WO2015058185A1 - Separase inhibitors and uses thereof - Google Patents

Separase inhibitors and uses thereof Download PDF

Info

Publication number
WO2015058185A1
WO2015058185A1 PCT/US2014/061353 US2014061353W WO2015058185A1 WO 2015058185 A1 WO2015058185 A1 WO 2015058185A1 US 2014061353 W US2014061353 W US 2014061353W WO 2015058185 A1 WO2015058185 A1 WO 2015058185A1
Authority
WO
WIPO (PCT)
Prior art keywords
groups
phenyl
combinations
group
separase
Prior art date
Application number
PCT/US2014/061353
Other languages
French (fr)
Inventor
Debananda PATI
Nenggang ZHANG
Original Assignee
Baylor College Of Medicine
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 Baylor College Of Medicine filed Critical Baylor College Of Medicine
Priority to US15/030,070 priority Critical patent/US10052310B2/en
Publication of WO2015058185A1 publication Critical patent/WO2015058185A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic 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
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole

Definitions

  • compositions that inhibit Separase activity include one or more compounds with one or more of the following structures:
  • R (in structures 1-13) and R 1A (in structures 3 and 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR 3 , N(R 3 ) 2 , C 2 R 3 , C 4 H 9 , phenyl, CH 2 -phenyl, S0 3 H, and combinations thereof.
  • R in structures 1-13 is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof.
  • R 2 in structures 1-4 and 6-13 is selected from the group consisting of alkanes, C(CH 3 ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7. s), C(CH 3 )(R 7.
  • R 7 and R 7 5 are each selected from the group consisting of alkanes, CH , CH 2 CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • R 4 in structure 5) is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH 3 , phenyl, O-phenyl, NH-phenyl, and combinations thereof.
  • R 5 in structure 5) is selected from the group consisting of NH, NR 6 , S, O, and combinations thereof.
  • R 6 in structure 5) is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof.
  • compositions of the present disclosure include one or more compounds with the following structure:
  • R in structure 1 is N0 2
  • R 2 in structure 1 is C(CH ) 2
  • Ri in structure 1 is Br and R 2 in structure 1 is CS.
  • compositions of the present disclosure include one or more compounds with the following structure:
  • R in structure 4 is Br
  • R 2 in structure 4 is CS
  • compositions of the present disclosure include more compounds with the following structure:
  • Rg is selected from the group consisting of alkanes, CH 3 , CH 2 CH 3 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • compositions of the present disclosure include one or more compounds with the following structure:
  • the compounds of the present disclosure are water soluble. In some embodiments, the compounds of the present disclosure are selective inhibitors of Separase. In some embodiments, the compounds of the present disclosure are non-competitive inhibitors of Separase. [0015] In additional embodiments, the present disclosure pertains to methods of treating a tumor in a subject by administering one or more compositions of the present disclosure to the subject.
  • the tumor comprises Separase overexpressed tumor cells, such as Separase overexpressed mammary tumor cells or Separase overexpressed aneuploid cells.
  • the tumor to be treated by the compositions of the present disclosure is associated with at least one of breast cancer, ovarian cancer, leukemia, thyroid cancer, neuroblastoma, brain cancer, lung cancer, colon cancer, gastrointestinal cancer, prostate cancer, osteosarcoma, glioblastoma, and combinations thereof.
  • the tumor to be treated by the compositions of the present disclosure is associated with breast cancer, such as triple negative breast cancer and luminal-B subtype endocrine resistant breast cancer.
  • the subjects to be treated by the compositions of the present disclosure are human beings, such as human beings suffering from cancer.
  • the compositions of the present disclosure are administered to subjects by intravenous administration.
  • FIGURE 1 provides structures of various separase inhibitors and potential separase inhibitors.
  • FIG. 1A provides structures of 2,2-dimethyl-5-nitro-2H-benzimidazole trihydrate (left panel), SYC-764 (center), and SYC-763 (right panel).
  • FIGS. 1B-C provides structures of various derivatives of the compounds in FIG. 1A.
  • FIG. ID provides the structure of Sepin-1.
  • FIG. IE provides structures of various Sepin-1 derivatives.
  • FIGURE 2 provides data relating to the development of a fluorogenic Separase assay using (Rad21)2-Rhl l0 as a substrate.
  • FIG. 2A shows a Rhl lO standard curve. The fluorescence intensity expressed by the relative fluorescent units (RFU) is linearly proportional to the amount of Rhl lO, which can be used to determine the amount of Rhl lO formation in Separase assay.
  • FIG. 2B shows that the amount of free Rhl 10 released from (Rad21)2-Rhl 10 is a function of the substrate concentration. The amount of Separase used in this experiment was 5ng/well.
  • FIG. 2C shows the amount of free Rhl lO released from (Rad21)2-Rhl l0 was correlated to the amount of Separase enzyme concentration.
  • the amount of Rhl 10 used in this experiment was 1 nmol/well.
  • FIGURE 3 shows results relating to the kinetics of the Separase assay.
  • the cleavage reaction was carried out in the presence of the increased concentration (0.015-2 mM) of (Rad21)2-Rhl l0 and 5ng of Separase. The total reaction volume was 25 ⁇ 1.
  • the plates were incubated at 37 °C for 3h before the fluorescence intensity was determined. The fluorescence intensity was converted to pmol of Rhl lO.
  • Michaelis-Menten curves were plotted (FIG. 3A), and V m and K m were calculated using Lineweaver-Burt plot (FIG. 3B).
  • FIGURE 4 shows results relating to the stability of Separase enzyme and substrate and the effect of TEV and DMSO on Separase assay.
  • FIG. 4A shows aliquots of diluted Separase in working concentration were kept on ice for 8h and then stored at -80, -20 or 4 °C for 24h. Their activity was analyzed in comparison to the same concentration of Separase that was freshly prepared from stock solutions.
  • FIG. 4B shows diluted (Rad21)2-Rhl l0 in working concentration was stored at -80 or 4 °C for 24h. The activity was analyzed using Separase assay. The same concentration of (Rad21)2-Rhl l0 that was freshly prepared from stock solution was used as control.
  • FIG. 4A shows aliquots of diluted Separase in working concentration were kept on ice for 8h and then stored at -80, -20 or 4 °C for 24h. Their activity was analyzed in comparison to the same concentration of Separase that was freshly prepared from stock solutions
  • FIG. 4C shows results where TEV protease and Separase were used to hydrolyze (Rad21)2-Rhl l0. The enzyme activity was determined after incubation at 37 °C for 3h.
  • FIG. 4D shows the effect of DMSO concentration on Separase assay.
  • FIGURE 5 shows high throughput screening for Separase inhibitors.
  • FIG. 5A shows the reproducibility of the screening. Each data point was repeated in two plates (A and B) and plotted with the Separase activity of A plate as x-axis and that of B plate as y-axis. A total of 9 representative duplicated 384-well plates with 3,456 data points are shown.
  • FIG. 5B shows RFU of Positive controls is well separated from that of the negative controls. 576 data points of positive controls and negative controls, respectively, from 18 384-well plates are shown, ⁇ is STDEV.
  • FIG. 5C is a chart summarizing the robustness of high throughput screening (HTS).
  • FIG. 5D shows results demonstrating the inhibition of Separase activity by the test compounds.
  • the test compounds that inhibited at least 50% of Separase activity in both A plates and B plates were identified.
  • the data points in the two upper right boxes showed the compounds that inhibited Separase activity by >50% and >80%, respectively.
  • 5E illustrates that, among the 14,400 compounds assayed, 97 are found to inhibit Separase activity by >50%.
  • the hit rate is 0.67%. 24 out of the 97 compounds inhibit Separase activity by >80%. Five of the 97 compounds were confirmed.
  • FIGURE 6 provides results relating to the characterization of Sepin-1 in inhibiting Separase activity.
  • FIG. 6A provides a structure of Sepin-1.
  • FIG. 6B shows that Sepin-1 inhibits the cleavage of Separase substrate Rad21. The in vitro transcribed and translated Myc-Rad21 was used as the substrate for activated Separase in the presence of or without Sepin-1. The final concentration of Sepin-1 in the reaction mixture was 50 ⁇ . molecular weight).
  • FIG. 6C shows the inhibition of Separase activity by Sepin-1. The IC 50 was 14.8 ⁇ , calculated using the KaleidaGraph program. The Inset shows a four-parameter dose -response curve fit with an estimated IC 50 of 15.8 ⁇ .
  • FIG. 6D shows the non-competitive inhibition Separase enzymatic activity by Sepin-1.
  • Sepin-1 and Separase were mixed 30 min before substrate (Rad21)2-Rhl l0 was added. After 3h incubation, the fluorescence intensity of Rhl lO was determined. The data were shown in Lineweaver-Burt plot.
  • Sepin-1 reduced the speed (V max ) of substrate conversion by Separase, but not the K m .
  • FIGURE 7 shows that Sepin- 1 selectively inhibits the growth of breast cancer cells and patient-derived triple negative breast cancer xenografts overexpressing Separase protein.
  • FIG. 7A shows an immunoblot of the Separase protein in breast cancer cell lines, MCF7 and MCFlOf.
  • FIG. 7B shows results of an MTT assay where MCF7 is more sensitive to Sepin-1 in inhibiting cell growth than MCFlOf ex vivo.
  • FIG. 7C shows that Sepin-1 inhibits the growth of xenograft tumor derived from MCF-7 in SCID-beige mice.
  • FIG. 7D shows the immuno staining of Separase in triple negative breast cancer tumor xenografts MCI and BCM-5471.
  • FIGS. 7E-F shows results where Sepin-1 inhibits the growth of xenograft tumors derived from breast cancer tumors in SCID-beige mice.
  • BCM-5471 xenograft tumors are more sensitive to Sepin-1 than MC-1 tumors.
  • FIG. 7G-H shows that Sepin-1- treated MCF-7 xenograft tumors have high level of apoptosis.
  • Tumor sections from MCF-7 xenografts treated with Sepin-1 or vehicle were stained with H&E (FIG. 7G) or immunostained with cleaved caspase-3 mAb (FIG. 7H). Apoptotic bodies in H&E staining (black arrows) and immuno staining (green) were observed in Sepin-1 -treated tumors.
  • FIGURE 8 shows that Sepin-1 inhibits cancer cell growth and induces apoptosis.
  • FIGS. 8A-C shows that Sepin-1 inhibits cell growth of leukemia cell lines (FIG. 8A), breast cancer cell lines (FIG. 8B), and Neuroblastoma cell lines (FIG. 8C). Cells were treated with serially diluted Sepin-1 for 72h. The cell viability was assessed using MTT assay.
  • FIG. 8D shows a western blot of full length (FL) and N-terminal (NT, surrogate for the active protein) Separase in cancer cell lines.
  • FIG. 8E shows a positive correlation between Separase protein level and sensitivity to Sepin-1 in inhibiting cell growth (IC 50 ).
  • FIG. 8F shows immunoblotting shown Sepin-1 - induced activation of caspase-3 and cleavage of poly (ADP-ribose) polymerase (Parp). Molt4 cells were treated with various concentrations of Sepin-1 for 24 h. Etoposide (Etop) was used as a control.
  • CI Parp refers to cleaved Parp.
  • CI Casp-3 refers to cleaved Caspase-3. * indicates nonspecific bands.
  • FIGURE 9 provides additional data relating to the identification of leads in HTS for Separase inhibitors.
  • FIG. 9A provides structures of the 5 lead compounds.
  • FIG. 9B provides dose-dependent inhibition of Separase activity by Sepin 1 and 2.
  • FIGURE 10 provides data relating to separase overexpression in Pediatric glioblastoma (GBM) and the effect of Sepin-1 in inhibiting the growth of glioblastoma cells in vitro and in vivo.
  • GBM Pediatric glioblastoma
  • FIGURE 11 provides additional data relating to structure- activity studies of Sepin-1 derivatives.
  • FIG. 11A shows the structures of Sepin-1 derivatives that were tested.
  • FIG. 11B shows data relating to the separase inhibition activities of the Sepin-1 derivatives.
  • the numbers (28-31 in FIG. 11B) correspond to structures shown in FIG. 11A.
  • Separase is a protease evolutionarily conserved from yeast to human. Its primary function is to cleave the cohesin subunit Sccl/Mcdl/Rad21 at the onset of anaphase, resulting in the dissolution of cohesin rings and independent segregation of sister chromatids into the two daughter cells.
  • Separase is overexpressed and mislocalized in a number of human tumors, including breast, prostate and osteosarcoma.
  • Separase overexpression is reported in a broad range of human tumors.
  • Separase overexpression in mouse models results in tumorigenesis.
  • Separase is significantly overexpressed in over 60% of human breast tumors compared to the matched normal breast tissue.
  • conditional overexpression of Separase in mammary epithelial cells is sufficient to induce separation of sister chromatids, aneuploidy and tumorigenesis.
  • Separase activity is tightly regulated during the cell cycle and is inactive when it binds to its inhibitory chaperone, securin. Separase is also inhibited via phosphorylation at Serl l26 by Cyclin B-Cdkl kinase and binding to Cyclin B. With the progression of cell cycle to the onset of anaphase, the anaphase promoting complex/cyclosome polyubiquitinates mitotic cyclin and securin, which are rapidly degraded by the 26S proteasome. Once enzymatically activated, Separase auto-cleaves, resulting in N-terminal and C-terminal fragments, which still associate with each other. The main function of Separase is to proteolytically cleave the cohesin subunit
  • Separase is overexpressed in human tumors, and its overexpression results in chromosomal missegregation and aneuploidy, pharmacological inhibition of Separase presents a novel strategy to target chromosomal missegregation induced tumorigenesis. Pharmacologic modulation of Separase activity remains elusive, however, due to a lack of small molecular compound inhibitors.
