[go: up one dir, main page]

WO2024158242A1 - Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient - Google Patents

Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient Download PDF

Info

Publication number
WO2024158242A1
WO2024158242A1 PCT/KR2024/001239 KR2024001239W WO2024158242A1 WO 2024158242 A1 WO2024158242 A1 WO 2024158242A1 KR 2024001239 W KR2024001239 W KR 2024001239W WO 2024158242 A1 WO2024158242 A1 WO 2024158242A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
kras
group
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/KR2024/001239
Other languages
French (fr)
Korean (ko)
Inventor
이대희
이지윤
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nbios Inc
Industry Academy Cooperation Foundation of Gangneung Wonju National University
Original Assignee
Nbios Inc
Industry Academy Cooperation Foundation of Gangneung Wonju National University
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 Nbios Inc, Industry Academy Cooperation Foundation of Gangneung Wonju National University filed Critical Nbios Inc
Priority claimed from KR1020240011679A external-priority patent/KR20240117500A/en
Publication of WO2024158242A1 publication Critical patent/WO2024158242A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4

Definitions

  • the present invention relates to a compound for inhibiting KRAS G12D mutation and a composition for preventing or treating cancer diseases containing the same as an active ingredient. More specifically, it relates to a compound capable of inhibiting cell proliferation by targeting the KRAS G12D mutation and a composition containing the same as an active ingredient. It relates to a composition for preventing or treating cancer disease, comprising:
  • Cancer is a disease caused by immature, overproliferating cells that contain genetic mutations. According to Statistics Korea, the three leading causes of death in 2021 are cancer, heart disease, and pneumonia, with cancer accounting for 26% of all deaths. This value is increasing by 0.6% every year, leading to an increase in cancer-related mortality (Statistics Korea, 2021). The cancer incidence rate was 214.2 per 100,000 population in 1999 and 445.3 in 2012. The risk of developing cancer is 37.3% for people who survive to the average life expectancy (age 81), 37.5% for men (age 77), and 34.9% for women (age 84).
  • Cancer refers to a phenomenon in which abnormalities occur in the DNA of the cell proliferation control system. This DNA synthesizes cancer-related proteins due to genetic mutations and abnormal active genetic information in the DNA, so it is called a cancer gene (oncogene). Under normal conditions, oncogenes do not induce cancer development but rather regulate proliferation; These genes cause cancer when activated by genetic mutation or abnormality. Mutations in cancer genes can cause cells to lose their ability to inhibit cell growth. Uncontrolled cell proliferation causes cancer. These cells can be moved to other parts of the body through processes such as angiogenesis and metastasis.
  • RAS mutations occur at a high rate in colon cancer, lung cancer, and pancreatic cancer.
  • RAS is a family of small GTPase proteins that play important roles in cell growth, differentiation, proliferation, and survival.
  • HRAS high-density polystyrene
  • KRAS KRAS
  • NRAS NRAS
  • the RAS gene is the most commonly found oncogene in human cancer.
  • KRAS is known to have the highest mutation rate compared to HRAS and NRAS in various cancer types.
  • KRAS is also known to exhibit a unique enzymatic activity that binds GTP in its active state and converts it to GDP, thereby inactivating KRAS. The rate of turnover is generally slow but can be dramatically increased by auxiliary GTPase activating proteins (GAPs).
  • GAPs auxiliary GTPase activating proteins
  • KRAS releases GDP through a guanine nucleotide exchange factor (GEF), like SOS.
  • GTP binding of KRAS causes several residues in the switch I region (residues 30-40) and switch II region (residues 60-70) to bind to the RAS effector protein. These switches can be regulated by GAP and GEF.
  • GAP guanine nucleotide exchange factor
  • KRAS is mutated, GAP binding is disrupted and remains in the GTP state, resulting in an abnormal signaling pathway that helps tumor formation and maintenance.
  • KRAS mutations have been reported at glycine 12 (G12), glycine 13 (G13), or glutamine 61.
  • G12 mutations account for approximately 80% of KRAS mutations, including G12D (41%), G12C (28%), and G12C (14%).
