Treating cancer with ATR inhibitors

Information

  • Patent Grant
  • 10813929
  • Patent Number
    10,813,929
  • Date Filed
    Friday, February 28, 2014
    10 years ago
  • Date Issued
    Tuesday, October 27, 2020
    3 years ago
Abstract
This invention relates to methods and compositions for treating pancreatic cancer. More specifically, this invention relates to treating pancreatic cancer with certain ATR inhibitors in combination with gemcitabine and/or radiation therapy. This invention also relates to methods and compositions for treating non-small cell lung cancer. More specifically, this invention relates to treating non-small cell lung cancer with an ATR inhibitor in combination with cisplatin or carboplatin, etoposide, and ionizing radiation.
Description
BACKGROUND OF THE INVENTION

Pancreatic cancer is the tenth most common site of new cancers and is responsible for 6% of all cancer related deaths. The 5-year survival rate is less than 5%.


Current therapies involve either neoadjuvant treatment with chemotherapy (e.g., with gemcitabine) and/or radiation therapy or surgical removal followed by either adjuvant chemotherapy (e.g., with gemcitabine) or radiation therapy. Although the survival rate with treatment of gemcitabine increases the 5-year survival from 10% to 20%, there still is a strong need for better therapies for treating pancreatic cancer.


Several therapeutics have been tested in phase II and phase III trials though results have not been too promising. Tipifarnib, an oral farnesyltransferase inhibitor, did not show significant improvement in overall survival when combined with gemcitabine. Similarly, cetuximab, an epidermal growth factor receptor (EGRF), also showed no clinical benefit when combined with gemcitabine. Only a small increase in overall survival (6.24 months versus 5.91 months) was observed.


Lung cancer is the second most common form of cancer and is the leading cause of cancer-related mortality. Non-small cell lung cancer (NSCLC) is the most common form of lung cancer, accounting for about 85% of all lung cancer cases. Most patients present with advanced stage III or IV NSCLC with a 5-year survival of 24% and 4% respectively. Because of the advanced nature of disease on presentation, surgical resection is often not an option. For the majority of patients therapy involves chemotherapy and/or radiation treatment. The selection of chemotherapy is highly variable based on disease stage, patient performance criteria and geographical regional preference. In most cases chemotherapy is based on a doublet that includes a platinating agent such as Cisplatin or carboplatin and a second cytotoxic drug such as gemcitabine, etoposide or taxotere. For a small number of patients, therapy can include treatment with agents that target specific proteins that are mutated or disregulated such as ALK and EGFR (eg crizotinib, gefitinib and erlotinib). Patients are selected for these targeted treatments based on genetic or proteomic markers. A great number of agents have been assessed in late stage NSCLC clinical studies, however most have shown very little benefit over chemotherapy based treatments, with median overall survival typically less than 11 months.


Accordingly, there is a tremendous need for new strategies to improve pancreatic and non-small cell lung cancer treatments.


ATR (“ATM and Rad3 related”) kinase is a protein kinase involved in cellular responses to certain forms of DNA damage (eg double strand breaks and replication stress). ATR kinase acts with ATM (“ataxia telangiectasia mutated”) kinase and many other proteins to regulate a cell's response to double strand DNA breaks and replication stress, commonly referred to as the DNA Damage Response (“DDR”). The DDR stimulates DNA repair, promotes survival and stalls cell cycle progression by activating cell cycle checkpoints, which provide time for repair. Without the DDR, cells are much more sensitive to DNA damage and readily die from DNA lesions induced by endogenous cellular processes such as DNA replication or exogenous DNA damaging agents commonly used in cancer therapy.


Healthy cells can rely on a host of different proteins for DNA repair including the DDR kinases ATR and ATM. In some cases these proteins can compensate for one another by activating functionally redundant DNA repair processes. On the contrary, many cancer cells harbour defects in some of their DNA repair processes, such as ATM signaling, and therefore display a greater reliance on their remaining intact DNA repair proteins which include ATR.


In addition, many cancer cells express activated oncogenes or lack key tumour suppressors, and this can make these cancer cells prone to dysregulated phases of DNA replication which in turn cause DNA damage. ATR has been implicated as a critical component of the DDR in response to disrupted DNA replication. As a result, these cancer cells are more dependent on ATR activity for survival than healthy cells. Accordingly, ATR inhibitors may be useful for cancer treatment, either used alone or in combination with DNA damaging agents, because they shut down a DNA repair mechanism that is more important for cellular survival in many cancer cells than in healthy normal cells.


In fact, disruption of ATR function (e.g. by gene deletion) has been shown to promote cancer cell death both in the absence and presence of DNA damaging agents. This suggests that ATR inhibitors may be effective both as single agents and as potent sensitizers to radiotherapy or genotoxic chemotherapy.


Furthermore, solid tumors often contain regions that are hypoxic (low oxygen levels). This is significant because hypoxic cancer cells are known to be resistant to treatment, most notably IR treatment, and are highly aggressive. One reason for this observation is that components of the DDR can be activated under hypoxic conditions and it has also been shown that hypoxic cells are more reliant on components of the DDR for survival.


For all of these reasons, there is a need for the development of potent and selective ATR inhibitors for the treatment of pancreatic cancer, for the treatment of lung cancer, and for the development of agents that are effective against both hypoxic and normoxic cancer cells.


SUMMARY OF THE INVENTION

This invention relates to uses of ATR inhibitors for treating pancreatic cancer and non-small cell lung cancer. With respect to pancreatic cancer, this invention relates to methods of treating pancreatic cancer in a patient (e.g., a human) with an ATR inhibitor in combination with gemcitabine and/or radiation therapy. Applicants have demonstrated synergistic efficacy of ATR inhibitors in combination with gemcitabine and/or radiation therapy in clonogenic and viability assays on the pancreatic cancer cell lines, (e.g. PSN-1, MiaPaCa-2 and Panc-1) as well as in a primary tumor line (e.g., Panc-M). Disruption of ATR activity was measured by assessing DNA damage induced phosphorylation of Chk1 (Ser 345) and by assessing DNA damage foci and RAD51 foci following irradiation.


With respect to non-small cell lung cancer, his invention relates to methods of treating non-small cell lung cancer with an ATR inhibitor in combination with cisplatin or carboplatin, etoposide, and ionizing radiation. Applicants have demonstrated synergy of ATR inhibitors in combination with cisplatin, etoposide, gemcitabine, oxaplatin and irinotecan in viability assays against a panel of 35 human lung cancer cell lines as well as demonstrated in vivo efficacy in a lung cancer mouse model in combination with cisplatin.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1. VE-821 radiosensitises pancreatic tumour cells.


a) Western Blot Analysis of Chk1 Inhibition.


Cells were treated with 100 nM gemcitabine for 1 h, 1 μM VE-821 was added 1 h later and cells were irradiated (6 Gy) 1 h after that. Drugs were left for the duration of the experiment and cells were lysed at 2 h post-irradiation and subjected to Western blot analysis.


B) VE-821 Radiosensitizes Pancreatic Tumour Cells but not Normal Fibroblasts.


PSN-1, Panc-1, MiaPaCa-2 pancreatic cancer cell lines and MRCS fibroblasts were treated with increasing concentrations of VE-821 for 96 h combined with or without 4 Gy radiation at 1 h after VE-821 addition. Cell viability was measured after 8 days and shown as normalized to DMSO-treated cells.


C) Scheduling of VE-821 Affects Radiosensitivity.


PSN-1 cells were plated as single cells, treated with 1 μM VE-821 at different time points in relation to 4 Gy irradiation and assessed for colony formation after 10 days. The survival fraction at 4 Gy for each of the treatment schedules was determined by taking into account the relevant plating efficiency of unirradiated cells.


D) Clonogenic Survival of Cells Pancreatic Cancer Cells in Response to ATR Inhibition.


Cells were treated with 1 μM VE-821 4 h after plating and 1 h prior to irradiation. Drug was removed after 72 h and colony-forming ability was assessed after 10 to 21 days. (n=3). *, P<0.05; **, P<0.01 over DMSO-treated control.



FIG. 2. VE-821 radiosensitises pancreatic tumour cells under hypoxic conditions.


A) clonogenic survival curves of cells treated with 1 μM VE-821 and irradiation under hypoxic conditions. Plated cells were transferred to hypoxia (0.5% O2) and acclimatised for 6 h. VE-821 (1 μM) was then added at 1 h prior to irradiation and left for 72 h upon which the medium was replaced. Cells were transferred to normoxia at 1 h post-irradiation.


B) clonogenic survival of cells after irradiation with 6 Gy and treatment with 1 μM VE-821 in oxic and hypoxic (0.5% O2) conditions, as described above and in FIG. 1 (n=3). *, P<0.05; **, P<0.01; ***, P<0.001 over DMSO-treated control.



FIG. 3. VE-821 sensitises pancreatic cancer cells to gemcitabine treatment.


A) clonogenic survival of cells treated with gemcitabine and 1 μM VE-821. Cells were treated with increasing concentrations of gemcitabine for 24 h followed by 72 h treatment of 1 μM VE-821. Colony forming ability was assessed after 10 to 21 days.


B) clonogenic survival of cells treated with gemcitabine in hypoxia. Plated cells were transferred to hypoxia (0.5% O2) and acclimatised for 6 h. Cells were then treated with increasing concentrations of gemcitabine for 24 h followed by 72 h treatment of 1 μM VE-821. Hypoxic cells were transferred to normoxia 1 h after VE-821 addition.


C) clonogenic survival after treatment with 20 nM gemcitabine and VE-821 in oxic and hypoxic (0.5% O2) conditions, as described above.


D) clonogenic survival of cells treated with gemcitabine and irradiation. PSN-1 and MiaPaCa-2 cells were treated with 5 nM or 10 nM gemcitabine, respectively, for 24 h, medium was then replaced and 1 μM VE-821 was added from 1 h prior to 72 h post 4 Gy irradiation. Colony forming ability was assessed after 10 to 21 days (n=3). *, P<0.05; **, P<0.01; ***, P<0.001 over DMSO-treated control.



