boston Biomedical, Inc.
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 Boston Biomedical, Inc. filed Critical Boston Biomedical, Inc.
Priority to JP2018568329A priority Critical patent/JP2019519573A/en
Priority to CA3029596A priority patent/CA3029596A1/en
Publication of WO2018005444A2 publication Critical patent/WO2018005444A2/en
Publication of WO2018005444A3 publication Critical patent/WO2018005444A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • 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
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • 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

Definitions

  • Disclosed herein are methods for treating cancer in a subject comprising administering a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors can be, for example, at least one compound chosen from the compounds having Formula A:
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors can be, for example, at least one compound chosen from the compounds having Formula B:
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one second compound chosen from kinase inhibitors can be, for example, at least one compound chosen from the compounds having Formula C:
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • chemotherapeutic agents cause cytotoxicity to both healthy noncancerous as well as cancerous cells.
  • the therapeutic index of these chemotherapeutic compounds i.e., a measure of the therapy’s ability to distinguish between cancerous and normal cells
  • a dose of a chemotherapy drug that is effective at killing cancer cells will also kill normal cells, especially those normal cells (such as epithelial cells and cells of the bone marrow) that undergo frequent cell division.
  • side effects such as hair loss, suppression of hematopoiesis causing anemia and immunodeficiency, and nausea often occur.
  • HCC Advanced hepatocarcinoma
  • the Sorafenib Hepatocellular Carcinoma Assessment Protocol (SHARP) trial remains the only randomized controlled trial of a systemic chemotherapeutic agent to demonstrate a statistically significant survival benefit in patients with advanced, unresectable HCC.
  • 602 patients were randomly assigned to receive sorafenib (400 mg twice daily) or placebo.
  • a median Overall Survival (OS) of 10.7 months was observed in the treatment arm compared to 7.9 months for placebo.
  • Time to Tumor Progression (TTP) for the sorafenib and placebo arms was 5.5 months and 2.8 months, respectively.
  • sorafenib monotherapy The benefits achieved with sorafenib monotherapy are important, but modest in magnitude. Many patients do not experience disease control, and disease control can be short-lived in those that do achieve it with therapy. Unfortunately, no other agent has proved to be more effective. Recent Phase III trials with other regimens (e.g., FOLFOX, brivanib, sunitinib and linifanib) have all failed to demonstrate a statistically significant improvement in OS when compared to treatment with sorafenib alone.
  • CSCs Cancer stem cells
  • stemness-high cancer cells cancer stem cells
  • CSCs are believed to possess at least the following four characteristics:
  • stemness means the capacity for a stem cell population to self-renew and transform into cancer cells (Gupta PB et al., Nat. Med.2009; 15(9):1010-1012). While CSCs form only a small percentage of the total cancer cell population in a tumor (Clarke MF, Biol. Blood Marrow Transplant. 2009; 11(2 suppl. 2):14-16), they give rise to heterogeneous lineages of differentiated cancer cells that make up the bulk of the tumor (see Gupta et al.2009). In addition, CSCs possess the capacity to spread to other sites in the body by metastasis where they seed the growth of the new tumors (Jordan CT et al. N. Engl. J. Med.2006; 355(12):1253-1261).
  • CSC stemness can also be associated with dysregulation of signaling pathways, which may contribute to their ability to metastasize.
  • stemness signaling pathways are tightly controlled and genetically intact.
  • the aberrant regulation of stemness signaling pathways in CSCs plays a key role in the uncontrolled self-renewal of these cells and their transformation into cancer cells (see Ajani et al. Semin. Oncol. (2015) 42 Suppl. 1:S3-17).
  • Dysregulation of stemness signaling pathways also contributes to CSC resistance to chemotherapy and radiotherapy and to cancer recurrence and metastasis.
  • Exemplary stemness signaling pathways involved in the induction and maintenance of stemness properties in CSCs include, but are not limited to, Janus kinase/ signal transducers and activators of transcription (JAK/STAT), Hedgehog (Desert (DHH), Indian (IHH), and Sonic (SHH))/PATCHED/(PTCH1)/ SMOOTHENED (SMO), NOTCH/DELTA-LIKE (DLL1, DLL3, DLL4)/JAGGED (JAG1, JAG2)/ CSL (CBF1/Su(H)/Lag- 1), WNT/APC/GSK3/ ⁇ -CATENIN/TCF4 and NANOG (Boman BM et al., J. Clin. Oncol.2008; 26(17):2828-2838).
  • Chemotherapy and radiation kills the majority of rapidly dividing cancer cells in a tumor but not CSCs that survive by acquiring resistance (see Jordan et al. 2006). Radiation / chemotherapy-resistant CSCs may also acquire the ability to metastasize to different sites in the body and maintain stemness at these locations through interactions with the microenvironment, thereby allowing for the spread of metastatic tumor growth (see Boman et al.2008). Interestingly, this enhanced tumorigenicity of CSCs correlates with the expression of genes normally expressed in adult stem cells, such as cell surface markers like CD44, CD133, and CD166.
  • anti-cancer therapies that specifically target CSC’s aberrant signaling pathways may help prevent tumor metastasis and provide a viable treatment option for patients with recurrent disease that is no longer treatable using traditional therapies. Such an approach may therefore improve the survival and quality of life of cancer patients, especially those patients suffering from metastatic disease. Unlocking this untapped potential involves the identification and validation of pathways that are essential for CSC self-renewal and survival. While many of the signaling pathways regulating embryonic or adult stem cell proliferation and differentiation are known, it remains to be seen if these same pathways are required for cancer stem cell self-renewal and survival.
  • the transcription factor Signal Transducer and Activator of Transcription 3 (also known as Acute-Phase Response Factor, APRF, DNA-Binding Protein APRF, ADMIO 3, HIES; referred to herein as STAT3) is a member of a family of seven transcription factors, STAT1 to STAT6, including STAT5a and STAT5b.
  • STATs are activated either by receptor associated tyrosine kinases like Janus kinases (JAKs) or by receptors with intrinsic tyrosine kinase activity such as PDGFR, EGFR, FLT3, EGFR, ABL, KDR, c-MET, or HER2.
  • the phosphorylated STAT protein (“pSTAT”) dimerizes, as a homo- or heterodimer, and translocates from the cytoplasm to the nucleus, where it binds to specific DNA-response elements in the promoters of target genes and induces gene expression.
  • STAT 2, 4, & 6 regulate primarily immune responses, while STAT3, along with STAT1 and STAT5, regulate the expression of genes controlling cell cycle (CYCLIN D1, D2, and c-MYC), cell survival (BCL-XL, BCL-2, MCL-1), and angiogenesis (HIF1 ⁇ , VEGF) (Furqan et al. Journal of Hematology & Oncology (2013) 6:90).
  • STAT3 activation is transient and tightly regulated, lasting for example, from about 30 minutes to several hours.
  • STAT3 is found to be aberrantly active.
  • Persistently active STAT3 is present in more than half of all breast and lung cancers as well as colorectal cancer (CRC), ovarian cancer, hepatocellular carcinoma, multiple myeloma and in more than 95% of all head/neck cancers.
  • STAT3 therefore seems to play a pivotal role in cancer progression and may be one of the principal mechanisms by which cancer cells acquire drug resistance.
  • STAT3 is a potent transcription regulator that targets genes involved in cell cycle, cell survival, oncogenesis, tumor invasion, and metastasis, including, but limited to, BCL-XL, c-MYC, CYCLIN D1, VEGF, MMP-2, and SURVIVIN.
  • STAT3 is also a key negative regulator of tumor immune surveillance and immune cell recruitment.
  • STAT3 may enable the survival and self- renewal capacity of CSCs across a broad spectrum of cancers.
  • the at least one compound chosen from compounds having Formula A can be, for example, an inhibitor of CSC growth and survival.
  • U.S. Patent No. 8,877,803 describes a compound having Formula A that inhibits STAT3 pathway activity with a cellular IC50 of ⁇ 0.25 ⁇ M.
  • Example 13 in the‘803 patent provides exemplary methods of synthesizing at least one compound having Formula A.
  • the at least one compound chosen from compounds having Formula A can be used, for example, in a method for treating cancers.
  • Example 6 of PCT Patent Application No. PCT/US2014/033566 the at least one compound chosen from compounds having Formula A was chosen to enter a clinical trial for patients with advanced cancers.
  • the disclosures of U.S. Patent No. 8,877,803 and PCT Patent Application No. PCT/US2014/033566 are incorporated herein by reference in their entireties.
  • Protein kinases are a family of enzymes that regulate a wide variety of cellular processes, including cell growth, cell proliferation, cell differentiation, and metabolism. Protein kinases communicate cell growth signals through the sequential phosphorylation of pathway partners. Pharmacologic inhibition of any kinase on a given signal transduction cascade may therefore block communication along the entire pathway. In addition, it is known that protein kinases play a role in disease states and disorders, for example, a kinase mutation and/or overexpression of the kinase are frequently present in many cancers, resulting in hyper-activated activity that often correlates with uncontrolled cell growth.
  • cancer stem cell pathway kinases are important therapeutic targets for the treatment of a variety of cancers (see, for example, U.S. Patent No. 8,299,106).
  • CSCPKs include STK33, MELK, AXL, p70S6K, and PDGFR ⁇ .
  • Activation of the CSCPK PDGFR ⁇ , a receptor tyrosine kinase (RTK), by a PDGF ligand triggers a signal transduction cascade that activates gene expression required for cell proliferation, angiogenesis, and apoptosis.
  • PDGFR ⁇ is a class III receptor tyrosine kinase that is related to the CFS-1 receptor/c-fms and the stem cell growth factor/c-kit proto-oncogene family.
  • the PDGFR ⁇ pathway which is active in early fetal development is also reactivated in many cancers including hepatocellular cancer (HCC), head and neck cancer, brain tumors, gastrointestinal tumors, skin cancer, prostate cancer, ovarian cancer, breast cancer, sarcoma, and leukemia.
  • HCC hepatocellular cancer
  • PDGFR ⁇ activation has recently been shown to play a key role in bone metastasis of prostate cancer.
  • Specifically targeting PDGFR ⁇ using a monoclonal antibody leads to significant reduction in tumor cell proliferation and survival with minimal toxicity.
  • Disruption of PDGFR ⁇ signaling may a viable target for the treatment of a broad spectrum of cancers with minimal toxicity.
  • the CSCPK STK33 or human serine/threonine kinase 33 is a member of the Ca 2+ /calmodulin-dependent kinase family (CAMK). Although its function in normal biology is not fully understood, STK33 may play a specific role in the dynamic behavior of the intermediate filament cytoskeleton through the phosphorylation of vimentin. STK33 knockdown decreases tumor-related gene expression and inhibits cell migration, invasion, and EMT, indicating that STK33 may be a mediator in signaling pathways that are involved in cancer. For example, STK33 has been implicated in the tumorigenesis and progression of hypopharyngeal squamous cell carcinoma (HSCC) (Huang et al. BMC Cancer. (2015)15:13).
  • HSCC hypopharyngeal squamous cell carcinoma
  • MELK Maternal Embryonic Leucine Zipper Kinase
  • MELK is a serine/threonine-CSCPK involved in cell cycle regulation, self-renewal of stem cells, apoptosis and splicing regulation.
  • MELK has broad substrate specificity. For example, MELK acts as an activator of apoptosis by phosphorylating and activating MAP3K5/ASK1.
  • MELK also regulates the cell cycle through phosphorylation of CDC25B which triggers the localization of CDC25B to the centrosome and the spindle poles during mitosis.
  • MELK also phosphorylates and inhibits BCL2L14 indicating a possible role in mammary carcinogenesis by inhibiting the pro-apoptotic function of BCL2L14.
  • the CSCPK AXL Receptor Tyrosine Kinase (or AXL) is a member of the Tyro3-Axl-Mer (TAM) receptor tyrosine kinase subfamily.
  • TAM Tyro3-Axl-Mer
  • the encoded protein possesses an extracellular domain which is composed of two immunoglobulin-like (IgL) motifs at the N-terminus, followed by two fibronectin type-III motifs (FNIII). It transduces signals from the extracellular matrix into the cytoplasm by binding to the vitamin K-dependent protein growth arrest-specific 6 (Gas6).
  • Downregulation of AXL suppresses brain tumor cell growth and invasion (Vajkoczy et al., Proc. Natl. Acad. Sci. U S A. 2006 11; 103(15):5799-804).
  • AXL is therefore a potential mediator of motility and it may control the invasiveness of breast cancer cells (Zhang et al.,
  • p70S6K (or p70 S6 Kinase) is a serine/threonine CSCPK that is a part of the PI3K/Akt/mTOR/P70S6K signaling pathway. Activation of p70S6K triggers S6 ribosomal protein phosphorylation and induces protein synthesis.
  • p70 S6 kinase (p70S6K) is frequently activated in high-grade malignant human ovarian cancer indicating a possible role in metastasis.
  • the at least one compound chosen from compounds having Formula B can be, for example, an inhibitor of a CSCPK (for example, the compound BBI503).
  • a CSCPK for example, the compound BBI503
  • the compounds of Formula B inhibit CSC proliferation and/or survival. Examples 1-5 of U.S. Patent No.8,299,106 further provide exemplary methods of synthesizing the at least one compound chosen from compounds having Formula B.
  • U.S. Patent No.8,299,106 is incorporated herein by reference in its entirety.
  • the present disclosure reports on the surprising discovery that a treatment combination of at least one compound chosen from the compounds of Formula A and at least one compound chosen from the compounds of Formula C had a greater effect at reducing the number cancer cells, including cancer stem cells, than the added effects of each compound acting alone.
  • the treatment combination of the present disclosure resulted in an enhanced inhibition of the expression of cancer cell stemness-associated factors, both in vitro and in vivo, as well as an enhanced decline in the number of cancer stem cells, both in vitro and in vivo, as compared to the added effect observed after treatment with the compound of Formula A alone and after treatment with the compound of Formula C alone.
  • treatment of HCC cell lines with the combination of at least one compound chosen from the compounds of Formula A (e.g., BBI608) and at least one compound chosen from the compounds of Formula C (e.g., sorafenib) resulted in enhanced decrease in the number of bulk tumor cells (FIG.1) and a reduction in the number of spheres formed by CSCs after culture in non- adherent stem cell media (FIG.2).
  • FIG.3 also shows that the volume of tumors in a HepG2 murine xenograft mouse model declined as a result of treatment with at least one compound of Formula A (e.g., BBI608) when compared to treatment with a control.
  • the present disclosure further reports on the surprising discovery that a treatment combination of at least one compound chosen from the compounds having Formula B and at least one compound chosen from the compounds having Formula C (sorafenib) had a greater effect at reducing the proliferation and/or survival of cancer cells, including cancer stem cells, than the added effects of either compound acting alone.
  • the treatment combination of the present disclosure resulted in an enhanced inhibition of the expression of cancer cell stemness- associated factors in vitro and in vivo, and an enhanced reduction in the number of cancer stem cells in vitro and in vivo, as compared to the added effects from the treatment of the tumors with the compound of Formula B (e.g. BBI503) alone and the compound of Formula C (e.g., sorafenib) alone.
  • a compound of Formula B was active against CSCs derived from HCC cell lines, both alone and in combination with a compound of Formula C (e.g., sorafenib).
  • the compound of Formula B not only inhibited cancer stem cell proliferation and/or survival but also reduced the ability of CSCs to form characteristic spheres in non-adherent stem cell media.
  • the equivalent treatment with compound of Formula C did not have any significant inhibitory activity on CSC sphere-forming ability or on the proliferation and/or survival of cancer cells.
  • treatment of cancer cells with the combination of a compound of Formula B and a compound of Formula C enhanced the reduction in colony formation (FIG.4, FIG.5, and FIG. 6).
  • the compound of Formula B e.g. BBI503, was also shown to inhibit the expression of cancer stem cell markers, such as, CD133 and CD44.
  • FIG. 7 shows that the average tumor volume in animals treated with a compound of Formula B was less than 50% of the average tumor volume obtained in controls.
  • a treatment combination of the present disclosure are believed to work on different pathways that are associated with cancer cells (e.g., CSCs).
  • CSCs cancer cells
  • the treatment combination of a compound of Formula A and a compound of Formula C or a compound of Formula B and a compound of Formula C results in an inhibition of cancer cell proliferation and/or survival that is greater than the added effects observed after treatment with either compound alone (sometimes referred to as “enhanced” or “synergistic” effects).
  • a compound of Formula A can work by inhibiting the STAT3 signaling pathway.
  • a compound of Formula A can directly bind and inhibit the activity of activated STAT3 (e.g., phosphorylated STAT3), thereby preventing transcription of ST AT3 -dependent target genes including the sternness-associated transcription factors c-MYC, OCT4, SOX2, and ⁇ -CATENIN.
  • a compound of Formula B can inhibit the activity of multiple malignancy-associated serine-threonine kinases (or cancer stem cell pathway kinases (CSCPKs)).
  • disclosed herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound chosen from compounds having Formula A:
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • disclosed herein are methods for treating cancer comprising administering to a subject in need thereof:
  • methods of treating cancer comprising administering to a subject in need thereof (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the cancer stemness inhibitor can be, for example, a STAT3 pathway inhibitor.
  • the cancer stemness inhibitor can be, for example, chosen from 2- (1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro-naphtho[2,3-b]furan-4,9- dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2-acetylnaphtho[2,3-b]furan-4,9-dione, 2-ethyl-naphtho[2,3-b]furan-4,9-dione, prodrugs of any of the foregoing, derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the cancer stemness inhibitor can be, for example, a cancer stem cell pathway kinase (CASPK) inhibitor.
  • CASPK cancer stem cell pathway kinase
  • the cancer stemness inhibitor can be chosen, for example, from the following compounds having the Formulae of:
  • prodrugs of any of the foregoing derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • a kit comprises (1) at least one first compound chosen from compounds having Formula A, compounds having Formula B, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
  • a kit comprises (1) at least one first compound chosen from compounds having Formula A, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
  • a kit comprises (1) at least one first compound chosen from compounds having Formula B, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
  • a kit comprises (1) at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (2) at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
  • FIG.1 illustrates the enhanced inhibition of HCC colony formation by a Hep3B cell line after treatment of the cells with a combination of a compound of Formula A and a compound of Formula C.
  • FIG.2 shows an example of the enhanced inhibition of Sk-Hep1, Huh7, and HepG2 CSC viability after treatment of the cells with a combination of a compound of Formula A and a compound of Formula C.
  • FIG.3 shows an example of the inhibition of tumor growth in a murine xenograft model of HCC after treatment with a compound of Formula A.
  • FIG.4 illustrates the enhanced inhibition of HCC colony formation by a Hep3B cell line after the treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
  • FIG. 5 illustrates the enhanced inhibition of HCC colony formation by a Hub7 cell line after treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
  • FIG.6 shows an example of the enhanced inhibition of Skhep1, Hep3B, and HepG2 CSC viability after treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
  • FIG.7 shows an example of the inhibition of tumor growth in a murine xenograft model of HCC after treatment with a compound of Formula B.
  • FIG. 8A shows an assessment of patients with advanced HCC treated with napabucasin (BBI608, Compound A) and sorafenib (Compound C) evaluated according to RECIST 1.1 and modified RECIST (mRECIST).
  • FIG. 8B shows an assessment of patients with advanced HCC treated with amcasertib (BBI503, Compound B) and sorafenib (Compound C) evaluated according to RECIST 1.1 and modified RECIST (mRECIST).
  • BBI608 napabucasin