  • a high proportion of lung (32%), pancreatic (86%), and colorectal (41%) cancers contain mutations in codon 12 of KRAS.
  • glycine is mutated to cysteine.
  • glycine is mutated to D-aspartic acid.
  • Targeted therapy using epidermal growth factor receptor (EGFR) in patients with KRAS mutations is known to have lower treatment efficacy and lower survival rates than patients without these mutations.
  • EGFR epidermal growth factor receptor
  • the EGFR family of receptor tyrosine kinases activates RAS by promoting GDP-GTP exchange. Inhibiting EGFR prevents RAS activation, while inhibiting SOS or SHP2 reduces GDP-GTP exchange rate and thus reduces GTP binding to RAS. Because EGFR is upstream of RAS, inactivation of this receptor tyrosine kinase can inhibit RAS activity.
  • Monoclonal antibodies against EGFR such as cetuximab or panitumumab, can increase overall survival and progression-free survival in patients with wild-type KRAS metastatic colorectal cancer. However, this antibody is not effective against KRAS mutant tumors, especially those with codon 12 or 13 mutations.
  • EGFR tyrosine kinase inhibitors such as erlotinib and gefitinib, are approved for the treatment of EGFR-mutant non-small cell lung cancer, but have no single effect in KRAS-mutant non-small cell lung cancer. .
  • Wild-type KRAS colorectal cancer responds to cetuximab treatment.
  • metastatic colorectal cancer acquires KRAS mutations that make the cancer resistant to anti-EGFR therapy. Therefore, an inhibitor that directly targets KRAS mutations is needed rather than an anticancer drug that targets EGFR.
  • inhibitors of mutant KRAS G12C in lung cancer are currently being developed and evaluated, inhibitors of KRAS G12D mutations in colon and pancreatic cancer have not been extensively evaluated.
  • Targeted therapy using epidermal growth factor receptor (EGFR) in cancer patients with KRAS G12D mutation of the present invention selectively proliferates KRAS G12D mutant cells to solve the problem of lower treatment efficacy compared to patients without this mutation.
  • the aim is to provide a pharmaceutical composition for preventing or treating cancer that contains an inhibitory compound and the same as an active ingredient and can be used for targeted treatment against KRAS G12D mutant cells.
  • a compound for inhibiting proliferation of KRAS G12D mutant cells represented by the following formula (1) is provided.
  • n is the number of repeat units, which is an integer from 1 to 10,
  • n is the number of repeat units that is an integer from 1 to 10,
  • X 1 is an oxygen atom or a sulfur atom
  • R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,
  • R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,
  • R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
  • n is the number of repeat units that is an integer from 1 to 4,
  • n is the number of repeating units that is an integer from 1 to 4,
  • X 1 is an oxygen atom
  • R 1 is a 1C to 10C alkyl group
  • R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group
  • R 5 is ego
  • X 2 is an oxygen atom or a sulfur atom
  • R 6 may be a hydrogen atom or a 1C to 10C alkyl group.
  • the compound represented by Formula 1 may be a compound represented by Formula 2 below.
  • a pharmaceutical composition for preventing or treating cancer disease comprising a compound represented by the following formula (1) or a salt thereof as an active ingredient is provided.
  • n is the number of repeat units, which is an integer from 1 to 10,
  • n is the number of repeat units that is an integer from 1 to 10,
  • X 1 is an oxygen atom or a sulfur atom
  • R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,
  • R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,
  • R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
  • n is the number of repeat units that is an integer from 1 to 4,
  • n is the number of repeating units that is an integer from 1 to 4,
  • X 1 is an oxygen atom
  • R 1 is a 1C to 10C alkyl group
  • R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group
  • R 5 is ego
  • X 2 is an oxygen atom or a sulfur atom
  • R 6 may be a hydrogen atom or a 1C to 10C alkyl group.
  • the compound represented by Formula 1 may be a compound represented by Formula 2 below.
  • the pharmaceutical composition for preventing or treating cancer disease may be used to inhibit proliferation of KRAS G12D mutant cells.
  • the KRAS G12D mutant cells may be one or more types selected from LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D, and LS174T KRAS G12D.