FIG. 4. VE-821 perturbs the irradiation-induced cell cycle checkpoint in pancreatic cancer cells.


VE-821 (1 μM) was added 1 h prior to 6 Gy irradiation and left for the duration of the experiment. Cells were lifted and fixed at 12 h or 24 h after irradiation, stained with propidium iodide and analysed for cell cycle distribution by flow cytometry (n=3)



FIG. 5. VE-821 increases 53BP1 and γH2AX foci number and reduces RAD51 foci formation.


Cells were treated with 1 μM VE-821 at various time points in relation to 6 Gy irradiation, as indicated, and fixed at 24 h post-irradiation. Subsequently, cells were stained for (A) γH2AX and (B) 53BP1 foci and the percentage of cells with more than 7 and 5 foci per cell was quantitated, respectively. C, for analysing Rad51 foci formation, cells were fixed at 6 h post-irradiation and the percentage of cells with more than 9 foci per cell was quantitated. Representative images are shown on the right (n=3). *, P<0.05



FIG. 6. Effect of VE-821 incubation time on plating efficiency.


PSN-1 cells were plated as single cells, treated with luM VE-821 for various time periods and assessed for colony formation after 10 days.



FIG. 7.


VE-821 perturbs the irradiation-induced G2/M checkpoint in pancreatic cancer cells in hypoxic conditions.


Cells were pre-incubated under hypoxic (0.5% O2) conditions for 6 h and 1 μM VE-821 was added 1 h prior to irradiation (6 Gy). Cells were transferred to normoxia 1 h after irradiation and were lifted and fixed at 12 h or 24 h after irradiation, stained with propidium iodide and analysed for cell cycle distribution by flow cytometry (n=3).



FIG. 8. Dose response relationship for radiosensitivity induced by Compounds 821, 822, 823, and 824.


Small scale clonogenic survival assays were performed on HeLa cells treated with the different ATR inhibitors at increasing concentrations followed by irradiation at 6 Gy. Data is plotted as decrease in clonogenic survival in relation to the DMSO-treated cells for both irradiated (SF 6 Gy, pink line) and unirradiated cells (plating efficiency, PE; blue line). A high degree of increased radiosensitivity can be seen as a large decrease in survival after irradiation accompanied by a small decrease in unirradiated survival at a specific drug concentration.



FIG. 9. Assessment of radiosensitivity in tumour cells and normal cells.

    • A) Clonogenic survival after drug treatment in the absence of irradiation. PSN1 and MiaPaca cells were plated at low densities, treated with the drugs indicated and assessed for clonogenic survival.
    • B) Clonogenic survival of PSN1, MiaPaca, and MRCS cells pretreated with Compounds 821, 822, 823 and 824 drugs followed by irradiation. Cells were plated at low densities, treated with drugs indicated 1 h prior to irradiation and assessed for clonogenic survival.



FIG. 10. Assessment of dependency of drug addition and removal timing on radiosensitivity.


MiaPaca cells were plated at low densities and drug was added at various time points in relation to the 4 Gy radiation treatment: 1 h prior to IR, 5 min after IR, 2 h or 4 h after IR; and removed at various time points. 5 min after, 1 h after, or 19 h after IR. Clonogenic survival was assessed after 14 days. Results are shown as the surviving fraction at 4 Gy (top panel) or the percentage radiosensitisation (middle panel) compared to the DMSO-treated cells. The different treatment schedules did not cause differences in plating efficiency (bottom panel).



FIG. 11. DNA damage foci analysis after Compound 822 treatment and irradiation.

    • A) Assessment of gH2AX, 53BP1 foci at 24 h after IR at 6 Gy and of RAD51 foci at 6 h after IR. MiaPaca cells were treated with 80 nM Compound 822 1 h prior or 1 h post irradiation and drug was washed away at 5 min after or 1 h after IR. Cells were fixed after 6 h (for RAD51 foci) or 24 h (for gH2AX and 53BP1 foci). The percentage of cells containing more than a certain number of foci was quantitated.
    • B) Time course of DNA damage foci. Cells were treated as in A and fixed at the time points shown followed by staining for gH2AX, 53BP1 and RAD51 foci. Data is shown as the mean number of foci at a particular time point (upper panels) or the percentage of cells containing more than a certain number of foci (lower panels).



FIG. 12. Cell cycle analysis of Compound 822-treated cells after 6 Gy irradiation.


PSN1 cells were treated with 40 nM Compound 822 1 h prior to 6 Gy irradiation in triplicate wells. Cells were lifted and fixed at several time points after IR, stained with propidium iodide and analysed by flow cytometry.

    • A) Cell cycle histogram plots. Fitted peaks are coloured red for G1 phase, shaded for S-phase, and green for G2/M phase. One out of three wells is shown for each time point and treatment.
    • B) Average cell cycle percentages over time. Cell cycle percentage values were obtained from fitted histogram plots (n=3) and plotted for control-treated and Compound 822-treated cells.



FIG. 13. MiaPaCa Tumor Volume over Time for Compound 822.



FIGS. 14 and 15. PSN-1 Tumor Volume over Time for Compound 822.



FIG. 16. Lung Cancer Cell Screen: VE-822 Synergizes with Chemotoxics Across a Panel of Lung Cancer Cell Lines in Lung Cell Viability Assay



FIG. 17. Lung Cancer Cell Screen: VE-822 Exhibits Greater than 3-fold Synergy with Chemotoxics in Lung Cancer Cell Lines in a Cell Viability Assay



FIG. 18. Pancreatic Cancer Cell Screen: VE-822 Synergizes with Cisplatin and Gemcitabine in Pancreatic Cancer Cell Lines in a Cell Viability Assay



FIG. 19. Pancreatic Cancer Cell Screen: VE-822 Exhibits Greater than 3-fold Synergy with Chemotoxics in Pancreatic Cancer Cell Lines a Cell Viability Assay



FIG. 20. Effect of VE-822 and cisplatin on tumor volume and body weight in a primary adenocarcinoma NSCLC xenograft in SCID mice.



FIG. 21: Effect of VE-822 administered PO q2d at 10, 30 or 60 mg/kg in combination with gemcitabine (15 mg/kg IP q3d) on the tumor volume of mice bearing PSN1 pancreatic cancer xenografts.





DETAILED DESCRIPTION OF THE INVENTION

One aspect of this invention provides methods for treating pancreatic cancer in a patient by administering to the patient an ATR inhibitor in combination with another known pancreatic cancer treatment. One aspect of the invention includes administering the ATR inhibitor in combination with gemcitabine. In some embodiments, the pancreatic cancer comprises one of the following cell lines: PSN-1, MiaPaCa-2 or Panc-1. According to another aspect, the cancer comprises the primary tumor line Panc-M.


Another aspect of the invention provides methods for treating cancer (e.g., pancreatic or non-small cell lung) in a patient by administering to the patient an ATR inhibitor in combination with radiation therapy.


Another aspect of the invention provides methods for treating non-small cell lung cancer in a patient by administering to the patient an ATR inhibitor in combination with cisplatin or carboplatin, etoposide, and/or ionizing radiation. Applicants have demonstrated synergy of ATR inhibitors in combination with cisplatin, etoposide, gemcitabine, oxaliplatin and irinotecan in viability assays against a panel of 35 human lung cancer cell lines as well as demonstrated in vivo efficacy in a lung cancer mouse model in combination with cisplatin. This invention also relates to the use of ATR inhibitors in combination with cisplatin or carboplatin, etoposide, and/or ionizing radiation for treating non-small cell lung cancer.


Examples of ATR inhibitors are shown in Table 1 below:











TABLE 1










embedded image


821








embedded image


822









The terms referring to compounds 821 and 822 are interchangeable with VE-821 and VE-822, respectively.


Another aspect provides a method of treating pancreatic cancer by administering to pancreatic cancer cells an ATR inhibitor selected from a compound in Table 1 in combination with one or more cancer therapies. In some embodiments, the ATR inhibitor is combined with chemoradiation, chemotherapy, and/or radiation therapy. As would be understood by one of skill in the art, chemoradiation refers to a treatment regime that includes both chemotherapy (such as gemcitabine) and radiation. In some embodiments, the chemotherapy is gemcitabine.


Yet another aspect provides a method of increasing the sensitivity of pancreatic cancer cells to a cancer therapy selected from gemcitabine or radiation therapy by administering an ATR inhibitor selected from a compound in Table 1 in combination with the cancer therapy.


In some embodiments, the cancer therapy is gemcitabine. In other embodiments, the cancer therapy is radiation therapy. In yet another embodiment the cancer therapy is chemoradiation.


Another aspect provides a method of inhibiting phosphorylation of Chk1 (Ser 345) in a pancreatic cancer cell comprising administering an ATR inhibitor selected from a compound in Table 1 after treatment with gemcitabine (e.g., 100 nM) and/or radiation (e.g., 6 Gy) to a pancreatic cancer cell.


Another aspect provides method of radiosensitizing hypoxic PSN-1, MiaPaCa-2 or PancM tumor cells by administering an ATR inhibitor selected from a compound in Table 1 to the tumor cell in combination with radiation therapy.


Yet another aspect provides a method of sensitizing hypoxic PSN-1, MiaPaCa-2 or PancM tumor cells by administering an ATR inhibitor selected from a compound in Table 1 to the tumor cell in combination with gemcitabine.


Another aspect provides a method of sensitizing PSN-1 and MiaPaCa-2 tumor cells to chemoradiation by administering an ATR inhibitor selected from a compound in Table 1 to the tumor cells in combination with chemoradiation.


Another aspect provides a method of disrupting damage-induced cell cycle checkpoints by administering an ATR inhibitor selected from a compound in Table 1 in combination with radiation therapy to a pancreatic cancer cell.


Another aspect provides a method of inhibiting repair of DNA damage by homologous recombination in a pancreatic cancer cell by administering an ATR inhibitor selected from a compound in Table 1 in combination with one or more of the following treatments: chemoradiation, chemotherapy, and radiation therapy.


In some embodiments, the chemotherapy is gemcitabine.