  • FIG. 8B shows an assessment of patients with advanced HCC treated with amcasertib (BBI503, Compound B) and sorafenib (Compound C) evaluated according to RECIST 1.1 and modified RECIST (mRECIST).
  • the term“about” when used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below those numerical values.
  • the term“about” is used herein to modify a numerical value above and below the stated value by a variance of 20%, 10%, 5%, or 1%.
  • the term“about” is used to modify a numerical value above and below the stated value by a variance of 10%.
  • the term“about” is used to modify a numerical value above and below the stated value by a variance of 5%.
  • the term“about” is used to modify a numerical value above and below the stated value by a variance of 1%.
  • “1-5 mg” is intended to encompass 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 1-2 mg, 1-3 mg, 1-4 mg, 1-5 mg, 2-3 mg, 2-4 mg, 2-5 mg, 3-4 mg, 3-5 mg, and 4-5 mg.
  • administer refers to any method of introducing to a subject a compound or pharmaceutical composition described herein and can include, for example, introducing the compound systemically, locally, or in situ to the subject.
  • a compound of the present disclosure produced in a subject from a composition is encompassed by these terms.
  • systemic or“systemically,” they generally refer to in vivo systemic absorption or accumulation of the compound or composition in the blood stream followed by distribution throughout the entire body.
  • the term“subject” generally refers to an organism to which a compound or pharmaceutical composition described herein can be administered.
  • a subject can be a mammal or mammalian cell, including a human or human cell.
  • the term also refers to an organism, which includes a cell or a donor or recipient of such cell.
  • the term“subject” refers to any animal (e.g., a mammal), including, but not limited to humans, mammals and non-mammals, such as non- human primates, mice, rabbits, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, fish, nematode, and insects, which is to be the recipient of a compound or pharmaceutical composition described herein.
  • the terms“subject” and“patient” are used interchangeably herein in reference to a human subject.
  • the terms“effective amount” and“therapeutically effective amount” refer to that amount of a compound or pharmaceutical composition described herein that is sufficient to effect the intended result including, but not limited to, disease treatment, as illustrated below.
  • the“therapeutically effective amount” is the amount that is effective for detectable killing or inhibition of the growth or spread of cancer cells, the size or number of tumors, and/or other measure of the level, stage, progression and/or severity of the cancer.
  • the“therapeutically effective amount” refers to the amount that is administered systemically, locally, or in situ (e.g., the amount of compound that is produced in situ in a subject).
  • the therapeutically effective amount can vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of cell migration.
  • the specific dose may vary depending on, for example, the weight of the subject, the particular pharmaceutical composition, subject and their age and existing health conditions or risk for health conditions, the dosing regimen to be followed, the severity of the disease, whether it is administered in combination with other agents, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • A“therapeutically effective amount” in reference to the treatment of a subject’s cancer means an amount capable of invoking, for example, one or more of the following effects: (1) inhibition, to some extent, of cancer or tumor growth, including a decrease or cessation in the progression of the subject’s cancer; (2) reduction in the number of cancer or tumor cells; (3) reduction in tumor size; (4) inhibition, e.g., a decrease or a cessation, of cancer or tumor cell infiltration into peripheral organs; (5) inhibition, e.g., a decrease or a cessation, of metastasis; (6) enhancement of anti-tumor immune response, which may, but is not required to, result in the regression or rejection of the tumor, or (7) relief, to some extent, of one or more symptoms associated with the cancer or tumor.
  • the therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual and the ability of one or more anti-cancer agents to elicit a desired response in the individual.
  • A“therapeutically effective amount” can be, for example, an amount of a compound where any toxic or detrimental effects resulting from the administration of the compound are outweighed by the therapeutically beneficial effects.
  • the terms“treatment,”“treating,”“ameliorating,” and“encouraging” are used interchangeably herein. These terms refer to an approach for obtaining beneficial or desired results including, but not limited to, a therapeutic benefit and/or prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject can still be afflicted with the underlying disorder.
  • the pharmaceutical composition may be administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • a treatment combination comprises a therapeutically effective amount of at least one first compound chosen from compounds having Formula A and compounds having Formula B and a therapeutically effective amount of at least one second compound chosen from compounds having Formula C.
  • the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors can have different mechanisms of action.
  • a combination treatment improves the prophylactic or therapeutic effect of the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors by functioning together to have an additive, synergistic, or enhanced effect.
  • a combination treatment of the present disclosure reduces the side effects associated with the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors.
  • the administrations of the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors may be separated in time by up to several weeks, but more commonly within 48 hours, and most commonly within 24 hours.
  • prodrug refers to a derivative of a parent drug molecule that requires biotransformation, either spontaneous or enzymatic, within the organism to release the active drug.
  • prodrugs are variations or derivatives of the compounds of Formula A, B or C that have groups cleavable under certain metabolic conditions, and which, when cleaved, become the compounds of Formula A, B or C respectively.
  • Such prodrugs then are pharmaceutically inactive in vivo, until they become active as a result of, for example, solvolysis or enzymatic degradation under physiological conditions.
  • Prodrug compounds, described herein may be called single, double, triple, etc., depending on the number of biotransformation steps required to release the active drug within the organism, and the number of functionalities present in a precursor-type form.
  • Prodrug forms often offer advantages of solubility, tissue compatibility, or delayed release in the mammalian organism.
  • Prodrugs commonly known in the art include, but are not limited to, well-known acid derivatives, such as, for example, esters prepared by reaction of the parent acids with a suitable alcohol, amides prepared by reaction of the parent acid compound with an amine, or basic groups reacted to form an acylated base derivative, etc.
  • Other prodrug derivatives may also be combined with other features disclosed herein to enhance bioavailability.
  • Those of skill in the art will appreciate that certain of the presently disclosed compounds having free amino, amido, hydroxy, or carboxylic groups can be converted into prodrugs.
  • Prodrugs include compounds having an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy, or carboxylic acid groups of the presently disclosed compounds.
  • the amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols as well as, for example, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine, and methionine sulfone.
  • Prodrugs may also include compounds having a carbonate, carbamate, amide, or alkyl ester moiety covalently bonded to any of the above substituents disclosed herein.
  • synergy refers to an effect of interaction or combination of two or more components to produce a combined effect greater than the sum of their separate effects (or“additive effects”).
  • a synergistic effect may be attained when the compounds are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. in separate tablets, pills or capsules, or by different injections in separate syringes.
  • an effective dosage of each active ingredient can be administered, for example, sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • a synergistic anticancer effect denotes an anticancer effect which is greater than the predicted purely additive effects of the individual compounds of the combination administered separately.
  • cancer stemness inhibitor means a molecule that can target, reduce, inhibit, interfere with, or modulate at least one of a plurality of pathways involved in cancer stemness or the expression (e.g., the production of a functional product, e.g., a protein) of at least one of a plurality of cancer stemness genes.
  • the expression or the expressed proteins can be used as biomarkers of the corresponding cancer stemness genes. Examples of these biomarkers include, but are not limited to, ⁇ -CATENIN, NANOG, SMO, SOX2, STAT3, AXL, ATM, c-MYC, KLF4, SURVIVIN, or BMI-1.
  • a cancer stemness inhibitor may alter cancer stem cell growth as well as heterogeneous cancer cell growth.
  • a cancer stemness inhibitor can be, for example, a small molecule that binds a protein encoded by a cancer stemness gene.
  • a cancer stemness inhibitor can be, for example, a biologic, e.g., a recombinant binding protein or peptide (e.g. APTSTAT3; see Kim et al. Cancer Res. (2014) 74(8):2144-51) or nucleic acid (e.g. STAT3 aiRNA; see U.S. Patent No.9,328,345, the content of which is incorporated herein in its entirety), or conjugate thereof, that binds to a protein encoded by a cancer stemness gene.
  • a biologic e.g., a recombinant binding protein or peptide (e.g. APTSTAT3; see Kim et al. Cancer Res. (2014) 74(8):2144-51) or nucleic acid (e.g. STAT3 aiRNA; see U.S. Patent No.9,328,345, the
  • a cancer stemness inhibitor can be, for example, a cell.
  • a cancer stemness inhibitor can be, for example, a STAT3 inhibitor (for example, that binds to and inhibits a biological activity of STAT3 (see Furtek et al., ACS Chem. Biol., 2016, 11 (2), pp 308– 318)).
  • a cancer stemness inhibitor can be, for example, a CSCPK inhibitor.
  • a cancer stemness inhibitor can be, for example, at least one compound chosen from 2-(1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro- naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2- acetylnaphtho[2,3-b]furan-4,9-dione, or 2-ethyl-naphtho[2,3-b]furan-4,9-dione, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • a cancer stemness inhibitor can be, for example, at least one compound chosen, for example, from the following compounds having the Formulae of:
  • prodrugs of any of the foregoing derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • a cancer stemness inhibitor of the present disclosure may be administered in an amount ranging from about 300 mg to about 700 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 700 mg to about 1200 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 800 mg to about 1100 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 850 mg to about 1050 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 960 mg to about 1000 mg.
  • the total amount of the cancer stemness inhibitor can be administered once daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 480 mg daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 960 mg daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 1000 mg daily. In certain embodiments, the total amount of the cancer stemness inhibitor can be administered in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 240 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 480 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 500 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered orally.
  • a cancer stemness inhibitor can be, for example, at least one compound chosen from compounds having Formula A.
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • prodrugs and derivatives of compounds having Formula A can be, for example, STAT3 inhibitors.
  • Non-limiting examples of prodrugs of compounds having Formula A include, for example, the phosphoric ester and phosphoric diester described in U.S. pre-grant Publication No. 2012/0252763 as compound numbers 4011 and 4012 and also suitable compounds described in in U.S. Patent No. 9,150,530.
  • Non-limiting examples of derivatives of compounds having Formula A include, for example, the derivatives disclosed in U.S. Patent No. 8,977,803. The disclosures of U.S. pre-grant Publication No. 2012/0252763 and U.S. Patent Nos. 9,150,530 and 8,977,803 are incorporated herein by reference in their entireties.
  • BB608 may also be known as 2-acetylnaphtho[2,3-b]furan-4,9-dione, napabucasin, BBI608, or BB608, and include tautomers thereof.
  • Compound A may be administered in an amount ranging from about 80 mg to about 1500 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 160 mg to about 1000 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 300 mg to about 700 mg a day. In certain embodiments, Compound A may be administered in an amount ranging from about 700 mg to about 1200 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 800 mg to about 1100 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 850 mg to about 1050 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 960 mg to about 1000 mg.
  • the total amount of Compound A can be administered, for example, once daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 480 mg daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 960 mg daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 1000 mg daily. In certain embodiments, the total amount of Compound A can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound A may be administered in an amount ranging from about 80 mg twice daily to about 750 mg twice daily.
  • Compound A may be administered in an amount ranging from about 80 mg twice daily to about 500 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 240 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 480 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 500 mg twice daily. In certain embodiments, Compound A may be administered, for example, orally.
  • a cancer stemness inhibitor can be, for example, at least one compound chosen from compounds having Formula B.
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • a cancer stemness inhibitor can be, for example, a cancer stem cell pathway kinase (CSCPK) inhibitor, a STK33 inhibitor, a MELK inhibitor, an AXL inhibitor, a p70S6K inhibitor, a PDGFR ⁇ inhibitor, or a NANOG inhibitor.
  • a cancer stemness inhibitor can be, for example, a cancer stem cell pathway kinase (CSCPK) inhibitor.
  • CSCPK cancer stem cell pathway kinase
  • a cancer stemness inhibitor can be, for example, a STK33 inhibitor.
  • a cancer stemness inhibitor can be, for example, a MELK inhibitor.
  • a cancer stemness inhibitor can be, for example, an AXL inhibitor.
  • a cancer stemness inhibitor can be, for example, a p70S6K inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a PDGFR ⁇ inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a NANOG inhibitor.
  • compounds having Formula B, and derivatives thereof can be, for example, cancer stem cell pathway kinase (CSCPK) inhibitors.
  • compounds having Formula B, and derivatives thereof can include inhibitors of, for example, STK33, MELK, AXL, p70S6K, and PDGFR ⁇ .
  • the at least one compound chosen from compounds having Formula B, and derivatives thereof can be, for example,STK33 inhibitors.
  • the at least one compound chosen from compounds having Formula B, and derivatives thereof can be, for example, MELK inhibitors.
  • the at least one compound chosen from compounds having Formula B, and derivatives thereof can be, for example, AXL inhibitors.
  • the at least one compound chosen from compounds having Formula B, and derivatives thereof can be, for example, p70S6K inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example, PDGFR ⁇ inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can inhibit NANOG expression.
  • Non-limiting examples of compounds having Formula B, and derivatives thereof can include, for example, the derivatives disclosed in U.S. Patent No. 8,299,106 and PCT Patent Application Publication No. WO2014160401. The disclosures of U.S. Patent No. 8,299,106 and PCT Patent Application Publication No. WO2014160401 are incorporated herein by reference in their entireties.
  • the CSCPK inhibitor can be chosen, for example, from the following compounds having the Formulae of:
  • prodrugs thereof derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the CSCPK inhibitor can be, for example, at least one compound chosen from compounds having Formula B.
  • the CSCPK inhibitor may be administered in an amount ranging from about 20 mg to about 600 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 50 mg to about 500 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 80 mg to about 400 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 80 mg to about 300 mg. In certain embodiments, the CSCPK inhibitor can be administered, for example, once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 100 mg daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 200 mg daily.
  • the CSCPK inhibitor can be administered, for example, in a dose of about 300 mg daily. In certain embodiments, the total amount of the CSCPK inhibitor can be administered, for example, in a single daily dose. In certain embodiments, the total amount of the CSCPK inhibitor can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 100 mg once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 200 mg once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, orally.
  • Compound B may be administered in an amount ranging from about 20 mg to about 600 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 50 mg to about 500 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 80 mg to about 400 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 80 mg to about 300 mg. In certain embodiments, Compound B can be administered, for example, once daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 100 mg daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 200 mg daily.
  • Compound B can be administered, for example, in a dose of about 300 mg daily. In certain embodiments, the total amount of Compound B can be administered, for example, in a single daily dose. In certain embodiments, the total amount of Compound B can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound B can be administered, for example, in a dose of about 100 mg once daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 200 mg once daily. In certain embodiments, Compound B can be administered, for example, orally.
  • kinase inhibitor refers to a molecule that can target, reduce, inhibit, interfere with, or modulate the activities of at least one of a plurality of protein kinases.
  • protein kinases include, but are not limited to, intracellular kinases (e.g., c-CRAF, BRAF, and mutant BRAF) and cell surface kinases (e.g., KIT, FLT-3, RET, RET/PTC, VEGFR-1, VEGFR-2, VEGFR-3, and PDGFR-ß).
  • kinase inhibitors include, but are not limited to, afatinib (an EGFR/ErbB2 inhibitor), axitinib, (a VEGFR1/VEGFR2/VEGFR3/PDGFRB/c-KIT inhibitor), bosutinib (a Bcr-Abl/SRC inhibitor), cetuximab (an EGFR inhibitor), cobimetinib (a MEK inhibitor), crizotinib (an ALK/Met inhibitor), cabozantinib (a RET/MET/VEGFR2 inhibitor), dasatinib (a multiple kinase inhibitor), entrectinib (a TrkA/TrkB/TrkC/ROS1/ALK inhibitor), erlotinib (an EGFR inhibitor), fostamatinib (a Syk inhibitor), gefitinib (an EGFR inhibitor), ibrutinib (a BTK inhibitor), imatinib (a Bcr-Abl inhibitor),
  • a kinase inhibitor can be, for example, at least one compound chosen from compounds having Formula C.
  • Compound C may also be known as 4-[4-[[4-chloro-3- (trifluoromethyl)phenyl]carbamoylamino]phenoxy]-N-methyl-pyridine-2-carboxamide, Nexavar, or sorafenib.
  • Compound C may be administered in an amount ranging from about 80 mg to about 1500 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 100 mg to about 1200 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 200 mg to about 1000 mg a day. In certain embodiments, Compound C may be administered in an amount ranging from about 400 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 600 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 700 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount of about 800 mg.
  • the total amount of Compound C can be administered, for example, once daily. In certain embodiments, the total amount of Compound C can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound C may be administered in an amount ranging from about 80 mg twice daily to about 750 mg twice daily. In certain embodiments, Compound C may be administered in an amount ranging from about 80 mg twice daily to about 600 mg twice daily. In certain embodiments, Compound C can be administered, for example, in a dose of about 200 mg twice daily. In certain embodiments, Compound C can be administered, for example, in a dose of about 400 mg twice daily. In certain embodiments, Compound C can be administered, for example, orally. In certain embodiments, Compound C can be administered, for example, on an empty stomach. In certain embodiments, Compound C can be administered, for example, either about 1 hour prior to a meal or about 2 hours after a meal.