  • composition for preventing or treating cancer disease may be for targeted treatment against the KRAS G12D mutant cells.
  • composition for preventing or treating cancer disease may be for inhibiting the MAPK signaling pathway.
  • Inhibition of the MAPK signaling pathway can be performed by inhibiting C-RAF phosphorylation in the RAS-RAF-MEK-ERK pathway.
  • composition for preventing or treating cancer disease may be for inhibiting the PI3K signaling pathway.
  • the inhibition of the PI3K signaling pathway can be performed by inhibiting AKT phosphorylation in the PI3K-AKT-mTOR pathway.
  • the cancer disease may be any one selected from colon cancer, colon cancer, rectal cancer, lung cancer, melanoma, thyroid cancer, uterine cancer, ovarian cancer, cervix, pancreatic cancer, stomach cancer, and liver cancer.
  • the compound of the present invention selectively inhibits cell proliferation in KRAS G12D mutant cells, it can solve the problem of low efficacy of conventional targeted therapy using epidermal growth factor receptor (EGFR), and the compound of the present invention and its salt are effective.
  • EGFR epidermal growth factor receptor
  • the pharmaceutical composition containing the composition as an ingredient can be used for targeted treatment against KRAS G12D mutant cells.
  • Figure 1 shows the cell growth measurement results of compounds Q1a to Q11a of Experimental Example 1 on wild-type cells of LIM1215 and KRAS mutant cells.
  • Figure 2 shows the molecular structure and three-dimensional model structure of the Q2a compound selected in Experimental Example 1.
  • FIG. 3 shows the results of protein thermal transfer (PTS) analysis to confirm whether the Q2a compound of Experimental Example 1 bound to KRAS.
  • Figure 4 shows the results of confirming whether compound Q2a has specific activity only in the KRAS G12D mutation in Experimental Example 1.
  • Figure 5 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of colon cancer cell line LIM1215 according to Experimental Example 2 when treated with Q2a compound.
  • Figure 6 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of colon cancer cell line SW48 according to Experimental Example 2 when treated with Q2a compound.
  • Figure 7 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of lung cancer cell line NCI-H838 according to Experimental Example 2 when treated with Q2a compound.
  • Figure 8 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of lung cancer cell line NCI-H1975 according to Experimental Example 2 when treated with Q2a compound.
  • FIG 9 shows the results of colony formation analysis for wild type (WT) LIM1215 cells and KRAS G12D mutant cells in Experimental Example 2.
  • FIG 10 shows the results of colony formation analysis for wild type (WT) SW48 cells and KRAS G12D mutant cells in Experimental Example 2.
  • FIG 11 shows the results of colony formation analysis for wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 2.
  • Figure 12 shows the results of colony formation analysis for wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 2.
  • Figure 13 shows the results of colony formation analysis of LS174T KRAS G12D mutant cells in Experimental Example 2.
  • Figure 14 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of LIM1215 cells in Experimental Example 3.
  • Figure 15 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of SW48 cells in Experimental Example 3.
  • Figure 16 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 3.
  • Figure 17 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 3.
  • Figure 18 shows the results of Western blotting of wild type (WT) LIM1215 cells and KRAS G12D mutant cells in Experimental Example 4 when treated with Q2a for 48.72 hours.
  • Figure 19 shows the results of Western blotting of wild type (WT) SW48 cells and KRAS G12D mutant cells in Experimental Example 4 when treated with Q2a for 48.72 hours.
  • Figure 20 shows the Western blot results of wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 4 when treated with Q2a for 48 and 72 hours.
  • Figure 21 shows the results of Western blotting of wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 4 when treated with Q2a for 48.72 hours.
  • Figure 22 shows the results of Western blotting of LIM1215 and SW48 cells in Experimental Example 4 when wild type (WT) and KRAS G12D mutant cells were treated with Q2a for 24 hours.
  • Figure 23 shows the results of Western blotting of wild type (WT) and KRAS G12D mutant cells of NCI-H1975 and NCI-H838 cells in Experimental Example 4 when treated with Q2a for 24 hours.
  • Figure 24 is a heatmap according to microarray analysis in Experimental Example 5.
  • Figure 25 shows the results of Western blot analysis in Experimental Example 5.