Another aspect provides a method of inhibiting repair of DNA damage by homologous recombination in a pancreatic cancer cell by administering an ATR inhibitor selected from a compound in Table 1 in combination with gemcitabine and radiation therapy.


In some embodiments, the pancreatic cancer cells are derived from a pancreatic cell line selected from PSN-1, MiaPaCa-2 or Panc-1.


In other embodiments, the pancreatic cancer cells are in a cancer patient. In other embodiments, the cancer cells are part of a tumor.


Another embodiment provides methods for treating non-small cell lung cancer in a patient by administering to the patient an ATR inhibitor in combination with other known non-small cell lung cancer treatments. One aspect of the invention includes administering to a patient an ATR inhibitor in combination with cisplatin or carboplatin, etoposide, and/or ionizing radiation.


Another aspect provides a method of treating non-small cell lung cancer by administering to a patient an ATR inhibitor selected from a compound in Table 1 in combination with one or more cancer therapies. In some embodiments, the ATR inhibitor is combined with chemoradiation, chemotherapy, and/or radiation therapy. As would be understood by one of skill in the art, chemoradiation refers to a treatment regime that includes both chemotherapy (such as cisplatin, carboplatin, or etoposide) and radiation. In some embodiments, the chemotherapy comprises Cisplatin or carboplatin, and etoposide.


Yet another aspect provides a method of increasing the sensitivity of non-small cell lung cancer cells to a cancer therapy selected from cisplatin or carboplatin, etoposide, and ionizing radiation by administering to a patient an ATR inhibitor selected from a compound in Table 1 in combination with one or more cancer therapy.


In some embodiments, the cancer therapy is cisplatin or carboplatin. In other embodiments, the cancer therapy is radiation therapy. In yet another embodiment the cancer therapy is etoposide.


In some embodiments, the cancer therapy is a combination of cisplatin or carboplatin, etoposide, and ionizing radiation. In some embodiments the cancer therapy is cisplatin or carboplatin and etoposide. In other embodiments the cancer therapy is cisplatin or carboplatin and etoposide and ionizing radiation. In yet other embodiments the cancer therapy is cisplatin or carboplatin and ionizing radiation.


Another aspect provides a method of inhibiting phosphorylation of Chk1 (Ser 345) in a non-small cell lung cancer cell comprising administering to a patient an ATR inhibitor selected from a compound in Table 1. In some embodiments, the ATR inhibitor is administered in combination with gemcitabine (e.g., 100 nM), cisplatin or carboplatin, etoposide, ionizing radiation or radiation (e.g., 6 Gy) to a non-small cell lung cancer cell.


In other embodiments, the non-small cell lung cancer cells are in a cancer patient.


In some embodiments, the ATR inhibitor is




embedded image


In other embodiments, the ATR inhibitor is




embedded image



Uses


Another aspect provides use of an ATR inhibitor selected from a compound in Table 1 in combination with gemcitabine and radiation therapy for treating pancreatic cancer.


Another aspect provides use of an ATR inhibitor selected from a compound in Table 1 in combination with cisplatin or carboplatin, etoposide, and ionizing radiation for treating non-small cell lung cancer.


In some embodiments, the ATR inhibitor is Compound VE-821. In other embodiments, the ATR inhibitor is Compound VE-822.


Manufacture of Medicaments


Another aspect provides use of an ATR inhibitor selected from a compound in Table 1 in combination with gemcitabine and radiation therapy for the manufacture of a medicament for treating pancreatic cancer.


Another aspect provides use of an ATR inhibitor selected from a compound in Table 1 in combination with cisplatin or carboplatin, etoposide, and ionizing radiation for the manufacture of a medicament for treating non-small cell lung cancer.


In some embodiments, the ATR inhibitor is Compound VE-821. In other embodiments, the ATR inhibitor is Compound VE-822.


EXAMPLES

The examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention in any way.


Cell Viability Assays


MiaPaCa-2, PSN-1, Panc1 and MRCS cells (5×104) were plated in 96-well plates and after 4 h treated with increasing concentrations of VE-821 at 1 h before irradiation with a single dose of 6 Gy. Medium was replaced 96 h post-irradiation at which point viability was measured using the using the Alamar Blue assay (Resazurin substrate, SIGMA). Cells were allowed to proliferate and cell viability was again analyzed at day 8 for the different treatment conditions. Cell viability and surviving fraction were normalized to the untreated (control) group.


Clonogenic Survival Assay


Logarithmically growing cells were plated in triplicate in 6-well tissue culture dishes under oxic (21% O2) or hypoxic conditions (0.5% O2) using an InVivo2 300 chamber (Ruskinn Technology, UK). Cells were incubated for 6 hours before irradiation under oxia or hypoxia using tightly sealed chambers. The target O2 level was achieved within 6 h of gassing and maintained during irradiation, as confirmed by an OxyLite oxygen probe (Oxford Optronix). Cells irradiated under hypoxia were exposed to normoxia at 1 h post-irradiation. As standard, VE-821 (1 μM) was added 1 h prior to irradiation (6 Gy) and was washed away 72 h after irradiation. For the chemotherapy experiments, cells were initially exposed to increasing concentrations of gemcitabine (5, 10 and 20 nM) for 24 h before addition of the VE-821 (1 μM) for another 72 h. The effect of triple combination of irradiation with VE-821 and gemcitabine was examined as well. Cells were incubated for 10-21 days until colonies were stained with 0.5% crystal violet and counted in a CellCount automated colony counter (Oxford Optronix). Clonogenic survival was calculated and data were fitted in the GraphPad Prism 4.0 (GraphPad Software, CA).


Western Blot


MiaPaCa-2 and PSN-1 cells were exposed to gemcitabine and/or 1 μM VE-821 drug 1 h prior to irradiation with a single dose of 6 Gy. Cells were lysed in RIPA buffer 2 h post-irradiation and subjected to SDS-PAGE electrophoresis and immunoblotting. Chemoluminescence (SuperSignal, Millipore) and film exposure was used to detect antibody binding. Exposed film was digitized and figures were assembled using Microsoft PowerPoint.


Nuclear Foci Analysis


Cells growing in 96-well plates were treated with 1 μM VE-821 drug 1 h prior to 6 Gy irradiation and fixed in 3% formaldehyde at multiple time points. Cells were subsequently pearmeabalised and blocked in PBS with 0.1% Triton 1% BSA (w/v). Cells were incubated with primary antibody overnight at 4° C. and after a PBS wash incubated with fluorescently labeled secondary antibody followed gy a PBS wash and nuclear staining with DAPI. Images were acquired and foci quantitated using an IN Cell Analyzer 1000 automated epifluorescence microscope and analysis software (GE Healthcare, Cahlfont St. Giles, UK)


Cell Cycle Analysis


Cells growing in 6-well dishes were treated with 1 μM VE-821 drug 1 h prior to 6 Gy irradiation. Cells were incubated for 6 h before irradiation under oxia (21% O2) or hypoxia (0.5% O2) using tightly sealed chambers. At multiple time points, cells were lifted in trypsin and fixed in 70% ethanol and stored at 4° C. Cells were incubated with propidium iodide (50 μg/ml in PBS containing 200 μg/ml RNAse) for 1 h at room temperature and analysed by flow cytometry (FACSort, Becton Dickinson). Cell cycle phase was quantitated using ModFit Cell Cycle Analysis software.


Cell Seeding and Compound Addition for Lung Cancer Cell Screen


All cell lines were seeded in 30 μl of tissue culture medium containing 10% FBS into 384-well opaque-bottom assay plates. The seeding density was based on the logarithmic growth rate of each cell line. After 24 hours, compound stock solutions were added to each well to afford a matrix consisting of 5 concentrations for VE-822 and 6 concentrations for chemotoxics. Each well contains either, agent alone or a combination of both agents. The final concentration range for VE-822 was 25 nM-2 μM. The concentration ranges for the chemotoxics were as follows: Etoposide, 10 nM-10 μM; Gemcitabine, 0.16 nM-160 nM; Cisplatin, 20 nM-20 μM; Oxaliplatin, 40 nM-40 μM; Irinotecan (SN-38), 0.12 nM-120 nM. The cells were then incubated for 96 hours at 37° C. in an atmosphere of 5% CO2 and 95% humidity.


Cell Seeding and Compound Addition for the Pancreatic Cancer Cell Screen


All cell lines were seeded in 30 μl of tissue culture medium containing 10% FBS into 384-well opaque-bottom plates. The seeding density was based on the logarithmic growth rate of each cell line. After 24 hours, compound stock solutions were added to each well to afford a matrix consisting of 9 concentrations for VE-822 and 7 concentrations for Gemcitabine and Cisplatin. Each well contains either, agent alone or a combination of both agents. The final concentration ranges were as follows: VE-822, 0.3 nM-2 μM; Gemcitabine, 0.3 nM-0.22 μM; Cisplatin, 30 nM-20 μM. The cells were then incubated for 96 hours at 37° C. in an atmosphere of 5% CO2 and 95% humidity.


Cell Viability Assay


This assay measures the number of viable cells in a culture based on the quantitation of ATP, which is present in metabolically active cells.


CellTiter-Glo Reagent (Promega, Madison, Wis., USA) was prepared according to the manufacturer's instructions and added 96 hours after compound addition (25 μl/well) to measure cell viability. Luminescence signal was measured with the PHERAStarFS (BMG Labtech, Cary, N.C., USA) automated plate reader. All cell lines were screened in duplicate.


Raw luminescence CellTiter-Glo (CTG) values were normalized to the mean CTG value for the negative control DMSO-treated samples on each assay plate. IC50 values for chemotoxic alone were calculated using DMSO-normalized cell survival values for the samples treated with chemotoxic compound alone. To determine fraction of cell survival in the presence of VE-822, raw CTG values were normalized to the mean CTG value for the samples exposed to the same concentration of VE-822 in the absence of the chemotoxic compound. VE-822-treated chemotoxic IC50 values were calculated using VE-822-normalized cell survival values for all samples treated with the chemotoxic at a given concentration of VE-822. A 3× or greater reduction in IC50 was used to identify strongly synergistic effects between VE-822 and chemotoxics.


