Method for determining responsiveness to chk1 inhibitors

A technology of CHK1 and inhibitors, applied in the field of determining the response to CHK1 inhibitors, can solve the problems of limited practicality, strong side effects of normal tissues, tumor cell resistance, etc.

Inactive Publication Date: 2008-02-06
ASTRAZENECA AB
View PDF12 Cites 1 Cited by
  • Summary
  • Abstract
  • Description
  • Claims
  • Application Information

AI Technical Summary

Problems solved by technology

[0002] Chemotherapy and radiation exposure are currently the main options for cancer treatment, but the utility of both approaches is severely limited by strong side effects on normal tissues and the frequent development of tumor cell resistance

Method used

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
View more

Image

Smart Image Click on the blue labels to locate them in the text.
Viewing Examples
Smart Image
  • Method for determining responsiveness to chk1 inhibitors
  • Method for determining responsiveness to chk1 inhibitors
  • Method for determining responsiveness to chk1 inhibitors

Examples

Experimental program
Comparison scheme
Effect test

Embodiment 1

[0059] Example 1: Demonstration of CHK1 activation following DNA damage in tumor cell line S and xenograft samples

[0060] A. In vitro studies:

[0061] In short, the 3×10 6 SW620 and / or HT29 cells were seeded into 10 cm tissue culture dishes and allowed to attach for 24 hours at 37°C. Cells were then treated with titrations of DNA damaging reagents (SN-38 (CQ International, China), doxorubicin (Sigma-Aldrich, MO) or gemcitabine (CQ International, China)). Then, after 8 hours of incubation with DNA damaging reagents, cells were harvested in Onyx lysis buffer (20 mM Tris, 137 mM NaCl, 1 mM EGTA, 1% Triton-X, 10% glycerol, 1.5 mM MgCl2, 1:100 dilution of CalbiochemPhophatase Inhibitor Cocktail Set II, 10 mM β-glycerol phosphate, 1 mM DTT, 7 μg / ml PMSF, 20 KIU / ml aprotinin, 1 mM Pefabloc, 0.1 mg / ml leupeptin) . Protein lysates were then generated, protein concentration was determined, an equal amount of protein was loaded, and phospho-CHK rabbit monoclonal antibody (Ser 345)...

Embodiment 2

[0065] Example 2: Demonstration of increased phospho-CHK following combination treatment of gemcitabine and CHK1 inhibitors in tumor cell lines, xenograft samples and substitute tissues

[0066] A. In Vitro Studies

[0067] In short, the 3×10 6 SW620 cells were seeded in 10 cm tissue culture dishes and allowed to attach for 24 hours at 37°C. Cells were then treated for 8 hours with 100 nM gemcitabine or 30 nM, 100 nM or 500 nM CHK inhibitor, or 100 nM gemcitabine and 30 nM, 100 nM or 500 nM in combination. A panel of cells was then harvested for Western blot analysis. The medium containing the compound is removed from the remaining cells and fresh tissue culture medium is added back. Cells were then harvested 22 hours later and protein lysates were generated as described above and analyzed by Western blot analysis using a phospho-CHK antibody. Western blot analysis indicated an increase in phospho-CHK following gemcitabine treatment alone, and a dose-dependent increase in ...

Embodiment 3

[0073]Similar combination studies have been done with irinotecan and Chk inhibitors. Data again indicated increased phospho-CHK staining compared to either reagent alone (data not shown). Example 3: Demonstration of Reduction of DNA Damage Nuclear Foci Following Inhibition of CHK1 Kinase

[0074] A. In vitro studies:

[0075] HeLa cells were seeded on coverslips and allowed to attach for 24 h at 37 °C. Then with 100nM Adriamycin TM The cells were treated with 500 nM CHK inhibitor for 5 hours. Cells were then treated as described in Example 1. The primary antibody used for the study was rabbit polyclonal anti-53BP1 (Novus, CO) diluted 1:120 and the secondary antibody was Alexa Fluor 488 anti-rabbit IgG (Molecular Probes, OR) diluted 1:300. Nuclear foci were quantified using Metaphorph analysis by measuring the fluorescence intensity per unit number of nuclei (determined by Hoescht staining). Figure 5 shows that when CHK inhibitors and doxorubicin hydrochloride (Adriamycin...

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

PUM

No PUM Login to view more

Abstract

The invention is directed at a method for predicting a patient's responsiveness to a CHK1 inhibitor. The method includes providing a sample from a test patient who is being treated with a DNA damaging agent; and determining for the presence of an activated CHK1 signal transduction pathway molecule, wherein the presence of an activated CHK1 signal transduction pathway molecule is indicative that a CHK1 inhibitor should be administered to the patient.

Description

field of invention [0001] The present invention relates to the use of a biomarker as an indicator of the activity of a therapeutic agent in a subject cancer patient. The biomarkers of the invention can be used, for example, to determine when to administer a therapeutic agent to a patient or to select a therapeutically effective dose of a therapeutic agent for a patient. Background of the invention [0002] Chemotherapy and radiation exposure are currently the main options for cancer treatment, but the utility of both approaches is severely limited by strong side effects on normal tissues and the frequent development of tumor cell resistance. It is thus highly desirable to improve the efficacy of these treatments in a manner that does not increase the toxicity associated with them. One way to achieve this is through the use of specific synergists. Typically, these agents are designed to be used in combination with existing or new agents. [0003] A single cell replicates b...

Claims

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

Application Information

Patent Timeline
no application Login to view more
Patent Type & Authority Applications(China)
IPC IPC(8): C12Q1/48G01N33/68G01N33/574
CPCG01N33/57496G01N33/6872C12Q1/485A61P29/00A61P35/00A61P35/02C12Q1/00G01N33/53G01N33/68G01N33/574
Inventor S·扎布卢多夫L·伯格朗
Owner ASTRAZENECA AB
Who we serve
  • R&D Engineer
  • R&D Manager
  • IP Professional
Why Eureka
  • Industry Leading Data Capabilities
  • Powerful AI technology
  • Patent DNA Extraction
Social media
Try Eureka
PatSnap group products