  • Figure 26 shows the change in tumor size over time according to Q2a administration in mice administered LIM1215 wild-type cells and KRAS G12D mutant cells in Experimental Example 5.
  • Figure 27 is a graph of mouse body weight change measured while administering compound Q2a of Experimental Example 5.
  • Figure 28 is a photo comparing the size of the tumor after sacrificing the mouse and removing the tumor 21 days after the treatment in Experimental Example 5.
  • Figure 29 shows the results of measuring the weight of the tumor before and after treatment according to Experimental Example 5.
  • Figure 30 shows the mechanism of cancer cell inhibition according to Q2a administration in the KRAS pathway.
  • substituted means that at least one hydrogen atom is deuterium, C1 to C30 alkyl group, C3 to C30 cycloalkyl group, C2 to C30 heterocycloalkyl group, C1 to C30 halogenated alkyl group, C6 to C30 aryl group, C1 to C30 heteroaryl group, C1 to C30 alkoxy group, C3 to C30 cycloalkoxy group, C1 to C30 heterocycloalkoxy group, C2 to C30 alkenyl group, C2 to C30 alkynyl group, C6 to C30 aryloxy group, C1 to C30 heteroaryloxy group, silyl oxide Group (-OSiH 3 ), -OSiR 1 H 2 (R 1 is a C1 to C30 alkyl group or C6 to C30 aryl group), -OSiR 1 R 2 H (R 1 and R 2 are each independently a C1 to C30 alkyl group or C6 to C30 halogenated alky
  • two adjacent substituents among the above substituents may be fused to form a saturated or unsaturated ring.
  • the carbon number range of the alkyl group or aryl group in the “substituted or unsubstituted C1 to C30 alkyl group” or “substituted or unsubstituted C6 to C30 aryl group” does not take into account the portion on which the substituent is substituted and is not substituted. It refers to the total number of carbon atoms constituting the alkyl portion or aryl portion when viewed as being formed. For example, a phenyl group substituted with a butyl group at the para position corresponds to an aryl group with 6 carbon atoms substituted with a butyl group with 4 carbon atoms.
  • hetero means that one functional group contains 1 to 4 hetero atoms selected from the group consisting of N, O, S, and P, and the remainder is carbon.
  • hydrogen means single hydrogen, double hydrogen, or tritium hydrogen, unless otherwise defined.
  • alkyl group means an aliphatic hydrocarbon group, unless otherwise defined.
  • the alkyl group may be a “saturated alkyl group” that does not contain any double or triple bonds.
  • the alkyl group may be an “unsaturated alkyl group” containing at least one double or triple bond.
  • Alkyl groups whether saturated or unsaturated, may be branched, straight-chain, or cyclic.
  • the alkyl group may be a C1 to C30 alkyl group. More specifically, it may be a C1 to C20 alkyl group, a C1 to C10 alkyl group, or a C1 to C4 alkyl group.
  • C1 to C4 alkyl groups have 1 to 4 carbon atoms in the alkyl chain, i.e., the alkyl chain is methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and t-butyl.
  • the alkyl chain is methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and t-butyl.
  • the alkyl group includes methyl group, ethyl group, propyl group, isopropyl group, butyl group, isobutyl group, t-butyl group, pentyl group, hexyl group, ethenyl group, propenyl group, butenyl group, cyclopropyl group, and cyclopropyl group. It means butyl group, cyclopentyl group, cyclohexyl group, etc.
  • Cycloalkyl group includes monocyclic or fused-ring polycyclic (i.e., rings splitting adjacent pairs of carbon atoms) functional groups.
  • Heterocycloalkyl group means that the cycloalkyl group contains 1 to 4 heteroatoms selected from the group consisting of N, O, S, and P, and the remainder is carbon.
  • the heterocycloalkyl group is a fused ring, at least one ring of the fused ring may include 1 to 4 heteroatoms.
  • Aryl groups include monocyclic or fused ring polycyclic (i.e., rings splitting adjacent pairs of carbon atoms) functional groups.
  • Heteroaryl group means that the aryl group contains 1 to 4 heteroatoms selected from the group consisting of N, O, S, and P, and the remainder is carbon.