  • progress refers to at least one of the following: (1) a response to prior therapy (e.g., chemotherapy) of progressive disease (PD); (2) the appearance of one or more new lesions after treatment with prior therapy (e.g., chemotherapy); and (3) at least a 5% (e.g., 10%, 20%) increase in the sum of diameters of target lesions, taking as a reference the smallest sum on study (this includes the baseline sum if that is the smallest on study).
  • a therapy e.g., chemotherapy
  • a therapy e.g., chemotherapy
  • that therapy e.g., chemotherapy
  • cancer in a subject refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain morphological features. Often, cancer cells can be in the form of a tumor or mass, but such cells may exist alone within a subject, or may circulate in the blood stream as independent cells, such as leukemic or lymphoma cells.
  • cancer examples include, but are not limited to, lung cancer, pancreatic cancer, bone cancer, skin cancer, head or neck cancer, cutaneous or intraocular melanoma, breast cancer, uterine cancer, ovarian cancer, peritoneal cancer, colon cancer, rectal cancer, colorectal adenocarcinoma, cancer of the anal region, stomach cancer, gastric cancer, gastrointestinal cancer, gastric adenocarcinoma, adrenocorticoid carcinoma, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, chondrosarcoma, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue
  • urological cancer a general term
  • hepatobiliary cancer another general term, includes liver cancers (itself a general term that includes hepatocellular carcinoma or cholangiocarcinoma), gallbladder cancer, biliary cancer, or pancreatic cancer.
  • liver cancers itself a general term that includes hepatocellular carcinoma or cholangiocarcinoma
  • gallbladder cancer biliary cancer
  • pancreatic cancer pancreatic cancer
  • solid tumor can include the term,“solid tumor.”
  • solid tumor refers to those conditions, such as cancer, that form an abnormal tumor mass, such as sarcomas, carcinomas, and lymphomas.
  • solid tumors include, but are not limited to, non-small cell lung cancer (NSCLC), neuroendocrine tumors, thyomas, fibrous tumors, metastatic colorectal cancer (mCRC), and the like.
  • NSCLC non-small cell lung cancer
  • mCRC metastatic colorectal cancer
  • the solid tumor disease can be, for example, an adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and the like.
  • the cancer can be, for example, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, gastric cancer, chondrosarcoma, colorectal adenocarcinoma, breast cancer, ovarian cancer, head and neck cancer, melanoma, gastric adenocarcinoma, lung cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, cervical cancer, brain tumor, multiple myeloma, leukemia, lymphoma, prostate cancer, cholangiocarcinoma, endometrial cancer, small bowel adenocarcinoma, uterine sarcoma, or adrenocorticoid carcinoma.
  • gastroesophageal junction cancer gastroesophageal adenocarcinoma
  • gastric cancer chondrosarcoma
  • colorectal adenocarcinoma breast cancer
  • ovarian cancer head and neck cancer
  • the cancer can be, for example, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, colorectal adenocarcinoma, breast cancer, ovarian cancer, head and neck cancer, melanoma, gastric adenocarcinoma, lung cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, cervical cancer, brain tumor, multiple myeloma, leukemia, lymphoma, prostate cancer, cholangiocarcinoma, endometrial cancer, small bowel adenocarcinoma, uterine sarcoma, or adrenocorticoid carcinoma.
  • gastroesophageal junction cancer gastroesophageal adenocarcinoma
  • colorectal adenocarcinoma breast cancer
  • ovarian cancer head and neck cancer
  • melanoma gastric adenocarcinoma
  • lung cancer pancreatic cancer
  • the cancer can be, for example, breast cancer. In certain embodiments, the cancer can be, for example, colorectal adenocarcinoma. In certain embodiments, the cancer can be, for example, small bowel adenocarcinoma. In certain embodiments, the cancer can be, for example, hepatocellular carcinoma. In certain embodiments, the cancer can be, for example, head and neck cancer. In certain embodiments, the cancer can be, for example, renal cell carcinoma. In certain embodiments, the cancer can be, for example, ovarian cancer. In certain embodiments, the cancer can be, for example, prostate cancer. In certain embodiments, the cancer can be, for example, lung cancer.
  • the cancer can be, for example, uterine sarcoma. In certain embodiments, the cancer can be, for example, esophageal cancer. In certain embodiments, the cancer can be, for example, endometrial cancer. In certain embodiments, the cancer can be, for example, cholangiocarcinoma. In certain embodiments, each of the cancers can be, for example, unresectable, advanced, refractory, recurrent, or metastatic.
  • salt(s), includes acidic and/or basic salts formed with inorganic and/or organic acids and bases.
  • pharmaceutically acceptable salt refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response and/or the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19.
  • compositions may be formed with inorganic or organic acids.
  • suitable inorganic acids include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid.
  • suitable organic acids include acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, and malonic acid.
  • Suitable pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, besylate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate
  • organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, lactic acid, trifluoracetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicylic acid. Salts may be prepared in situ during the isolation and purification of the disclosed compound, or separately, such as by reacting the compound with a suitable base or acid, respectively.
  • Non-limiting examples of pharmaceutically acceptable salts derived from bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • suitable alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts.
  • suitable pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.
  • Non- limiting examples of suitable organic bases from which salts may be derived include primary amines, secondary amines, tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • pharmaceutically acceptable base addition salts can be chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
  • solvate represents an aggregate that comprises one or more molecules of a compound of the present disclosure with one or more molecules of a solvent or solvents.
  • Solvates of the compounds of the present disclosure include, for example, hydrates.
  • the at least one compound disclosed herein may be in the form of a pharmaceutical composition.
  • the pharmaceutical compositions may comprise the at least one compound having Formula A and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical compositions may comprise the at least one compound having Formula B and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical compositions may comprise the at least one compound having Formula C and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula A in a subject (i.e., a prodrug).
  • the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula B in a subject (i.e., a prodrug).
  • the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula C in a subject (i.e., a prodrug).
  • carrier means a pharmaceutically acceptable material, composition or vehicle, such as, for example, a liquid or solid filler, diluent, excipient, solvent or encapsulating material involved in or capable of carrying or transporting the subject pharmaceutical compound from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically acceptable material, composition or vehicle such as, for example, a liquid or solid filler, diluent, excipient, solvent or encapsulating material involved in or capable of carrying or transporting the subject pharmaceutical compound from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the Formulation and not injurious to the patient.
  • Non-limiting examples of pharmaceutically acceptable carriers, carriers, and/or diluents include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water;
  • wetting agents such as sodium lauryl sulfate, magnesium stearate, and polyethylene oxide-polypropylene oxide copolymer as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives, and antioxidants can also be present in the compositions.
  • compositions disclosed herein that are suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, a solution in an aqueous or non-aqueous liquid, a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, a water-in-oil emulsion, an elixir, a syrup, pastilles (using an inert base, such as gelatin, glycerin, sucrose, and/or acacia) and/or mouthwashes, each containing a predetermined amount of the at least one compound of the present disclosure.
  • an inert base such as gelatin, glycerin, sucrose, and/or acacia
  • a pharmaceutical composition disclosed herein may be administered as a bolus, electuary, or paste.
  • Solid dosage forms for oral administration may be mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium carbonate, and sodium starch glycolate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol, glycerol
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type also may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.
  • cyclodextrins e.g., hydroxypropyl- ⁇ -cyclodextrin, may be
  • compositions also may include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • Suspensions in addition to the compounds according to the disclosure, may contain suspending agents as, such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar, and tragacanth, and mixtures thereof.
  • compositions disclosed herein, for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds according to the present disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the compounds of the present disclosure.
  • Pharmaceutical compositions which are suitable for vaginal administration also may include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing carriers that are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a pharmaceutical composition or pharmaceutical tablet of the present disclosure may include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants.
  • the pharmaceutical composition or pharmaceutical tablet may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to the pharmaceutical composition or pharmaceutical tablet of the present disclosure, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • Powders and sprays may contain, in addition to a pharmaceutical composition or a pharmaceutical tablet of the present disclosure, excipients such as lactose, talc, silicic acid, aluminium hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances. Additionally, sprays may contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Ophthalmic formulations are also contemplated as being within the scope of the present disclosure.
  • compositions suitable for parenteral administration may comprise at least one more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions, emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • a composition described herein may include at least one compound chosen from compounds of Formula A and pharmaceutically acceptable salts and solvates thereof and one or more surfactants.
  • the surfactant can be, for example, sodium lauryl sulfate (SLS), sodium dodecyl sulfate (SDS), or one or more polyoxylglycerides.
  • the polyoxylglyceride can be lauroyl polyoxylglycerides (sometimes referred to as Gelucire TM ) or linoleoyl polyoxylglycerides (sometimes referred to as Labrafil TM ). Examples of such compositions are shown in PCT Patent Application No. PCT/US2014/033566, the contents of which are incorporated herein in their entireties.
  • the present invention provides further embodiments of suitable pharmaceutical formulations having selected particle size distribution and methods for identifying an optimum particle size distribution, suitable drug regimen, dosage and interval, suitable methods of preparing 2-acetylnaphtho[2,3-b]furan-4,9-dione including their crystalline forms, and further specific suitable cancer stemness inhibitors and kinase inhibitors as described in the co-owned PCT applications published as WO 2009/036099, WO 2009/036101, WO 2011/116398, WO 2011/116399, WO 2014/169078, and WO 2009/033033, the contents of which are incorporated by reference herein in their entirety.
  • a composition described herein may include at least one compound chosen from compounds of Formula B and pharmaceutically acceptable salts and solvates thereof and one or more surfactants.
  • the surfactant can be, for example, sodium lauryl sulfate (SLS), sodium dodecyl sulfate (SDS), or one or more polyoxylglycerides.
  • the polyoxylglyceride can be lauroyl polyoxylglycerides (sometimes referred to as Gelucire TM ) or linoleoyl polyoxylglycerides (sometimes referred to as Labrafil TM ).
  • the compounds or pharmaceutical compositions described herein may be administered in combination with any of a variety of known therapeutics, including for example, chemotherapeutic and other anti-neoplastic agents, anti-inflammatory compounds, and/or immunosuppressive compounds.
  • the compounds, products, and/or pharmaceutical compositions described herein are useful in conjunction with any of a variety of known treatments including, by way of non-limiting example, surgical treatments and methods, radiation therapy, chemotherapy, and/or hormone or other endocrine-related treatment.
  • methods of treating cancer in a subject in need thereof comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • methods of treating cancer cells comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the cancer cells are in a subject.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • methods of inhibiting, reducing, and/or diminishing cancer stem cell’s survival and/or self-renewal comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the cancer stem cells are in a subject.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • methods of treating cancer that is refractory to conventional chemotherapies and/or targeted therapies in a subject comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • the conventional chemotherapies and/or targeted therapies may comprise administering at least one compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutical acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the conventional chemotherapies and/or target therapies may comprise administering sorafenib.
  • methods of treating recurrent cancer in a subject that has failed surgery, oncology therapy (e.g., chemotherapy), and/or radiation therapy comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • oncology therapy e.g., chemotherapy
  • radiation therapy e.g., chemotherapy
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • methods of treating or preventing cancer metastasis in a subject comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • methods of preventing relapse or suppressing regrowth or recurrent of cancer in a subject comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • the method is a part of an adjuvant therapy.
  • the method comprises administering a treatment combination of the present disclosure after or currently with a primary treatment of cancer.
  • the primary treatment is chosen from chemotherapies, radiation therapies, hormone therapies, targeted therapies, or biological therapies.
  • methods of sensitizing or resensitizing cells to at least one kinase inhibitor comprising administering a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one kinase inhibitor is sorafenib.
  • the methods comprise administering a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
  • the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • At least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
  • the cancer can be, for example, hepatocellular carcinoma. In certain embodiments, the cancer can be, for example, cholangiocarcinoma.
  • the cancer may be unresectable. In certain embodiments, the cancer may be advanced. In certain embodiments, the cancer may be refractory. In certain embodiments, the cancer may be recurrent. In certain embodiments, the cancer may be metastatic. In certain embodiments, the cancer may be associated with overexpression of STAT3. In certain embodiments, the cancer may be associated with nuclear ⁇ -catenin localization.
  • the methods disclosed herein comprise administering to a subject in need thereof a therapeutically effective amount of at least one compound chosen from cancer stemness inhibitors in combination with a therapeutically effective amount of at least one compound chosen from kinase inhibitors.
  • treatment with the combination (“combo”) of Compound A and Compound C resulted in an enhanced inhibition of HCC colony formation in comparison to treatment with Compound A (BBI608) alone or with Compound C (sorafenib) alone.
  • EXAMPLE 3 Treatment of a murine xenograft model of human cancer with Compound A
  • HepG2 cells were inoculated subcutaneously into male athymic nude mice (8 x 10 6 cells/mouse) and allowed to form palpable tumors. Once the tumors reached approximately 500 mm 3 , the animals were treated orally with either vehicle (control), or Compound A (BBI608; 10 mg/kg, i.v.) as indicated in FIG. 3. The animals received a total of 5 doses. Treatment with Compound A (BBI608) inhibited tumor growth as compared to control animals.
  • the treatment with the combination of Compound B (BBI503) and Compound C (sorafinib) resulted in an enhanced inhibition of HCC colony formation in comparison to treatment with Compound A (BBI503) alone or with Compound C (sorafenib) alone.
  • EXAMPLE 6 Treatment of a murine xenograft model of human cancer with Compound B
  • BBI503 In the mouse xenograft model of human HCC, HepG2 cells were inoculated subcutaneously into male athymic nude mice (8 x 10 6 cells/mouse) and allowed to form palpable tumors. Once the tumors reached approximately 500 mm 3 , the animals were treated orally with either vehicle (control), or Compound B (BBI503; 100 mg/kg, p.o.) as indicated in FIG. 7. The animals received a total of 5 doses. Treatment with Compound B (BBI503) inhibited tumor growth as compared to control animals.
  • EXAMPLE 7 Clinical trials with treatment combinations of Compound A (BBI608) and
  • Compound C (sorafenib) or Compound B (BBI503) and Compound C (sorafenib)
  • the effects of two treatment combinations in patients with advanced hepatocellular carcinoma were studied in a Phase IB/II study to assess the safety, tolerability, and preliminary anti-cancer activity of the drug combinations disclosed herein.
  • the first treatment combination included Compound A (BBI608) and Compound C (sorafenib)
  • the second treatment combination included Compound B (BBI503) and Compound C (sorafenib).
  • Phase IB/II study determined the safety and efficacy of the aforementioned drug combinations on a population of adult patients with advanced hepatocellular carcinoma who had not received systemic chemotherapy.
  • the Phase IB portion involved dose-escalation of Compound A (BBI608) administered in combination with a fixed starting dose of Compound C (sorafenib) (Arm 1), and dose escalation of Compound B (BBI503) administered in combination with a fixed starting dose of Compound C (sorafenib) (Arm 2).
  • the fixed starting dose-level of sorafenib for both arms was 400 mg twice daily (800 mg total daily dose). Eligible patients were randomized to either Arm 1 or Arm 2.
  • escalating doses of Compound A were administered to cohorts of three to six patients until recommended Phase II dose (RP2D) was determined according to the established criteria for determining dose-limiting toxicity (DLT) and dose-escalation.
  • PK pharmacokinetic
  • R2D Phase II dose
  • the Phase II portion was an open-label, 3-arm, randomized Phase II trial of patients with advanced HCC who had not received prior systemic treatment. Patients were randomized to receive either, Arm 1: Compound C (sorafenib) administered in combination with Compound A (BBI608) (at the RP2D determined for BBI608 plus sorafenib during the Phase IB portion); Arm 2: Compound C (sorafenib) in combination with Compound B (BBI503) (at the RP2D determined for BBI503 plus sorafenib during the Phase IB portion), or Arm 3: Compound C (sorafenib) alone at a starting dose of 400 mg twice daily. The starting dose for sorafenib was the same for all study arms.
  • a patient was allowed to continue protocol therapy beyond progression that is determined by RECIST criteria (either RECIST 1.1 or mRECIST for patients with HCC), provided that the patient is obtaining potential clinical benefit in the opinion of the investigator. If sorafenib was discontinued due to sorafenib-related toxicities, a patient was allowed to continue receiving the study drug (either BBI608 or BBI503) provided there were no signs of toxicity and the patient obtained a clinical benefit from the drug.
  • RECIST criteria either RECIST 1.1 or mRECIST for patients with HCC
  • Napabucasin and amcasertib were safely combined with sorafenib at full dose with no unexpected adverse events. Encouraging anti-cancer activity in patients with HCC who have not received prior systemic therapy was observed. RP2D dose was determined to be 240mg BID for napabucasin and 100mg QD for amcasertib. A randomized phase II part of this study is currently enrolling patients into three arms. Arm I: napabucasin+sorafenib, Arm II: amcasertib+sorafenib and Arm III: sorafenib alone.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Methods comprising administering and kits comprising a therapeutically effective amount of at least one first compound chosen from compounds having Formula A: and compounds having Formula B: prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and a therapeutically effective amount of at least one second compound chosen from compounds having Formula C: prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.