Primary Adenocarcinoma NSCLC Xenograft Model


Tumor tissue was excised from a patient with a poorly differentiated adenocarcinoma. This tumor tissue was implanted subcutaneously in the flank of a SCID mouse and passaged twice before compound testing. For compound testing passage-two tumor tissue was implanted subcutaneously in the flank of SCID mice and tumors grown to a volume of about 200 mm3 Cisplatin was dosed alone at either 1 or 3 mg/kg ip, once per week (ip, q7d, on day 2 of each week) for two weeks. VE-822 was dosed as a solution alone at 60 mg/kg po on 4 consecutive days per weekly cycle (qd4, dosed on days 1, 2, 3 and 4 each week). Two combination groups received cisplatin at 1 or 3 mg/kg plus VE-822 at 60 mg/kg po on the same schedule as the single agent group. A control group received vehicle alone (10% Vitamin E TPGS in water, po qd4). All drug treatment was stopped on Day 28. Vehicle, cisplatin (1 mg/kg) and VE-822 (60 mg/kg) groups were sacrificed and the remainder monitored for a further 40 days to assess tumor re-growth.


PSN1 Pancreatic Cancer Xenograft Model


PSN1 cells (1×106 cells per mouse) were implanted as a mixture in Matrigel (100 μl per mouse) into the flank of female nude MF1 mice and grown to a volume of about 200 mm3 prior to compound administration. Gemcitabine was dosed alone at 15 mg/kg ip, once every three days (ip, q3d) in 0.5% methylcellulose in water for a maximum of 10 cycles. VE-822 was dosed, as a suspension in 0.5% methylcellulose in water, alone at either 10, 30 or 60 mg/kg po every other day for 28 days (po q2d). Three combination groups received gemcitabine at 15 mg/kg plus VE-822 either at 10, 30 or at 60 mg/kg po on the same schedule as the single agent groups. A control group received vehicle alone (0.5% methylcellulose ip q3d). All drug treatment was stopped on Day 30. Vehicle and VE-822 groups were sacrificed on day 13 due to excessive tumor volumes.


Results


Compounds VE-821 and VE-822 Sensitize Pancreatic Cancer Cells to Radiation Therapy


Compound VE-821 inhibits phosphorylation of Chk1 (Ser 345) after treatment with gemcitabine (100 nM), radiation (6 Gy) or both (see FIG. 1, panel A). Compound VE-821 radiosensitises pancreatic tumour cells but not normal cells. When cells were irradiated in the presence of Compound VE-821, a decrease in surviving fraction was observed and this radiosensitising effect increased as the drug incubation time after irradiation was extended (see FIG. 1, panel C).


Compound VE-821 radiosensitises tumour PSN-1, MiaPaCa-2 and PancM cells under hypoxic conditions (see FIG. 2, panels A-B). Compound VE-821 also sensitises normoxic and hypoxic cancer cells to gemcitabine (see FIG. 3, panels B-C). Compound VE-821 potentiates the effect of chemoradiation in both PSN-1 and MiaPaCa-2 cancer cells (see FIG. 3, panel D). Compound VE-821 disrupts damage-induced cell cycle checkpoints (see FIG. 7). Compound VE-821 inhibits repair of DNA damage by homologous recombination (see FIG. 5, panels A, B, and C).


Results for Compounds 821 and 822 are shown in FIGS. 8 to 15 and FIGS. 16 to 21. VE-821 and VE-822 sensitize cancer cells to radiation therapy (see FIGS. 8 to 12).


VE-822 Enhances the Antitumor Effects of Cancer Therapies in Xenograft Models


VE-822 enhances the antitumor effects of ionizing radiation in a MiaPaCa pancreatic cancer xenograft model (see FIG. 13) and in a PSN-1 pancreatic cancer xenograft model (see FIGS. 14 and 15).


VE-822 enhances the antitumor effects of cisplatin in a primary adenocarcinoma NSCLC xenograft model. FIG. 20 shows the effect of VE-822 and cisplatin on tumor volume and body weight in a primary adenocarcinoma NSCLC xenograft in SCID mice. Data are mean±sem, n=9-10. Black filled circles are vehicle treatment; Red filled diamonds are Cisplatin treatment (1 mg/kg q7d); Blue filled diamonds are Cisplatin treatment (3 mg/kg q7d); Green filled squares are VE-822 treatment (60 mg/kg qd4); Green empty triangles are Cisplatin (1 mg/kg) and VE-822 (60 mg/kg qd4); Blue empty triangles are Cisplatin (3 mg/kg) and VE-822 (60 mg/kg qd4) (see FIG. 20).


VE-822 also enhances the antitumor effects of gemcitabine in a PSN1 pancreatic cancer xenograft model. FIG. 21 shows the effect of VE-822 administered PO q2d at 10, 30 or 60 mg/kg in combination with gemcitabine (15 mg/kg IP q3d) on the tumor volume of mice bearing PSN1 pancreatic cancer xenografts. Data shown are mean tumor volume±SEM (n=8 per group). Red filled circles are VE-822 treatment; Black filled squares are vehicle treatment; Green filled circles are gemcitabine treatment; Blue filled circles are gemcitabine and VE-822 (10 mg/kg) treatment; Red filled circles are gemcitabine and VE-822 (30 mg/kg) treatment; Pink filled circles are gemcitabine and VE-822 (60 mg/kg) treatment;


VE-822 Synergizes with Chemotoxics Across a Panel of Lung Cancer Cell Lines


The heat map represents the maximum shift in IC50 of each chemotoxic achieved when combined with VE-822 for 96 hours. Colors represent an IC50 shift range from −10 (antagonism, blue) to 10 (synergy, red) (see FIG. 16). VE-822 exhibits greater than 3-fold synergy with cisplatin, etoposide, gemcitabine, oxaplatin and irinotecan in lung cancer cell lines (see FIG. 17).


VE-822 Synergizes with Cisplatin and Gemcitabine in Pancreatic Cancer Cell Lines.


The heat map represents the maximum shift in IC50 of each chemotoxic achieved when combined with VE-822 for 96 hours. Colors represent an IC50 shift range from −10 (antagonism, blue) to 10 (synergy, red) (see FIG. 18).


While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds, methods, and processes of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example herein.

Claims
  • 1. A method of treating non-small cell lung cancer in a patient, comprising administering to a patient with non-small cell lung cancer a therapeutic amount of compound 822:
CROSS REFERENCE TO RELATED APPLICATIONS

The present application is a continuation of U.S. patent application Ser. No. 13/633,114, filed on Oct. 1, 2012, which claims the benefit of U.S. provisional application No. 61/542,084 filed on Sep. 30, 2011, the entire contents of each of which are incorporated herein by reference.