  • the heteroaryl group is a fused ring, at least one ring of the fused ring may include 1 to 4 heteroatoms.
  • the number of ring atoms is the sum of the number of carbon atoms and the number of non-carbon atoms.
  • the present invention provides a compound for inhibiting proliferation of KRAS G12D mutant cells represented by the following formula (1).
  • n is the number of repeating units, which is an integer from 1 to 10,
  • n is the number of repeat units that is an integer from 1 to 10,
  • X 1 is an oxygen atom or a sulfur atom
  • R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,
  • R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,
  • R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
  • n is the number of repeat units that is an integer from 1 to 4,
  • n is the number of repeating units that is an integer from 1 to 4,
  • X 1 is an oxygen atom
  • R 1 is a 1C to 10C alkyl group
  • R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group
  • R 5 is ego
  • X 2 is an oxygen atom or a sulfur atom
  • R 6 may be a hydrogen atom or a 1C to 10C alkyl group.
  • the compound represented by Formula 1 may be a compound represented by Formula 2 below.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer disease, comprising a compound represented by the following formula (1) or a salt thereof as an active ingredient.
  • n is the number of repeating units, which is an integer from 1 to 10,
  • n is the number of repeat units that is an integer from 1 to 10,
  • X 1 is an oxygen atom or a sulfur atom
  • R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,
  • R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,
  • R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
  • n is the number of repeat units that is an integer from 1 to 4,
  • n is the number of repeating units that is an integer from 1 to 4,
  • X 1 is an oxygen atom
  • R 1 is a 1C to 10C alkyl group
  • R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group
  • R 5 is ego
  • X 2 is an oxygen atom or a sulfur atom
  • R 6 may be a hydrogen atom or a 1C to 10C alkyl group.
  • the compound represented by Formula 1 may be a compound represented by Formula 2 below.
  • Another example provides a method for preventing and/or treating cancer disease, comprising administering a pharmaceutically effective amount of the compound represented by Formula 1 or a salt thereof to a subject in need thereof.
  • the method may further include the step of identifying a subject in need of prevention and/or treatment of a cancer disease.
  • the terms “subject,” “patient,” “individual,” and “host” and their variants are interchangeable and refer to any mammalian subject to which a compound or salt or composition thereof described herein is administered.
  • Non-limiting examples include humans, livestock (e.g. dogs, cats, etc.), farm animals (e.g. cattle, sheep, pigs, horses, etc.), and laboratory animals (e.g. monkeys, rats, etc.) in need of diagnosis, treatment or treatment. , mice, rabbits, guinea pigs, etc.), especially humans.
  • the methods described herein are applicable to both human prophylactic or therapeutic and veterinary applications.
  • the phrase “subject in need” includes subjects such as mammalian subjects who would benefit from administration of the compositions described herein.
  • Another example provides the use of the compound represented by Formula 1 or a salt thereof for the treatment and/or prevention of cancer diseases.
  • the pharmaceutical composition for preventing or treating cancer disease may be used to inhibit proliferation of KRAS G12D mutant cells.
  • the KRAS G12D mutant cells may be one or more types selected from LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D, and LS174T KRAS G12D.
  • LIM1215 KRAS G12D, SW48 KRAS G12D, and LS174T KRAS G12D are colon cancer cell lines
  • NCI-H1975 KRAS G12D and NCI-H838 KRAS G12D are lung cancer cell lines.
  • composition for preventing or treating cancer disease can be used for targeted treatment against the KRAS G12D mutant cells. This is because, although it has little cytostatic effect on KRAS wild-type cells, it can selectively exhibit anti-proliferation effect on KRAS G12D mutant cells.
  • composition for preventing or treating cancer diseases can be used to inhibit the MAPK signaling pathway.
  • Inhibition of the MAPK signaling pathway can be performed by inhibiting C-RAF phosphorylation in the RAS-RAF-MEK-ERK pathway.
  • composition for preventing or treating cancer diseases can be used to inhibit the PI3K signaling pathway.
  • the inhibition of the PI3K signaling pathway can be performed by inhibiting AKT phosphorylation in the PI3K-AKT-mTOR pathway.