  • the present disclosure pertains to compositions that inhibit Separase activity. In some embodiments, the present disclosure pertains to methods of treating a tumor in a subject by administering the compositions of the present disclosure to the subject.
  • compositions of the present disclosure include one or more compounds that inhibit Separase activity.
  • the compounds include one or more of the following structures:
  • structures 1-13 can resonate in different manners.
  • structures 7-10 may represent different resonating structures of the same compounds.
  • structures 11-13 may resonate to resemble the compounds disclosed in structures 7-10. Therefore, various embodiments of the present disclosure also cover resonating versions of structures 1-13 that may not be disclosed in the aforementioned structures.
  • R (in structures 1-13) and R 1A (in structures 3 and 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR , N(R 3 ) 2 , C 2 R 3 , C 4 H 9 , phenyl, CH 2
  • R (in structures 1-13) is N0 2 .
  • Ri and R 1A (in structures 3 and 11-13) are CI.
  • Ri (in structures 1-13) is N0 2
  • R 1A (in structures 3 and 11-13) is Br.
  • R (in structures 1-13) is selected from the group consisting of H, OH, CH 3 , C 4 Hg, alkyl groups, and combinations thereof. In some embodiments, R 3 (in structures 1-13) is H.
  • R 2 in structures 1-4 and 6-13 is selected from the group consisting of alkanes, C(CH ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7. s), C(CH 3 )(R 7 5 ), C(CH 3 )(CH 2 CH 3 ), C(CH 3 )(Phenyl), C(CH 3 )(CF 3 ), C(C 4 H 9 ) 2 , CO, CS, CH 2 , O, and combinations thereof.
  • R 7 and R 7 5 are each selected from the group consisting of alkanes, CH 3 , CH 2 CH 3 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 Hg, CO, CS, and combinations thereof.
  • R 2 in structures 1-4 and 6-13) is C(CH 3 ) 2 .
  • R 4 in structure 5) is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH 3 , phenyl, O-phenyl, NH-phenyl, and combinations thereof.
  • R 4 (in structure 5) is CH 3 .
  • R 5 in structure 5) is selected from the group consisting of NH, NR 6 , S, O, and combinations thereof. In some embodiments, R 5 (in structure 5) is NH. In some embodiments, R 6 (in structure 5) is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof.
  • compositions of the present disclosure include one or more compounds with the following structure:
  • R in structure 1 is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR , N(R 3 ) 2 , C 2 R 3 , C 4 Hg, phenyl, CH 2 -phenyl, S0 3 H, and combinations thereof.
  • R 3 in structure 1 is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof.
  • R in structure 1 is H.
  • R 2 in structure 1 is selected from the group consisting of alkanes, C(CH ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7 . 5 ), C(CH 3 )(R 7 .
  • R 7 and R 7 5 are each selected from the group consisting of alkanes, CH 3 , CH 2 CH 3 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • Ri in structure 1 is N0 2
  • R 2 in structure 1 is C(CH ) 2 .
  • Ri in structure 1 is Br
  • R 2 in structure 1 is CS.
  • compositions of the present disclosure include more compounds with the following structure:
  • R in structure 4 is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR , N(R 3 ) 2 , C 2 R 3 , C 4 H 9 , phenyl, CH 2 -phenyl, S0 3 H, and combinations thereof.
  • R in structure 4 is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof. In some embodiments, R in structure 4 is H. In some embodiments, R 2 in structure 4 is selected from the group consisting of alkanes, C(CH 3 ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7 . 5 ), C(CH 3 )(R 7 .
  • R 7 and R 7 5 are each selected from the group consisting of alkanes, CH , CH 2 CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • Ri in structure 4 is Br
  • R 2 in structure 4 is CS.
  • compositions of the present disclosure include one or more compounds with one or more of the following structures:
  • Ri (in structures 7-13) and RI A (in structures 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo- reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR , N(R 3 ) 2 , C 2 R 3 , C 4 H 9 , phenyl, CH 2 -phenyl, S0 3 H, and combinations thereof.
  • R 3 is selected from the group consisting of H, OH, CH 3 , C 4 Hg, alkyl groups, and combinations thereof.
  • R 2 is selected from the group consisting of alkanes, C(CH ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7 . 5 ), C(CH 3 )(R 7 .
  • R 7 and R 7 5 are each selected from the group consisting of alkanes, CH 3 , CH 2 CH 3 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • compositions of the present disclosure include one or more compounds with the following structure:
  • R and R 1A in structure 12 are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH 2 , N0 2 , Br, F, CI, I, CF 3 , CF 3 S0 2 , C(N 2 )CF 3 , CN, CH 3 , CH 3 0, C0 2 H, CONH 2 , CONHR 3 , NHR , N(R 3 ) 2 , C 2 R 3 , C 4 H 9 , phenyl, CH 2 -phenyl, S0 3 H, and combinations thereof.
  • R in structure 12 is selected from the group consisting of H, OH, CH , C 4 H 9 , alkyl groups, and combinations thereof.
  • R 3 in structure 12 is H.
  • R 2 in structure 12 is selected from the group consisting of alkanes, C(CH 3 ) 2 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R 7 )(R 7.5 ), C(CH 3 )(R 7.
  • R 7 and R 5 are each selected from the group consisting of alkanes, CH 3 , CH 2 CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • R in structure 12 is N0 2
  • Ri A in structure 12 is Br
  • R 2 in structure 12 is C(CH 3 ) 2 .
  • compositions of the present disclosure include one or more compounds with the following structure:
  • Rg is selected from the group consisting of alkanes, CH 3 , CH 2 CH 3 , cycloalkanes, cyclopentane, cyclohexane, phenyl, C 4 H 9 , CO, CS, and combinations thereof.
  • compositions of the present disclosure include one or more compounds with the following structure (referred to as Sepin-1 and also shown in FIG. ID):
  • compositions of the present disclosure include one or more derivatives of Sepin-1.
  • the Sepin-1 derivatives include, without limitation, one or more of the structures shown in FIG. IE.
  • the compositions of the present disclosure can contain one or more of the aforementioned compounds that inhibit Separase activity.
  • the compositions of the present disclosure can also have one or more physiologically acceptable carriers or excipients.
  • the compositions of the present disclosure can also include formulation materials for modifying, maintaining, or preserving various conditions, including pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, and/or adsorption or penetration of the compounds.
  • Suitable formulation materials include, but are not limited to: amino acids (e.g., glycine); antimicrobials; antioxidants (e.g., ascorbic acid); buffers (e.g., Tris-HCl); bulking agents (e.g., mannitol and glycine); chelating agents (e.g., EDTA); complexing agents (e.g., hydroxypropyl-beta-cyclodextrin); and the like.
  • amino acids e.g., glycine
  • antimicrobials e.g., ascorbic acid
  • buffers e.g., Tris-HCl
  • bulking agents e.g., mannitol and glycine
  • chelating agents e.g., EDTA
  • complexing agents e.g., hydroxypropyl-beta-cyclodextrin
  • the compounds in the compositions of the present disclosure can have various properties. For instance, in some embodiments, the compounds of the present disclosure are water soluble. In some embodiments, the compounds of the present disclosure inhibit Separase activity. In some embodiments, the compounds of the present disclosure are non-competitive inhibitors of Separase.
  • the compounds of the present disclosure are selective inhibitors of Separase.
  • the compounds of the present disclosure inhibit Separase activity without significantly inhibiting the activity of other enzymes.
  • the compounds of the present disclosure inhibit Separase activity without significantly inhibiting the activity of cysteine proteases other than Separase (e.g., Caspases, tissue kallikrein, coagulation factor Xa, and other proteases that cleave after arginine at PI position).
  • cysteine proteases other than Separase e.g., Caspases, tissue kallikrein, coagulation factor Xa, and other proteases that cleave after arginine at PI position.
  • the compounds of the present disclosure inhibit the growth of tumor cells.
  • the compounds of the present disclosure can inhibit the growth of Separase overexpressed tumor cells.
  • the compounds of the present disclosure can selectively inhibit the growth of Separase overexpressed tumor cells without significantly affecting the growth of normal cells.
  • the compounds of the present disclosure can selectively inhibit the growth of Separase overexpressed aneuploid cells.
  • the compounds of the present disclosure inhibit the growth of tumor cells at IC 50 concentrations that range from about 0.5 ⁇ to about 65 ⁇ .
  • the compounds of the present disclosure inhibit the growth of tumor cells at IC 50 concentrations that range from about 1 ⁇ to about 10 ⁇ .
  • the compounds of the present disclosure can have numerous applications in treating tumors.
  • additional embodiments of the present disclosure pertain to methods of treating various tumors in various subjects by administering one or more compositions of the present disclosure to the subject.
  • compositions of the present disclosure can be utilized to treat various types of tumors.
  • the tumors to be treated include Separase overexpressed tumor cells.
  • the tumors to be treated include Separase overexpressed mammary tumor cells, Separase overexpressed aneuploid cells, and combinations thereof.
  • the tumors to be treated by the compositions of the present disclosure may be associated with various types of cancers.
  • the tumors to be treated may be associated with at least one of breast cancer, leukemia, thyroid cancer, neuroblastoma, brain cancer, lung cancer, colon cancer, prostate cancer, osteosarcoma, glioblastoma, ovarian cancer, gastrointestinal cancer and combinations thereof.
  • the tumors to be treated are associated with breast cancer.
  • the tumors to be treated are associated with triple negative breast cancer and luminal-B subtype endocrine resistant breast cancer.
  • the compositions of the present disclosure can treat tumors by various mechanisms. For instance, in some embodiments, the compounds in the compositions of the present disclosure inhibit the growth of tumor cells associated with a tumor. In some embodiments, the compounds in the compositions of the present disclosure selectively inhibit the growth of Separase overexpressed tumor cells associated with the tumor. In some embodiments, the compounds in the compositions of the present disclosure selectively inhibit the growth of Separase overexpressed aneuploid cells associated with the tumor.
  • compositions of the present disclosure can be used to treat tumors in various subjects.
  • the subject is a human being.
  • the subject is a human being suffering from cancer.
  • the subjects may be non-human animals, such as mice, rats, other rodents, or larger mammals, such as dogs, monkeys, pigs, cattle and horses.
  • compositions of the present disclosure can be administered to subjects by various methods.
  • the compositions of the present disclosure can be administered by oral administration (including gavage), inhalation, subcutaneous administration (sub-q), intravenous administration (I.V.), intraperitoneal administration (LP.), intramuscular administration (I.M.), intrathecal injection, and combinations of such modes.
  • the therapeutic compositions of the present disclosure can be administered by topical application (e.g, transderm, ointments, creams, salves, eye drops, and the like).
  • the compositions of the present disclosure can be administered intravenously. Additional modes of administration can also be envisioned.
  • compositions of the present disclosure may also be administered in a single dose or multiple doses throughout a time period.
  • the compositions of the present disclosure may be administered to a subject in two separate doses.
  • compositions of the present disclosure may be administered to localized sites in a subject, such as tissue or vasculature that contain a tumor.
  • a subject such as tissue or vasculature that contain a tumor.
  • the compositions of the present disclosure may be injected directly into an area of a subject that displays tumor growth.
  • compositions of the present disclosure may be coadministered with other therapies.
  • the compositions of the present disclosure may be co-administered along with anti-cancer drugs.
  • Applicants report a high throughput screening for Separase inhibitors (Sepins).
  • Applicants developed a fluorogenic Separase assay using Rhodamine 110 conjugated Rad21 peptide as substrate and screened a small molecule compound library.
  • Applicants have screened a chemical library with 14,400 small compounds and identified five compounds that inhibited Separase activity.
  • Sepin-1 a noncompetitive inhibitor of Separase, which can inhibit the growth of cancer cell lines and mammary xenograft tumors in mice by inducing apoptosis.
  • Sepin-1 inhibits Separase enzymatic activity with an IC 50 of 14.8 ⁇ .
  • Applicants have also found that the sensitivity to Sepin-1 in most cases is positively correlated to the level of Separase in both cancer cell lines and tumors.
  • Example 1.1 Development of Separase activity assay using (Rad21)2-Rhl l0 as a substrate
  • AMC 7-amido-4-mrthyl coumaric acid
  • Rhodamine 110 Rhodamine 110
  • Rad21 peptide Asp-Arg-Glu-Ile-Nle-Arg
  • Glu-X-X-Arg Glu-X-X-Arg
  • Rhl lO released was linearly correlated with the increase of (Rad21)2-Rhl l0 concentration between 0-2nmol/well (FIG. 2B). Similarly, the amount of Rhl lO released was a linear function with the increase of Separase concentration between 0- lOng/well (FIG. 2C).
  • incubation temperature significantly affects the kinetics of Separase.
  • the release of Rhl lO catalyzed by Separase was much faster at 37 °C than that at room temperature (25 °C) (FIG. 2D).
  • the amount of Rhl lO released from (Rad21)2-Rhl l0 linearly increased between 0 and 3h (FIG. 2D).