US Referenced Citations (148)
Number Name Date Kind
4309430 Bock et al. Jan 1982 A
5143824 Yamakawa et al. Sep 1992 A
6469002 Ohshima et al. Oct 2002 B1
6660753 Van Wagenen et al. Dec 2003 B2
6790935 Mutter et al. Sep 2004 B1
6858600 Hamilton et al. Feb 2005 B2
6992087 Verhoest et al. Jan 2006 B2
7041672 Verhoest et al. May 2006 B2
7043079 Malvar et al. May 2006 B2
7145002 Brands et al. Dec 2006 B2
7199123 Munchhof Apr 2007 B2
7277118 Foote Oct 2007 B2
7385626 Aggarwal et al. Jun 2008 B2
7394926 Bryll et al. Jul 2008 B2
7452993 Arnold et al. Nov 2008 B2
7574131 Chang et al. Aug 2009 B2
7622583 Ungashe et al. Nov 2009 B2
7626021 Arnold et al. Dec 2009 B2
7700601 Chan et al. Apr 2010 B2
7704995 Buhr et al. Apr 2010 B2
7829558 Arnold et al. Nov 2010 B2
7872031 Lauffer et al. Jan 2011 B2
7902197 Elworthy et al. Mar 2011 B2
7932254 DuBois et al. Apr 2011 B2
7939531 Bamberg et al. May 2011 B2
7981893 Mortensen et al. Jul 2011 B2
8063032 Chytil et al. Nov 2011 B2
8106197 Cui et al. Jan 2012 B2
8410112 Charrier et al. Apr 2013 B2
8492582 Yokotani et al. Jul 2013 B2
8765751 Charrier et al. Jul 2014 B2
8841308 Charrier et al. Sep 2014 B2
8841337 Charrier et al. Sep 2014 B2
8841449 Charrier et al. Sep 2014 B2
8841450 Charrier et al. Sep 2014 B2
8846686 Charrier et al. Sep 2014 B2
8846917 Charrier et al. Sep 2014 B2
8846918 Charrier et al. Sep 2014 B2
8853217 Charrier et al. Oct 2014 B2
8877759 Charrier et al. Nov 2014 B2
8912198 Charrier et al. Dec 2014 B2
8962631 Charrier et al. Feb 2015 B2
8969356 Charrier et al. Mar 2015 B2
8999632 Falcon et al. Apr 2015 B2
9035053 Charrier et al. May 2015 B2
9062008 Charrier et al. Jun 2015 B2
9096584 Charrier et al. Aug 2015 B2
9334244 Charrier et al. May 2016 B2
9340546 Ahmad et al. May 2016 B2
9365557 Charrier et al. Jun 2016 B2
9650381 Ahmad et al. May 2017 B2
9670215 Ahmad et al. Jun 2017 B2
9701674 Charrier et al. Jul 2017 B2
9718827 Ahmad et al. Aug 2017 B2
9791456 Falcon et al. Oct 2017 B2
9862709 Charrier et al. Jan 2018 B2
20020064314 Comaniciu et al. May 2002 A1
20020158984 Brodsky et al. Oct 2002 A1
20020180759 Park et al. Dec 2002 A1
20020195563 Iida et al. Dec 2002 A1
20030008882 Hamilton et al. Jan 2003 A1
20030055085 Wagenen et al. Mar 2003 A1
20030199511 Li et al. Oct 2003 A1
20040034037 Harbeson et al. Feb 2004 A1
20040075741 Berkey et al. Apr 2004 A1
20040100560 Stavely et al. May 2004 A1
20040175042 Kroeker et al. Sep 2004 A1
20040180905 Munchhof Sep 2004 A1
20040202382 Pilu Oct 2004 A1
20040252193 Higgins Dec 2004 A1
20040264793 Okubo Dec 2004 A1
20050116968 Barrus et al. Jun 2005 A1
20050123902 Meneses et al. Jun 2005 A1
20050207487 Monroe Sep 2005 A1
20060083440 Chen Apr 2006 A1
20060211709 Buhr et al. Sep 2006 A1
20070037794 Ungashe et al. Feb 2007 A1
20070092245 Bazakos et al. Apr 2007 A1
20070120954 Allen et al. May 2007 A1
20070149547 Bonnefous et al. Jun 2007 A1
20070254868 Lauffer et al. Nov 2007 A1
20070270420 Harbeson et al. Nov 2007 A1
20070287711 Arnold et al. Dec 2007 A1
20080132698 Fagnou et al. Jun 2008 A1
20090001843 Enomoto et al. Jan 2009 A1
20090005381 Brown et al. Jan 2009 A1
20090143410 Patel Jun 2009 A1
20090215724 DuBois et al. Aug 2009 A1
20090215750 Bamberg et al. Aug 2009 A1
20090215785 DuBois et al. Aug 2009 A1
20090215788 Elworthy et al. Aug 2009 A1
20090306087 Ibrahim et al. Dec 2009 A1
20100036118 Arnold et al. Feb 2010 A1
20100168138 DeGoey et al. Jul 2010 A1
20100204214 Chytil et al. Aug 2010 A1
20100222318 Charrier Sep 2010 A1
20100233091 Neumann et al. Sep 2010 A1
20100249387 Inouye Sep 2010 A1
20110015231 Al-Abed et al. Jan 2011 A1
20110275797 Yokotani et al. Nov 2011 A1
20110288067 Hendricks et al. Nov 2011 A1
20110288097 Hendricks et al. Nov 2011 A1
20120025805 Matsushita et al. Feb 2012 A1
20120027874 Charrier et al. Feb 2012 A1
20120035407 Charrier et al. Feb 2012 A1
20120035408 Charrier et al. Feb 2012 A1
20120040020 Charrier et al. Feb 2012 A1
20120046295 Charrier et al. Feb 2012 A1
20120065247 Thompson et al. Mar 2012 A1
20120115874 Wang et al. May 2012 A1
20120122884 Charrier et al. May 2012 A1
20120177748 Charrier et al. Jul 2012 A1
20120178756 Charrier et al. Jul 2012 A1
20130017273 Everitt et al. Jan 2013 A1
20130018035 MacCormick et al. Jan 2013 A1
20130034616 Storck et al. Feb 2013 A1
20130089624 Charrier et al. Apr 2013 A1
20130089625 Charrier et al. Apr 2013 A1
20130089626 Pollard et al. Apr 2013 A1
20130095193 Charrier et al. Apr 2013 A1
20130096139 Charrier et al. Apr 2013 A1
20130115310 Charrier et al. May 2013 A1
20130115311 Charrier et al. May 2013 A1
20130115312 Charrier et al. May 2013 A1
20130115313 Charrier et al. May 2013 A1
20130115314 Charrier et al. May 2013 A1
20130172273 Aizpurua Iparraguirre et al. Jul 2013 A1
20130184292 Charrier et al. Jul 2013 A1
20140044802 Pollard et al. Feb 2014 A1
20140107093 Charrier et al. Apr 2014 A1
20140113005 Charrier et al. Apr 2014 A1
20140134596 Falcon et al. May 2014 A1
20140163000 Ahmad et al. Jun 2014 A1
20140249157 Ahmad et al. Sep 2014 A1
20150031661 Charrier et al. Jan 2015 A1
20150051187 Charrier et al. Feb 2015 A1
20150158872 Charrier et al. Jun 2015 A1
20150239874 Charrier et al. Aug 2015 A1
20150247866 Falcon et al. Sep 2015 A1
20150274710 Charrier et al. Oct 2015 A1
20150353560 Ahmad et al. Dec 2015 A1
20150359797 Helleday et al. Dec 2015 A1
20160030424 Pollard et al. Feb 2016 A1
20160271129 Charrier et al. Sep 2016 A1
20160311809 Charrier et al. Oct 2016 A1
20160347754 Ahmad et al. Dec 2016 A1
20170349596 Ahmad et al. Dec 2017 A1
20180072735 Ahmad et al. Mar 2018 A1
Foreign Referenced Citations (188)
Number Date Country
1551869 Dec 2004 CN
101001606 Jul 2007 CN
101479255 Jul 2009 CN
101537007 Sep 2009 CN
101652354 Feb 2010 CN
101671336 Mar 2010 CN
103373996 Oct 2013 CN
313724 May 1989 EP
1217000 Jun 2002 EP
2157090 Feb 2010 EP
62270623 Nov 1987 JP
63208520 Aug 1988 JP
2002072370 Mar 2002 JP
2002072372 Mar 2002 JP
2002518389 Jun 2002 JP
2003074370 Mar 2003 JP
2003516974 May 2003 JP
2005511531 Apr 2005 JP
2005530760 Oct 2005 JP
2006156445 Jun 2006 JP
2006516124 Jun 2006 JP
2006519232 Aug 2006 JP
2006519833 Aug 2006 JP
2006520794 Sep 2006 JP
2006521357 Sep 2006 JP
2006526031 Nov 2006 JP
2007524682 Aug 2007 JP
2008510790 Apr 2008 JP
2008510792 Apr 2008 JP
2008517945 May 2008 JP
2008525453 Jul 2008 JP
2008543754 Dec 2008 JP
2009503103 Jan 2009 JP
2009027904 Feb 2009 JP
2009530233 Aug 2009 JP
2009532356 Sep 2009 JP
2009533327 Sep 2009 JP
2009541247 Nov 2009 JP
2009541268 Nov 2009 JP
2010506934 Mar 2010 JP
2010509356 Mar 2010 JP
2010077286 Apr 2010 JP
2010513433 Apr 2010 JP
2010180180 Aug 2010 JP
2011500778 Jan 2011 JP
2011042639 Mar 2011 JP
2012508260 Apr 2012 JP
2012513398 Jun 2012 JP
2012533248 Dec 2012 JP
2013501720 Jan 2013 JP
2013505900 Feb 2013 JP
2013517264 May 2013 JP
2013525476 Jun 2013 JP
2014510072 Apr 2014 JP
2014518545 Jul 2014 JP
2014165380 Sep 2014 JP
593316 Jun 2013 NZ
593969 Nov 2013 NZ
1997043267 Nov 1997 WO
1998042701 Oct 1998 WO
WO 9944609 Sep 1999 WO
2000004014 Jan 2000 WO
WO 0076982 Dec 2000 WO
2001044206 Jun 2001 WO
2002009648 Feb 2002 WO
WO 02080899 Oct 2002 WO
2003004472 Jan 2003 WO
2003004475 Jan 2003 WO
WO 03032971 Apr 2003 WO
2003045924 Jun 2003 WO
2003076422 Sep 2003 WO
2003080610 Oct 2003 WO
2003087057 Oct 2003 WO
2003092686 Nov 2003 WO
2003093297 Nov 2003 WO
2003101968 Dec 2003 WO
2004000318 Dec 2003 WO
WO 2004000820 Dec 2003 WO
2004033431 Apr 2004 WO
2004055005 Jul 2004 WO
2004055006 Jul 2004 WO
2004080982 Sep 2004 WO
WO 2004076412 Sep 2004 WO
2004084813 Oct 2004 WO
2004084824 Oct 2004 WO
2004085409 Oct 2004 WO
2004103279 Dec 2004 WO
WO 2004103369 Dec 2004 WO
WO 2004103991 Dec 2004 WO
2005028475 Mar 2005 WO
WO 2005058876 Jun 2005 WO
2005079802 Sep 2005 WO
2005123672 Dec 2005 WO
2006015124 Feb 2006 WO
WO 2006021886 Mar 2006 WO
2006053342 May 2006 WO
WO 2006047504 May 2006 WO
2006058074 Jun 2006 WO
2006067462 Jun 2006 WO
2006071548 Jul 2006 WO
2006075152 Jul 2006 WO
2006088837 Aug 2006 WO
2006114180 Nov 2006 WO
2006120573 Nov 2006 WO
WO 2006124874 Nov 2006 WO
WO 2006135604 Dec 2006 WO
2007015632 Feb 2007 WO
WO 2007016674 Feb 2007 WO
2007058850 May 2007 WO
2007063012 Jun 2007 WO
2007066805 Jun 2007 WO
2007076360 Jul 2007 WO
2007096151 Aug 2007 WO
2007096764 Aug 2007 WO
2007096765 Aug 2007 WO
WO 2007095588 Aug 2007 WO
2007102770 Sep 2007 WO
2007111904 Oct 2007 WO
2007126964 Nov 2007 WO
2007147874 Dec 2007 WO
WO 2007147746 Dec 2007 WO
WO 2008025820 Mar 2008 WO
2008037477 Apr 2008 WO
2008038010 Apr 2008 WO
2008040651 Apr 2008 WO
2008060907 May 2008 WO
WO 2008051493 May 2008 WO
2008071456 Jun 2008 WO
2008074997 Jun 2008 WO
2008079291 Jul 2008 WO
2008079903 Jul 2008 WO
2008079906 Jul 2008 WO
2008103277 Aug 2008 WO
2008106692 Sep 2008 WO
2008122375 Oct 2008 WO
2008124850 Oct 2008 WO
2008141065 Nov 2008 WO
2008144463 Nov 2008 WO
2008144464 Nov 2008 WO
2008157191 Dec 2008 WO
WO 2008156174 Dec 2008 WO
2009007390 Jan 2009 WO
2009012482 Jan 2009 WO
2009014637 Jan 2009 WO