  • the cancer disease may be any one selected from colon cancer, colon cancer, rectal cancer, lung cancer, melanoma, thyroid cancer, uterine cancer, ovarian cancer, cervix, pancreas cancer, stomach cancer, and liver cancer, but the scope of the present invention is not limited thereto and KRAS Any carcinoma that can cause a G12D mutation is possible.
  • the term ‘including as an active ingredient’ means containing a sufficient amount to achieve the efficacy or activity of the compound represented by Formula 1 or a salt thereof.
  • the compound represented by Formula 1 or its salt in the composition of the present invention is, for example, 0.001 mg/kg or more, preferably 0.1 mg/kg or more, more preferably 10 mg/kg. It may contain more than kg, more preferably more than 100 mg/kg, even more preferably more than 250 mg/kg, and most preferably more than 1 g/kg.
  • the quantitative lower limit and/or upper limit of the compound represented by Formula 1 or its salt can be selected within an appropriate range by a person skilled in the art.
  • salt refers to a compound that does not cause significant irritation to the organism to which the compound is administered and does not impair the biological activity and physical properties of the compound. It means dosage form.
  • the pharmaceutical salts include the compounds of the present invention, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid, sulfonic acids such as methanesulfonic acid, ethanesulfonic acid, and p-toluenesulfonic acid, tartaric acid, formic acid, citric acid, acetic acid, and trichloroacid.
  • organic carboxylic acids such as loacetic acid, trifluoroacetic acid, capric acid, isobutanoic acid, malonic acid, succinic acid, phthalic acid, gluconic acid, benzoic acid, lactic acid, fumaric acid, maleic acid, salicylic acid, etc.
  • the compound of the present invention can be reacted with a base to produce salts such as alkali metal salts such as ammonium salts, sodium or potassium salts, alkaline earth metal salts such as calcium or magnesium salts, dicyclohexylamine, and N-methyl-D-glue. It may be obtained by forming salts of organic bases such as carmine, tris(hydroxymethyl) methylamine, and amino acid salts such as arginine and lysine, but is not limited thereto.
  • the pharmaceutical composition of the present invention can be prepared using pharmaceutically suitable and physiologically acceptable auxiliaries in addition to the active ingredients, and the auxiliaries include excipients, disintegrants, sweeteners, binders, coating agents, swelling agents, lubricants, and lubricants. Agents or flavoring agents can be used.
  • the pharmaceutical composition may be preferably formulated as a pharmaceutical composition containing one or more pharmaceutically acceptable carriers in addition to the active ingredients described above.
  • the pharmaceutical composition may be in the form of granules, powders, tablets, coated tablets, capsules, suppositories, solutions, syrups, juices, suspensions, emulsions, drops, or injectable solutions.
  • the active ingredient may be combined with an oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, etc.
  • suitable binders, lubricants, disintegrants and coloring agents may also be included in the mixture.
  • Suitable binders include, but are not limited to, starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tracacance or sodium oleate, sodium stearate, magnesium stearate, sodium Includes benzoate, sodium acetate, sodium chloride, etc.
  • Disintegrants include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, etc.
  • Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and these.
  • saline solution sterile water
  • Ringer's solution buffered saline solution
  • albumin injection solution dextrose solution
  • maltodextrin solution glycerol
  • glycerol glycerol
  • ethanol ethanol
  • diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, or tablets.
  • the pharmaceutical composition of the present invention can be administered orally or parenterally, and in the case of parenteral administration, it can be administered by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, transdermal administration, etc., and is preferably parenteral administration. .
  • the appropriate dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration method, patient's age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity, and is usually A skilled doctor can easily determine and prescribe an effective dosage for desired treatment or prevention.
  • the daily dosage of the pharmaceutical composition of the present invention is 0.001-10 g/kg.
  • the pharmaceutical composition of the present invention is manufactured in unit dosage form by formulating using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person skilled in the art. Alternatively, it can be manufactured by placing it in a multi-capacity container. At this time, the formulation may be in the form of a solution, suspension, or emulsion in an oil or aqueous medium, or may be in the form of an extract, powder, granule, tablet, or capsule, and may additionally contain a dispersant or stabilizer.