  • Both di- and mono-substituted Rad21-Rhl l0 can be cleaved by Separase.
  • the fluorescence intensity of mono-substituted Rad21-Rhl l0 is about 10% of free Rhl lO.
  • V max (4.37 pmol Rhl lO/h, or 201 pmol Rhl lO/h/pmol Separase) and K m (123 ⁇ ) were calculated using a Lineweaver-Burk plot (FIG. 3B).
  • the stability of enzyme and substrate at the working conditions is important because the process of distributing different components in high throughput screening is complicated and time-consuming.
  • Applicants diluted Separase enzyme and (Rad21)2-Rhl l0 to working concentration, and kept them at 4 °C for 8h.
  • the diluted enzyme and substrate were frozen at -20 °C or -80 °C or stored at 4 °C for 24h before their activities were determined.
  • the activity of Separase at the working concentration had no significant difference when it was kept at 4 °C, -20°C, or -80 °C for 24h.
  • the activity of diluted Separase was reduced by 20% (FIG. 4A).
  • (Rad21)2-Rhl l0 at working concentration was unaffected by storage by storage at 4 °C or -80 °C for 24h (FIG. 4B).
  • the construct that was used to express Separase in 293T cells contains two IgG binding domains of protein A (Z-domain), followed by four TEV-pro tease cleavage sequences on the N terminus of Separase (ZZ-TEV4-Separase).
  • TEV protease was used to release the Separase from IgG-Agarose beads after activation by Xenopus cytostatic factor extract.
  • the assay was not affected by 2-14% of DMSO, but it was inhibited with the increase of DMSO concentration of >14%.
  • the IC 50 was -27% (FIG. 5D). Because the DMSO concentration in Separase assay used in HTS was less than 3%, Applicants conclude that DMSO has no significant effect in this assay.
  • Example 1.4 High throughput screening for Separase inhibitors
  • Applicants have identified 97 compounds that inhibited Separase activity by more than 50%. Among the 97 compounds, 24 reduced more than 80% of Separase activity (FIG. 5E). Applicants ordered all of these 97 compounds and verified the activities of these compounds in inhibiting Separase enzymatic activity. Five of them were confirmed, which were named Separase inhibitors (Sepin-1 to 5, shown in FIG. 9A). Among them, Sepin-1 had the highest activity, which was further characterized below. Also see FIG. 9B.
  • Sepin-1 is 2,2-dimethyl-5-nitro-2H-benzimidazole-l,3 dioxide with molecular weight 223 Da and melting point 228°C (FIGS. ID and 6A). Applicants used 13C NMR to confirm the identity of Sepin-1 before use for further experimentation. Sepin-1 could inhibit the Rad21 cleavage by activated Separase in vitro when in vitro transcribed and translated Rad21 was used as the substrate (FIG. 6B). In the fluorogenic Separase activity assay using (Rad21)2-Rhl l0 as the substrate, the concentration of Sepin-1 to inhibit 50% of Separase activity (IC 50 ) was 14.8 ⁇ (FIG. 6C).
  • Separase has at least two sites interacting with Rad21. One is the catalytic site, and the other is outside the catalytic site. Although blocking the catalytic site does not affect Rad21 binding to Separase, it is unclear whether Rad21 binding to the domain outside the catalytic site has any effect on its cleavage by Separase. Non-competitive inhibition of Separase by Sepin-1 suggests that Sepin-1 might bind to a third place on Separase, which is different from the two sites that Rad21 protein binds because Sepin-1 can inhibit Separase to cleave the peptide substrate (Rad21)-Rhl 10 that is small and might only bind to the catalytic site of Separase.
  • Example 1.6 Toxicity of Sepin-1 in mice
  • Applicants injected Sepin-1 at a single dose in the range of 50-100mg/kg body weight into C57B6 mice via tail vein. While the mice at the highest dose of 90mg/kg died within 30-90 minutes of injection, the mice injected with a dose of ⁇ 80mg/kg had no apparent effect, and have now survived for over 6 months. Blood analysis suggested that the mice receiving 90 mg/kg of Sepin-1 had high lactate dehydrogenase (LDH) (>500IU), suggesting muscle damage in these animals. However, the heart and skeletal muscles did not show light microscopic evidence of morphologic damage.
  • LDH lactate dehydrogenase
  • histopathology indicated no significant lesions in major organs, including heart, lungs, liver, kidney, eye, gall bladder, pancreas, spleen, brain, adrenal gland, cervical lymph nodes, salivary gland, stomach, small intestine, skeletal muscles, bone marrow, and ear.
  • mice [00103] Based on the above results, Applicants expanded this study to identify the Severely Toxic Dose (i.e., dose that causes death or irreversible severe toxicity, in 10% of rodents, STD10) using more mice injected at 75 to 90 mg/Kg range. A dose of 80mg/Kg was estimated as STD10 for Sepin-1 in these mice.
  • Sepin-1 was introduced to a cohort of 12 mice with a dose of lOmg/kg via intraperitoneal injection (IP) daily for six weeks, no weight loss or apparent adverse effects on the well-being of the mice were found.
  • IP intraperitoneal injection
  • Example 1.7 Sepin-1 inhibits the growth of cancer cells and tumors
  • FIG. 7 Breast epithelial cell line MCFlOf is not tumorigenic and has lower Separase level compared to isogenic MCF7 line (FIG. 7A). When treated with Sepin-1, MCF7 cells were over 3.3 times more sensitive than MCFlOf cells (FIG. 7B). The sensitivity of MCF7 cells to Sepin-1 treatment was also further verified from the growth of xenograft tumors derived from MCF7 cells in mice (FIG. 7C).
  • Applicants also investigated Sepin-1 activity against patient derived xenografts from two triple-negative (ER- PR- HER2-) breast tumors (MCI and BCM-5471). Compared to the BCM-5471 tumor, Separase is overexpressed in the MCI tumor. While Separase is primarily found in the mitotic cells of the BCM-5471 tumor, in MCI tumor it was found in majority of non-cycling (Ki67 staining negative) cells (FIG. 7D), as Applicants have previously described (Clin. Cancer Res., 2009, 15, 2703-2710). Once the tumor sizes reached ⁇ 200mg, mice were treated with Sepin-1 at a dose of lOmg/kg by intraperitoneal injection every day for 3 weeks.
  • Sepin-1 treatment inhibited the growth of xenograft MCI tumors by 70% over the three week treatment (FIG. 7E). However, there was no significant difference in growth between vehicle control and Sepin-1 treatment in xenograft BCM-5471 (FIG. 7F). Collectively, these data suggest that Sepin-1 inhibits tumor cell growth through selective inhibition of Separase.
  • Applicants used the MTT assay to test the inhibitory effect of Sepin- 1 on the growth of a variety of human cancer cell lines, including leukemia, breast cancer and neuroblastoma. The results are summarized in Table 1 and FIGS. 8A-C.
  • Parp-1 a chromatin-associated enzyme, catalyzes the poly ADP-ribosylation of proteins that are involved in chromatin architecture, DNA damage repair, and DNA metabolism. Parp-1 plays an important role in mediating the normal cellular response to DNA damage in non- apoptotic cells. However, it is cleaved by caspases in apoptotic cells, which is a marker of apoptosis. Although Sepin-1 induced cell growth inhibition appears to be via apoptosis, how Sepin-1 triggers the apoptosis remains to be determined.
  • Applicants also investigated Sepin-1 activity against patient derived xenografts from two triple-negative (ER ⁇ PR " HER2 " ) breast tumors (MCI and BCM-5471). Compared to the BCM-5471 tumor, Separase is overexpressed in the MCI tumor (FIG. 7D). While Separase is primarily found in the mitotic cells of the BCM-5471 tumor, it was found in majority of non- cycling (Ki67 staining negative) cells in MCI tumor (FIG. 7D), as Applicants have previously described.
  • mice were treated with Sepin-1 at a dose of 10 mg/kg by intraperitoneal injection every day for 3 weeks.
  • Sepin-1 treatment inhibited the growth of xenograft MCI tumors by 70% over the three week treatment (FIG. 7E).
  • FIG. 7F there was no significant difference in growth between vehicle control and Sepin-1 treatment in xenograft BCM-5471 (FIG. 7F), possibly due to low Separase level that causes less sensitivity to Sepin-1.
  • Sepin-1 -treated MFC7 xenograft tumors showed increase of apoptotic bodies in H&E stained sections (FIG.
  • HTS is an alternate approach that can be used to find potential Separase inhibitors.
  • Separase is an ideal drug target. Pharmacologic inhibition of Separase is a novel strategy to treat Separase overexpressed aneuploid tumors, at least because of the following reasons: 1) while homozygous deletion of Separase is embryonically lethal, mice with Separase haploinsufficiency live a normal life with no disease phenotype compared to the WT animal, indicating that reduction in Separase level has no adverse effect on an organism's wellbeing; 2) Separase, a promoter of aneuploidy, is a highly specific protease that is overexpressed in a large percentage (>60 ) of human breast tumors, as well as in prostate tumors and osteosarcoma, suggesting that attenuation of Separase level pharmacologically in human cancers may kill Separase-addicted aneuploid cells; and 3) Separase is not only overexpressed but also constitutively mislocalized to the nucleus of the human tumors, providing an opportunity to target the nuclear bound Sep
  • titrating down Separase level therapeutically may not only inhibit tumor cell proliferation, but also effectively reverse the aneuploid phenotype in Separase overexpressed tumor cells, while sparing the normal cells.
  • Separase mislocalization to nucleus can facilitate its targeting in the aneuploid tumor cells.
  • Applicants also reason that cells with knockdown nuclear Separase will initiate mitotic arrest and apoptosis, and may therefore be more sensitive to traditional chemotherapy.
  • the sources of the antibodies used in this Example are as follows: Actin mAb (Sigma, St. Louis, MO), Parp mAb (BD Bioscience, San Jose, CA), Separase mAb (Abnova, Taiwan), Caspase-3 pAb (Cell Signaling technology, Danvers, MA), Ki67 pAb (Vector Labs, Burlingame, CA), Rad21 pAb (Mol. Cell Biol., 200, 22, 8267-8277).
  • the substrate used in this Example is (Rad21)2-Rhl l0, which has two Rad21 peptide molecules (Asp-Arg-Glu-Ile-Nle-Arg) conjugated to one molecule of Rhodamine 110 (Rhl lO) (CPC Scientific, Sunnyvale CA).
  • the assay was set up using 384-well low volume black polystyrene plates (Corning, NY). Fifteen micro liter of activated Separase ( ⁇ 5ng) was mixed with 5 ⁇ 1 of test compound.
  • the cleavage buffer (CB) used to dilute reagents was (30mM Hepes- KOH pH7.7, 50mM NaCl, 25mM NaF, 25mM KC1, 5mM MgC12, 1.5mM ATP, and ImM EGTA). The mixture was incubated at room temperature for lh, and then 5 ⁇ 1 of 0.2mM substrate (Rad21)2-Rhl l0 was added.
  • the positive control contained Separase, substrate and CB without the test compound.
  • the negative control contained substrate and CB only.
  • HTS was performed at the John S. Dunn Gulf Coast Consortium for Chemical Genomics at the University of Texas Medical School at Houston.
  • the compound library used in HTS was the Maybridge HitFinderTM Collection that contains 14,400 compounds.
  • the compounds were stored in 180 96-well plates, 80 compounds per plate.
  • the concentration of the stock solution of each compound was lOmM in DMSO.
  • Each compound was diluted to 500 ⁇ in CB using a Biomek FX automation workstation (Beckman Coulter, Inc. Brea, CA) immediately before the assay was performed.
  • HTS was performed on 384-well plates using a Biomek NX Automation Workstation (Beckman Coulter, Inc.) to dispense the compounds and Separase enzyme.
  • the percent inhibition of Separase enzyme activity by compounds was calculated as 100* ⁇ l-[(RFIcompound-RFInegative)/(RFIpositive-RFInegative)] ⁇ . The compounds that inhibited more than 50% of Separase activity on both plate A and B were identified as hits.
  • Sepin-1 Applicants applied 200 ⁇ Sepin-1 (diluted in PBS) one time at an acute dose range of
  • MCF7 xenografts (ER+ PR+ HER2-) were generated by injection of one million cultured cells grown under standard in vitro conditions as previously described (Proc. Natl. Acad. Sci. U. S. A 105, 2008, 13033-13038).
  • Donor tumors for PDX lines MCI and BCM-5471 were divided into small fragments and retransplanted into the fat pat of recipient SCID Beige mice (5 mice per group). When the tumors reached approximately ⁇ 200mg, mice were treated with either vehicle or Sepin-1. Sepin-1 was injected at dose of lOmg/kg daily for 5 days a week for 3 weeks.
  • mice were washed with PBS for 3x30min; dehydrated for lh at 70%, 80%, 90% and 100% two times; and immerged in Xylene several times until the tissues became clear.
  • the samples were embedded in wax and sectioned with 5 ⁇ in thickness. All mice were maintained in accordance with the NIH Guide for the Care and Use of Laboratory Animals with approval from the Baylor College of Medicine Institutional Animal Care and Use Committee.
  • Myc-Rad21 protein was prepared by using pCS2MT Rad21 plasmid ⁇ Cancer Res., 1959, 19, 515-520) as the template and the TNT SP6 High- Yield Wheat Germ Protein Expression kit (Promega, Madison, WI). Myc-Rad21 protein was mixed with Separase with or without the presence of Sepin-1. The final concentration of Sepin-1 was 50 ⁇ . The reaction mixture was incubated in a 37°C water bath for 1 h. The cleavage of myc-Rad21 was immunoblotted with Rad21 pAb.
  • FIG. 10 provides data relating to separase overexpression in Pediatric glioblastoma (GBM) and the effect of Sepin-1 in inhibiting the growth of glioblastoma cells in vitro and in vivo.
  • FIG. 10A shows representative immunofluorescence pictures of normal brain (bottom panel) and a pediatric glioblastoma samples (#4687,top panel) showing Separase expression and nuclear localization in non-mitotic GBM cells but not in normal brain.
  • FIG. IOC provides MTT cytotoxicity assays showing efficient cell death of two Separase overexpressing primary pediatric glioblastoma samples.
  • FIG. 11A illustrates the Sepin-1 derivatives that were synthesized and examined.
  • the separase activity of the Sepin-1 derivatives shown in the box in FIG. 11A (compounds 27-31) were tested. The results are summarized in FIG. 11B.