Description

METHODS FOR TREATING CANCER
The present application claims the benefit of priority under 35 U.S.C. §119 of U.S. Provisional Patent Application Nos. 62/355,410 filed June 28, 2016, 62/506,929 filed May 16, 2017 and 62/514,059 filed June 2, 2017; the contents of which are incorporated by reference herein in their entireties.
Disclosed herein are methods for treating cancer in a subject comprising administering a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors can be, for example, at least one compound chosen from the compounds having Formula A:
Figure imgf000003_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors can be, for example, at least one compound chosen from the compounds having Formula B:
Figure imgf000004_0001
(B)
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the at least one second compound chosen from kinase inhibitors can be, for example, at least one compound chosen from the compounds having Formula C:
Figure imgf000004_0002
(C)
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
The National Cancer Institute estimates 1,685,210 new cases of cancer will be diagnosed in the United States and 595,690 people will die from the disease in 2016. The most common cancers are projected to be breast cancer, lung and bronchus cancer, prostate cancer, colon and rectum cancer, bladder cancer, melanoma of the skin, non-Hodgkin lymphoma, thyroid cancer, kidney and renal pelvis cancer, leukemia, endometrial cancer, and pancreatic cancer. Despite advances in the treatment of certain forms of cancer through surgery, radiotherapy, and chemotherapy, many types of cancer are essentially incurable. Even when an effective treatment is available for a particular cancer, the side effects from the treatment can have a significant adverse impact on the patient’s quality of life. Most conventional chemotherapy agents have toxicity and limited efficacy, especially for patients with advanced solid tumors. Conventional chemotherapeutic agents cause cytotoxicity to both healthy noncancerous as well as cancerous cells. The therapeutic index of these chemotherapeutic compounds (i.e., a measure of the therapy’s ability to distinguish between cancerous and normal cells) can be quite low. Frequently, a dose of a chemotherapy drug that is effective at killing cancer cells will also kill normal cells, especially those normal cells (such as epithelial cells and cells of the bone marrow) that undergo frequent cell division. When normal cells are subject to chemotherapy, side effects such as hair loss, suppression of hematopoiesis causing anemia and immunodeficiency, and nausea often occur. Depending on the general health of the patient, such side effects can preclude the administration of chemotherapy all together, or, at least, inflict significant discomfort on cancer patients that diminishes their quality of life. Even for cancer patients who respond to chemotherapy with tumor regression, cancers often quickly relapse, progress, and spread by metastasis after the initial response to chemotherapy. Such recurrent cancers are often highly refractory to additional rounds of chemotherapy treatment.
Advanced hepatocarcinoma (HCC) remains a great clinical challenge because of the chemoresistant nature of the disease, the toxicity profile of currently available systemic chemotherapeutic agents, and the general poor health and underlying liver dysfunction of HCC patients. The Sorafenib Hepatocellular Carcinoma Assessment Protocol (SHARP) trial remains the only randomized controlled trial of a systemic chemotherapeutic agent to demonstrate a statistically significant survival benefit in patients with advanced, unresectable HCC. In that study, 602 patients were randomly assigned to receive sorafenib (400 mg twice daily) or placebo. A median Overall Survival (OS) of 10.7 months was observed in the treatment arm compared to 7.9 months for placebo. Time to Tumor Progression (TTP) for the sorafenib and placebo arms was 5.5 months and 2.8 months, respectively.
The benefits achieved with sorafenib monotherapy are important, but modest in magnitude. Many patients do not experience disease control, and disease control can be short-lived in those that do achieve it with therapy. Unfortunately, no other agent has proved to be more effective. Recent Phase III trials with other regimens (e.g., FOLFOX, brivanib, sunitinib and linifanib) have all failed to demonstrate a statistically significant improvement in OS when compared to treatment with sorafenib alone.
There have also been multiple studies of combination therapy using sorafenib plus an additional agent or regimen. Few combinations, however, have proved robust enough for further evaluation in the Phase III, pivotal setting. Those that have been evaluated in Phase III trials, such as Brivanib, FOLFOX, and Erlotinib, have failed to show superiority to sorafenib alone. In the BRISK-FL study, increased toxicity and a poorer quality of life were observed, as well as an increased incidence of hand-foot syndrome was observed in combination studies with 5-FU or 5- FU derivative therapies. An enormous unmet medical need exists for active systemic therapies in HCC.
Cancer stem cells (CSCs) or cancer cells with high stemness (stemness-high cancer cells) are believed to be responsible for the rapid tumor recurrence and resistance. CSCs are believed to possess at least the following four characteristics:
1. Stemness
As used herein, stemness means the capacity for a stem cell population to self-renew and transform into cancer cells (Gupta PB et al., Nat. Med.2009; 15(9):1010-1012). While CSCs form only a small percentage of the total cancer cell population in a tumor (Clarke MF, Biol. Blood Marrow Transplant. 2009; 11(2 suppl. 2):14-16), they give rise to heterogeneous lineages of differentiated cancer cells that make up the bulk of the tumor (see Gupta et al.2009). In addition, CSCs possess the capacity to spread to other sites in the body by metastasis where they seed the growth of the new tumors (Jordan CT et al. N. Engl. J. Med.2006; 355(12):1253-1261).
2. Aberrant signaling pathways
CSC stemness can also be associated with dysregulation of signaling pathways, which may contribute to their ability to metastasize. In normal stem cells, stemness signaling pathways are tightly controlled and genetically intact. In contrast, the aberrant regulation of stemness signaling pathways in CSCs plays a key role in the uncontrolled self-renewal of these cells and their transformation into cancer cells (see Ajani et al. Semin. Oncol. (2015) 42 Suppl. 1:S3-17). Dysregulation of stemness signaling pathways also contributes to CSC resistance to chemotherapy and radiotherapy and to cancer recurrence and metastasis. Exemplary stemness signaling pathways involved in the induction and maintenance of stemness properties in CSCs include, but are not limited to, Janus kinase/ signal transducers and activators of transcription (JAK/STAT), Hedgehog (Desert (DHH), Indian (IHH), and Sonic (SHH))/PATCHED/(PTCH1)/ SMOOTHENED (SMO), NOTCH/DELTA-LIKE (DLL1, DLL3, DLL4)/JAGGED (JAG1, JAG2)/ CSL (CBF1/Su(H)/Lag- 1), WNT/APC/GSK3/β-CATENIN/TCF4 and NANOG (Boman BM et al., J. Clin. Oncol.2008; 26(17):2828-2838).
3. Resistance to traditional therapies
Unfortunately, cancers that initially respond to chemotherapy and radiation treatment often relapse in a form that is resistant to these traditional therapies. While the detailed mechanism underlying such resistance is not well understood, aberrant regulation of CSC stemness signaling pathways (see Boman et al.2008) in the context of a tumor’s microenvironment (Borovski T. et al., Cancer Res.2011; 71(3):634-639) may play a key role in the acquisition of such resistance. 4. Ability to contribute to tumor recurrence and metastasis
Chemotherapy and radiation kills the majority of rapidly dividing cancer cells in a tumor but not CSCs that survive by acquiring resistance (see Jordan et al. 2006). Radiation / chemotherapy-resistant CSCs may also acquire the ability to metastasize to different sites in the body and maintain stemness at these locations through interactions with the microenvironment, thereby allowing for the spread of metastatic tumor growth (see Boman et al.2008). Interestingly, this enhanced tumorigenicity of CSCs correlates with the expression of genes normally expressed in adult stem cells, such as cell surface markers like CD44, CD133, and CD166.
Because the survival of CSCs may be the principal reason why cancers relapse after treatment with chemotherapy and/or radiation, anti-cancer therapies that specifically target CSC’s aberrant signaling pathways may help prevent tumor metastasis and provide a viable treatment option for patients with recurrent disease that is no longer treatable using traditional therapies. Such an approach may therefore improve the survival and quality of life of cancer patients, especially those patients suffering from metastatic disease. Unlocking this untapped potential involves the identification and validation of pathways that are essential for CSC self-renewal and survival. While many of the signaling pathways regulating embryonic or adult stem cell proliferation and differentiation are known, it remains to be seen if these same pathways are required for cancer stem cell self-renewal and survival.
The transcription factor Signal Transducer and Activator of Transcription 3 (also known as Acute-Phase Response Factor, APRF, DNA-Binding Protein APRF, ADMIO 3, HIES; referred to herein as STAT3) is a member of a family of seven transcription factors, STAT1 to STAT6, including STAT5a and STAT5b. STATs are activated either by receptor associated tyrosine kinases like Janus kinases (JAKs) or by receptors with intrinsic tyrosine kinase activity such as PDGFR, EGFR, FLT3, EGFR, ABL, KDR, c-MET, or HER2. Upon tyrosine phosphorylation by receptor associated kinases, the phosphorylated STAT protein (“pSTAT”) dimerizes, as a homo- or heterodimer, and translocates from the cytoplasm to the nucleus, where it binds to specific DNA-response elements in the promoters of target genes and induces gene expression. STAT 2, 4, & 6 regulate primarily immune responses, while STAT3, along with STAT1 and STAT5, regulate the expression of genes controlling cell cycle (CYCLIN D1, D2, and c-MYC), cell survival (BCL-XL, BCL-2, MCL-1), and angiogenesis (HIF1α, VEGF) (Furqan et al. Journal of Hematology & Oncology (2013) 6:90).
In normal cells, STAT3 activation is transient and tightly regulated, lasting for example, from about 30 minutes to several hours. However, in a wide variety of human cancers, including all of the major carcinomas as well as some hematologic tumors, STAT3 is found to be aberrantly active. Persistently active STAT3 is present in more than half of all breast and lung cancers as well as colorectal cancer (CRC), ovarian cancer, hepatocellular carcinoma, multiple myeloma and in more than 95% of all head/neck cancers. STAT3 therefore seems to play a pivotal role in cancer progression and may be one of the principal mechanisms by which cancer cells acquire drug resistance. STAT3 is a potent transcription regulator that targets genes involved in cell cycle, cell survival, oncogenesis, tumor invasion, and metastasis, including, but limited to, BCL-XL, c-MYC, CYCLIN D1, VEGF, MMP-2, and SURVIVIN. STAT3 is also a key negative regulator of tumor immune surveillance and immune cell recruitment. Thus, STAT3 may enable the survival and self- renewal capacity of CSCs across a broad spectrum of cancers. A pharmaceutical compound with activity against CSCs, for example, through STAT3 inhibition, holds great promise as a treatment option for cancer patients with advanced disease.
In certain embodiments, the at least one compound chosen from compounds having Formula A can be, for example, an inhibitor of CSC growth and survival. U.S. Patent No. 8,877,803 describes a compound having Formula A that inhibits STAT3 pathway activity with a cellular IC50 of ~0.25 µM. Example 13 in the‘803 patent provides exemplary methods of synthesizing at least one compound having Formula A. In certain embodiments, the at least one compound chosen from compounds having Formula A can be used, for example, in a method for treating cancers. In Example 6 of PCT Patent Application No. PCT/US2014/033566 the at least one compound chosen from compounds having Formula A was chosen to enter a clinical trial for patients with advanced cancers. The disclosures of U.S. Patent No. 8,877,803 and PCT Patent Application No. PCT/US2014/033566 are incorporated herein by reference in their entireties.
Protein kinases are a family of enzymes that regulate a wide variety of cellular processes, including cell growth, cell proliferation, cell differentiation, and metabolism. Protein kinases communicate cell growth signals through the sequential phosphorylation of pathway partners. Pharmacologic inhibition of any kinase on a given signal transduction cascade may therefore block communication along the entire pathway. In addition, it is known that protein kinases play a role in disease states and disorders, for example, a kinase mutation and/or overexpression of the kinase are frequently present in many cancers, resulting in hyper-activated activity that often correlates with uncontrolled cell growth. Thus, cancer stem cell pathway kinases (CSCPK) are important therapeutic targets for the treatment of a variety of cancers (see, for example, U.S. Patent No. 8,299,106). Non-limiting examples of CSCPKs include STK33, MELK, AXL, p70S6K, and PDGFRα. Activation of the CSCPK PDGFRα, a receptor tyrosine kinase (RTK), by a PDGF ligand triggers a signal transduction cascade that activates gene expression required for cell proliferation, angiogenesis, and apoptosis. PDGFRα is a class III receptor tyrosine kinase that is related to the CFS-1 receptor/c-fms and the stem cell growth factor/c-kit proto-oncogene family. The PDGFRα pathway which is active in early fetal development is also reactivated in many cancers including hepatocellular cancer (HCC), head and neck cancer, brain tumors, gastrointestinal tumors, skin cancer, prostate cancer, ovarian cancer, breast cancer, sarcoma, and leukemia. In addition, PDGFRα activation has recently been shown to play a key role in bone metastasis of prostate cancer. Specifically targeting PDGFRα using a monoclonal antibody leads to significant reduction in tumor cell proliferation and survival with minimal toxicity. Disruption of PDGFRα signaling may a viable target for the treatment of a broad spectrum of cancers with minimal toxicity.
The CSCPK STK33 or human serine/threonine kinase 33 is a member of the Ca2+/calmodulin-dependent kinase family (CAMK). Although its function in normal biology is not fully understood, STK33 may play a specific role in the dynamic behavior of the intermediate filament cytoskeleton through the phosphorylation of vimentin. STK33 knockdown decreases tumor-related gene expression and inhibits cell migration, invasion, and EMT, indicating that STK33 may be a mediator in signaling pathways that are involved in cancer. For example, STK33 has been implicated in the tumorigenesis and progression of hypopharyngeal squamous cell carcinoma (HSCC) (Huang et al. BMC Cancer. (2015)15:13).
Maternal Embryonic Leucine Zipper Kinase (or MELK) is a serine/threonine-CSCPK involved in cell cycle regulation, self-renewal of stem cells, apoptosis and splicing regulation. MELK has broad substrate specificity. For example, MELK acts as an activator of apoptosis by phosphorylating and activating MAP3K5/ASK1. MELK also regulates the cell cycle through phosphorylation of CDC25B which triggers the localization of CDC25B to the centrosome and the spindle poles during mitosis. MELK also phosphorylates and inhibits BCL2L14 indicating a possible role in mammary carcinogenesis by inhibiting the pro-apoptotic function of BCL2L14.
The CSCPK AXL Receptor Tyrosine Kinase (or AXL) is a member of the Tyro3-Axl-Mer (TAM) receptor tyrosine kinase subfamily. The encoded protein possesses an extracellular domain which is composed of two immunoglobulin-like (IgL) motifs at the N-terminus, followed by two fibronectin type-III motifs (FNIII). It transduces signals from the extracellular matrix into the cytoplasm by binding to the vitamin K-dependent protein growth arrest-specific 6 (Gas6). Downregulation of AXL suppresses brain tumor cell growth and invasion (Vajkoczy et al., Proc. Natl. Acad. Sci. U S A. 2006 11; 103(15):5799-804). AXL is therefore a potential mediator of motility and it may control the invasiveness of breast cancer cells (Zhang et al., Cancer Res. (2008) 15; 68(6):1905-15).
p70S6K (or p70 S6 Kinase) is a serine/threonine CSCPK that is a part of the PI3K/Akt/mTOR/P70S6K signaling pathway. Activation of p70S6K triggers S6 ribosomal protein phosphorylation and induces protein synthesis. p70 S6 kinase (p70S6K) is frequently activated in high-grade malignant human ovarian cancer indicating a possible role in metastasis.
In certain embodiments, the at least one compound chosen from compounds having Formula B can be, for example, an inhibitor of a CSCPK (for example, the compound BBI503). As disclosed in U.S. Patent No.8,299,106, the compounds of Formula B inhibit CSC proliferation and/or survival. Examples 1-5 of U.S. Patent No.8,299,106 further provide exemplary methods of synthesizing the at least one compound chosen from compounds having Formula B. U.S. Patent No.8,299,106 is incorporated herein by reference in its entirety.
The present disclosure reports on the surprising discovery that a treatment combination of at least one compound chosen from the compounds of Formula A and at least one compound chosen from the compounds of Formula C had a greater effect at reducing the number cancer cells, including cancer stem cells, than the added effects of each compound acting alone. For example, the treatment combination of the present disclosure resulted in an enhanced inhibition of the expression of cancer cell stemness-associated factors, both in vitro and in vivo, as well as an enhanced decline in the number of cancer stem cells, both in vitro and in vivo, as compared to the added effect observed after treatment with the compound of Formula A alone and after treatment with the compound of Formula C alone.
In one example, treatment of HCC cell lines with the combination of at least one compound chosen from the compounds of Formula A (e.g., BBI608) and at least one compound chosen from the compounds of Formula C (e.g., sorafenib) resulted in enhanced decrease in the number of bulk tumor cells (FIG.1) and a reduction in the number of spheres formed by CSCs after culture in non- adherent stem cell media (FIG.2). FIG.3 also shows that the volume of tumors in a HepG2 murine xenograft mouse model declined as a result of treatment with at least one compound of Formula A (e.g., BBI608) when compared to treatment with a control.
The present disclosure further reports on the surprising discovery that a treatment combination of at least one compound chosen from the compounds having Formula B and at least one compound chosen from the compounds having Formula C (sorafenib) had a greater effect at reducing the proliferation and/or survival of cancer cells, including cancer stem cells, than the added effects of either compound acting alone. For example, the treatment combination of the present disclosure resulted in an enhanced inhibition of the expression of cancer cell stemness- associated factors in vitro and in vivo, and an enhanced reduction in the number of cancer stem cells in vitro and in vivo, as compared to the added effects from the treatment of the tumors with the compound of Formula B (e.g. BBI503) alone and the compound of Formula C (e.g., sorafenib) alone.
In another example, a compound of Formula B was active against CSCs derived from HCC cell lines, both alone and in combination with a compound of Formula C (e.g., sorafenib). The compound of Formula B not only inhibited cancer stem cell proliferation and/or survival but also reduced the ability of CSCs to form characteristic spheres in non-adherent stem cell media. In contrast, the equivalent treatment with compound of Formula C did not have any significant inhibitory activity on CSC sphere-forming ability or on the proliferation and/or survival of cancer cells. In contrast, treatment of cancer cells with the combination of a compound of Formula B and a compound of Formula C enhanced the reduction in colony formation (FIG.4, FIG.5, and FIG. 6). The compound of Formula B, e.g. BBI503, was also shown to inhibit the expression of cancer stem cell markers, such as, CD133 and CD44.