  • the small molecule quinazoline (Q2a) compound was obtained from KIST (Korea Institute of Science and Technology). KRAS mutant cells were purchased from Horizon Discovery. PARP, C-RAF, P-CRAF, AKT, P-AKT, ERK, P-ERK, HSP90, C-MYC, NOTCH1, and ⁇ -actin antibodies were purchased from Cell Signaling Technology. Secondary antibodies anti-rabbit Ig horseradish peroxidase (HRP) and anti-mouse Ig horseradish peroxidase (HRP) were purchased from GeneTex, and EZ-Cytox for cell proliferation measurement was purchased from Dogen Bio.
  • HRP horseradish peroxidase
  • HRP anti-mouse Ig horseradish peroxidase
  • ANNEXIN V-FITC apoptosis detection kit for measuring apoptosis was purchased from Koma Biotech. Protein Thermal ShiftTM dye kit and Raf-1-RBD beads for pulling down active RAS and detection kit were purchased from Thermo Fisher Scientific and Cell Signaling Technology.
  • Wild-type cancer cells and KRAS G12D mutant cancer cell lines purchased from Horizon Discovery were used.
  • Colon cell lines LIM1215 and SW48, and non-small cell lung cancer (NSCLC) cell lines NCI-H1975 and NCI-H838 were cultured in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin.
  • COLO205, LOVO, HCT8, HCT15, SW480, and SW620 were cultured in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin. All cells were cultured in a 5% CO 2 incubator at 37°C.
  • Cells were seeded in a 60 mm 2 cell culture dish overnight in a 5% CO 2 incubator at 37°C. Compounds were treated at concentrations of 0 ⁇ M, 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M. After obtaining the cells using trypsin-EDTA, the cells were centrifuged at 1500 rpm for 5 minutes to remove the supernatant, and washed with cold PBS for 5 minutes at 1500 rpm. After removing the supernatant, cells were lysed using 2x sample buffer (100 ⁇ l). After heating for 5 to 7 minutes using a heating block, it was quantified using a BCA protein analysis kit (Thermal Fisher Scientific).
  • Equal amounts of protein samples were separated by SDS-PAGE and transferred to PVDF membranes using an electroblotting device (Bio-Rad). Membranes were blocked in blocking buffer (TBS-T + 5% skim milk) for 1 h and incubated with primary antibodies overnight at 4°C. The membrane was washed with TBS-T and incubated with horseradish peroxidase-conjugated secondary antibody for 1 hour at room temperature. Protein expression was visualized using the ECL kit (Amersham Biosciences).
  • KRAS G12D mutant cancer cells and wild-type cancer cells were seeded in a 96-well plate at 5 ⁇ 10 3 to 1 ⁇ 10 4 cells per well and grown overnight. Afterwards, the cells were treated with quinazoline derivative Q2a compound at concentrations of 0 ⁇ M, 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M. After compound treatment, WST analysis using Ez-cytox was performed to measure cell proliferation. After treatment with 10% Ez-cytox, the reaction was performed in a 5% CO 2 incubator at 37°C for 2 hours. Optical density (OD) values were determined by measuring absorbance at 490-500 nm using a microplate reader.
  • Cells were seeded in 6-well plates at 1 ⁇ 10 3 cells per well and grown overnight. Cells were treated with Q2a compound at concentrations of 0 ⁇ M, 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M, and the medium was replaced with new medium containing Q2a compound every 3 days. After 21 days of Q2a compound treatment, the medium was removed and washed with cold PBS. Cell colonies were fixed with methanol for 10 min and stained with crystal violet (0.1% in 20% methanol) for 20 min. After staining, it was carefully washed with distilled water. After at least three independent experiments, the number of surviving colonies was calculated, and the number of surviving colonies (SF) was calculated according to Equation 1 below.
  • Fluorescence-activated cell sorting was performed to measure apoptosis in KRAS G12D mutant cells upon treatment with Q2a compound.