Landscapes

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

Abstract

In some embodiments, the present disclosure pertains to compositions with compounds that inhibit Separase activity. In additional embodiments, the present disclosure pertains to methods of treating a tumor in a subject by administering one or more compositions of the present disclosure to the subject.

Description

TITLE
SEPARASE INHIBITORS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Patent Application No. 61/892,911, filed on October 18, 2013; and U.S. Provisional Patent Application No. 61/936,976, filed on February 7, 2014. The entirety of each of the aforementioned applications is incorporated herein by reference.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[0002] This invention was made with government support under Grant No. 1RO1 CA109330, awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND
[0003] Currently, there is no method or composition to target human tumors with abnormal chromosome numbers (aneuploidy). Moreover, current methods and compositions for treating various types of tumors have limitations in terms of efficacy and specificity. Therefore, more effective compositions and methods are needed to address the above limitations, especially for tumors with aneuploidy.
SUMMARY
[0004] In some embodiments, the present disclosure pertains to compositions that inhibit Separase activity. In some embodiments, the compositions of the present disclosure include one or more compounds with one or more of the following structures:
Figure imgf000003_0001
(i) (2) (3)
Figure imgf000004_0001
(4) (5) (6)
Figure imgf000004_0002
(7) (8) (9)
Figure imgf000004_0003
(10) (11) (12)
Figure imgf000004_0004
O
(13) [0005] In some embodiments, R (in structures 1-13) and R1A (in structures 3 and 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof. In some embodiments, R (in structures 1-13) is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof. In some embodiments, R2 (in structures 1-4 and 6-13) is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.s), C(CH3)(R7.s), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R7 5 (in structures 1-4 and 6-13) are each selected from the group consisting of alkanes, CH , CH2CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
[0006] In some embodiments, R4 (in structure 5) is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH3, phenyl, O-phenyl, NH-phenyl, and combinations thereof. In some embodiments, R5 (in structure 5) is selected from the group consisting of NH, NR6, S, O, and combinations thereof. In some embodiments, R6 (in structure 5) is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof.
[0007] In more specific embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000005_0001
[0008] In some embodiments, R in structure 1 is N02, and R2 in structure 1 is C(CH )2 . In some embodiments, Ri in structure 1 is Br and R2 in structure 1 is CS.
[0009] In more specific embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000006_0001
[0010] In some embodiments, R in structure 4 is Br, and R2 in structure 4 is CS.
[0011] In more specific embodiments, the compositions of the present disclosure include more compounds with the following structure:
Figure imgf000006_0002
[0012] In some embodiments, Rg is selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
[0013] In further embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000006_0003
[0014] In some embodiments, the compounds of the present disclosure are water soluble. In some embodiments, the compounds of the present disclosure are selective inhibitors of Separase. In some embodiments, the compounds of the present disclosure are non-competitive inhibitors of Separase. [0015] In additional embodiments, the present disclosure pertains to methods of treating a tumor in a subject by administering one or more compositions of the present disclosure to the subject. In some embodiments, the tumor comprises Separase overexpressed tumor cells, such as Separase overexpressed mammary tumor cells or Separase overexpressed aneuploid cells.
[0016] In some embodiments, the tumor to be treated by the compositions of the present disclosure is associated with at least one of breast cancer, ovarian cancer, leukemia, thyroid cancer, neuroblastoma, brain cancer, lung cancer, colon cancer, gastrointestinal cancer, prostate cancer, osteosarcoma, glioblastoma, and combinations thereof. In some embodiments, the tumor to be treated by the compositions of the present disclosure is associated with breast cancer, such as triple negative breast cancer and luminal-B subtype endocrine resistant breast cancer.
[0017] In some embodiments, the subjects to be treated by the compositions of the present disclosure are human beings, such as human beings suffering from cancer. In some embodiments, the compositions of the present disclosure are administered to subjects by intravenous administration.
DESCRIPTION OF THE FIGURES
[0018] FIGURE 1 provides structures of various separase inhibitors and potential separase inhibitors. FIG. 1A provides structures of 2,2-dimethyl-5-nitro-2H-benzimidazole trihydrate (left panel), SYC-764 (center), and SYC-763 (right panel). FIGS. 1B-C provides structures of various derivatives of the compounds in FIG. 1A. FIG. ID provides the structure of Sepin-1. FIG. IE provides structures of various Sepin-1 derivatives.
[0019] FIGURE 2 provides data relating to the development of a fluorogenic Separase assay using (Rad21)2-Rhl l0 as a substrate. FIG. 2A shows a Rhl lO standard curve. The fluorescence intensity expressed by the relative fluorescent units (RFU) is linearly proportional to the amount of Rhl lO, which can be used to determine the amount of Rhl lO formation in Separase assay. FIG. 2B shows that the amount of free Rhl 10 released from (Rad21)2-Rhl 10 is a function of the substrate concentration. The amount of Separase used in this experiment was 5ng/well. FIG. 2C shows the amount of free Rhl lO released from (Rad21)2-Rhl l0 was correlated to the amount of Separase enzyme concentration. The amount of Rhl 10 used in this experiment was 1 nmol/well. FIG. 2D shows optimization of incubation time for the fluorogenic Separase assay. After Separase and (Rad21)2-Rhl l0 were mixed, the reaction mixtures were incubated at 25°C or 37°C. The fluorescence intensity was determined every 30 min. Bar = mean of 3 samples +SD. pmol Rhl 10/h on the second y-axis in FIGS. 2B and 2C was calculated using the standard curve shown in FIG. 2A.
[0020] FIGURE 3 shows results relating to the kinetics of the Separase assay. The cleavage reaction was carried out in the presence of the increased concentration (0.015-2 mM) of (Rad21)2-Rhl l0 and 5ng of Separase. The total reaction volume was 25μ1. The plates were incubated at 37 °C for 3h before the fluorescence intensity was determined. The fluorescence intensity was converted to pmol of Rhl lO. Michaelis-Menten curves were plotted (FIG. 3A), and Vm and Km were calculated using Lineweaver-Burt plot (FIG. 3B).
[0021] FIGURE 4 shows results relating to the stability of Separase enzyme and substrate and the effect of TEV and DMSO on Separase assay. FIG. 4A shows aliquots of diluted Separase in working concentration were kept on ice for 8h and then stored at -80, -20 or 4 °C for 24h. Their activity was analyzed in comparison to the same concentration of Separase that was freshly prepared from stock solutions. FIG. 4B shows diluted (Rad21)2-Rhl l0 in working concentration was stored at -80 or 4 °C for 24h. The activity was analyzed using Separase assay. The same concentration of (Rad21)2-Rhl l0 that was freshly prepared from stock solution was used as control. FIG. 4C shows results where TEV protease and Separase were used to hydrolyze (Rad21)2-Rhl l0. The enzyme activity was determined after incubation at 37 °C for 3h. FIG. 4D shows the effect of DMSO concentration on Separase assay.
[0022] FIGURE 5 shows high throughput screening for Separase inhibitors. FIG. 5A shows the reproducibility of the screening. Each data point was repeated in two plates (A and B) and plotted with the Separase activity of A plate as x-axis and that of B plate as y-axis. A total of 9 representative duplicated 384-well plates with 3,456 data points are shown. FIG. 5B shows RFU of Positive controls is well separated from that of the negative controls. 576 data points of positive controls and negative controls, respectively, from 18 384-well plates are shown, σ is STDEV. FIG. 5C is a chart summarizing the robustness of high throughput screening (HTS). 14,400 compounds in 180 96-well plates were screened using 384-well plates in duplicates (A and B), with a total of 90 384-well plates. The value of signal to background ratio (S:B), signal to noise ratio (S:N) and the Z' factor is the mean of 90 plates, while the value of S:B range (A:B), S:N range (A:B) and Z' range (A:B) is the mean of 45 A plates or B plates. FIG. 5D shows results demonstrating the inhibition of Separase activity by the test compounds. The test compounds that inhibited at least 50% of Separase activity in both A plates and B plates were identified. The data points in the two upper right boxes showed the compounds that inhibited Separase activity by >50% and >80%, respectively. FIG. 5E illustrates that, among the 14,400 compounds assayed, 97 are found to inhibit Separase activity by >50%. The hit rate is 0.67%. 24 out of the 97 compounds inhibit Separase activity by >80%. Five of the 97 compounds were confirmed.
[0023] FIGURE 6 provides results relating to the characterization of Sepin-1 in inhibiting Separase activity. FIG. 6A provides a structure of Sepin-1. FIG. 6B shows that Sepin-1 inhibits the cleavage of Separase substrate Rad21. The in vitro transcribed and translated Myc-Rad21 was used as the substrate for activated Separase in the presence of or without Sepin-1. The final concentration of Sepin-1 in the reaction mixture was 50μιη.
Figure imgf000009_0001
molecular weight). FIG. 6C shows the inhibition of Separase activity by Sepin-1. The IC50 was 14.8μΜ, calculated using the KaleidaGraph program. The Inset shows a four-parameter dose -response curve fit with an estimated IC50 of 15.8 μΜ. (n=3+SE). FIG. 6D shows the non-competitive inhibition Separase enzymatic activity by Sepin-1. Sepin-1 and Separase were mixed 30 min before substrate (Rad21)2-Rhl l0 was added. After 3h incubation, the fluorescence intensity of Rhl lO was determined. The data were shown in Lineweaver-Burt plot. Sepin-1 reduced the speed (Vmax) of substrate conversion by Separase, but not the Km.
[0024] FIGURE 7 shows that Sepin- 1 selectively inhibits the growth of breast cancer cells and patient-derived triple negative breast cancer xenografts overexpressing Separase protein. FIG. 7A shows an immunoblot of the Separase protein in breast cancer cell lines, MCF7 and MCFlOf. FIG. 7B shows results of an MTT assay where MCF7 is more sensitive to Sepin-1 in inhibiting cell growth than MCFlOf ex vivo. FIG. 7C shows that Sepin-1 inhibits the growth of xenograft tumor derived from MCF-7 in SCID-beige mice. FIG. 7D shows the immuno staining of Separase in triple negative breast cancer tumor xenografts MCI and BCM-5471. Tumor sections were stained with Separase mAb (green) and Ki67 pAb (red). DNA was stained with DAPI (blue). FIGS. 7E-F shows results where Sepin-1 inhibits the growth of xenograft tumors derived from breast cancer tumors in SCID-beige mice. BCM-5471 xenograft tumors are more sensitive to Sepin-1 than MC-1 tumors. The xenograft tumors' weight was assessed over three week period of Sepin-1 or vehicle treatment in SCID-beige mice at a dose of 10 mg/kg daily via intraperitoneal injection for 5 days a week (N=5/group). FIGS. 7G-H shows that Sepin-1- treated MCF-7 xenograft tumors have high level of apoptosis. Tumor sections from MCF-7 xenografts treated with Sepin-1 or vehicle were stained with H&E (FIG. 7G) or immunostained with cleaved caspase-3 mAb (FIG. 7H). Apoptotic bodies in H&E staining (black arrows) and immuno staining (green) were observed in Sepin-1 -treated tumors.
[0025] FIGURE 8 shows that Sepin-1 inhibits cancer cell growth and induces apoptosis. FIGS. 8A-C shows that Sepin-1 inhibits cell growth of leukemia cell lines (FIG. 8A), breast cancer cell lines (FIG. 8B), and Neuroblastoma cell lines (FIG. 8C). Cells were treated with serially diluted Sepin-1 for 72h. The cell viability was assessed using MTT assay. FIG. 8D shows a western blot of full length (FL) and N-terminal (NT, surrogate for the active protein) Separase in cancer cell lines. FIG. 8E shows a positive correlation between Separase protein level and sensitivity to Sepin-1 in inhibiting cell growth (IC50). The Separase bands from (D) were quantified and normalized with beta actin. FIG. 8F shows immunoblotting shown Sepin-1 - induced activation of caspase-3 and cleavage of poly (ADP-ribose) polymerase (Parp). Molt4 cells were treated with various concentrations of Sepin-1 for 24 h. Etoposide (Etop) was used as a control. CI Parp refers to cleaved Parp. CI Casp-3 refers to cleaved Caspase-3. * indicates nonspecific bands.
[0026] FIGURE 9 provides additional data relating to the identification of leads in HTS for Separase inhibitors. FIG. 9A provides structures of the 5 lead compounds. FIG. 9B provides dose-dependent inhibition of Separase activity by Sepin 1 and 2.
[0027] FIGURE 10 provides data relating to separase overexpression in Pediatric glioblastoma (GBM) and the effect of Sepin-1 in inhibiting the growth of glioblastoma cells in vitro and in vivo.
[0028] FIGURE 11 provides additional data relating to structure- activity studies of Sepin-1 derivatives. FIG. 11A shows the structures of Sepin-1 derivatives that were tested. FIG. 11B shows data relating to the separase inhibition activities of the Sepin-1 derivatives. The numbers (28-31 in FIG. 11B) correspond to structures shown in FIG. 11A.
DETAILED DESCRIPTION
[0029] It is to be understood that both the foregoing general description and the following detailed description are illustrative and explanatory, and are not restrictive of the subject matter, as claimed. In this application, the use of the singular includes the plural, the word "a" or "an" means "at least one", and the use of "or" means "and/or", unless specifically stated otherwise. Furthermore, the use of the term "including", as well as other forms, such as "includes" and "included", is not limiting. Also, terms such as "element" or "component" encompass both elements or components comprising one unit and elements or components that comprise more than one unit unless specifically stated otherwise.
[0030] The section headings used herein are for organizational purposes and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated herein by reference in their entirety for any purpose. In the event that one or more of the incorporated literature and similar materials defines a term in a manner that contradicts the definition of that term in this application, this application controls.
[0031] Separase is a protease evolutionarily conserved from yeast to human. Its primary function is to cleave the cohesin subunit Sccl/Mcdl/Rad21 at the onset of anaphase, resulting in the dissolution of cohesin rings and independent segregation of sister chromatids into the two daughter cells.
[0032] Recent studies indicate that Separase is overexpressed and mislocalized in a number of human tumors, including breast, prostate and osteosarcoma. In fact, Separase overexpression is reported in a broad range of human tumors. In addition, Separase overexpression in mouse models results in tumorigenesis. Furthermore, Separase is significantly overexpressed in over 60% of human breast tumors compared to the matched normal breast tissue. In addition, the conditional overexpression of Separase in mammary epithelial cells is sufficient to induce separation of sister chromatids, aneuploidy and tumorigenesis. These data suggest that abnormal Separase expression and mislocalization is a driver of aneuploidy and tumorigenesis. [0033] Separase activity is tightly regulated during the cell cycle and is inactive when it binds to its inhibitory chaperone, securin. Separase is also inhibited via phosphorylation at Serl l26 by Cyclin B-Cdkl kinase and binding to Cyclin B. With the progression of cell cycle to the onset of anaphase, the anaphase promoting complex/cyclosome polyubiquitinates mitotic cyclin and securin, which are rapidly degraded by the 26S proteasome. Once enzymatically activated, Separase auto-cleaves, resulting in N-terminal and C-terminal fragments, which still associate with each other. The main function of Separase is to proteolytically cleave the cohesin subunit
Rad21.
[0034] Because Separase is overexpressed in human tumors, and its overexpression results in chromosomal missegregation and aneuploidy, pharmacological inhibition of Separase presents a novel strategy to target chromosomal missegregation induced tumorigenesis. Pharmacologic modulation of Separase activity remains elusive, however, due to a lack of small molecular compound inhibitors.
[0035] In some embodiments, the present disclosure pertains to compositions that inhibit Separase activity. In some embodiments, the present disclosure pertains to methods of treating a tumor in a subject by administering the compositions of the present disclosure to the subject.