In another example, treatment of tumors in a mouse xenograft model of human liver cancer (HepG2) with a compound of Formula B (e.g. BBI503) significantly inhibited tumor growth compared to controls. FIG. 7 shows that the average tumor volume in animals treated with a compound of Formula B was less than 50% of the average tumor volume obtained in controls.
Without being limited to any particular observation or hypothesis, the components of a treatment combination of the present disclosure are believed to work on different pathways that are associated with cancer cells (e.g., CSCs). The treatment combination of a compound of Formula A and a compound of Formula C or a compound of Formula B and a compound of Formula C results in an inhibition of cancer cell proliferation and/or survival that is greater than the added effects observed after treatment with either compound alone (sometimes referred to as "enhanced" or "synergistic" effects). A compound of Formula A can work by inhibiting the STAT3 signaling pathway. Specifically, a compound of Formula A can directly bind and inhibit the activity of activated STAT3 (e.g., phosphorylated STAT3), thereby preventing transcription of ST AT3 -dependent target genes including the sternness-associated transcription factors c-MYC, OCT4, SOX2, and β-CATENIN. A compound of Formula B can inhibit the activity of multiple malignancy-associated serine-threonine kinases (or cancer stem cell pathway kinases (CSCPKs)).
In certain embodiments, disclosed herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound chosen from compounds having Formula A:
Figure imgf000013_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and a therapeutically effective amount of at least one compound chosen from compounds having Formula C:
Figure imgf000013_0002
(C)
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, disclosed herein are methods for treating cancer comprising administering to a subject in need thereof:
a therapeutically effective amount of at least one compound chosen from compounds having Formula B:
Figure imgf000014_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and
a therapeutically effective amount of at least one compound chosen from compounds having Formula C:
Figure imgf000014_0002
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, methods of treating cancer are disclosed comprising administering to a subject in need thereof (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the cancer stemness inhibitor can be, for example, a STAT3 pathway inhibitor.
In certain embodiments, the cancer stemness inhibitor can be, for example, chosen from 2- (1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro-naphtho[2,3-b]furan-4,9- dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2-acetylnaphtho[2,3-b]furan-4,9-dione, 2-ethyl-naphtho[2,3-b]furan-4,9-dione, prodrugs of any of the foregoing, derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the cancer stemness inhibitor can be, for example, a cancer stem cell pathway kinase (CASPK) inhibitor. In certain embodiments, the cancer stemness inhibitor can be chosen, for example, from the following compounds having the Formulae of:
Figure imgf000015_0001
Figure imgf000016_0001
prodrugs of any of the foregoing, derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, a kit is disclosed that comprises (1) at least one first compound chosen from compounds having Formula A, compounds having Formula B, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
In certain embodiments, a kit is disclosed that comprises (1) at least one first compound chosen from compounds having Formula A, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
In certain embodiments, a kit is disclosed that comprises (1) at least one first compound chosen from compounds having Formula B, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, and (2) at least one second compound chosen from compounds having Formula C, prodrugs, derivatives, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
In certain embodiments, a kit is disclosed that comprises (1) at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (2) at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing, together with instructions for administration and/or use.
Aspects and embodiments of the present disclosure are set forth or will be readily apparent from the following detailed description. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only, and are not intended to be restrictive of the claims. BRIEF DESCRIPTION OF THE DRAWINGS FIG.1 illustrates the enhanced inhibition of HCC colony formation by a Hep3B cell line after treatment of the cells with a combination of a compound of Formula A and a compound of Formula C.
FIG.2 shows an example of the enhanced inhibition of Sk-Hep1, Huh7, and HepG2 CSC viability after treatment of the cells with a combination of a compound of Formula A and a compound of Formula C.
FIG.3 shows an example of the inhibition of tumor growth in a murine xenograft model of HCC after treatment with a compound of Formula A.
FIG.4 illustrates the enhanced inhibition of HCC colony formation by a Hep3B cell line after the treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
FIG. 5 illustrates the enhanced inhibition of HCC colony formation by a Hub7 cell line after treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
FIG.6 shows an example of the enhanced inhibition of Skhep1, Hep3B, and HepG2 CSC viability after treatment of the cells with a combination of a compound of Formula B and a compound of Formula C.
FIG.7 shows an example of the inhibition of tumor growth in a murine xenograft model of HCC after treatment with a compound of Formula B. FIG. 8A shows an assessment of patients with advanced HCC treated with napabucasin (BBI608, Compound A) and sorafenib (Compound C) evaluated according to RECIST 1.1 and modified RECIST (mRECIST). FIG. 8B shows an assessment of patients with advanced HCC treated with amcasertib (BBI503, Compound B) and sorafenib (Compound C) evaluated according to RECIST 1.1 and modified RECIST (mRECIST). The following are definitions of terms used in the present specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification individually or as part of another group, unless otherwise indicated.
When the term“about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below those numerical values. In general, the term“about” is used herein to modify a numerical value above and below the stated value by a variance of 20%, 10%, 5%, or 1%. In certain embodiments, the term“about” is used to modify a numerical value above and below the stated value by a variance of 10%. In certain embodiments, the term“about” is used to modify a numerical value above and below the stated value by a variance of 5%. In certain embodiments, the term“about” is used to modify a numerical value above and below the stated value by a variance of 1%.
When a range of values is listed herein, it is intended to encompass each value and sub- range within that range. For example,“1-5 mg” is intended to encompass 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 1-2 mg, 1-3 mg, 1-4 mg, 1-5 mg, 2-3 mg, 2-4 mg, 2-5 mg, 3-4 mg, 3-5 mg, and 4-5 mg.
The terms“administer,”“administering,” or“administration” are used herein in their broadest sense. These terms refer to any method of introducing to a subject a compound or pharmaceutical composition described herein and can include, for example, introducing the compound systemically, locally, or in situ to the subject. Thus, a compound of the present disclosure produced in a subject from a composition (whether or not it includes the compound) is encompassed by these terms. When these terms are used in connection with the term“systemic” or“systemically,” they generally refer to in vivo systemic absorption or accumulation of the compound or composition in the blood stream followed by distribution throughout the entire body.
The term“subject” generally refers to an organism to which a compound or pharmaceutical composition described herein can be administered. A subject can be a mammal or mammalian cell, including a human or human cell. The term also refers to an organism, which includes a cell or a donor or recipient of such cell. In certain embodiments, the term“subject” refers to any animal (e.g., a mammal), including, but not limited to humans, mammals and non-mammals, such as non- human primates, mice, rabbits, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, fish, nematode, and insects, which is to be the recipient of a compound or pharmaceutical composition described herein. Under some circumstances, the terms“subject” and“patient” are used interchangeably herein in reference to a human subject.
The terms“effective amount” and“therapeutically effective amount” refer to that amount of a compound or pharmaceutical composition described herein that is sufficient to effect the intended result including, but not limited to, disease treatment, as illustrated below. In certain embodiments, the“therapeutically effective amount” is the amount that is effective for detectable killing or inhibition of the growth or spread of cancer cells, the size or number of tumors, and/or other measure of the level, stage, progression and/or severity of the cancer. In certain embodiments, the“therapeutically effective amount” refers to the amount that is administered systemically, locally, or in situ (e.g., the amount of compound that is produced in situ in a subject). The therapeutically effective amount can vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of cell migration. The specific dose may vary depending on, for example, the weight of the subject, the particular pharmaceutical composition, subject and their age and existing health conditions or risk for health conditions, the dosing regimen to be followed, the severity of the disease, whether it is administered in combination with other agents, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
A“therapeutically effective amount” in reference to the treatment of a subject’s cancer, means an amount capable of invoking, for example, one or more of the following effects: (1) inhibition, to some extent, of cancer or tumor growth, including a decrease or cessation in the progression of the subject’s cancer; (2) reduction in the number of cancer or tumor cells; (3) reduction in tumor size; (4) inhibition, e.g., a decrease or a cessation, of cancer or tumor cell infiltration into peripheral organs; (5) inhibition, e.g., a decrease or a cessation, of metastasis; (6) enhancement of anti-tumor immune response, which may, but is not required to, result in the regression or rejection of the tumor, or (7) relief, to some extent, of one or more symptoms associated with the cancer or tumor. The therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual and the ability of one or more anti-cancer agents to elicit a desired response in the individual. A“therapeutically effective amount” can be, for example, an amount of a compound where any toxic or detrimental effects resulting from the administration of the compound are outweighed by the therapeutically beneficial effects.
As used herein, the terms“treatment,”“treating,”“ameliorating,” and“encouraging” are used interchangeably herein. These terms refer to an approach for obtaining beneficial or desired results including, but not limited to, a therapeutic benefit and/or prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject can still be afflicted with the underlying disorder. For prophylactic benefit, the pharmaceutical composition may be administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
The terms“combination,”“combinatorial,” or“combination treatment,” as used herein, mean the administration of at least two different agents (e.g., at least one first compound chosen from compounds having Formula A and compounds having Formula B and at least one second compound chosen from compounds having Formula C, as well as one or more additional agents) to treat a disorder, condition, or symptom, e.g., a cancer condition. Such combination/combination treatment may involve the administration of one agent before, during, and/or after the administration of a second agent. The first agent and the second agent can be administered concurrently, separately, or sequentially to a subject in separate pharmaceutical compositions. The first agent and the second agent may be administered to a subject by the same or different routes of administration. In certain embodiments, a treatment combination comprises a therapeutically effective amount of at least one first compound chosen from compounds having Formula A and compounds having Formula B and a therapeutically effective amount of at least one second compound chosen from compounds having Formula C.
For example, the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors can have different mechanisms of action. In certain embodiments, a combination treatment improves the prophylactic or therapeutic effect of the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors by functioning together to have an additive, synergistic, or enhanced effect. In certain embodiments, a combination treatment of the present disclosure reduces the side effects associated with the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors. The administrations of the at least one first compound chosen from cancer stemness inhibitors and the at least one second compound chosen from kinase inhibitors may be separated in time by up to several weeks, but more commonly within 48 hours, and most commonly within 24 hours.
As used herein, the term“prodrug” refers to a derivative of a parent drug molecule that requires biotransformation, either spontaneous or enzymatic, within the organism to release the active drug. For example, prodrugs are variations or derivatives of the compounds of Formula A, B or C that have groups cleavable under certain metabolic conditions, and which, when cleaved, become the compounds of Formula A, B or C respectively. Such prodrugs then are pharmaceutically inactive in vivo, until they become active as a result of, for example, solvolysis or enzymatic degradation under physiological conditions. Prodrug compounds, described herein, may be called single, double, triple, etc., depending on the number of biotransformation steps required to release the active drug within the organism, and the number of functionalities present in a precursor-type form.
Prodrug forms often offer advantages of solubility, tissue compatibility, or delayed release in the mammalian organism. Prodrugs commonly known in the art include, but are not limited to, well-known acid derivatives, such as, for example, esters prepared by reaction of the parent acids with a suitable alcohol, amides prepared by reaction of the parent acid compound with an amine, or basic groups reacted to form an acylated base derivative, etc. Other prodrug derivatives may also be combined with other features disclosed herein to enhance bioavailability. Those of skill in the art will appreciate that certain of the presently disclosed compounds having free amino, amido, hydroxy, or carboxylic groups can be converted into prodrugs. Prodrugs include compounds having an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy, or carboxylic acid groups of the presently disclosed compounds. The amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols as well as, for example, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine, and methionine sulfone. Prodrugs may also include compounds having a carbonate, carbamate, amide, or alkyl ester moiety covalently bonded to any of the above substituents disclosed herein.
The term“synergy,”“synergistic,”“synergistically,” or“enhanced” as used herein refers to an effect of interaction or combination of two or more components to produce a combined effect greater than the sum of their separate effects (or“additive effects”). A synergistic effect may be attained when the compounds are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient can be administered, for example, sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together. A synergistic anticancer effect denotes an anticancer effect which is greater than the predicted purely additive effects of the individual compounds of the combination administered separately.
As used herein, the term“cancer stemness inhibitor” means a molecule that can target, reduce, inhibit, interfere with, or modulate at least one of a plurality of pathways involved in cancer stemness or the expression (e.g., the production of a functional product, e.g., a protein) of at least one of a plurality of cancer stemness genes. The expression or the expressed proteins can be used as biomarkers of the corresponding cancer stemness genes. Examples of these biomarkers include, but are not limited to, β-CATENIN, NANOG, SMO, SOX2, STAT3, AXL, ATM, c-MYC, KLF4, SURVIVIN, or BMI-1. A cancer stemness inhibitor may alter cancer stem cell growth as well as heterogeneous cancer cell growth.
In certain embodiments, a cancer stemness inhibitor can be, for example, a small molecule that binds a protein encoded by a cancer stemness gene. In certain embodiments, a cancer stemness inhibitor can be, for example, a biologic, e.g., a recombinant binding protein or peptide (e.g. APTSTAT3; see Kim et al. Cancer Res. (2014) 74(8):2144-51) or nucleic acid (e.g. STAT3 aiRNA; see U.S. Patent No.9,328,345, the content of which is incorporated herein in its entirety), or conjugate thereof, that binds to a protein encoded by a cancer stemness gene. In certain embodiments, a cancer stemness inhibitor can be, for example, a cell. In certain embodiments, a cancer stemness inhibitor can be, for example, a STAT3 inhibitor (for example, that binds to and inhibits a biological activity of STAT3 (see Furtek et al., ACS Chem. Biol., 2016, 11 (2), pp 308– 318)). In certain embodiments, a cancer stemness inhibitor can be, for example,a CSCPK inhibitor.
In certain embodiments, a cancer stemness inhibitor can be, for example, at least one compound chosen from 2-(1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro- naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2- acetylnaphtho[2,3-b]furan-4,9-dione, or 2-ethyl-naphtho[2,3-b]furan-4,9-dione, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, a cancer stemness inhibitor can be, for example, at least one compound chosen, for example, from the following compounds having the Formulae of:
Figure imgf000024_0001
Figure imgf000025_0001
prodrugs of any of the foregoing, derivatives of any of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, a cancer stemness inhibitor of the present disclosure may be administered in an amount ranging from about 300 mg to about 700 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 700 mg to about 1200 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 800 mg to about 1100 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 850 mg to about 1050 mg. In certain embodiments, the cancer stemness inhibitor may be administered in an amount ranging from about 960 mg to about 1000 mg.
In certain embodiments, the total amount of the cancer stemness inhibitor can be administered once daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 480 mg daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 960 mg daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 1000 mg daily. In certain embodiments, the total amount of the cancer stemness inhibitor can be administered in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 240 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 480 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered in a dose of about 500 mg twice daily. In certain embodiments, the cancer stemness inhibitor can be administered orally.
In certain embodiments, a cancer stemness inhibitor can be, for example, at least one compound chosen from compounds having Formula A. As used herein, the terms“a compound having Formula A,”“compounds having Formula A,” and“Compound A,” which can be used interchangeably, each means a compound chosen from compounds having Formula A:
Figure imgf000027_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, prodrugs and derivatives of compounds having Formula A can be, for example, STAT3 inhibitors. Non-limiting examples of prodrugs of compounds having Formula A include, for example, the phosphoric ester and phosphoric diester described in U.S. pre-grant Publication No. 2012/0252763 as compound numbers 4011 and 4012 and also suitable compounds described in in U.S. Patent No. 9,150,530. Non-limiting examples of derivatives of compounds having Formula A include, for example, the derivatives disclosed in U.S. Patent No. 8,977,803. The disclosures of U.S. pre-grant Publication No. 2012/0252763 and U.S. Patent Nos. 9,150,530 and 8,977,803 are incorporated herein by reference in their entireties.
Compounds having Formula A, shown below,
Figure imgf000027_0002