WO 2009005638 Jan 2009 WO
2009016460 Feb 2009 WO
2009024825 Feb 2009 WO
2009037247 Mar 2009 WO
2009053737 Apr 2009 WO
WO 2009099982 Aug 2009 WO
2009106885 Sep 2009 WO
WO 2009111280 Sep 2009 WO
WO 2009115517 Sep 2009 WO
2010015803 Feb 2010 WO
WO 2010016005 Feb 2010 WO
WO 2010017055 Feb 2010 WO
2010048131 Apr 2010 WO
2010054398 May 2010 WO
2010063634 Jun 2010 WO
2010068483 Jun 2010 WO
2010071837 Jun 2010 WO
WO 2010071837 Jun 2010 WO
WO 2010073034 Jul 2010 WO
WO 2010075200 Jul 2010 WO
2011006074 Jan 2011 WO
2011008830 Jan 2011 WO
WO 2011017513 Feb 2011 WO
WO 2011035855 Mar 2011 WO
WO 2011044157 Apr 2011 WO
WO 2011086531 Jul 2011 WO
2011117145 Sep 2011 WO
2011124998 Oct 2011 WO
2011130689 Oct 2011 WO
2011143399 Nov 2011 WO
2011143419 Nov 2011 WO
2011143422 Nov 2011 WO
2011143423 Nov 2011 WO
2011143425 Nov 2011 WO
2011143426 Nov 2011 WO
2011144584 Nov 2011 WO
2011144585 Nov 2011 WO
WO 2011138751 Nov 2011 WO
WO 2011143425 Nov 2011 WO
2012138938 Oct 2012 WO
2012158785 Nov 2012 WO
2013049726 Apr 2013 WO
WO 2013049722 Apr 2013 WO
WO 2013049859 Apr 2013 WO
Non-Patent Literature Citations (216)
Entry
Rosen et al., Premetrexed combination therapy in the treatment of non-small cell lung cancer, 2002, Semin Oncol, 2, Suppl 5, pp. 23-29.
Wagner et al., Prospects for the Use of ATR Inhibitors to Treat Cancer, 2010, Pharmaceuticals, 3, pp. 1311-1334 (Year: 2010).
Abdel-Magid, A., “Inhibitors of ATR Kinase for Treatment on Cancer”, ACS Medicinal Chemistry Letters, 4(8), (2013), pp. 688-689.
Ammar, Y.A., et al., “3-Ethoxycarbonylmethylenequinoxalin-2-one in Heterocyclic Synthesis. Part 1: Synthesis of New Substituted and Condensed Quinoxalines”, Afinidad (2005), 62, pp. 151-160.
Charrier, J.D., et al, “Discovery of Potent and Selective Inhibitors of Ataxia Telangiesctasia Mutated and Rad3 Related (ATR) Protein Kinase as Potential Anticancer Agents” J. Med. Chem. (Mar. 17, 2011), 54(7), pp. 2320-2330 (DOI: 10.1021/jm101488z).
Charrier, J.D., et al., “Discovery of Potent and Selective Inhibitors of ATR (Ataxia Telangiectasia Mutated and Rad3 Related) as Potential Anticancer Agents”, Supplementary Information, Apr. 14, 2011.
Charrier, J.D., “Discovery of potent and selective inhibitors of Ataxia Telangiectasia mutated and Rad3 related (ATR) protein kinase as potential anticancer agents”, Presentation, ACS Denver 2011, Aug. 28, 2011.
Clark, B.A.J., et al., “Mass Spectrometry of Pyrroloä2, 3-Büpyrazines and Pyrazinoä2,3-Büindole”, Organic Mass Spectrometry, 12(7), (1997), pp. 421-423.
Curtin, N.J., “Inhibiting the DNA damage response as a therapeutic manoeuvre in cancer”, British Journal of Pharmacology, (2013), pp. 1-52.
El-Emary, T.I., “Synthesis and Biological Activity of Some New Pyrazolo[3,4-b]pyrazines”, J. Chin. Chem. Soc. (2006), 53, pp. 391-401.
Fernandes, P.S., et al., “Synthesis and Biological Activity of Heterocyclic Derivatives derived from Ethyl-2-hydroxy-quinoxaline-3-carboxylate”, J. Indian Chem. Soc. (1986), 63, pp. 427-429.
Finlay, M.R., et al., “Modulation of DNA repair by pharmacological inhibitors of the PIKK protein kinase family”, Bioorg. Med. Chem. Letters, 22(17) (2012), pp. 5352-5359.
Fokas, E., et al., “Targeting ATR in vivo using the novel inhibitor VE-822 results in selective sensitization of pancreatic tumors to radiation”, Cell Death and Disease, 3 (2012), pp. 1-5 (DOI: 10.1038/cddis.2012.181).
Fokas, E., et al., “Targeting ATR in DNA damage response and cancer therapeutics”, Cancer Treatment Reviews (2013), (DOI: 10.1016/j.ctrv.2013.03.002).
Gentili, F., et al., “Alpha2-Adrenoreceptors Profile Modulation. 4. From Antagonist to Agonist Behavior”, J. Med. Chem., 51(14), Jun. 25, 2008), pp. 4289-4299.
Hall-Jackson, C.A., et al., “ATR is a caffeine-sensitive, DNA-activated protein kinase with a substrate specificity distinct from DNA-PK”, Oncogene, 18(48) (1999), pp. 6707-6713.
Hickson, I., et al., “Identification and Characterization of a Novel and Specific Inhibitor of the Ataxia-Telangiectasia Mutated Kinase ATM”, Cancer Research (2004), 64, pp. 9152-9159.
Hilton, S., et al., “Identification and characterisation of 2-aminopyridine inhibitors of checkpoint kinase 2”, Bioorg. Med. Chem., (2010) 18, pp. 707-718.
Jiang, B., et al., “Synthesis and cytotoxicity evaluation of novel indolylpyrimidiens and indolylpyrazines as potential antitummor agents”, Bioorganic & Medicinal Chemistry, 9 (2001), pp. 1149-1154.
Kim, S.T., et al., “Substrate Specificities and Identification of Putative Substrates of ATM Kinase Family Members”, J. Biol. Chem. (1999) 274, pp. 37538-37543.
Klicnar, J., et al., “Studien in der Chinoxalinreihe III. Syntheses, Reaktionen und ir-spektren einiger 3-hydroxy-2-carboxymethylchinoxalin-derivative”, Collection Czechoslay. Chem. Commun. (1965), 30, pp. 3092-3101.
Kurasawa, Y., et al., “Revised Structure for the Product from the Reaction of 3-Hydrazinocarbonylmethylene-2-oxo-1,2,3,4-tetrahydroquinoxaline with Nitrous Acid”, Chem. Pharm. Bull. (1984), 32(10), pp. 4140-4143.
Luo, H., et al., “Molecular dynamics-based self-organizing molecular field analysis on 3-amino-6-arypyrazines as the ataxia telangiectasia mutated and Rad3 related (ATR) protein kinase inhibitors”, Medicinal Chemistry Research, (2013), pp. 1-12.
McKenna, G., et al., “Evaluation of the first potent and highly selective inhibitor of ATR kinase: an approach to selectively sensitize cancer cells to ionizing radiation and hypoxia”, Abstract, Mar. 31, 2012.
McKenna, G., et al., “Evaluation of the first potent and highly selective inhibitor of ATR inhibitor, VE-821: an approach to selectively sensitize cancer cells to ionizing radiation and hypoxia”, Poster, Mar. 31, 2012.
Middleton, F., et al., “ATR as a Therapeutic Target”, Advances in DNA Repair in Cancer, Northern Institute for Cancer Research, Newcastle University (2013), pp. 211-228.
Nakamura, H., et al., “Bimodal chemiluminescence of 8-chlorostyryl-6-phenylethynylimidazopyrazinone: Large bathochromic shift caused by a styryl group at 8-position”, Tetrahedron Letters, 39, (1998), pp. 301-304.
Pires, I.M., et al., “Targeting radiation-resisitant hypoxic tumour cells thorugh ATR inhibition”, British Journal of Cancer, Jun. 19, 2012, pp. 1-9.
Pollard, J., “Inhibition of the DNA Damage Response Kinase, ATR, as a Promising Anti-Cancer Approach”, Presentation, Mar. 8, 2012.
Qi, et al., “Chemi- and bio-luminescence of coelenterazine analogs with phenyl homologs at the C-2 position”, Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry, 13, (1992), pp. 1607-1611.
Reaper, P.M., et al., “Selective Killing of ATM- or p53-deficient cancer cells through inhibition of ATR”, Supplementary Information, Nature Chemical Biology, Apr. 13, 2011, DOI: 10.1038/NCHEMBIO.573.
Reaper, P.M., et al., “Selective Killing of ATM- or p53-deficient cancer cells through inhibition of ATR”, Presentation, Nov. 21, 2011.
Reaper, P.M., et al., “Selective Killing of ATM- or p53-deficient cancer cells through inhibition of ATR”, Presentation, Nov. 29, 2011.
Reaper, P.M., et al., “Evaluation of a potent and highly selective inhibitor of ATR kinase: an approach to selectively sensitize cancer cells to genotoxic drugs”, Abstract, Mar. 31, 2012.
Reaper, P.M., et al., “Evaluation of a Potent and Highly Selective Inhibitor of ATR Kinase: An Approach to Selectively Sensitize Cancer Cells to Genotoxic Drugs”, Poster, Mar. 31, 2012.
Sarkaria, J.N., et al., “Inhibition of ATM and ATR Kinase Activities by the Radiosensitizing Agent, Caffeine”, Cancer Research (1999) 59, pp. 4375-4382.
Sugimoto, T., et al., “Imidazopteridines. I. Synthesis of Imidazo[1,2-c]pteridine and its Alkyl Derivatives”, Bull. Chem. Soc. Japan (1977) 50(10), pp. 2744-2747.
Ward, I.M., et al., “Histone H2AX is Phosphorylated in an ATR-dependent Manner in Response to Replicational Stress”, J. Biol. Chem. (2001), 51, pp. 47759-47762.
International Search Report and Written Opinion dated Jul. 19, 2011 for Application No. PCT/US2011/036246.
International Search Report and Written Opinion dated Aug. 23, 2013 for Application No. PCT/US2013/035466.
International Search Report and Written Opinion dated Apr. 23, 2013 for Application No. PCT/US2012/058127.
International Search Report and Written Opinion dated Jan. 11, 2012 for Application No. PCT/US2011/036243.
International Search Report and Written Opinion dated Oct. 12, 2011 for Application No. PCT/US2011/036239.
International Search Report and Written Opinion dated Dec. 20, 2013 for Application No. PCT/US2013/063254.
International Search Report and Written Opinion dated Dec. 28, 2011 for Application No. PCT/US2011/036245.
International Search Report and Written Opinion dated Jan. 8, 2013 for Application No. PCT/US2012/058374.
International Search Report and Written Opinion dated Jun. 28, 2011 for Application No. PCT/US2011/036242.
International Search Report and Written Opinion dated Mar. 4, 2010 for Application No. PCT/US2009/068827.