[0036] Compositions
[0037] In some embodiments, the compositions of the present disclosure include one or more compounds that inhibit Separase activity. In some embodiments, the compounds include one or more of the following structures:
Figure imgf000013_0001
(1) (2) (3)
Figure imgf000014_0001
(4) (5) (6)
Figure imgf000014_0002
II If
o o o
(7) (8) (9)
Figure imgf000014_0003
(10) (11) (12)
Figure imgf000014_0004
(13) [0038] One skilled in the art will recognize that the above structures can represent numerous different compounds. Exemplary compounds represented by the above structures are illustrated in FIGS. 1A-E.
[0039] One skilled in the art will also recognize that structures 1-13 can resonate in different manners. For instance, in some embodiments, structures 7-10 may represent different resonating structures of the same compounds. Likewise, in some embodiments, structures 11-13 may resonate to resemble the compounds disclosed in structures 7-10. Therefore, various embodiments of the present disclosure also cover resonating versions of structures 1-13 that may not be disclosed in the aforementioned structures.
[0040] Additional compounds that represent variations of the above structures can also be envisioned. For instance, in some embodiments, R (in structures 1-13) and R1A (in structures 3 and 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2- phenyl, S03H, and combinations thereof. In some embodiments R (in structures 1-13) is N02. In some embodiments, Ri and R1A (in structures 3 and 11-13) are CI. In some embodiments, Ri (in structures 1-13) is N02, and R1A (in structures 3 and 11-13) is Br.
[0041] In some embodiments, R (in structures 1-13) is selected from the group consisting of H, OH, CH3, C4Hg, alkyl groups, and combinations thereof. In some embodiments, R3 (in structures 1-13) is H.
[0042] In some embodiments, R2 (in structures 1-4 and 6-13) is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.s), C(CH3)(R7 5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R7 5 (in structures 1-4 and 6-13) are each selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4Hg, CO, CS, and combinations thereof. In some embodiments, R2 (in structures 1-4 and 6-13) is C(CH3)2. [0043] In some embodiments, R4 (in structure 5) is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH3, phenyl, O-phenyl, NH-phenyl, and combinations thereof. In some embodiments, R4 (in structure 5) is CH3.
[0044] In some embodiments, R5 (in structure 5) is selected from the group consisting of NH, NR6, S, O, and combinations thereof. In some embodiments, R5 (in structure 5) is NH. In some embodiments, R6 (in structure 5) is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof.
[0045] In more specific embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000016_0001
[0046] In some embodiments, R in structure 1 is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4Hg, phenyl, CH2-phenyl, S03H, and combinations thereof. In some embodiments, R3 in structure 1 is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof. In some embodiments, R in structure 1 is H. In some embodiments, R2 in structure 1 is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R7 5 are each selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof. In some embodiments illustrated in FIG. 1A (left panel), Ri in structure 1 is N02, and R2 in structure 1 is C(CH )2 . In some embodiments illustrated in FIG. 1A (middle panel), Ri in structure 1 is Br, and R2 in structure 1 is CS.
[0047] In more specific embodiments, the compositions of the present disclosure include more compounds with the following structure:
Figure imgf000017_0001
[0048] In some embodiments, R in structure 4 is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof. In some embodiments, R in structure 4 is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof. In some embodiments, R in structure 4 is H. In some embodiments, R2 in structure 4 is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R7 5 are each selected from the group consisting of alkanes, CH , CH2CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof. In some embodiments illustrated in FIG. 1A (right panel), Ri in structure 4 is Br, and R2 in structure 4 is CS.
[0049] In more specific embodiments, the compositions of the present disclosure include one or more compounds with one or more of the following structures:
Figure imgf000018_0001
(7) (8) (9)
Figure imgf000018_0002
(10) (ID (12)
Figure imgf000018_0003
(13)
[0050] In some embodiments, Ri (in structures 7-13) and RIA (in structures 11-13) are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo- reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof. In some embodiments, R3 (in structures 7-13) is selected from the group consisting of H, OH, CH3, C4Hg, alkyl groups, and combinations thereof. In some embodiments, R2 (in structures 7-13) is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R7 5 (in structures 7-13) are each selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
[0051] In more specific embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000019_0001
[0052] In some embodiments, R and R1A in structure 12 are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof. In some embodiments, R in structure 12 is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof. In some embodiments, R3 in structure 12 is H. In some embodiments, R2 in structure 12 is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.s), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof. In some embodiments, R7 and R 5 are each selected from the group consisting of alkanes, CH3, CH2CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof. In some embodiments, R in structure 12 is N02, RiA in structure 12 is Br, and R2 in structure 12 is C(CH3)2.
[0053] In further embodiments, the compositions of the present disclosure include one or more compounds with the following structure:
Figure imgf000020_0001
[0054] In some embodiments, Rg is selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
[0055] In more specific embodiments, the compositions of the present disclosure include one or more compounds with the following structure (referred to as Sepin-1 and also shown in FIG. ID):
Figure imgf000020_0002
[0056] In more specific embodiments, the compositions of the present disclosure include one or more derivatives of Sepin-1. In some embodiments, the Sepin-1 derivatives include, without limitation, one or more of the structures shown in FIG. IE.
[0057] Composition content
[0058] In various embodiments, the compositions of the present disclosure can contain one or more of the aforementioned compounds that inhibit Separase activity. In some embodiments, the compositions of the present disclosure can also have one or more physiologically acceptable carriers or excipients. In some embodiments, the compositions of the present disclosure can also include formulation materials for modifying, maintaining, or preserving various conditions, including pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, and/or adsorption or penetration of the compounds. Suitable formulation materials include, but are not limited to: amino acids (e.g., glycine); antimicrobials; antioxidants (e.g., ascorbic acid); buffers (e.g., Tris-HCl); bulking agents (e.g., mannitol and glycine); chelating agents (e.g., EDTA); complexing agents (e.g., hydroxypropyl-beta-cyclodextrin); and the like.
[0059] Compound properties
[0060] The compounds in the compositions of the present disclosure can have various properties. For instance, in some embodiments, the compounds of the present disclosure are water soluble. In some embodiments, the compounds of the present disclosure inhibit Separase activity. In some embodiments, the compounds of the present disclosure are non-competitive inhibitors of Separase.
[0061] In more specific embodiments, the compounds of the present disclosure are selective inhibitors of Separase. For instance, in some embodiments, the compounds of the present disclosure inhibit Separase activity without significantly inhibiting the activity of other enzymes. In more specific embodiments, the compounds of the present disclosure inhibit Separase activity without significantly inhibiting the activity of cysteine proteases other than Separase (e.g., Caspases, tissue kallikrein, coagulation factor Xa, and other proteases that cleave after arginine at PI position).
[0062] In some embodiments, the compounds of the present disclosure inhibit the growth of tumor cells. For instance, in some embodiments, the compounds of the present disclosure can inhibit the growth of Separase overexpressed tumor cells. In some embodiments, the compounds of the present disclosure can selectively inhibit the growth of Separase overexpressed tumor cells without significantly affecting the growth of normal cells. In some embodiments, the compounds of the present disclosure can selectively inhibit the growth of Separase overexpressed aneuploid cells. [0063] In some embodiments, the compounds of the present disclosure inhibit the growth of tumor cells at IC50 concentrations that range from about 0.5 μιη to about 65 μιη. In some embodiments, the compounds of the present disclosure inhibit the growth of tumor cells at IC50 concentrations that range from about 1 μιη to about 10 μιη.
[0064] Methods of treating Tumors
[0065] In view of the above properties, the compounds of the present disclosure can have numerous applications in treating tumors. As such, additional embodiments of the present disclosure pertain to methods of treating various tumors in various subjects by administering one or more compositions of the present disclosure to the subject.
[0066] Tumors
[0067] The compositions of the present disclosure can be utilized to treat various types of tumors. For instance, in some embodiments, the tumors to be treated include Separase overexpressed tumor cells. In more specific embodiments, the tumors to be treated include Separase overexpressed mammary tumor cells, Separase overexpressed aneuploid cells, and combinations thereof.
[0068] In some embodiments, the tumors to be treated by the compositions of the present disclosure may be associated with various types of cancers. For instance, in some embodiments, the tumors to be treated may be associated with at least one of breast cancer, leukemia, thyroid cancer, neuroblastoma, brain cancer, lung cancer, colon cancer, prostate cancer, osteosarcoma, glioblastoma, ovarian cancer, gastrointestinal cancer and combinations thereof. In some embodiments, the tumors to be treated are associated with breast cancer. In some embodiments, the tumors to be treated are associated with triple negative breast cancer and luminal-B subtype endocrine resistant breast cancer.
[0069] Without being bound by theory, the compositions of the present disclosure can treat tumors by various mechanisms. For instance, in some embodiments, the compounds in the compositions of the present disclosure inhibit the growth of tumor cells associated with a tumor. In some embodiments, the compounds in the compositions of the present disclosure selectively inhibit the growth of Separase overexpressed tumor cells associated with the tumor. In some embodiments, the compounds in the compositions of the present disclosure selectively inhibit the growth of Separase overexpressed aneuploid cells associated with the tumor.
[0070] Subjects
[0071] The compositions of the present disclosure can be used to treat tumors in various subjects. For instance, in some embodiments, the subject is a human being. In some embodiments, the subject is a human being suffering from cancer. In additional embodiments, the subjects may be non-human animals, such as mice, rats, other rodents, or larger mammals, such as dogs, monkeys, pigs, cattle and horses.
[0072] Administration
[0073] The compositions of the present disclosure can be administered to subjects by various methods. For instance, the compositions of the present disclosure can be administered by oral administration (including gavage), inhalation, subcutaneous administration (sub-q), intravenous administration (I.V.), intraperitoneal administration (LP.), intramuscular administration (I.M.), intrathecal injection, and combinations of such modes. In further embodiments, the therapeutic compositions of the present disclosure can be administered by topical application (e.g, transderm, ointments, creams, salves, eye drops, and the like). In more specific embodiments, the compositions of the present disclosure can be administered intravenously. Additional modes of administration can also be envisioned.
[0074] In various embodiments, the compositions of the present disclosure may also be administered in a single dose or multiple doses throughout a time period. For instance, in some embodiments, the compositions of the present disclosure may be administered to a subject in two separate doses.
[0075] In addition, the compositions of the present disclosure may be administered to localized sites in a subject, such as tissue or vasculature that contain a tumor. For instance, the compositions of the present disclosure may be injected directly into an area of a subject that displays tumor growth.
[0076] In various embodiments, the compositions of the present disclosure may be coadministered with other therapies. For instance, in some embodiments, the compositions of the present disclosure may be co-administered along with anti-cancer drugs. [0077] Additional Embodiments
[0078] Reference will now be made to more specific embodiments of the present disclosure and experimental results that provide support for such embodiments. However, Applicants note that the disclosure herein is for illustrative purposes only and is not intended to limit the scope of the claimed subject matter in any way.
[0079] Example 1. Identification and Characterization of Separase Inhibitors (Sepins) for Cancer Therapy
[0080] In this Example, Applicants report a high throughput screening for Separase inhibitors (Sepins). Applicants developed a fluorogenic Separase assay using Rhodamine 110 conjugated Rad21 peptide as substrate and screened a small molecule compound library. In particular, Applicants have screened a chemical library with 14,400 small compounds and identified five compounds that inhibited Separase activity. Applicants have further characterized Sepin-1, a noncompetitive inhibitor of Separase, which can inhibit the growth of cancer cell lines and mammary xenograft tumors in mice by inducing apoptosis. In particular, Applicants have observed that Sepin-1 inhibits Separase enzymatic activity with an IC50 of 14.8 μΜ. Applicants have also found that the sensitivity to Sepin-1 in most cases is positively correlated to the level of Separase in both cancer cell lines and tumors.
[0081] Example 1.1. Development of Separase activity assay using (Rad21)2-Rhl l0 as a substrate
[0082] 7-amido-4-mrthyl coumaric acid (AMC) is a commonly used dye in protease assays, and Applicants previously reported a fluorogenic Separase assay using AMC-labeled Rad21 peptide as the substrate (Anal. Biochem., 2009, 392, 133-138). One of the caveats of AMC-based protease assays is that the results from such assays can potentially be biased and misinterpreted due to the interference of label and compound auto fluorescence at the excitation and emission wavelength of 350 nm and 500nm, respectively. These interfering factors can be significantly reduced by using redshift dyes, such as Rhodamine 110 (Rhl lO), which has been widely used in various protease assays. [0083] Rad21 peptide (Asp-Arg-Glu-Ile-Nle-Arg) that contains Separase cleaving consensus motif (Glu-X-X-Arg) was conjugated to Rhl lO. Two Rad21 peptides were linked to one Rhl lO molecule forming a bisamide, and the product was named (Rad21)2-Rhl 10.
[0084] Because the fluorophore is in the lactone state, (Rad21)2-Rhl l0 virtually has no autofluorescence. Once the amide bonds between the Rad21 peptide and Rhl lO moiety is cleaved by Separase, the Rahl lO moiety in the Rad21-Rhl l0 switches its lactone state to a quinine state, which exhibits significant increase of fluorescence intensity. The fluorescence intensity (relative fluorescence unit, RFU) increases proportionally with the increase of the amount of free Rhl lO, measured at
Figure imgf000025_0001
(FIG. 2A). The standard curve of RFU versus Rhl 10 can be used to calculate the amount of Rhl 10 formed during the Separase assay.
[0085] Applicants tested the relationship of the increasing concentrations of substrate and enzyme combinations. The amount of Rhl lO released was linearly correlated with the increase of (Rad21)2-Rhl l0 concentration between 0-2nmol/well (FIG. 2B). Similarly, the amount of Rhl lO released was a linear function with the increase of Separase concentration between 0- lOng/well (FIG. 2C).