may also be known as 2-acetylnaphtho[2,3-b]furan-4,9-dione, napabucasin, BBI608, or BB608, and include tautomers thereof.
Suitable methods of preparing 2-acetylnaphtho[2,3-b]furan-4,9-dione, including its crystalline forms, and additional cancer stemness inhibitors are described in the co-owned PCT applications published as WO 2009/036099, WO 2009/036101, WO 2011/116398, WO 2011/116399, and WO 2014/169078; and the contents of each of these applications are incorporated herein by reference in their entireties.
In certain embodiments, Compound A may be administered in an amount ranging from about 80 mg to about 1500 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 160 mg to about 1000 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 300 mg to about 700 mg a day. In certain embodiments, Compound A may be administered in an amount ranging from about 700 mg to about 1200 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 800 mg to about 1100 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 850 mg to about 1050 mg. In certain embodiments, Compound A may be administered in an amount ranging from about 960 mg to about 1000 mg. In certain embodiments, the total amount of Compound A can be administered, for example, once daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 480 mg daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 960 mg daily. In certain embodiments, Compound A can be administered, for example, in a dose of about 1000 mg daily. In certain embodiments, the total amount of Compound A can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound A may be administered in an amount ranging from about 80 mg twice daily to about 750 mg twice daily. In certain embodiments, Compound A may be administered in an amount ranging from about 80 mg twice daily to about 500 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 240 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 480 mg twice daily. In certain embodiments, Compound A may be administered, for example, in a dose of about 500 mg twice daily. In certain embodiments, Compound A may be administered, for example, orally.
In certain embodiments, a cancer stemness inhibitor can be, for example, at least one compound chosen from compounds having Formula B. As used herein, the terms“a compound having Formula B,”“compounds having Formula B,” and“Compound B,” which can be used interchangeably, each means a compound chosen from compounds having Formula B:
Figure imgf000029_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, a cancer stemness inhibitor can be, for example, a cancer stem cell pathway kinase (CSCPK) inhibitor, a STK33 inhibitor, a MELK inhibitor, an AXL inhibitor, a p70S6K inhibitor, a PDGFRα inhibitor, or a NANOG inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example,a cancer stem cell pathway kinase (CSCPK) inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a STK33 inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a MELK inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, an AXL inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a p70S6K inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a PDGFRα inhibitor. In certain embodiments, a cancer stemness inhibitor can be, for example, a NANOG inhibitor.
In certain embodiments, compounds having Formula B, and derivatives thereof, can be, for example, cancer stem cell pathway kinase (CSCPK) inhibitors. In certain embodiments, compounds having Formula B, and derivatives thereof, can include inhibitors of, for example, STK33, MELK, AXL, p70S6K, and PDGFRα. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example,STK33 inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example, MELK inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example, AXL inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example, p70S6K inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can be, for example, PDGFRα inhibitors. In certain embodiments, the at least one compound chosen from compounds having Formula B, and derivatives thereof, can inhibit NANOG expression. Non-limiting examples of compounds having Formula B, and derivatives thereof, can include, for example, the derivatives disclosed in U.S. Patent No. 8,299,106 and PCT Patent Application Publication No. WO2014160401. The disclosures of U.S. Patent No. 8,299,106 and PCT Patent Application Publication No. WO2014160401 are incorporated herein by reference in their entireties.
In certain embodiments, the CSCPK inhibitor can be chosen, for example, from the following compounds having the Formulae of:
Figure imgf000030_0001
Figure imgf000031_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
In certain embodiments, the CSCPK inhibitor can be, for example, at least one compound chosen from compounds having Formula B.
Suitable methods of preparing compounds having Formula B, and derivatives thereof, are described in U.S. Patent No. 8,299,106 and PCT Patent Application Publication No. WO2014160401; the contents of each application are incorporated herein by reference in their entireties.
In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 20 mg to about 600 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 50 mg to about 500 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 80 mg to about 400 mg. In certain embodiments, the CSCPK inhibitor may be administered in an amount ranging from about 80 mg to about 300 mg. In certain embodiments, the CSCPK inhibitor can be administered, for example, once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 100 mg daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 200 mg daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 300 mg daily. In certain embodiments, the total amount of the CSCPK inhibitor can be administered, for example, in a single daily dose. In certain embodiments, the total amount of the CSCPK inhibitor can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 100 mg once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, in a dose of about 200 mg once daily. In certain embodiments, the CSCPK inhibitor can be administered, for example, orally.
In certain embodiments, Compound B may be administered in an amount ranging from about 20 mg to about 600 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 50 mg to about 500 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 80 mg to about 400 mg. In certain embodiments, Compound B may be administered in an amount ranging from about 80 mg to about 300 mg. In certain embodiments, Compound B can be administered, for example, once daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 100 mg daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 200 mg daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 300 mg daily. In certain embodiments, the total amount of Compound B can be administered, for example, in a single daily dose. In certain embodiments, the total amount of Compound B can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound B can be administered, for example, in a dose of about 100 mg once daily. In certain embodiments, Compound B can be administered, for example, in a dose of about 200 mg once daily. In certain embodiments, Compound B can be administered, for example, orally. As used herein, the term“kinase inhibitor” or“kinase inhibitors” refers to a molecule that can target, reduce, inhibit, interfere with, or modulate the activities of at least one of a plurality of protein kinases. Examples of these protein kinases include, but are not limited to, intracellular kinases (e.g., c-CRAF, BRAF, and mutant BRAF) and cell surface kinases (e.g., KIT, FLT-3, RET, RET/PTC, VEGFR-1, VEGFR-2, VEGFR-3, and PDGFR-ß). Examples of kinase inhibitors include, but are not limited to, afatinib (an EGFR/ErbB2 inhibitor), axitinib, (a VEGFR1/VEGFR2/VEGFR3/PDGFRB/c-KIT inhibitor), bosutinib (a Bcr-Abl/SRC inhibitor), cetuximab (an EGFR inhibitor), cobimetinib (a MEK inhibitor), crizotinib (an ALK/Met inhibitor), cabozantinib (a RET/MET/VEGFR2 inhibitor), dasatinib (a multiple kinase inhibitor), entrectinib (a TrkA/TrkB/TrkC/ROS1/ALK inhibitor), erlotinib (an EGFR inhibitor), fostamatinib (a Syk inhibitor), gefitinib (an EGFR inhibitor), ibrutinib (a BTK inhibitor), imatinib (a Bcr-Abl inhibitor), lapatinib (an EGFR/ErbB2 inhibitor), lenvatinib (a VEGFR2 inhibitor), mubritinib, nilotinib (a Bcr-Abl inhibitor), pazopanib (a VEGFR2/PDGFR/c-kit inhibitor), pegaptanib (a VEGF inhibitor), ruxolitinib (a JAK inhibitor), sorafenib (a multiple kinase inhibitor), sunitinib (a multiple kinase inhibitor), SU6656 (a Src inhibitor), vandetanib (a RET/VEGFR/EGFR inhibitor), or vemurafenib (a BRAF inhibitor).
In certain embodiments, a kinase inhibitor can be, for example, at least one compound chosen from compounds having Formula C. As used herein, the terms“a compound having Formula C,”“compounds having Formula C,” and“Compound C,” which can be used interchangeably, each means a compound chosen from compounds having Formula C:
Figure imgf000033_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. Compound C may also be known as 4-[4-[[4-chloro-3- (trifluoromethyl)phenyl]carbamoylamino]phenoxy]-N-methyl-pyridine-2-carboxamide, Nexavar, or sorafenib.
In certain embodiments, Compound C may be administered in an amount ranging from about 80 mg to about 1500 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 100 mg to about 1200 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 200 mg to about 1000 mg a day. In certain embodiments, Compound C may be administered in an amount ranging from about 400 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 600 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount ranging from about 700 mg to about 900 mg. In certain embodiments, Compound C may be administered in an amount of about 800 mg. In certain embodiments, the total amount of Compound C can be administered, for example, once daily. In certain embodiments, the total amount of Compound C can be administered, for example, in divided doses more than once daily, such as twice daily (BID) or more often. In certain embodiments, Compound C may be administered in an amount ranging from about 80 mg twice daily to about 750 mg twice daily. In certain embodiments, Compound C may be administered in an amount ranging from about 80 mg twice daily to about 600 mg twice daily. In certain embodiments, Compound C can be administered, for example, in a dose of about 200 mg twice daily. In certain embodiments, Compound C can be administered, for example, in a dose of about 400 mg twice daily. In certain embodiments, Compound C can be administered, for example, orally. In certain embodiments, Compound C can be administered, for example, on an empty stomach. In certain embodiments, Compound C can be administered, for example, either about 1 hour prior to a meal or about 2 hours after a meal.
The terms“progress,”“progressed,” and“progression,” as used herein, refer to at least one of the following: (1) a response to prior therapy (e.g., chemotherapy) of progressive disease (PD); (2) the appearance of one or more new lesions after treatment with prior therapy (e.g., chemotherapy); and (3) at least a 5% (e.g., 10%, 20%) increase in the sum of diameters of target lesions, taking as a reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). As used herein,“sensitizing” can mean making subjects who were previously resistant, non-responsive, or somewhat responsive to a therapy (e.g., chemotherapy) regimen sensitive, responsive, or more responsive to that therapy (e.g., chemotherapy) regimen.
The term“cancer” in a subject refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain morphological features. Often, cancer cells can be in the form of a tumor or mass, but such cells may exist alone within a subject, or may circulate in the blood stream as independent cells, such as leukemic or lymphoma cells. Examples of cancer, as used herein, include, but are not limited to, lung cancer, pancreatic cancer, bone cancer, skin cancer, head or neck cancer, cutaneous or intraocular melanoma, breast cancer, uterine cancer, ovarian cancer, peritoneal cancer, colon cancer, rectal cancer, colorectal adenocarcinoma, cancer of the anal region, stomach cancer, gastric cancer, gastrointestinal cancer, gastric adenocarcinoma, adrenocorticoid carcinoma, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, chondrosarcoma, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, Ewing’s sarcoma, cancer of the urethra, cancer of the penis, prostate cancer, bladder cancer, testicular cancer, cancer of the ureter, carcinoma of the renal pelvis, mesothelioma, hepatocellular cancer, biliary cancer, kidney cancer, renal cell carcinoma, chronic or acute leukemia, lymphocytic lymphomas, neoplasms of the central nervous system (CNS), spinal axis tumors, brain stem glioma, glioblastoma multiforme, astrocytomas, schwannomas, ependymomas, medulloblastomas, meningiomas, squamous cell carcinomas, pituitary adenomas, including refractory versions of any of the above cancers, or a combination of one or more of the above cancers. Some of the exemplified cancers are included in general terms and are included in this term. For example, urological cancer, a general term, can include bladder cancer, prostate cancer, kidney cancer, testicular cancer, and the like; and hepatobiliary cancer, another general term, includes liver cancers (itself a general term that includes hepatocellular carcinoma or cholangiocarcinoma), gallbladder cancer, biliary cancer, or pancreatic cancer. Both urological cancer and hepatobiliary cancer are contemplated by the present disclosure and included in the term“cancer.”
As used herein,“cancer” can include the term,“solid tumor.” As used herein, the term “solid tumor” refers to those conditions, such as cancer, that form an abnormal tumor mass, such as sarcomas, carcinomas, and lymphomas. Examples of solid tumors include, but are not limited to, non-small cell lung cancer (NSCLC), neuroendocrine tumors, thyomas, fibrous tumors, metastatic colorectal cancer (mCRC), and the like. In certain embodiments, the solid tumor disease can be, for example, an adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and the like.
In certain embodiments, the cancer can be, for example, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, gastric cancer, chondrosarcoma, colorectal adenocarcinoma, breast cancer, ovarian cancer, head and neck cancer, melanoma, gastric adenocarcinoma, lung cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, cervical cancer, brain tumor, multiple myeloma, leukemia, lymphoma, prostate cancer, cholangiocarcinoma, endometrial cancer, small bowel adenocarcinoma, uterine sarcoma, or adrenocorticoid carcinoma. In certain embodiments, the cancer can be, for example, esophageal cancer, gastroesophageal junction cancer, gastroesophageal adenocarcinoma, colorectal adenocarcinoma, breast cancer, ovarian cancer, head and neck cancer, melanoma, gastric adenocarcinoma, lung cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, cervical cancer, brain tumor, multiple myeloma, leukemia, lymphoma, prostate cancer, cholangiocarcinoma, endometrial cancer, small bowel adenocarcinoma, uterine sarcoma, or adrenocorticoid carcinoma. In certain embodiments, the cancer can be, for example, breast cancer. In certain embodiments, the cancer can be, for example, colorectal adenocarcinoma. In certain embodiments, the cancer can be, for example, small bowel adenocarcinoma. In certain embodiments, the cancer can be, for example, hepatocellular carcinoma. In certain embodiments, the cancer can be, for example, head and neck cancer. In certain embodiments, the cancer can be, for example, renal cell carcinoma. In certain embodiments, the cancer can be, for example, ovarian cancer. In certain embodiments, the cancer can be, for example, prostate cancer. In certain embodiments, the cancer can be, for example, lung cancer. In certain embodiments, the cancer can be, for example, uterine sarcoma. In certain embodiments, the cancer can be, for example, esophageal cancer. In certain embodiments, the cancer can be, for example, endometrial cancer. In certain embodiments, the cancer can be, for example, cholangiocarcinoma. In certain embodiments, each of the cancers can be, for example, unresectable, advanced, refractory, recurrent, or metastatic.
The term“salt(s),” as used herein, includes acidic and/or basic salts formed with inorganic and/or organic acids and bases. As used herein, the term“pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response and/or the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19.
Pharmaceutically acceptable salts may be formed with inorganic or organic acids. Non- limiting examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid. Non-limiting examples of suitable organic acids include acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, and malonic acid. Other non-limiting examples of suitable pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, besylate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, and valerate salts. In certain embodiments, organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, lactic acid, trifluoracetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicylic acid. Salts may be prepared in situ during the isolation and purification of the disclosed compound, or separately, such as by reacting the compound with a suitable base or acid, respectively. Non-limiting examples of pharmaceutically acceptable salts derived from bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4alkyl)4 salts. Non-limiting examples of suitable alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts. Further non-limiting examples of suitable pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate. Non- limiting examples of suitable organic bases from which salts may be derived include primary amines, secondary amines, tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In certain embodiments, pharmaceutically acceptable base addition salts can be chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
The term“solvate” represents an aggregate that comprises one or more molecules of a compound of the present disclosure with one or more molecules of a solvent or solvents. Solvates of the compounds of the present disclosure include, for example, hydrates.
The at least one compound disclosed herein may be in the form of a pharmaceutical composition. In certain embodiments, the pharmaceutical compositions may comprise the at least one compound having Formula A and at least one pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical compositions may comprise the at least one compound having Formula B and at least one pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical compositions may comprise the at least one compound having Formula C and at least one pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula A in a subject (i.e., a prodrug). In certain embodiments, the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula B in a subject (i.e., a prodrug). In certain embodiments, the pharmaceutical compositions may comprise one or more compounds and at least one pharmaceutically acceptable carrier, where the one or more compounds are capable of being converted into the at least one compound having Formula C in a subject (i.e., a prodrug).
The term“carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as, for example, a liquid or solid filler, diluent, excipient, solvent or encapsulating material involved in or capable of carrying or transporting the subject pharmaceutical compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the Formulation and not injurious to the patient. Non-limiting examples of pharmaceutically acceptable carriers, carriers, and/or diluents include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical Formulations. Wetting agents, emulsifiers, and lubricants, such as sodium lauryl sulfate, magnesium stearate, and polyethylene oxide-polypropylene oxide copolymer as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives, and antioxidants can also be present in the compositions.
Pharmaceutical compositions disclosed herein that are suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, a solution in an aqueous or non-aqueous liquid, a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, a water-in-oil emulsion, an elixir, a syrup, pastilles (using an inert base, such as gelatin, glycerin, sucrose, and/or acacia) and/or mouthwashes, each containing a predetermined amount of the at least one compound of the present disclosure.
A pharmaceutical composition disclosed herein may be administered as a bolus, electuary, or paste.
Solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like) may be mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium carbonate, and sodium starch glycolate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and polyethylene oxide-polypropylene oxide copolymer; absorbents, such as kaolin and bentonite clay; lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type also may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof. Additionally, cyclodextrins, e.g., hydroxypropyl-β-cyclodextrin, may be used to solubilize compounds.
The pharmaceutical compositions also may include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents. Suspensions, in addition to the compounds according to the disclosure, may contain suspending agents as, such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar, and tragacanth, and mixtures thereof.
Pharmaceutical compositions disclosed herein, for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds according to the present disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the compounds of the present disclosure. Pharmaceutical compositions which are suitable for vaginal administration also may include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing carriers that are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a pharmaceutical composition or pharmaceutical tablet of the present disclosure may include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants. The pharmaceutical composition or pharmaceutical tablet may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to the pharmaceutical composition or pharmaceutical tablet of the present disclosure, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
Powders and sprays may contain, in addition to a pharmaceutical composition or a pharmaceutical tablet of the present disclosure, excipients such as lactose, talc, silicic acid, aluminium hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances. Additionally, sprays may contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of the present disclosure.
Compositions suitable for parenteral administration may comprise at least one more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions, emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
In certain embodiments, a composition described herein may include at least one compound chosen from compounds of Formula A and pharmaceutically acceptable salts and solvates thereof and one or more surfactants. In certain embodiments, the surfactant can be, for example, sodium lauryl sulfate (SLS), sodium dodecyl sulfate (SDS), or one or more polyoxylglycerides. For example, the polyoxylglyceride can be lauroyl polyoxylglycerides (sometimes referred to as GelucireTM) or linoleoyl polyoxylglycerides (sometimes referred to as LabrafilTM). Examples of such compositions are shown in PCT Patent Application No. PCT/US2014/033566, the contents of which are incorporated herein in their entireties.
The present invention provides further embodiments of suitable pharmaceutical formulations having selected particle size distribution and methods for identifying an optimum particle size distribution, suitable drug regimen, dosage and interval, suitable methods of preparing 2-acetylnaphtho[2,3-b]furan-4,9-dione including their crystalline forms, and further specific suitable cancer stemness inhibitors and kinase inhibitors as described in the co-owned PCT applications published as WO 2009/036099, WO 2009/036101, WO 2011/116398, WO 2011/116399, WO 2014/169078, and WO 2009/033033, the contents of which are incorporated by reference herein in their entirety. In certain embodiments, a composition described herein may include at least one compound chosen from compounds of Formula B and pharmaceutically acceptable salts and solvates thereof and one or more surfactants. In certain embodiments, the surfactant can be, for example, sodium lauryl sulfate (SLS), sodium dodecyl sulfate (SDS), or one or more polyoxylglycerides. For example, the polyoxylglyceride can be lauroyl polyoxylglycerides (sometimes referred to as GelucireTM) or linoleoyl polyoxylglycerides (sometimes referred to as LabrafilTM).
In certain embodiments, the compounds or pharmaceutical compositions described herein may be administered in combination with any of a variety of known therapeutics, including for example, chemotherapeutic and other anti-neoplastic agents, anti-inflammatory compounds, and/or immunosuppressive compounds. In certain embodiments, the compounds, products, and/or pharmaceutical compositions described herein are useful in conjunction with any of a variety of known treatments including, by way of non-limiting example, surgical treatments and methods, radiation therapy, chemotherapy, and/or hormone or other endocrine-related treatment.
In certain embodiments, methods of treating cancer in a subject in need thereof are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, methods of treating cancer cells are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the cancer cells are in a subject. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
In certain embodiments, methods of inhibiting, reducing, and/or diminishing cancer stem cell’s survival and/or self-renewal are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the cancer stem cells are in a subject. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, methods of treating cancer that is refractory to conventional chemotherapies and/or targeted therapies in a subject are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, the conventional chemotherapies and/or targeted therapies may comprise administering at least one compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutical acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the conventional chemotherapies and/or target therapies may comprise administering sorafenib.
In certain embodiments, methods of treating recurrent cancer in a subject that has failed surgery, oncology therapy (e.g., chemotherapy), and/or radiation therapy are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
In certain embodiments, methods of treating or preventing cancer metastasis in a subject are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
In certain embodiments, methods of preventing relapse or suppressing regrowth or recurrent of cancer in a subject are disclosed herein, comprising administering (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, the method is a part of an adjuvant therapy. In certain embodiments, the method comprises administering a treatment combination of the present disclosure after or currently with a primary treatment of cancer. In certain embodiments, the primary treatment is chosen from chemotherapies, radiation therapies, hormone therapies, targeted therapies, or biological therapies.
In certain embodiments, methods of sensitizing or resensitizing cells to at least one kinase inhibitor are disclosed herein comprising administering a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one kinase inhibitor is sorafenib. In certain embodiments, the methods comprise administering a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. In certain embodiments, the at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition. In certain embodiments, at least one second compound chosen from kinase inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing may be included in a pharmaceutical composition.
In certain embodiments, the cancer can be, for example, hepatocellular carcinoma. In certain embodiments, the cancer can be, for example, cholangiocarcinoma.
In certain embodiments, the cancer may be unresectable. In certain embodiments, the cancer may be advanced. In certain embodiments, the cancer may be refractory. In certain embodiments, the cancer may be recurrent. In certain embodiments, the cancer may be metastatic. In certain embodiments, the cancer may be associated with overexpression of STAT3. In certain embodiments, the cancer may be associated with nuclear β-catenin localization. EXAMPLES Examples are provided below to further illustrate different features of the present invention. The examples also illustrate useful methodology for practicing the invention. These examples do not limit the claimed invention.
The methods disclosed herein comprise administering to a subject in need thereof a therapeutically effective amount of at least one compound chosen from cancer stemness inhibitors in combination with a therapeutically effective amount of at least one compound chosen from kinase inhibitors.
EXAMPLE 1: Treatment with the combination of Compound A (BBI608) and Compound C
(sorafenib) enhanced the inhibition of bulk cancer cell colony formation in vitro The ability of bulk cancer cells to undergo clonogenic expansion after treatment with the combination of Compound A and Compound C was examined using a colony formation assay. For these studies, human HCC cell lines (HepG2, 1000 cells per well) were seeded in 6-well plates. 24 hours after seeding, the cells were treated with either DMSO (as control), Compound A alone (0.15 µM), Compound C alone (2 µM), or with the combination of Compound A (0.15 µM) and Compound C (2 µM). Cells were then cultured for 7-10 days until visible colonies formed.
As shown in FIG. 1, treatment with the combination (“combo”) of Compound A and Compound C resulted in an enhanced inhibition of HCC colony formation in comparison to treatment with Compound A (BBI608) alone or with Compound C (sorafenib) alone.
EXAMPLE 2: Treatment with the combination of Compound A (BBI608) and Compound C
(sorafenib) enhanced the inhibition of cancer stem cell sphere formation in vitro Cancer stem cell sphere formation (i.e. spherogenesis) was examined after treatment of cancer cells with the combination (“combo) of Compound A (BBI608) and Compound C (sorafenib). Single Sk-Hep1, Huh7, and HepG2 cells were grown in suspension for 48 h before being cultured for 24 hours in the presence of either Compound A alone (BBI608; 1.6 µM), Compound C alone (sorafenib; 2 µM), or with the combination of Compound A (BBI608) and Compound C (sorafenib) at a concentrations of 1.6 µM and 2 µM, respectively. The cells were then washed out and cultured in standard cell culture medium for an additional 24 hours. The cells were then examined and photographed using a Differential interference contrast (DIC) microscopy. Blue coloration of cells is due to Tryphan Blue and indicates the presence of dead cells.
As shown in FIG.2, treatment with the combination (“combo”) of Compound A (BBI608) and Compound C (sorafenib) resulted in a significant increase in the inhibition of Sk-Hep1, Huh7, and HepG2 CSC viability as compared to treatment with Compound A (BBI608) alone or with Compound C (sorafenib) alone.
In summary, the studies described in Examples 1 and 2 demonstrated that Compound A and Compound C act synergistically in vitro, and these data suggest significant potential for combined therapy using Compound A and Compound C for HCC.
EXAMPLE 3: Treatment of a murine xenograft model of human cancer with Compound A
(BBI608)
In the mouse xenograft model of human HCC, HepG2 cells were inoculated subcutaneously into male athymic nude mice (8 x 106 cells/mouse) and allowed to form palpable tumors. Once the tumors reached approximately 500 mm3, the animals were treated orally with either vehicle (control), or Compound A (BBI608; 10 mg/kg, i.v.) as indicated in FIG. 3. The animals received a total of 5 doses. Treatment with Compound A (BBI608) inhibited tumor growth as compared to control animals.
EXAMPLE 4: Treatment with the combination of Compound B (BBI503) and Compound C
(sorafenib) enhanced the inhibition of bulk cancer cell colony formation in vitro The ability of bulk cancer cells to undergo clonogenic expansion after treatment with the combination of Compound B (BBI503) and Compound C (sorafinib) was examined using a colony formation assay. For these studies, human HCC cell lines (Hep3B and Hub7, 1000 cells per well) were seeded in 6-well plates.24 hours after seeding, the cells were treated with either DMSO (as control), Compound B alone (0.3 µM), Compound C alone (2 µM), or with the combination of Compound B (0.3 µM) and Compound C (2 µM). Cells were then cultured for 7-10 days until visible colonies formed.
As shown in FIG. 4 and FIG. 5, the treatment with the combination of Compound B (BBI503) and Compound C (sorafinib) resulted in an enhanced inhibition of HCC colony formation in comparison to treatment with Compound A (BBI503) alone or with Compound C (sorafenib) alone.
EXAMPLE 5: Treatment with the combination of Compound B (BBI503) and Compound C
(sorafenib) enhanced the inhibition of cancer stem cell sphere formation in vitro Cancer stem cell sphere formation (i.e. spherogenesis) was examined after treatment of cancer cells with the combination (“combo) of Compound B (BBI503) and Compound C (sorafenib). Single Sk-Hep1, Huh7, and HepG2 cells were grown in suspension for 48 h before being cultured for 24 hours in the presence of either Compound B alone (BBI503; 1 µM), Compound C alone (sorafenib ; 2 µM), or with the combination of Compound A (BBI608) and Compound C (sorafenib) at a concentrations of 1 µM and 2 µM, respectively. The cells were then washed out and cultured in standard cell culture medium for an additional 24 hours. The cells were then examined and photographed using a Differential interference contrast (DIC) microscopy. Blue coloration of cells is due to Tryphan Blue and indicates the presence of dead cells.
As shown in FIG.6, treatment with the combination (“combo”) of Compound B (BBI503) and Compound C (sorafenib) resulted in a significant increase in the inhibition of Sk-Hep1, Huh7, and HepG2 CSC viability as compared to treatment with Compound B (BBI503) alone or with Compound C (sorafenib) alone.
In summary, the studies described in Examples 4 and 5 demonstrated that Compound B and Compound C act synergistically in vitro, and these data suggest significant potential for combined therapy using Compound A and Compound C for HCC.
EXAMPLE 6: Treatment of a murine xenograft model of human cancer with Compound B
(BBI503) In the mouse xenograft model of human HCC, HepG2 cells were inoculated subcutaneously into male athymic nude mice (8 x 106 cells/mouse) and allowed to form palpable tumors. Once the tumors reached approximately 500 mm3, the animals were treated orally with either vehicle (control), or Compound B (BBI503; 100 mg/kg, p.o.) as indicated in FIG. 7. The animals received a total of 5 doses. Treatment with Compound B (BBI503) inhibited tumor growth as compared to control animals.
EXAMPLE 7: Clinical trials with treatment combinations of Compound A (BBI608) and
Compound C (sorafenib) or Compound B (BBI503) and Compound C (sorafenib) The effects of two treatment combinations in patients with advanced hepatocellular carcinoma were studied in a Phase IB/II study to assess the safety, tolerability, and preliminary anti-cancer activity of the drug combinations disclosed herein. The first treatment combination included Compound A (BBI608) and Compound C (sorafenib) and the second treatment combination included Compound B (BBI503) and Compound C (sorafenib).
This open label, Phase IB/II study determined the safety and efficacy of the aforementioned drug combinations on a population of adult patients with advanced hepatocellular carcinoma who had not received systemic chemotherapy. The Phase IB portion involved dose-escalation of Compound A (BBI608) administered in combination with a fixed starting dose of Compound C (sorafenib) (Arm 1), and dose escalation of Compound B (BBI503) administered in combination with a fixed starting dose of Compound C (sorafenib) (Arm 2). The fixed starting dose-level of sorafenib for both arms was 400 mg twice daily (800 mg total daily dose). Eligible patients were randomized to either Arm 1 or Arm 2.
In Arm 1, escalating doses of Compound A (BBI608) were administered to cohorts of three to six patients until recommended Phase II dose (RP2D) was determined according to the established criteria for determining dose-limiting toxicity (DLT) and dose-escalation.
In Arm 2, escalating doses of Compound B (BBI503) were administered to cohorts of three to six patients until recommended Phase II dose (RP2D) was determined according to the established criteria for determining dose-limiting toxicity (DLT) and dose-escalation. Prior to initiation of combination therapy in each arm, Compound C (sorafenib) was administered as monotherapy starting on Cycle 1, Day 1 for 14 days. Dose-adjustment of sorafenib according to the approved product label was allowed. Following the sorafenib run-in period, the combination regimen began on Cycle 1, Day 15. Protocol therapy continued in repeating 28-day cycles until disease progression, unacceptable toxicity, or another discontinuation criterion was met.
For both arms, pharmacokinetic (PK) studies were performed on Cycle 1, Day 15 and Cycle 2, Day 15. An additional pharmacokinetic study was also performed as necessary to confirm exposure following dose-modification. Once recommended Phase II dose (RP2D) was established for both study arms, the Phase II portion began.
The Phase II portion was an open-label, 3-arm, randomized Phase II trial of patients with advanced HCC who had not received prior systemic treatment. Patients were randomized to receive either, Arm 1: Compound C (sorafenib) administered in combination with Compound A (BBI608) (at the RP2D determined for BBI608 plus sorafenib during the Phase IB portion); Arm 2: Compound C (sorafenib) in combination with Compound B (BBI503) (at the RP2D determined for BBI503 plus sorafenib during the Phase IB portion), or Arm 3: Compound C (sorafenib) alone at a starting dose of 400 mg twice daily. The starting dose for sorafenib was the same for all study arms.
A total of approximately 90 patients were enrolled in the Phase II portion of the study, or approximately 30 patients in each study arm.
Pharmacodynamic assessments were performed in patients with readily accessible tumors through an optional on-study tumor biopsy. Archival tissue, if available, was collected from all patients.
Throughout the study, safety and tolerability of Compound A (BBI608) in combination with Compound C (sorafenib) and of Compound B (BBI503) in combination with Compound C (sorafenib) were continually assessed for the duration of study treatment and up to 30 days after discontinuation of the study drug (either Compound A or Compound B). Evaluation of anti-tumor activity was performed at regular 8-week intervals, with the first assessment 8 weeks (56-days) after Cycle 1, Day 1. The radiologic assessment was evaluated according to RECIST 1.1 and modified RECIST (mRECIST) for patients with HCC. Alpha- fetoprotein (AFP) measurement was performed at baseline, at the end of the 2-week sorafenib monotherapy Run-In, and at the start of each subsequent study cycle.
A patient was allowed to continue protocol therapy beyond progression that is determined by RECIST criteria (either RECIST 1.1 or mRECIST for patients with HCC), provided that the patient is obtaining potential clinical benefit in the opinion of the investigator. If sorafenib was discontinued due to sorafenib-related toxicities, a patient was allowed to continue receiving the study drug (either BBI608 or BBI503) provided there were no signs of toxicity and the patient obtained a clinical benefit from the drug.
Patient characteristics
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Of the 17 pts evaluable by RECIST, 9 were in Arm 1 and 8 were in Arm 2. DCR was observed in 100 % of the evaluable pts in both Arm 1 and 2, 9 SD in Arm 1 and 8 SD in Arm2. The initial results of RECIST 1.1. and mRECIST for napabucasin + sorafenib and amcasertib + sorafenib are shown in FIGs.8A and 8B respectively.
Figure imgf000056_0001
Napabucasin and amcasertib were safely combined with sorafenib at full dose with no unexpected adverse events. Encouraging anti-cancer activity in patients with HCC who have not received prior systemic therapy was observed. RP2D dose was determined to be 240mg BID for napabucasin and 100mg QD for amcasertib. A randomized phase II part of this study is currently enrolling patients into three arms. Arm I: napabucasin+sorafenib, Arm II: amcasertib+sorafenib and Arm III: sorafenib alone.
The many features and advantages of the present disclosure are apparent from the detailed specification, and thus it is intended by the appended claims to cover all such features and advantages of the present disclosure that fall within the true spirit and scope of the present disclosure. Further, since numerous modifications and variations will readily occur to those skilled in the art, it is not desired to limit the present disclosure to the exact construction and operation illustrated and described accordingly, all suitable modifications and equivalents may be resorted to, falling within the scope of the present disclosure.