International Search Report and Written Opinion dated Mar. 15, 2010 for Application No. PCT/US2009/063922.
International Search Report and Written Opinion dated Jan. 30, 2013 for Application No. PCT/US2012/058117.
International Search Report and Written Opinion dated Feb. 15, 2013 for Application No. PCT/US2012/064421.
International Search Report and Written Opinion dated Jun. 17, 2011 for Application No. PCT/US2011/036214.
International Search Report and Written Opinion dated Aug. 10, 2012 for Application No. PCT/US2012/032438.
International Search Report and Written Opinion dated Nov. 12, 2012 for Application No. PCT/US2012/058121.
International Search Report and Written Opinion dated Nov. 12, 2012 for Application No. PCT/US2012/058119.
International Search Report and Written Opinion dated Feb. 1, 2013 for Application No. PCT/US2012/064426.
International Search Report and Written Opinion dated Feb. 1, 2013 for Application No. PCT/US2012/064430.
International Search Report and Written Opinion dated Feb. 26, 2013 for Application No. PCT/US2012/064433.
International Search Report and Written Opinion dated Jan. 30, 2013 for Application No. PCT/US2012/064435.
International Search Report and Written Opinion dated Feb. 27, 2014 for Application No. PCT/US2013/064920.
Non-Final Office Action dated Aug. 8, 2013 in U.S. Appl. No. 13/631,732.
Non-Final Office Action dated Aug. 8, 2013 in U.S. Appl. No. 13/631,727.
Adamczyk et al., Synthesis of 3,7-dihydroimidazo[1,2a]pyrazine-3-ones and their chemiluminescent properties. Tetrahedron. 2003;59(41):8129-42.
Bracher et al., Total Synthesis of the Indolizidinium Alkaloid Ficuseptine. Eur J Org Chem. 2002:2288-91.
Buscemi et al., DNA damage-induced cell cycle regulation and function of novel Chk2 phosphoresidues. Mol Cell Biol. Nov. 2006;26(21):7832-45. Epub Aug. 28, 2006.
Caira, Crystalline Polymorphism of Organic Compounds. Topics in Current Chemistry. Design of Organic Solids. 1998;198:163-208.
Campone et al., Phase I and pharmacokinetic trial of AP5346, a DACH-platinum-polymer conjugate, administered weekly for three out of every 4 weeks to advanced solid tumor patients. Cancer Chemother Pharmacol. Sep. 2007;60(4):523-33. Epub Feb. 17, 2007.
Chen et al., Development of biomarker of ATR activity in surrogate human tissues. Newcastle University. Poster. Nov. 2012. 1 page.
Chen et al., Targeting the S and G2 checkpoint to treat cancer. Drug Discov Today. Mar. 2012;17(5-6):194-202. doi: 10.1016/j.drudis.2011.12.009. Epub Dec. 15, 2011.
Curtin, Inhibiting the DNA damage response as a therapeutic manoeuvre in cancer. Br J Pharmacol. Aug. 2013;169(8):1745-65. doi: 10.1111/bph.12244.
Darabantu et al., Synthesis of new polyaza heterocycles. Part 42: Diazines. Tetrahedron. 2005;61(11):2897-905.
De Wergifosse et al., Coelenterazine: a two-stage antioxidant in lipid micelles. Free Radical Biol Med. 2004;36(3):278-87.
Dias et al., Synthesis of 2,6-diphenylpyrazine derivatives and their DNA binding and cytotoxic properties. Eur J Med Chem. 2005;40:1206-13.
Erickson et al., Structure-guided expansion of kinase fragment libraries driven by support vector machine models. Biochim Biophys Acta. Mar. 2010;1804(3):642-52. doi: 10.1016/j.bbapap.2009.12.002. Epub Dec. 11, 2009.
Goto et al.,Squid bioluminescence I. Structure of watasenia oxyluciferin, a possible light-emitter in the bioluminescence of watasenia scintillans. Tetrahedron Lett. 1974;15(26):2321-4.
Hart et al., Renilla Reinformis Bioluminescence: Luciferase-Catalyzed Production of Nonradiating Excited States from Luciferin Analogues and Elucidation of the Excited State Species Involved in Energy Transfer to Renilla Green Fluorescent Protein. Biochemistry. 1979;18:2204-10.
Hilfiker et al., Relevance of Solid-state Properties for Pharmaceutical Products. Polymorphism: in the Pharmaceutical Industry. 2006;1-19.
Hirano et al., Bioluminescent properties of fluorinated semi-synthetic aequorins. Tetrahedron Lett. 1998;39(31):5541-4.
Jia et al., A Facile Preparation of 2,6-Diarylpyrazines. Heteroatom Chemistry. 1998;9(3):341-5.
Jones et al., A Suzuki Coupling Approach to Pyrazines Related to Coelenterazine. Synlett. 1996;(6):509-10.
Kao et al., Inhibition of λ-H2AX after ionizing radiation as a biological surrogate of impaired upstream DNA damage signaling and radiosensitivity. J Cancer Mol. 2010;5(2):49-54.
Lima et al., Bioisosterism: a useful strategy for molecular modification and drug design. Curr Med Chem. 2005;12(1):23-49.
Ling et al., Mechanism of Cell Cycle G2/M Arrest in Human Gastric Cancer BGC823 Cells Induced by Diallyl Disulfide. Chinese J Clin Oncol. Feb. 28, 2010;(3):121-5.
Liu et al., Chemical Biology Foundation. Science Press. Sep. 30, 2010;213-8.
Middleton et al., ATR as a Therapeutic Target. Cancer Drug Discovery and Development. 2013. Author's Proof. 20 pages.
Middleton et al., Chemosensitisation by, and Single Agent Activity of, ATR Inhibitor VE-821 in Human Breast Cancer Cells. Eur J Canc. Nov. 1, 2012;85-6.
Muslimovic et al., An optimized method for measurement of gamma-H2AX in blood mononuclear and cultured cells. Nat Protoc. 2008;3(7):1187-93. doi: 10.1038/nprot.2008.93.
Nowotnik et al., ProLindac (AP5346): a review of the development of an HPMA DACH platinum Polymer Therapeutic. Adv Drug Deliv Rev. Nov. 12, 2009;61(13):1214-9. doi: 10.1016/j.addr.2009.06.004. Epub Aug. 9, 2009. Review.
Patani et al., Bioisosterism: A Rational Approach in Drug Design. Chem Rev. Dec. 19, 1996;96(8):3147-76.
Prevo et al., The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther. Sep. 2012;13(11):1072-81. doi: 10.4161/cbt.21093. Epub Jul. 24, 2012.
Reaper, et al., Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nat Chem Biol. Apr. 13, 2011;7(7):428-30. doi: 10.1038/nchembio.573. Supplementary Information.
Redon et al., λ-H2AX as a biomarker of DNA damage induced by ionizing radiation in human peripheral blood lymphocytes and artificial skin. Adv Space Res. 2009;43(8):1171-8.
Registry (STN), 2004, RN 726138-31-4.
Richards et al., An Autoinhibitory Tyrosine Motif in the Cell-Cycle-Regulated Nek7 Kinase is Released through Binding of Nek9. Molec Cell. 2009;36:560-70.
Schultheiss et al., Facile Synthesis of Diarylpyrazines Using Suzuki Coupling of Dichloropyrazines with Aryl Boronic Acids. Heterocycles. 2003;60(8):1891-7.
Sevilla et al., Microwave-assisted synthesis of 1,3-dihydro-[1,2,5]thiadiazolo[3,4-b]pyrazine-2,2-dioxides. Tetrahedron Letters. 2006;47(48):8603-6.
Shimomura et al., Semi-synthetic aequorins with improved sensitivity to Ca2+ ions. Biochem J. Aug. 1, 1989;261(3):913-20.
Teranishi et al., Synthesis and Chemiluminescence of Coelenterazine (Oplophorus Luciferin) Analogues . Bulletin Chem Soc Japan. 1990;63(11):3132-40.
Tutin, CCLVII.-Syntheses in the epinephrine series. Part II. The formation and properties of some 2 : 5- and 2 : 6-substituted pyrazines and their conversion into amino-ketones and imino-diketones. J Chem Soc Trans. 1910;97:2495-524.
Vicent, Polymer Anticancer Ding Conjugates: Use as Single Agents and as Combination Therapy. 2007 AACR Annual Meeting. Apr. 14-18, 2007:56-62.
Wu et al., Chemi- and bioluminescence of coelenterazine analogues with a conjugated group at the C-8 position. Tetrahedron Lett. 2001;42(16):2997-3000.
U.S. Appl. No. 13/106,476, filed May 12, 2011, Charrier et al.
U.S. Appl. No. 14/816,432, filed Aug. 3, 2015, Pollard et al.
U.S. Appl. No. 14/678,489, filed Apr. 3, 2015, Charrier et al.
U.S. Appl. No. 14/633,394, filed Feb. 27, 2015, Falcon et al.
U.S. Appl. No. 13/106,184, filed May 12, 2011, Charrier et al.
U.S. Appl. No. 14/562,965, filed Dec. 8, 2014, Charrier et al.
U.S. Appl. No. 14/223,109, filed Mar. 24, 2014, Charrier et al.
U.S. Appl. No. 14/467,175, filed Aug. 25, 2014, Charrier et al.
PCT/US2011/036246, Jul. 19, 2011, International Search Report and Written Opinion.
PCT/US2012/035466, Aug. 23, 2013, International Search Report and Written Opinion.
PCT/US2012/058127, Apr. 23, 2013, International Search Report and Written Opinion.
PCT/US2011/036243, Jan. 11, 2012, International Search Report and Written Opinion.
PCT/US2011/036239, Oct. 12, 2012, International Search Report and Written Opinion.
PCT/US2013/063254, Dec. 20, 2013, International Search Report and Written Opinion.
PCT/US2011/036245, Dec. 28, 2011, International Search Report and Written Opinion.
PCT/US2012/058374, Jan. 8, 2013, International Search Report and Written Opinion.
PCT/US2011/036242, Jun. 28, 2011, International Search Report and Written Opinion.
PCT/US2009/068827, Mar. 4, 2010, International Search Report and Written Opinion.
PCT/US2009/063922, Mar. 15, 2010, International Search Report and Written Opinion.
PCT/US2012/058117, Jan. 30, 2013, International Search Report and Written Opinion.
PCT/US2012/064421, Feb. 15, 2013, International Search Report and Written Opinion.
PCT/US2011/036214, Jun. 17, 2011, International Search Report and Written Opinion.