[0086] In addition, incubation temperature significantly affects the kinetics of Separase. The release of Rhl lO catalyzed by Separase was much faster at 37 °C than that at room temperature (25 °C) (FIG. 2D). The amount of Rhl lO released from (Rad21)2-Rhl l0 linearly increased between 0 and 3h (FIG. 2D). Both di- and mono-substituted Rad21-Rhl l0 can be cleaved by Separase. The fluorescence intensity of mono-substituted Rad21-Rhl l0 is about 10% of free Rhl lO. Taking the assay efficiency and the cost effectiveness into consideration, Applicants developed the Separase assay using 5ng (~21.7fmol) of Separase and lnmol of (Rad21)2-Rhl l0 in 25μ1 reaction volume with 3h incubation at 37 °C. This assay condition was adapted for the high throughput screening for Separase inhibitors that is described below.
[0087] Example 1.2. Kinetics of hydrolysis of (Rad21 2-Rhl l0 by Separase
[0088] To investigate the kinetics of (Rad21)2-Rhl l0 cleavage by Separase, Applicants incubated increasing concentrations of the substrate with 5 ng of Separase for 3h at 37 °C. Plotting the rate of (Rad21)2-Rhl l0 cleavage by Separase as determined by the formation of Rhl lO versus the substrate concentration could be fit to the classic Michaelis-Menten hyperbola. The rate of Rhl lO production at lower concentration of the substrate was proportional to the concentration of substrate, while that at higher concentration of substrate was approaching saturation (FIG. 3A). The Vmax (4.37 pmol Rhl lO/h, or 201 pmol Rhl lO/h/pmol Separase) and Km (123μΜ) were calculated using a Lineweaver-Burk plot (FIG. 3B).
[0089] Example 1.3. Optimization of the assay conditions
[0090] The stability of enzyme and substrate at the working conditions is important because the process of distributing different components in high throughput screening is complicated and time-consuming. To test stability of the enzyme and substrate, Applicants diluted Separase enzyme and (Rad21)2-Rhl l0 to working concentration, and kept them at 4 °C for 8h. The diluted enzyme and substrate were frozen at -20 °C or -80 °C or stored at 4 °C for 24h before their activities were determined. The activity of Separase at the working concentration had no significant difference when it was kept at 4 °C, -20°C, or -80 °C for 24h. However, compared to the freshly prepared Separase, the activity of diluted Separase was reduced by 20% (FIG. 4A). In contrast, (Rad21)2-Rhl l0 at working concentration was unaffected by storage by storage at 4 °C or -80 °C for 24h (FIG. 4B).
[0091] The construct that was used to express Separase in 293T cells contains two IgG binding domains of protein A (Z-domain), followed by four TEV-pro tease cleavage sequences on the N terminus of Separase (ZZ-TEV4-Separase). TEV protease was used to release the Separase from IgG-Agarose beads after activation by Xenopus cytostatic factor extract. To eliminate the possibility that TEV interferes with the Separase assay by cleaving (Rad21)2-Rhl l0, Applicants tested the activity of TEV and Separase in hydro lyzing (Rad21)2-Rhl l0 in parallel. The results showed that TEV could not cleave (Rad21)2-Rhl l0 (FIG. 4C), suggesting TEV does not interfere with the Separase assay.
[0092] Because Separase is a caspase-like protease, Applicants also tested whether caspases can cleave (Rad21)2-Rhl l0. Results indicated that caspase-3 and -7, two major effectors, could not cleave (Rad21)2-Rhl l0 (data not shown). [0093] The stock solution of the substrate and small compounds used in high throughput screen (HTS) was prepared in DMSO. To investigate the effect of DMSO on the Separase assay, Applicants tested different concentrations of DMSO in the reaction mixture with final concentration ranging from 2% to 38%. The assay was not affected by 2-14% of DMSO, but it was inhibited with the increase of DMSO concentration of >14%. The IC50 was -27% (FIG. 5D). Because the DMSO concentration in Separase assay used in HTS was less than 3%, Applicants conclude that DMSO has no significant effect in this assay.
[0094] Example 1.4. High throughput screening for Separase inhibitors
[0095] After two successful test runs in the HTS setting, Applicants performed screening for Separase inhibitors using the Maybridge HitFinder™ Collection that contains 14,400 compounds. Each compound was assayed in duplicate on two different plates (Plate A and Plate B). The reproducibility of the two plates could be assessed by plotting RFU of A plate versus RFU of B plate (FIG. 5A). Applicants analyzed 9 representative duplicated plates with 3,456 data points, including 288 positive controls, 288 background controls and 2,880 compound assay samples. They were highly reproducible with R2=0.9324 (FIG. 5B). Except for a few data points (6 at the upper left corner and 1 at the middle right in FIG. 5B), the majority of positive controls and all background controls were within the range of mean + 3σ (FIG. 5B). The overall quality of the screening was high and robust with Signal to Background ratio (S:B) 10.54, Signal to Noise ratio (S:N) 46.35 and Z' factor 0.55 (FIG. 5C). Z' factor reflects the signal dynamic range and the data variation associated with the signal measurements. The range of Z' factor is between 0 and 1. A Z' factor >0.5 indicates the assay was optimal.
[0096] After the percentage of Separase activity inhibited by the test compounds was calculated, those compounds that reduced Separase activity by more than 50% on both plate A and B were identified as active compounds (FIG. 5D). The majority of the compounds had very little effect on the activity of Separase, which were concentrated on the lower left corner of FIG. 5D, while a few compounds appeared on the upper right corner of FIG. 5D, inhibiting Separase activity at least by >50% (bigger square box) or by >80% (smaller square box).
[0097] Applicants have identified 97 compounds that inhibited Separase activity by more than 50%. Among the 97 compounds, 24 reduced more than 80% of Separase activity (FIG. 5E). Applicants ordered all of these 97 compounds and verified the activities of these compounds in inhibiting Separase enzymatic activity. Five of them were confirmed, which were named Separase inhibitors (Sepin-1 to 5, shown in FIG. 9A). Among them, Sepin-1 had the highest activity, which was further characterized below. Also see FIG. 9B.
[0098] Example 1.5. Characterization of Sepin-1 in inhibiting Separase activity
[0099] Sepin-1 is 2,2-dimethyl-5-nitro-2H-benzimidazole-l,3 dioxide with molecular weight 223 Da and melting point 228°C (FIGS. ID and 6A). Applicants used 13C NMR to confirm the identity of Sepin-1 before use for further experimentation. Sepin-1 could inhibit the Rad21 cleavage by activated Separase in vitro when in vitro transcribed and translated Rad21 was used as the substrate (FIG. 6B). In the fluorogenic Separase activity assay using (Rad21)2-Rhl l0 as the substrate, the concentration of Sepin-1 to inhibit 50% of Separase activity (IC50) was 14.8 μΜ (FIG. 6C). To examine the kinetics of Separase inhibition by Sepin-1, different amounts of Sepin-1 compound were used to inhibit the cleavage of (Rad21)2-Rhl l0 by Separase. The data showed that Sepin-1 did not affect the Km but reduced the Vmax (FIG. 6D), suggesting the binding site of Sepin-1 on Separase is different from that of the substrate. To further confirm the specificity of Sepin-1 binding to Separase, 0.1% Tween-20 or 1% BSA was included in the reaction mixture. The results indicated that Tween-20 and BSA did not affect the kinetics of Sepin-1 in inhibiting Separase activity (data not shown). Based on this result Applicants conclude that the mode of Sepin-1 inhibition of Separase enzymatic activity is non-competitive. The results also indicate that Sepin- 1 is not a promiscuous inhibitor.
[00100] In yeast, Separase has at least two sites interacting with Rad21. One is the catalytic site, and the other is outside the catalytic site. Although blocking the catalytic site does not affect Rad21 binding to Separase, it is unclear whether Rad21 binding to the domain outside the catalytic site has any effect on its cleavage by Separase. Non-competitive inhibition of Separase by Sepin-1 suggests that Sepin-1 might bind to a third place on Separase, which is different from the two sites that Rad21 protein binds because Sepin-1 can inhibit Separase to cleave the peptide substrate (Rad21)-Rhl 10 that is small and might only bind to the catalytic site of Separase.
[00101] Example 1.6. Toxicity of Sepin-1 in mice [00102] To investigate the toxicity in mice, Applicants injected Sepin-1 at a single dose in the range of 50-100mg/kg body weight into C57B6 mice via tail vein. While the mice at the highest dose of 90mg/kg died within 30-90 minutes of injection, the mice injected with a dose of <80mg/kg had no apparent effect, and have now survived for over 6 months. Blood analysis suggested that the mice receiving 90 mg/kg of Sepin-1 had high lactate dehydrogenase (LDH) (>500IU), suggesting muscle damage in these animals. However, the heart and skeletal muscles did not show light microscopic evidence of morphologic damage. In addition, histopathology indicated no significant lesions in major organs, including heart, lungs, liver, kidney, eye, gall bladder, pancreas, spleen, brain, adrenal gland, cervical lymph nodes, salivary gland, stomach, small intestine, skeletal muscles, bone marrow, and ear. In addition, there was no apparent distressing effect on the mice in clinical observations when the dose was 70mg/ml or lower.
[00103] Based on the above results, Applicants expanded this study to identify the Severely Toxic Dose (i.e., dose that causes death or irreversible severe toxicity, in 10% of rodents, STD10) using more mice injected at 75 to 90 mg/Kg range. A dose of 80mg/Kg was estimated as STD10 for Sepin-1 in these mice. When Sepin-1 was introduced to a cohort of 12 mice with a dose of lOmg/kg via intraperitoneal injection (IP) daily for six weeks, no weight loss or apparent adverse effects on the well-being of the mice were found.
[00104] Example 1.7. Sepin-1 inhibits the growth of cancer cells and tumors
[00105] Based on the Sepin-1 activity in vitro, Applicants reasoned that the level of Separase in cells may affect their sensitivity to Sepin-1 (i.e., higher the Separase greater the inhibition and vice versa). To test this hypothesis, Applicants selected two breast cell lines and two tumors that have differential level of Separase protein (FIG. 7). Breast epithelial cell line MCFlOf is not tumorigenic and has lower Separase level compared to isogenic MCF7 line (FIG. 7A). When treated with Sepin-1, MCF7 cells were over 3.3 times more sensitive than MCFlOf cells (FIG. 7B). The sensitivity of MCF7 cells to Sepin-1 treatment was also further verified from the growth of xenograft tumors derived from MCF7 cells in mice (FIG. 7C).
[00106] Applicants also investigated Sepin-1 activity against patient derived xenografts from two triple-negative (ER- PR- HER2-) breast tumors (MCI and BCM-5471). Compared to the BCM-5471 tumor, Separase is overexpressed in the MCI tumor. While Separase is primarily found in the mitotic cells of the BCM-5471 tumor, in MCI tumor it was found in majority of non-cycling (Ki67 staining negative) cells (FIG. 7D), as Applicants have previously described (Clin. Cancer Res., 2009, 15, 2703-2710). Once the tumor sizes reached ~200mg, mice were treated with Sepin-1 at a dose of lOmg/kg by intraperitoneal injection every day for 3 weeks. Compared to vehicle control, Sepin-1 treatment inhibited the growth of xenograft MCI tumors by 70% over the three week treatment (FIG. 7E). However, there was no significant difference in growth between vehicle control and Sepin-1 treatment in xenograft BCM-5471 (FIG. 7F). Collectively, these data suggest that Sepin-1 inhibits tumor cell growth through selective inhibition of Separase.
[00107] Applicants used the MTT assay to test the inhibitory effect of Sepin- 1 on the growth of a variety of human cancer cell lines, including leukemia, breast cancer and neuroblastoma. The results are summarized in Table 1 and FIGS. 8A-C.
Figure imgf000030_0001
Table 1. Half maximal inhibitory concentration (IC50) of Sepin-1 in growth inhibition of different cancer cell lines. Cancer cells were treated with serially diluted Sepin-1 for 72h. The cell viability was assessed using the MTT assay.
[00108] Different cell lines have different sensitivity to Sepin-1 in inhibiting cell growth. The half maximal inhibitory concentration of Sepin-1 (IC50) ranges from 1.0 μΜ to over 60 μΜ (Table 1 and FIGS. 8A-C).
[00109] To define the relationship of Separase level and its sensitivity to Sepin-1 in cell lines, Applicants investigated the Separase protein level in 14 cancer cell lines (2 leukemia, 8 neuroblastoma, 1 thyroid, 1 lung cancer and 2 breast cancer cells) with Western blot (FIGS. 8D and 7A). Both full length (FL) and auto-cleaved N-terminal (NT, as a surrogate for the active protein) Separase bands were quantified and normalized with loading control protein beta actin. When IC50 of Sepin- 1 inhibiting cell growth and the Separase protein level in the particular cell line were plotted side by side, both Separase FL and NT level in cells were inversely co-related to the IC50 values (FIG. 8E), indicating that cells overexpressing Separase are not only more sensitive to the inhibition of Separase, but may also be addicted to overexpressed Separase.
[00110] To determine the mechanism of cell growth inhibition induced by Sepin-1, Applicants performed immunoblotting to investigate if apoptosis is induced after Molt4 cells were treated with Sepin-1. Activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase (Parp-1) are two commonly used parameters in determination of apoptosis. Applicants used Etoposide as a positive control, which is a well-known chemotherapy agent that induces DNA damage by interacting with topoisomerase II. Western blot data indicated that caspase-3 proenzyme was induced and activated by the formation of cleaved caspase-3 with the increase of Sepin-1 concentration. Apoptosis induction following Sepin-1 treatment was also supported by Parp-1 cleavage (FIG. 8F). In addition, Applicants observed that the effect of Sepin-1 on the activation of caspase-3 and degradation of Parp-1 is similar to that of Etoposide (FIG. 8F).
[00111] Parp-1, a chromatin-associated enzyme, catalyzes the poly ADP-ribosylation of proteins that are involved in chromatin architecture, DNA damage repair, and DNA metabolism. Parp-1 plays an important role in mediating the normal cellular response to DNA damage in non- apoptotic cells. However, it is cleaved by caspases in apoptotic cells, which is a marker of apoptosis. Although Sepin-1 induced cell growth inhibition appears to be via apoptosis, how Sepin-1 triggers the apoptosis remains to be determined.
[00112] Example 1.8. Sepin-1 inhibits the growth of human tumor xenografts in mice
[00113] Based on the in vitro data shown above, Applicants reasoned that Sepin-1 effect in inhibiting tumor growth in vivo is also dependent on the level of Separase (i.e. the higher the Separase level, the greater the inhibition, and vice versa). To test this hypothesis, Applicants selected two human breast cell lines (MCF7 and MCFlOf) and two mammary tumors (MCI and BCM-5471) that have differential level of Separase protein (FIG. 7). Breast epithelial cell line MCFlOf is not tumorigenic and has lower Separase level compared to isogenic MCF7 line (FIG. 8A). When treated with Sepin-1, MCF7 cells were over 3.3 times more sensitive than MCFlOf cells (FIG. 8B). The sensitivity of MCF7 cells to Sepin-1 treatment was also further verified from the growth of xenograft tumors derived from MCF7 cells in mice (FIG. 7C).
[00114] Applicants also investigated Sepin-1 activity against patient derived xenografts from two triple-negative (ER~ PR" HER2") breast tumors (MCI and BCM-5471). Compared to the BCM-5471 tumor, Separase is overexpressed in the MCI tumor (FIG. 7D). While Separase is primarily found in the mitotic cells of the BCM-5471 tumor, it was found in majority of non- cycling (Ki67 staining negative) cells in MCI tumor (FIG. 7D), as Applicants have previously described.
[00115] Once the tumors size reached ~200mg, mice were treated with Sepin-1 at a dose of 10 mg/kg by intraperitoneal injection every day for 3 weeks. Compared to vehicle control, Sepin-1 treatment inhibited the growth of xenograft MCI tumors by 70% over the three week treatment (FIG. 7E). However, there was no significant difference in growth between vehicle control and Sepin-1 treatment in xenograft BCM-5471 (FIG. 7F), possibly due to low Separase level that causes less sensitivity to Sepin-1. Similar to leukemia cell line Molt4, Sepin-1 -treated MFC7 xenograft tumors showed increase of apoptotic bodies in H&E stained sections (FIG. 7G), which is consistent with the finding that cleaved caspase-3 staining was significantly increased (FIG. 7H). Collectively these data suggest that Sepin-1 inhibits tumor cell growth by induction of apoptosis through selective inhibition of Separase.
[00116] In the absence of any crystal structural information, it is difficult to develop inhibitors against the Separase enzymatic active site using a rational drug design approach. HTS is an alternate approach that can be used to find potential Separase inhibitors.