Claims

WHAT IS CLAIMED IS: 1. A method for treating cancer in a subject in need thereof, comprising administering:
(a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
2. A method for treating cancer refractory or resistant to a kinase inhibitor in a subject comprising administering:
(a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
3. A method for preventing cancer relapses in a subject comprising administering:
(a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
4. A method for suppressing regrowth or recurrence of cancer in a subject comprising administering: (a) a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (b) a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
5. The method according to any one of claims 1-4, wherein the cancer stemness inhibitors comprise STAT3 pathway inhibitors.
6. The method according to any one of claims 1-5, wherein the cancer stemness inhibitors comprise 2-(1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro- naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2- acetylnaphtho[2,3-b]furan-4,9-dione, and 2-ethyl-naphtho[2,3-b]furan-4,9-dione.
7. The method according to any one of claims 1-6, wherein the cancer stemness inhibitors comprise compounds having Formula A:
Figure imgf000058_0001
8. The method according to any one of claims 1-7, wherein the cancer stemness inhibitors comprise inhibitors of at least one kinase chosen from cancer stem cell pathway kinases (CSCPK).
9. The method according to claim 8, wherein the at least one kinase is chosen from STK33, MELK, AXL, p70S6K, and PDGFRα.
10. The method according to any one of claims 1-9, wherein the cancer stemness inhibitors comprise
Figure imgf000059_0001
Figure imgf000060_0001
11. The method according to any one of claims 1-10, wherein the cancer stemness inhibitors comprise compounds having Formula B:
Figure imgf000060_0002
12. The method according to any one of claims 1-11, wherein the kinase inhibitors comprise compounds having Formula C:
Figure imgf000060_0003
13. The method according to any one of claims 1-12, wherein the cancer is chosen from liver cancers.
14. The method according to any one of claims 1-13, wherein the cancer is chosen from hepatocellular carcinoma and cholangiocarcinoma.
15. The method according to any one of claims 1-14, wherein the cancer is advanced, refractory, recurrent, or metastatic.
16. A method for sensitizing or re-sensitizing cancer cells to a kinase inhibitor comprising administering to the cancer cells at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
17. A method for treating cancer cells comprising administering a therapeutically effective amount of at least one first compound chosen from cancer stemness inhibitors, prodrugs of the foregoing, derivatives of the foregoing, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
18. The method according to any one of claims 16-17, wherein the cancer cells are in a subject.
19. The method according to any one of claims 16-18, wherein the cancer stemness inhibitors comprise STAT3 pathway inhibitors.
20. The method according to any one of claims 16-19, wherein the cancer stemness inhibitors comprise 2-(1-hydroxyethyl)-naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-chloro- naphtho[2,3-b]furan-4,9-dione, 2-acetyl-7-fluoro-naphtho[2,3-b]furan-4,9-dione, 2- acetylnaphtho[2,3-b]furan-4,9-dione, and 2-ethyl-naphtho[2,3-b]furan-4,9-dione.
21. The method according to any one of claims 16-20, wherein the cancer stemness inhibitors comprise compounds having Formula A:
Figure imgf000062_0001
22. The method according to any one of claims 16-18, wherein the cancer stemness inhibitors comprise cancer stem cell pathway inhibitors.
23. The method according to any one of claims 16-18 and 22, wherein the cancer stemness comprise
Figure imgf000062_0002
Figure imgf000063_0001
24. The method according to any one of claims 16-18, 22, and 23, wherein the cancer stemness inhibitors comprise compounds having Formula B:
Figure imgf000063_0002
25. The method according to any one of claims 16-23, comprising administering a therapeutically effective amount of at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
The method according to any one of claims 16-25, wherein the kinase inhibitors comprise compounds having Formula C:
Figure imgf000064_0001
27. The method according to any one of claims 16-26, wherein the cancer is chosen from liver cancers.
28. The method according to any one of claims 16-27, wherein the cancer is chosen from hepatocellular carcinoma and cholangiocarcinoma.
29. The method according to any one of claims 16-28, wherein the cancer is advanced, refractory, recurrent, or metastatic.
30. A method for treating cancer in a subject comprising administering to a subject in need thereof a therapeutically effective amount of at least one first compound chosen from compounds having Formula A:
Figure imgf000064_0002
and compounds having Formula B:
Figure imgf000065_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and a therapeutically effective amount of at least one second compound chosen from compounds having Formula C:
Figure imgf000065_0002
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
31. A method for treating a cancer refractory or resistant to at least one kinase inhibitor in a subject comprising administering to a subject in need thereof a therapeutically effective amount of at least one first compound chosen from compounds having Formula A:
Figure imgf000066_0001
and compounds having Formula B:
Figure imgf000066_0002
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and a therapeutically effective amount of at least one second compound chosen from compounds having Formula C:
Figure imgf000066_0003
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. A method for sensitizing a cancer to at least one kinase inhibitor in a subject comprising administering to a subject in need thereof a therapeutically effective amount of at least one first compound chosen from compounds having Formula A:
Figure imgf000067_0001
and compounds having Formula B:
Figure imgf000067_0002
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing.
A method for re-sensitizing a cancer to at least one kinase inhibitor in a subject comprising administering to a subject in need thereof a therapeutically effective amount of at least one first compound chosen from compounds having Formula A:
Figure imgf000067_0003
and compounds having Formula B:
Figure imgf000068_0001
prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing. 34. A kit comprising (a) at least one first compound chosen from cancer stemness inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; (b) at least one second compound chosen from kinase inhibitors, prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, and solvates of any of the foregoing; and (c) instructions for administration and/or use of the at least one first compound and the at least one second compound.
PCT/US2017/039430 2016-06-28 2017-06-27 Methods for treating cancer WO2018005444A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2018568329A JP2019519573A (en) 2016-06-28 2017-06-27 Methods for treating cancer
CA3029596A CA3029596A1 (en) 2016-06-28 2017-06-27 Methods for treating cancer

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662355410P 2016-06-28 2016-06-28
US62/355,410 2016-06-28
US201762506929P 2017-05-16 2017-05-16
US62/506,929 2017-05-16
US201762514059P 2017-06-02 2017-06-02
US62/514,059 2017-06-02

Publications (2)

Publication Number Publication Date
WO2018005444A2 true WO2018005444A2 (en) 2018-01-04
WO2018005444A3 WO2018005444A3 (en) 2018-02-15

Family

ID=59325662

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/039430 WO2018005444A2 (en) 2016-06-28 2017-06-27 Methods for treating cancer

Country Status (4)

Country Link
JP (1) JP2019519573A (en)
CA (1) CA3029596A1 (en)
TW (1) TW201806592A (en)
WO (1) WO2018005444A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020061232A1 (en) * 2018-09-18 2020-03-26 1 Globe Biomedical Co., Ltd. Treatment for non-alcoholic fatty liver disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2021518532A (en) * 2018-03-16 2021-08-02 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー Analysis of response to therapeutic agents in cancer

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009033033A2 (en) 2007-09-06 2009-03-12 Boston Biomedical, Inc. Compositions of kinase inhibitors and their use for treatment of cancer and other diseases related to kinases
WO2009036099A1 (en) 2007-09-10 2009-03-19 Boston Biomedical, Inc. A novel group of stat3 pathway inhibitors and cancer stem cell pathway inhibitors
WO2011116398A1 (en) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Novel compounds and compositions for targeting cancer stem cells
WO2011116399A1 (en) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Novel methods for targeting cancer stem cells
WO2014160401A1 (en) 2013-03-13 2014-10-02 Boston Biomedical, Inc. 3-(aryl or heteroaryl) methyleneindolin-2-one derivatives as inhibitors of cancer stem cell pathway kinases for the treatment of cancer
WO2014169078A2 (en) 2013-04-09 2014-10-16 Boston Biomedical, Inc. Methods for treating cancer
US8977803B2 (en) 2011-11-21 2015-03-10 Western Digital Technologies, Inc. Disk drive data caching using a multi-tiered memory
US9150530B2 (en) 2011-03-04 2015-10-06 Zhoushan Haizhongzhou Xinsheng Pharmaceuticals Co., Ltd. Esters of 4, 9-dihydroxy-naphtho [2, 3-b] furans for disease therapies
US9328345B2 (en) 2007-08-27 2016-05-03 1 Globe Health Institute Llc Compositions of asymmetric interfering RNA and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180250261A1 (en) * 2015-04-27 2018-09-06 Boston Biomedical, Inc. Method for treating cancer with a stat3 pathway inhibitor and kinase inhibitor

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9328345B2 (en) 2007-08-27 2016-05-03 1 Globe Health Institute Llc Compositions of asymmetric interfering RNA and uses thereof
US8299106B2 (en) 2007-09-06 2012-10-30 Boston Biomedical, Inc. Compositions of kinase inhibitors and their use for treatment of cancer and other diseases related to kinases
WO2009033033A2 (en) 2007-09-06 2009-03-12 Boston Biomedical, Inc. Compositions of kinase inhibitors and their use for treatment of cancer and other diseases related to kinases
US8877803B2 (en) 2007-09-10 2014-11-04 Boston Biomedical, Inc. Stat3 pathway inhibitors and cancer stem cell inhibitors
US20120252763A1 (en) 2007-09-10 2012-10-04 Boston Biomedical, Inc. Novel group of stat3 pathway inhibitors and cancer stem cell pathway inhibitors
WO2009036101A1 (en) 2007-09-10 2009-03-19 Boston Biomedical, Inc. Novel compositions and methods for cancer treatment
WO2009036099A1 (en) 2007-09-10 2009-03-19 Boston Biomedical, Inc. A novel group of stat3 pathway inhibitors and cancer stem cell pathway inhibitors
WO2011116399A1 (en) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Novel methods for targeting cancer stem cells
WO2011116398A1 (en) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Novel compounds and compositions for targeting cancer stem cells
US9150530B2 (en) 2011-03-04 2015-10-06 Zhoushan Haizhongzhou Xinsheng Pharmaceuticals Co., Ltd. Esters of 4, 9-dihydroxy-naphtho [2, 3-b] furans for disease therapies
US8977803B2 (en) 2011-11-21 2015-03-10 Western Digital Technologies, Inc. Disk drive data caching using a multi-tiered memory
WO2014160401A1 (en) 2013-03-13 2014-10-02 Boston Biomedical, Inc. 3-(aryl or heteroaryl) methyleneindolin-2-one derivatives as inhibitors of cancer stem cell pathway kinases for the treatment of cancer
WO2014169078A2 (en) 2013-04-09 2014-10-16 Boston Biomedical, Inc. Methods for treating cancer

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
AJANI ET AL., SEMIN. ONCOL., vol. 42, no. 1, 2015, pages 3 - 17
BOMAN BM ET AL., J. CLIN. ONCOL., vol. 26, no. 17, 2008, pages 2828 - 2838
BOROVSKI T. ET AL., CANCER RES., vol. 71, no. 3, 2011, pages 634 - 639
CLARKE MF, BIOL. BLOOD MARROW TRANSPLANT., vol. 11, no. 2, 2009, pages 14 - 16
FURQAN ET AL., JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 6, 2013, pages 90
FURTEK ET AL., ACS CHEM. BIOL., vol. 11, no. 2, 2016, pages 308 - 318
GUPTA PB ET AL., NAT. MED, vol. 15, no. 9, 2009, pages 1010 - 1012
HUANG ET AL., BMC CANCER., vol. 15, 2015, pages 13
J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
JORDAN CT ET AL., N. ENGL. J. MED, vol. 355, no. 12, 2006, pages 1253 - 1261
KIM ET AL., CANCER RES., vol. 74, no. 8, 2014, pages 2144 - 51
VAJKOCZY ET AL., PROC. NATL. ACAD. SCI. USA., vol. 103, no. 15, 11 December 2005 (2005-12-11), pages 5799 - 804
ZHANG ET AL., CANCER RES., vol. 68, no. 6, 15 December 2007 (2007-12-15), pages 1905 - 15

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020061232A1 (en) * 2018-09-18 2020-03-26 1 Globe Biomedical Co., Ltd. Treatment for non-alcoholic fatty liver disease
TWI825177B (en) * 2018-09-18 2023-12-11 大陸商北京強新生物科技有限公司 Treatment of non-alcoholic fatty liver disease
US12318381B2 (en) 2018-09-18 2025-06-03 1Globe Biomedical Co., Ltd. Treatment for non-alcoholic fatty liver disease

Also Published As

Publication number Publication date
CA3029596A1 (en) 2018-01-04
TW201806592A (en) 2018-03-01
WO2018005444A3 (en) 2018-02-15
JP2019519573A (en) 2019-07-11

Similar Documents

Publication Publication Date Title
US20180085341A1 (en) Methods for treating cancer
US10646464B2 (en) Methods for treating cancer
JP2021121629A (en) Methods for treating cancer using stat3 pathway inhibitors and kinase inhibitors
US20190231735A1 (en) Methods for treating cancer
TW201731500A (en) Methods for treating cancer
US20180098959A1 (en) Methods for treating cancer
WO2018005444A2 (en) Methods for treating cancer
JP7513286B2 (en) New drug combination strategies for the treatment of chemotherapy-refractory cancers
HK40047114A (en) New combination solution for treating chemotherapy refractory cancer

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: 17739417

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2018568329

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3029596

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17739417

Country of ref document: EP

Kind code of ref document: A2