PCT/US2012/032438, Aug. 10, 2012, International Search Report and Written Opinion.
PCT/US2012/058121, Nov. 12, 2012, International Search Report and Written Opinion.
PCT/US2012/058119, Nov. 12, 2012, International Search Report and Written Opinion.
PCT/US2012/064426, Feb. 1, 2013, International Search Report and Written Opinion.
PCT/US2012/064430, Feb. 1, 2013, International Search Report and Written Opinion.
PCT/US2012/064433, Feb. 26, 2013, International Search Report and Written Opinion.
PCT/US2012/064435, Jan. 30, 2013, International Search Report and Written Opinion.
PCT/US2013/064920, Feb. 27, 2014, International Search Report and Written Opinion.
The relevance of CN1551869 is understood from its English-language abstract.
The relevance of CN101001606 is understood from its English-language abstract.
The relevance of CN101479255 is understood from its English-language abstract.
The relevance of CN101537007 is understood from its English-language abstract.
The relevance of CN101671336 is understood from its English-language abstract.
The relevance of CN103373996 is understood from its English-language abstract.
The relevance of JP2002-072370 is understood from its English-language abstract.
The relevance of JP2002-072372 is understood from its English-language abstract.
The relevance of JP2002-518389 is understood from its English-language abstract.
The relevance of JP2003-074370 is understood from its English-language abstract.
The relevance of JP2003-516974 is understood from its English-language abstract.
The relevance of JP2005-511531 is understood from its English-language abstract.
The relevance of JP2005-530760 is understood from its English-language abstract.
The relevance of JP2006-156445 is understood from its English-language abstract.
The relevance of JP2006-516124 is understood from its English-language abstract.
The relevance of JP2006-519232 is understood from its English-language abstract.
The relevance of JP2006-519833 is understood from its English-language abstract.
The relevance of JP2006-520794 is understood from its English-language abstract.
The relevance of JP2006-521357 is understood from its English-language abstract.
The relevance of JP2007-524682 is understood from its English-language abstract.
The relevance of JP2008-510790 is understood from its English-language abstract.
The relevance of JP2008-510792 is understood from its English-language abstract.
The relevance of JP2008-517945 is understood from its English-language abstract.
The relevance of JP2008-525453 is understood from its English-language abstract.
The relevance of JP2008-543754 is understood from its English-language abstract.
The relevance of JP2009-027904 is understood from its English-language abstract.
The relevance of JP2009-503103 is understood from its English-language abstract.
The relevance of JP2009-530233 is understood from its English-language abstract.
The relevance of JP2009-532356 is understood from its English-language abstract.
The relevance of JP2009-533327 is understood from its English-language abstract.
The relevance of JP2009-541247 is understood from its English-language abstract.
The relevance of JP2009-541268 is understood from its English-language abstract.
The relevance of JP2010-077286 is understood from its English-language abstract.
The relevance of JP2010-180180 is understood from its English-language abstract.
The relevance of JP2010-506934 is understood from its English-language abstract.
The relevance of JP2010-509356 is understood from its English-language abstract.
The relevance of JP2010-513433 is understood from its English-language abstract.
The relevance of JP2011-042639 is understood from its English-language abstract.
The relevance of JP2011-500778 is understood from its English-language abstract.
The relevance of JP2012-508260 is understood from its English-language abstract.
The relevance of JP2012-513398 is understood from its English-language abstract.
The relevance of JP2012-533248 is understood from its English-language abstract.
The relevance of JP2013-501720 is understood from its English-language abstract.
The relevance of JP2013-505900 is understood from its English-language abstract.
The relevance of JP2013-517264 is understood from its English-language abstract.
The relevance of JP2013-525476 is understood from its English-language abstract.
The relevance of JP2014-165380 is understood from its English-language abstract.
The relevance of JP2014-510072 is understood from its English-language abstract.
The relevance of JP2014-518545 is understood from its English-language abstract.
The relevance of JP62-270623 is understood from its English-language abstract.
The relevance of JP63-208520 is understood from its English-language abstract.
The relevance of NZ593316 is understood from its English-language abstract.
The relevance of NZ593969 is understood from its English-language abstract.
The relevance of WO2004/103991 is understood from its English-language abstract.
The relevance of WO2008/156174 is understood from its English-language abstract.
The relevance of WO2011/035855 is understood from its English-language abstract.
The relevance of “Liu et al.” is understood by the chemical structures.
The relevance of “Ling et al.” is understood from its English-language abstract.
Katritzky, A.R., et al., “Efficient synthesis of 3,5-functionalized isoxazoles and isoxazolines via 1,3-dipolar cycloaddition reactions of 1-propargyl- and 1-allylbenzotriazoles”, J. Heterocyclic Chem., 37(6), (2000), pp. 1505-1510.
Kumpaty, H.J., et al., “Synthesis of N-Methyl Secondary Amines”, Synth. Commun., 33(8), (2003), pp. 1411-1416.
March, J., March's Advanced Organic Chemistry, 2007, John Wiley and Sons, Chapter 16.
Wuts, P.G.M., Greene's Protective Groups in Organic Synthesis, 4th Edition, 2006, John Wiley and Sons, Chapter 4.
Wuts, P.G.M., Greene's Protective Groups in Organic Synthesis, 4th Edition, 2006, John Wiley and Sons, Chapter 7.
Saito, R., et al., “Synthesis and in vitro evaluation of botryllazine B analogues as a new class of inhibitor against human aldose reductase”, Tetrahedron, 65 (2009), pp. 3019-3026.
Non-Final Office Action dated Sep. 28, 2013 in U.S. Appl. No. 13/631,727.
Non-Final Office Action dated Sep. 28, 2013 in U.S. Appl. No. 13/631,732.
Bastin et al., Salt Selection and Optimisation Procedures for Pharmaceutical New Chemical Entities. Organic Process Research and Development. American Chemical Society. 2000;4(5):427-35.
Biss et al., Selective tumor killing based on specific DNA-damage response deficiencies. Cancer Biology & Therapy. Mar. 2012; 239-46.
Kumar et al., Salt selection in drug development. Pharmaceutical Technology. 2008;32(3):128-46.
Peasland et al., Identification and evaluation of a potent novel ATR inhibitor, NU6027, in breast and ovarian cancer cell lines. British Journal of Cancer. Jul. 2011; 105(3):372-81.
Serajuddin, Salt formation to improve drug solubility. Advanced Drug Delivery Reviews. 2007; 59(7):603-16.
Brittain, editor. Polymorphism in pharmaceutical solids. CRC Press; 2009, Chapters 7 (p. 233-281) and 12 (p. 436-480).
Hancock et al., Characteristics and significance of the amorphous state in pharmaceutical systems. J Pharm Sci. Jan. 1997;86(1):1-12.
Pollard et al. Defining optimal dose schedules for ATR inhibitors in combination with DNA damaging drugs: Informing clinical studies of VX-970, the first-in-class ATR inhibitor. Proceedings: AACR Annual Meeting. Apr. 16-20, 2016.
Morgan et al., 2010, “Mechanism of radiosensitization by the Chk1/2 inhibitor AZD7762 involves abrogation of the G2 checkpoint and inhibition of homologous recombinational DNA repair.” Cancer Res. 70(12):4972-81.
No Author Listed, “Brief course of molecular pharmacology,” P.V. Sergeev, ed., 1975, p. 10.
Cholodov et al., “Clinical pharmacokinetics.” Medicine, 1985, pp. 83-98, 134-138, 160, 378-380.
Yokoi et al., “Hypoxia Increases Resistance of Human Pancreatic Cancer Cells to Apoptosis Induced by Gemcitabine,” Clinical Cancer Research, 2004, vol. 10, pp. 2299-2306.
Shibamoto et al., “Radiosensitivity of Human Pancreatic Cancer Cells In Vitro and In Vivo, and the Effect of a New Hypoxic Cell Sensitizer, Doranidazole,” Radiother Oncol, 2000, vol. 56, No. 2, pp. 265-270, Abstract.
U.S. Appl. No. 15/967,110 of Charrier et al., filed Apr. 30, 2018.
U.S. Appl. No. 15/608,630 of Charrier et al., filed May 30, 2017.
U.S. Appl. No. 15/849,241 of Charrier et al., filed Dec. 20, 2017.
U.S. Appl. No. 15/633,477 of Ahmad et al., filed Jun. 26, 2017.
U.S. Appl. No. 15/763,366 of Pollard et al., filed Mar. 26, 2018.
U.S. Appl. No. 15/693,521 of Falcon et al., filed Sep. 1, 2017.
Related Publications (1)
Number Date Country
20140356456 A1 Dec 2014 US
Provisional Applications (1)
Number Date Country
61542084 Sep 2011 US
Continuations (1)
Number Date Country
Parent 13633114 Oct 2012 US
Child 14193845 US