[00117] Separase is an ideal drug target. Pharmacologic inhibition of Separase is a novel strategy to treat Separase overexpressed aneuploid tumors, at least because of the following reasons: 1) while homozygous deletion of Separase is embryonically lethal, mice with Separase haploinsufficiency live a normal life with no disease phenotype compared to the WT animal, indicating that reduction in Separase level has no adverse effect on an organism's wellbeing; 2) Separase, a promoter of aneuploidy, is a highly specific protease that is overexpressed in a large percentage (>60 ) of human breast tumors, as well as in prostate tumors and osteosarcoma, suggesting that attenuation of Separase level pharmacologically in human cancers may kill Separase-addicted aneuploid cells; and 3) Separase is not only overexpressed but also constitutively mislocalized to the nucleus of the human tumors, providing an opportunity to target the nuclear bound Separase.
[00118] Therefore, titrating down Separase level therapeutically may not only inhibit tumor cell proliferation, but also effectively reverse the aneuploid phenotype in Separase overexpressed tumor cells, while sparing the normal cells. Furthermore, Separase mislocalization to nucleus can facilitate its targeting in the aneuploid tumor cells. Applicants also reason that cells with knockdown nuclear Separase will initiate mitotic arrest and apoptosis, and may therefore be more sensitive to traditional chemotherapy.
[00119] Example 1.9. Materials and Methods
[00120] Cell lines
[00121] The cell lines used included leukemia: HL-60, JM1, Jurkat, Molt-4, and Raji; breast cancer: HBLIOO, MCF7, MCFIOF, MDA-MB-436, MDA-MB-231, SK-BR-3 and T47D; thyroid cancer: TT; neuroblastoma: CHP-212, IMR-32, KCNR, LAN- 5, NGP, SHEP, SK-N-AS, SK- NBE (2), and SH-SY5Y; brain tumor: CHLA-02-ATRT; lung cancer: A549; colon cancer: HCT116. The cells were cultured according to the protocols recommended by ATCC.
[00122] Antibodies
[00123] The sources of the antibodies used in this Example are as follows: Actin mAb (Sigma, St. Louis, MO), Parp mAb (BD Bioscience, San Jose, CA), Separase mAb (Abnova, Taiwan), Caspase-3 pAb (Cell Signaling technology, Danvers, MA), Ki67 pAb (Vector Labs, Burlingame, CA), Rad21 pAb (Mol. Cell Biol., 200, 22, 8267-8277).
[00124] Purification and activation of Separase [00125] Epitope-tagged Separase was expressed, purified and activated as described previously (Anal. Biochem., 2009, 392, 133-138).
[00126] Fluorogenic Separase activity assay
[00127] The substrate used in this Example is (Rad21)2-Rhl l0, which has two Rad21 peptide molecules (Asp-Arg-Glu-Ile-Nle-Arg) conjugated to one molecule of Rhodamine 110 (Rhl lO) (CPC Scientific, Sunnyvale CA). The assay was set up using 384-well low volume black polystyrene plates (Corning, NY). Fifteen micro liter of activated Separase (~5ng) was mixed with 5μ1 of test compound. The cleavage buffer (CB) used to dilute reagents was (30mM Hepes- KOH pH7.7, 50mM NaCl, 25mM NaF, 25mM KC1, 5mM MgC12, 1.5mM ATP, and ImM EGTA). The mixture was incubated at room temperature for lh, and then 5μ1 of 0.2mM substrate (Rad21)2-Rhl l0 was added. The positive control contained Separase, substrate and CB without the test compound. The negative control contained substrate and CB only. After the reaction mixture was incubated at 37 °C for 3h, the relative fluorescence intensity was measured (without stopping the enzymatic activity) at ex=390nrnAem=450nm for 7-amido-4-mrthyl coumaric acid (AMC) released from Rad21-MCA or Aex=490nrnAem=528nm for Rhl lO released from (Rad21)2-Rhl l0 using a Synergy™ 4 Multi- Mode Microplate Reader (BioTek Instruments Inc., Winooski, VT).
[00128] High throughput screening (HTS)
[00129] HTS was performed at the John S. Dunn Gulf Coast Consortium for Chemical Genomics at the University of Texas Medical School at Houston. The compound library used in HTS was the Maybridge HitFinder™ Collection that contains 14,400 compounds. The compounds were stored in 180 96-well plates, 80 compounds per plate. The concentration of the stock solution of each compound was lOmM in DMSO. Each compound was diluted to 500μΜ in CB using a Biomek FX automation workstation (Beckman Coulter, Inc. Brea, CA) immediately before the assay was performed. HTS was performed on 384-well plates using a Biomek NX Automation Workstation (Beckman Coulter, Inc.) to dispense the compounds and Separase enzyme. Three hundred and twenty compounds were assayed in every 384-well plate. In each assay well, 5μ1 of 500μΜ compound was mixed with 5ng of Separase in 15μ1 CB. The reaction mixture was incubated at 25 °C for lh. After 5μ1 of substrate was added to the reaction mixture using Thermo Scientific Multidrop Combi Reagent Dispenser (Thermo Fisher Scientific, Waltham MA), the plate was incubated at 37 °C for 3h. Each assay plate included 32 positive controls in which CB was used instead of the compounds, and 32 negative controls which did not have either compound or Separase. Each compound was assayed in duplicate in two separate plates (A and B). The RFI was determined with Tecan Infinite 200 Reader (Tecan Group Ltd, Switzerland). The quality of HTS was evaluated by Signal to Background ratio (S:B), Signal to Noise ratio (S:N), and Z' defined as S:B= RFIpositive /RFInegative, S:N= (RFIpositive- RFInegative)/STDEVnegative, and Z'=l-[(3*STDEVpositive+3*STDEVnegative)/(RFIpositive- RFInegative)] . The percent inhibition of Separase enzyme activity by compounds was calculated as 100*{ l-[(RFIcompound-RFInegative)/(RFIpositive-RFInegative)] }. The compounds that inhibited more than 50% of Separase activity on both plate A and B were identified as hits.
[00130] In vitro cytotoxicity assay
[00131] The inhibitory effect of Sepin-1 on cell growth was assayed using the 3-(4,5- dimethylthiazol-2yl)-2,5-diphenyl-tetrazolium bromide (MTT) colorimetric dye reduction method (Cancer Chemother. Pharmacol., 2006, 58, 13-23). One hundred thirty-five μΐ of exponentially growing cancer cells were plated at a density of 0.4-1.0 x 105 cells/well in a 96- well microtiter plate. After 16h, Sepin-1 at specified concentrations was added to each well. After 72h of continuous drug exposure, 15 μΐ of 5mg/ml MTT was added to each well and the plates were incubated for 4h at 37 °C. Medium was replaced with 150 μΐ of DMSO to solubilize the formazan, and the optical density (OD) was measured at 550 nm using a microplate spectrophotometer (Anthos Analytical, Durham, NC, USA). Cell viability was calculated by subtracting the background OD of media alone from the OD of test wells, then dividing by the OD of the control (untreated) wells. Replicates of 3 wells were used for each drug concentration. The in vitro cytotoxicity assay was repeated once and the IC50 reported was the average of two independent experiments.
[00132] Toxicity of Sepin- 1 in mice
[00133] To test Maximum Tolerated Dose (MTD) and No Observable Effect Level (NOEL) of
Sepin-1, Applicants applied 200 μΐ Sepin-1 (diluted in PBS) one time at an acute dose range of
50 to lOOmg/kg body weight to 8-10 weeks old C57B6 mice (~25g) through intravenous tail vein injection (n=3/group). Except for the MTD dose, after Sepin-1 injection, the mice were returned to the cage and their reaction to the drug was monitored from one week to several months. Complete necropsy, blood chemistry and pathology of the Sepin-1 injected mice were carried out by the Comparative Pathology Laboratory at the Baylor College of Medicine.
[00134] Growth inhibition of Xenograft tumors
[00135] Patient-derived xenograft (PDX) lines MCI (ER- PR- HER2-) (Proc Natl Acad Sci U S A. 100, 2003,3983-3988) and BCM-5471 (ER- PR- HER2-) (Cancer Res. 73, 2013,4885-4897) were maintained as described (Cancer Res. 73, 2013,4885-4897) by serial transplantation into SCID Beige immunocompromised host mice whose endogenous mammary epithelium was removed surgically (Cancer Res. 19, 1959, 515-20).
[00136] For treatment studies, MCF7 xenografts (ER+ PR+ HER2-) were generated by injection of one million cultured cells grown under standard in vitro conditions as previously described (Proc. Natl. Acad. Sci. U. S. A 105, 2008, 13033-13038). Donor tumors for PDX lines MCI and BCM-5471 were divided into small fragments and retransplanted into the fat pat of recipient SCID Beige mice (5 mice per group). When the tumors reached approximately ~200mg, mice were treated with either vehicle or Sepin-1. Sepin-1 was injected at dose of lOmg/kg daily for 5 days a week for 3 weeks. Guided by toxicity studies, Applicants chose this dose for SCID-beige mice with no apparent effect on body weight. Vehicle injected mice served as controls. Size of the xenografts were measured every four days using calipers and tumor weight was estimated using the guidelines by NCI for xenograft evaluation (http://ncifrederick.cancer.gov /Lasp /Acuc/Frederick /Media/ Documents /ACUC 14.pdf). After 3 weeks treatment, the tumors were harvested and fixed in 10% Buffed Formalin Phosphate (Fisher Scientific, Pittsburg, PA) overnight. The tumor tissues were washed with PBS for 3x30min; dehydrated for lh at 70%, 80%, 90% and 100% two times; and immerged in Xylene several times until the tissues became clear. The samples were embedded in wax and sectioned with 5μιη in thickness. All mice were maintained in accordance with the NIH Guide for the Care and Use of Laboratory Animals with approval from the Baylor College of Medicine Institutional Animal Care and Use Committee.
[00137] In vitro cleavage of Myc-Rad21 assay [00138] Myc-Rad21 protein was prepared by using pCS2MT Rad21 plasmid {Cancer Res., 1959, 19, 515-520) as the template and the TNT SP6 High- Yield Wheat Germ Protein Expression kit (Promega, Madison, WI). Myc-Rad21 protein was mixed with Separase with or without the presence of Sepin-1. The final concentration of Sepin-1 was 50 μΜ. The reaction mixture was incubated in a 37°C water bath for 1 h. The cleavage of myc-Rad21 was immunoblotted with Rad21 pAb.
[00139] Characterization of Sepin-1 mediated inhibition of Separase activity in vitro
[00140] Different amounts of Sepin-1 were mixed with activated Separase for 30 min before substrate (Rad21)2-Rhl l0 was added. After 3 h incubation at 37°C, the fluorescence intensity of Rhl lO was determined. IC50 was calculated using either KaleidaGraph program (Synerge software, Reading, PA) or four parameter logistic equation (4-parameter dose-response curve fit): Inhibition of Activity (%) = min + (max - min) / { 1 + (log[M] / IC50)}Hill Slop, where "min" and "max" represent minimal and maximal activity inhibition, respectively. Hill Slop is the slope of the curve at its midpoint. To identify the mode of Sepin-1 interacting with Separase, different amounts of both Sepin-1 and (Rad21)2-Rhl l0 were used in the Separase activity assay.
[00141] Immunofluorescence microscopy
[00142] Deparafinization of tumor tissue sections and immuno staining were performed using the protocol reported previously (Clin. Cancer Res., 2009, 15, 2703-2710).
[00143] Immunoblotting
[00144] Preparation of protein samples and Western blot were performed as described previously (J Cell Biol, 2008, 183, 1019-1031; PLoS. ONE., 2013, 8, e69458).
Screening Compound Signal; Bacgrotiinl S:B Range .Nohe S;N Ratige Z' Range
Z>
Plate No,'1 Plate o,b (S:B) (A s K) (S:N) (A : B) (A : B)
1 1-4 7.45 (7.7 : 7.2) 5S..26 (56.6 · 39.9) 0.34 (03 035)
5-S S.1 (8.5 : 7.8} 45.87 (44.4 : 47.3) 0.66 (0.06 0.67:
3 S>-!2 5.95 ί'5.1 : 6.7; 34.77 (24,6 : 44.9) 0.48 (0.59 0.56';
4 13-16 5.27 (5 A : 5.5) 1S.4S 06,5 : 20.5) 0.5 (0.3S 0.56;
.5 17-20 6..S9 (S.3 : 5.7) 2.9.47 (3S : 20.7; 0.53 (0.64 0.42)
6 21-24 6.55 (7.5 : 5.6) 3S.71 (4.3.4 : 34.0) 0.60 (0.71 0.48)
7 25-2% 8.23 (9.0 ; 7.5) 40.41 (52.5 :46,5; .5S (0.58 0.5S)
& 29- 2 6.19 (5.2 ; 7.1) 25.36 (36.4:26,3; 0.55 (0.35 0.44}
33-3*5 6.SS (76 : 5.7) 25.30 (26.4 : 56.2) 0.55 (055 0.4?)
10 37-40 6.12 (59 : 5.4} 49,93 (50.S : 49.1) 0.54 (0.62 0.46)
H 41-44 14,3? (14.5 : 14.5» 42.77 (42.7 ; 42.S) 0,47 (0.S6 0.47}
12 45-48 1319 (14.2 : 12.2» 50.17 (61.5 : 5S.S) 0,66 (0.66 0.65 >
13 49-52 1344 (14.9 : 12.0» 53.44 (62.1 :44.7) 0,67 (0.69 0.64}
14 53-56 13.31 (11.9 : 54.8) 55.51 (3S.5 ; 64,5; 0,71 (0,67 0.75,;
15 57-60 12.58 (11.5 : 13.7) 44.35 (41.5 :47,2 ; 0,69 (0,65 0.74;
1 61 -64 12.00 (140 ; 10,0) 45,44 (51.0 : 31.9) 0.65 (0,69 06S)
17 65-cS 15.45 (14.3 ; 12.S) 70.20 (4 .5 ; 98.9} 0.70 (0.70 0.70)
IS (50.72 13.46 f 12.9 : 14.0» 4S.il (47.1 : 49.1) 0.66 (0.64 0.67":
19 75-70 12.00 (11.7 : 12.5» 40.5S (36.S : 44.4) 0.57 (0.57 0.57)
20 77-80 70S (7.4 : 6.S.i 3 .82 (55. S : 27.S) 0.40 (0.42 0.38}
21 SI -84 174 (12.6 : lO.Si 59.46 ("5.4 :43.5) 0.51 (0.63 0.51
22 85-88 SSI (10.5 : 7.3} 29.09 (32.5 : 25.7; 0.53 (0.63 0.44) 5 S.9-92 14.5S (16.1 ; 13.1) 79,42 (113.6; 45.5 : 0.6 (0.70 0.62)
24 93-96 9.45 (15.4: 7.5) 26.56 (35.2 ; 17,9: 0.47 (0.51 043)
25 97-i 10.12 i'12.3 : S O) 3S.95 (49.1 ;2S 7} 0.37 (0.53 0.22)
26 101-104 12.77 (Ϊ2..3 : 13,5) 59.57 (53.0 : 66.1) 0.65 (056 0.65) 7 105-108 11.00 (9.6: 12.4) 55.56 (54,5 : 56.6) 0.65 (0.67 0.64)
2 3.09-3.12 12.93 (14.8 : 11.1» 50.40 (63,6 : 57.2) 0,68 (0.72 0.64}
29 113-115 11 ,53 (9.0; 14.1: 44.27 (376 ; 51.0) 0,66 i'O.SS 0.64}
30 117-129 13.77 Si 5.3 : 12,1) S5.1S (101.6 ; iSS.S; 0,68 (0.71 0.65;
31 121-124 13.13 (11.9 : 14.4) "9.1 (74.0 ; S4.3; 0,59 (0.59 0.60;
32 125-125 10.59 (8.2 : 13.0) 45.S1 (24.5 : 69.3) 0.40 (0.34 0.46)
35 12.9-132 12.55 (9,9 : 15.2: 66.61 (43.9 ; S9.3; 0.44 (034 0.53)
34 135-135 11.52 (12.S ; 10.5'» 51.14 (64.6 : 57.7) 0.50 (0. 7 0.625: 5 137-140 S.12 i'6.9 : 9.4; 45.53 (59.9 : 31.1) 0.55 (0.57 0.54;
36 141-144 13.S2 (15.7 : ll.&'i 5S.54 (63.6 : 55.1) 0.58 (0.62 0.54;
37 145-14S S...55 (9.2 : 7.9) 21.05 (23.6 : 1S.5; 0.49 (0.4S 0.50;
3S 349- 52 10.55 (10.1 : 11.0» 44.29 (466 : 41.5} 0.59 (0.59 0.60;
39 1 3-156 9.97 (7,9: 12.1) 55.56 (26.5 : 56,8 : 0.44 (0.33 0.56)
4<> 157-160 8. SI (10,6: 7.3) 29.09 (32.5 :25.-; 0.53 (0,63 0,44)
41 161-154 11.04 (502 : 11.9) 54.2? (59.5 : 6S.9) 0.5.3 (045 0.60)
42 165-16* 13.3S (149 : 11,5) 52.31 (65.3 ; 59.3) 0.55 (0.68 0.50)
43 169-172 9.S7 (12.6: 7,2) 65.59 (79.S : 43.4) 0.45 (060 0.3 S)
44 175-175 li.SS (9:6: 14.1; 32.04 (24.5 : 59.5) 0.46 ί'0.40 0.52; 5 377- IS 1102 (12.4-9.7: 1.57 (63.3 : 59.S) 0,51 (0.55 0.49}
Average 10.54 (10.75 · 46.35 (47.92 ; 44.77} .55 (β.57 6.54)
SB 2.69 (3.06 : 3.03) 15.77 (20.38 ; 18.70) cue (0.12 0.12)
Table 2. Summary of robustness of the HTS for Separase inhibitors, (a) 384-well plates were used for the HTS. (b) Aliquots of compound stocks were in 96-well plates, 80 compounds/plate. Compounds from every 4 plates were screened in one 384-well plate, (c) The screening was duplicated. Two duplicated 384-well plates were named A and B, respectively. [00145] Example 2. Inhibition of the growth of glioblastoma cells by Sepin-1
[00146] FIG. 10 provides data relating to separase overexpression in Pediatric glioblastoma (GBM) and the effect of Sepin-1 in inhibiting the growth of glioblastoma cells in vitro and in vivo. FIG. 10A shows representative immunofluorescence pictures of normal brain (bottom panel) and a pediatric glioblastoma samples (#4687,top panel) showing Separase expression and nuclear localization in non-mitotic GBM cells but not in normal brain. FIG. 10B shows Kaplan- Meier plot showing significantly lower survival in GBM patients with high Separase expression (n=12). FIG. IOC provides MTT cytotoxicity assays showing efficient cell death of two Separase overexpressing primary pediatric glioblastoma samples. The inset (Western blot) shows levels of Separase in these samples. FIG. 10D provides survival analysis showing a significant increase in the life span of Sepin-1 treated mice with orthotopic xenograft of Separase overexpressing pediatric GBM tumor (#4687). SCID-beige mice after two weeks of transplantation were treated at a dose of lOmg/kg daily ip for 5 days a week for 4 weeks with either Sepin-1 or vehicle (n=6/group).
[00147] Example 3. Optimization of Sepin-1
[00148] This Example pertains to the optimization of the structure of Sepin-1 to obtain Sepin-1 analogs with higher potency. Over 30 sepin-1 derivatives were synthesized and tested. FIG. 11A illustrates the Sepin-1 derivatives that were synthesized and examined. The separase activity of the Sepin-1 derivatives shown in the box in FIG. 11A (compounds 27-31) were tested. The results are summarized in FIG. 11B.
[00149] As shown in FIG. 11B, Applicants observed that compound 29 was approximately twice as active as lead compound 27. Applicants also observed that compound 30 was approximately 10 times more active than lead compound 27.
[00150] Without further elaboration, it is believed that one skilled in the art can, using the description herein, utilize the present disclosure to its fullest extent. The embodiments described herein are to be construed as illustrative and not as constraining the remainder of the disclosure in any way whatsoever. While the embodiments have been shown and described, many variations and modifications thereof can be made by one skilled in the art without departing from the spirit and teachings of the invention. Accordingly, the scope of protection is not limited by the description set out above, but is only limited by the claims, including all equivalents of the subject matter of the claims. The disclosures of all patents, patent applications and publications cited herein are hereby incorporated herein by reference, to the extent that they provide procedural or other details consistent with and supplementary to those set forth herein.

Claims

WHAT IS CLAIMED IS:
1. A composition for inhibiting Separase activity, wherein the composition comprises a compound selected from the group consisting of:
Figure imgf000041_0001
Figure imgf000041_0002
Figure imgf000041_0003
Figure imgf000042_0001
o
Figure imgf000042_0002
o
Figure imgf000042_0003
Figure imgf000043_0001
Figure imgf000043_0002
o-
and combinations thereof.
2. The composition of claim 1, wherein R and RiA are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof.
3. The composition of claim 2, wherein R3 is selected from the group consisting of H, OH, CH3, C4H9, alkyl groups, and combinations thereof.
4. The composition of claim 1, wherein R2 is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.s), C(CH )(R7 5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof.
5. The composition of claim 4, wherein R7 and R7 5 are each selected from the group consisting of alkanes, CH3, CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4Hg, CO, CS, and combinations thereof.
6. The composition of claim 1, wherein R4 is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH3, phenyl, O-phenyl, NH-phenyl, and combinations thereof.
7. The composition of claim 1, wherein R5 is selected from the group consisting of NH, NR6, S, O, and combinations thereof.
8. The composition of claim 7, wherein R6 is selected from the group consisting of H, OH, CH3, C4Hg, alkyl groups, and combinations thereof.
9. The composition of claim 1, wherein the compound comprises the following structure:
Figure imgf000044_0001
wherein R is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF , C(N2)CF3, CN, CH3, CF3S02, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, SO3H, and combinations thereof, and wherein R2 is selected from the group consisting of C(CH3)2, C(CH ) (phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof.
10. The composition of claim 9, wherein R3 is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof.
11. The composition of claim 1, wherein the compound is selected from the group consisting
Figure imgf000045_0001
o
Figure imgf000045_0002
Figure imgf000045_0003
il
O
Figure imgf000046_0001
Figure imgf000046_0002
and combinations thereof; wherein Ri and R1A are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3SO2, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof; wherein R3 is selected from the group consisting of H, OH, CH3, C4Hg, alkyl groups, and combinations thereof;
wherein R2 is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof; and wherein R7 and R7 5 are each selected from the group consisting of alkanes, CH , CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4Hg, CO, CS, and combinations thereof.
12. The composition of claim 1, wherein the compound comprises the following structure:
Figure imgf000047_0001
wherein Ri and R1A are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof;
wherein R is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof; wherein R2 is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof; and wherein R7 and R7 5 are each selected from the group consisting of alkanes, CH , CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4I¾, CO, CS, and combinations thereof.
13. The composition of claim 12, wherein R is N02, RiA is Br, and R2 is C(CH )2.
14. The composition of claim 1, wherein the compound comprises the following structure:
Figure imgf000048_0001
wherein Rg is selected from the group consisting of alkanes, CH3, CH2CH3, cycloalk cyclopentane, cyclohexane, phenyl, C4I¾, CO, CS, and combinations thereof.
15. The composition of claim 1, wherein the compound comprises the following structure:
Figure imgf000048_0002
16. The compositions of claim 1, wherein the compound is water soluble.
17. The composition of claim 1, wherein the compound is a selective inhibitor of Separase.
18. The composition of claim 1, wherein the compound is a non-competitive inhibitor of Separase.
19. A method of treating a tumor in a subject, wherein the method comprises: administering a composition to the subject, wherein the composition comprises a compound that inhibits Separase activity.
20. The method of claim 19, wherein the tumor comprises Separase overexpressed tumor cells.
21. The method of claim 19, wherein the tumor comprises Separase overexpressed mammary tumor cells.
22. The method of claim 19, wherein the tumor comprises Separase overexpressed aneuploid cells.
23. The method of claim 19, wherein the tumor is associated with at least one of breast cancer, leukemia, thyroid cancer, neuroblastoma, brain cancer, lung cancer, colon cancer, prostate cancer, osteosarcoma, glioblastoma, ovarian cancer, gastrointestinal cancer, and combinations thereof.
24. The method of claim 19, wherein the tumor is associated with breast cancer.
25. The method of claim 19, wherein the subject is a human being.
26. The method of claim 19, wherein the subject is a human being suffering from cancer.
27. The method of claim 19, wherein the administering of the composition comprises intravenous administration.
28. The method of claim 19, wherein the compound is a selective inhibitor of Separase.
29. The method of claim 19, wherein the compound is a non-competitive inhibitor of Separase.
30. The method of claim 19, wherein the compound selectively inhibits growth of Separase overexpressed tumor cells associated with the tumor.
31. The method of claim 19, wherein the composition comprises a compound selected from the group consisting of:
Figure imgf000050_0001
Figure imgf000051_0001
H
R
H
Figure imgf000051_0002
Figure imgf000051_0003
Figure imgf000051_0004
O"
Figure imgf000051_0005
Figure imgf000052_0001
Figure imgf000052_0002
Figure imgf000052_0003
and combinations thereof.
32. The method of claim 31, wherein R and RiA are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof.
33. The method of claim 32, wherein R3 is selected from the group consisting of H, OH, CH3, C4H9, alkyl groups, and combinations thereof.
34. The method of claim 31, wherein R2 is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.s), C(CH )(R7 5),
C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof.
35. The method of claim 34, wherein R7 and R7 5 are each selected from the group consisting of alkanes, CH , CH2CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
36. The method of claim 31, wherein R4 is selected from the group consisting of alkanes, cycloalkanes, cyclopentane, cyclohexane, CH , phenyl, O-phenyl, NH-phenyl, and combinations thereof.
37. The method of claim 31, wherein R5 is selected from the group consisting of NH, NR6, S, O, and combinations thereof.
38. The method of claim 37, wherein R6 is selected from the group consisting of H, OH, CH3, C4H9, alkyl groups, and combinations thereof.
39. The method of claim 31, wherein the compound comprises the following structure:
Figure imgf000054_0001
wherein R is selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, , CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR3, N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof, and wherein R2 is selected from the group consisting of C(CH3)2, C(CH3) (phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof.
40. The method of claim 39, wherein R is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof.
41. The method of claim 31, wherein the composition comprises a compound selected from the group consisting of:
Figure imgf000055_0001
o
Figure imgf000055_0002
Figure imgf000055_0003
Figure imgf000056_0001
O"
Figure imgf000056_0002
and combinations thereof; wherein R and R1A are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof; wherein R is selected from the group consisting of H, OH, CH , C4H9, alkyl groups, and combinations thereof;
wherein R2 is selected from the group consisting of alkanes, C(CH3)2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3), C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O and combinations thereof; and wherein R7 and R 5 are each selected from the group consisting of alkanes, CH , CH2CH3, cycloalkanes, cyclopentane, cyclohexane, phenyl, C4I¾, CO, CS, and combinations thereof.
42. The method of claim 31, wherein the compound comprises the following structure:
Figure imgf000057_0001
wherein Rg is selected from the group consisting of alkanes, CH3, CH2CH3, cycloalk cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
43. The method of claim 31, wherein the compound comprises the following structure:
Figure imgf000057_0002
44. The method of claim 31, wherein the compound comprises the following structure:
Figure imgf000058_0001
wherein R and RiA are each selected from the group consisting of alkanes, alkenes, alkynes, carboxyl groups, alkoxy groups, methoxy groups, ethers, nitro groups, nitriles, sulfates, sulfonates, halogens, primary amine groups, secondary amine groups, tertiary amine groups, alcohols, boronic acids, triazoles, photo-reactive groups, OH, NH2, N02, Br, F, CI, I, CF3, CF3S02, C(N2)CF3, CN, CH3, CH30, C02H, CONH2, CONHR3, NHR , N(R3)2, C2R3, C4H9, phenyl, CH2-phenyl, S03H, and combinations thereof;
wherein R3 is selected from the group consisting of H, OH, CH3, C4Hg, alkyl groups, and combinations thereof;
wherein R2 is selected from the group consisting of alkanes, C(CH )2, cycloalkanes, cyclopentane, cyclohexane, phenyl, C(R7)(R7.5), C(CH3)(R7.5), C(CH3)(CH2CH3),
C(CH3)(Phenyl), C(CH3)(CF3), C(C4H9)2, CO, CS, CH2, O, and combinations thereof; and
wherein R7 and R7 5 are each selected from the group consisting of alkanes, CH3, CH2CH , cycloalkanes, cyclopentane, cyclohexane, phenyl, C4H9, CO, CS, and combinations thereof.
45. The method of claim 44, wherein Ri is N02, RIA is Br, and R2 is C(CH3)2.
PCT/US2014/061353 2013-10-18 2014-10-20 Separase inhibitors and uses thereof WO2015058185A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/030,070 US10052310B2 (en) 2013-10-18 2014-10-20 Separase inhibitors and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361892911P 2013-10-18 2013-10-18
US61/892,911 2013-10-18
US201461936976P 2014-02-07 2014-02-07
US61/936,976 2014-02-07

Publications (1)

Publication Number Publication Date
WO2015058185A1 true WO2015058185A1 (en) 2015-04-23

Family

ID=52828787

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/061353 WO2015058185A1 (en) 2013-10-18 2014-10-20 Separase inhibitors and uses thereof

Country Status (1)

Country Link
WO (1) WO2015058185A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020263909A1 (en) * 2019-06-24 2020-12-30 The Regents Of The University Of California Methods for preparation of active separase
CN114456266A (en) * 2022-03-01 2022-05-10 中国科学院广州生物医药与健康研究院 Method for in-situ detection and separation of enzyme cleavage product

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4246196A (en) * 1978-12-23 1981-01-20 Hoechst Ag. Diaminophenyl ureas
US4343942A (en) * 1966-11-08 1982-08-10 Research Corporation Quinoxaline derivatives
US5435860A (en) * 1992-01-08 1995-07-25 Mec Co., Ltd. Benzimidazole derivative and composition for treating copper and copper alloy surfaces comprising the same
US20020164620A1 (en) * 2001-01-19 2002-11-07 Boehringer Ingelheim International Gmbh Method for identifying compounds modulating sister chromatid separation
US20090175873A1 (en) * 2005-11-29 2009-07-09 The Scripps Research Institute Inhibiting tumor cell invasion, metastasis and angiogenesis

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343942A (en) * 1966-11-08 1982-08-10 Research Corporation Quinoxaline derivatives
US4246196A (en) * 1978-12-23 1981-01-20 Hoechst Ag. Diaminophenyl ureas
US5435860A (en) * 1992-01-08 1995-07-25 Mec Co., Ltd. Benzimidazole derivative and composition for treating copper and copper alloy surfaces comprising the same
US20020164620A1 (en) * 2001-01-19 2002-11-07 Boehringer Ingelheim International Gmbh Method for identifying compounds modulating sister chromatid separation
US20090175873A1 (en) * 2005-11-29 2009-07-09 The Scripps Research Institute Inhibiting tumor cell invasion, metastasis and angiogenesis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MEYER ET AL.: "Overexpression and Mislocalization of the Chromosomal Segregation Protein Separase in Multiple Human Cancers", CLIN CANCER RES., vol. 15, no. 8, 2009, pages 2703 - 2710 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020263909A1 (en) * 2019-06-24 2020-12-30 The Regents Of The University Of California Methods for preparation of active separase
CN114456266A (en) * 2022-03-01 2022-05-10 中国科学院广州生物医药与健康研究院 Method for in-situ detection and separation of enzyme cleavage product

Similar Documents

Publication Publication Date Title
Biamonte et al. Heat shock protein 90: inhibitors in clinical trials
Bridges Chemical inhibitors of protein kinases
Li et al. PCC0208017, a novel small-molecule inhibitor of MARK3/MARK4, suppresses glioma progression in vitro and in vivo
Huang et al. Novel hybrid molecule overcomes the limited response of solid tumours to HDAC inhibitors via suppressing JAK1-STAT3-BCL2 signalling
Li et al. The dietary compound luteolin inhibits pancreatic cancer growth by targeting BCL-2
Fracasso et al. A Phase 1 study of UCN-01 in combination with irinotecan in patients with resistant solid tumor malignancies
Hashim et al. Targeted pancreatic cancer therapy with the small molecule drug conjugate SW IV-134
Wang et al. CHM-1, a novel synthetic quinolone with potent and selective antimitotic antitumor activity against human hepatocellular carcinoma in vitro and in vivo
Eastman et al. A novel indolocarbazole, ICP-1, abrogates DNA damage-induced cell cycle arrest and enhances cytotoxicity: similarities and differences to the cell cycle checkpoint abrogator UCN-01
US10946012B2 (en) Methods for inducing tumor regression, inhibiting tumor growth, and inducing apoptosis in breast tumors with geranylgeranyltransferase I inhibitors
Hsieh et al. Chamaecypanone C, a novel skeleton microtubule inhibitor, with anticancer activity by trigger caspase 8-Fas/FasL dependent apoptotic pathway in human cancer cells
CA2761253A1 (en) Combinations of therapeutic agents for treating melanoma
Guan et al. WX-132-18B, a novel microtubule inhibitor, exhibits promising anti-tumor effects
Chen et al. Anti-tumor effects of B-2, a novel 2, 3-disubstituted 8-arylamino-3H-imidazo [4, 5-g] quinazoline derivative, on the human lung adenocarcinoma A549 cell line in vitro and in vivo
Chen et al. A-432411, a novel indolinone compound that disrupts spindle pole formation and inhibits human cancer cell growth
Cui et al. Falnidamol and cisplatin combinational treatment inhibits non-small cell lung cancer (NSCLC) by targeting DUSP26-mediated signal pathways
Sun et al. ABT-737 synergizes with arsenic trioxide to induce apoptosis of gastric carcinoma cells in vitro and in vivo
Cuyàs et al. Revisiting silibinin as a novobiocin-like Hsp90 C-terminal inhibitor: computational modeling and experimental validation
Niso et al. Multifunctional thiosemicarbazones targeting sigma receptors: in vitro and in vivo antitumor activities in pancreatic cancer models
Guo et al. The disulfiram/copper complex induces apoptosis and inhibits tumour growth in human osteosarcoma by activating the ROS/JNK signalling pathway
Wang et al. Inhibition of USP1 activates ER stress through Ubi-protein aggregation to induce autophagy and apoptosis in HCC
Kim et al. Glucosamine is an effective chemo-sensitizer via transglutaminase 2 inhibition
Wang et al. Abemaciclib is synergistic with doxorubicin in osteosarcoma pre-clinical models via inhibition of CDK4/6–Cyclin D–Rb pathway
WO2015058185A1 (en) Separase inhibitors and uses thereof
Castaneda et al. Phase I and pharmacokinetic study of lonafarnib, SCH 66336, using a 2-week on, 2-week off schedule in patients with advanced solid tumors

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: 14853841

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15030070

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 14853841

Country of ref document: EP

Kind code of ref document: A1