  • KRAS G12D mutant cancer cells and wild-type cancer cells were seeded at 1 ⁇ 10 5 to 1.5 ⁇ 10 5 cells per well in a 6-well plate and grown overnight. Afterwards, the cells were treated with Q2a compound at concentrations of 0 ⁇ M, 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M, and performed according to the kit protocol. Cells were obtained using trypsin EDTA, centrifuged at 1000 xg for 5 minutes to remove the supernatant, and washed with cold PBS at 1000 xg for 5 minutes.
  • the LIM1215 KRAS G12D mutant cell line was seeded in a 60 mm 2 cell culture dish overnight in a 5% CO 2 incubator at 37°C.
  • Q2a compound was treated at concentrations of 0 ⁇ M, 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M.
  • Cellular activity of KRAS protein was measured using the active RAS pull-down and detection kit according to the manufacturer's instructions (Cell Signaling Technology).
  • GTP-bound form of KRAS was extracted from cell lysates using GST-Raf-1 RBD beads. After washing, loading buffer was added to the sample, boiled for 5 minutes, and then SDS-PAGE and Western blotting using KRAS antibody were performed.
  • mice Six-week-old female BALB/C nude mice were purchased from Nara Biotech. 3 ⁇ 10 6 LIM1215 wild type and KRAS G12D mutant cells were administered to the subcutaneous tissue of both flanks of nude mice. Tumors were allowed to grow to an average volume of approximately 100 mm 3 before starting treatment, and mice were treated with compound Q2a by intraperitoneal administration (ip). During the treatment period, tumor volume (V) was measured with calipers once every 2 days and calculated according to the following equation 2.
  • Tumor volume (V) ( L ⁇ S 2 ) / 2 (L is long axis, S is short axis)
  • FACS fluorescence-activated cell sorting
  • Tm Protein thermal shift
  • mutant cells other than KRAS G12D were treated with the Q2a compound and cell growth was measured, and the results are shown in Figure 4.
  • experiments were performed on colon cancer cell lines such as wild-type COLO205, KRAS-G12D mutant LS174T and SNUC2A, KRAS-G12V mutant SW620 and SW480, and KRAS-G13D mutant LOVO and HCT15 cells. After treating each cell with 5 ⁇ M of Q2a compound, cell growth was measured using Ez-cytox 48 and 72 hours later.
  • the wild type KRAS G12V and KRAS G13D mutants did not show any significant effect, but the KRAS G12D mutant cells LS174T and LOVO showed a cell growth inhibitory effect. In other words, it can be confirmed that the Q2a compound exhibits specific activity only in the KRAS G12D mutant.
  • WST-1 analysis was performed to compare the degree of cell growth inhibition following Q2a treatment of a pair of wild-type and KRAS G12D mutant cells in the same cell line.
  • Q2a compound was administered at various concentrations (0 ⁇ M, 1 ⁇ M, 5 ⁇ M, 10 ⁇ M) to wild-type and KRAS G12D mutant pairs of colon cancer cell lines LIM1215 and SW48 and lung cancer cell lines NCI-H838 and NCI-H1975 for 24, 48, and 72 hours. After each treatment, the cell growth inhibition rate was measured, and the results are shown in Figures 5 to 8. After reacting with Ez-cytox for 2 hours, the cell proliferation rate was measured using a microplate reader. As a result, both colon cancer cell lines LIM1214 and SW48 showed a tendency to inhibit cell growth in the KRAS G12D mutant compared to the wild type.
  • LIM1215 cells showed inhibition of cell proliferation compared to the wild type in the 5 ⁇ M and 10 ⁇ M treatment groups when treated for 24 and 48 hours, and cell proliferation was observed at all treatment concentrations of 1 ⁇ M, 5 ⁇ M, and 10 ⁇ M when treated for 72 hours. Inhibition appeared.
  • Lung cancer cell lines showed a better cell proliferation inhibition effect than colon cancer cell lines.
  • KRAS G12D mutant cells showed about 30% inhibition of cell proliferation in the 1 ⁇ M concentration group when treated for 48 hours, even when treated for 24 hours and 72 hours. It was confirmed that cell proliferation was inhibited in a concentration-dependent manner.
  • KRAS G12D mutant cells were shown to be inhibited in cell growth in a concentration-dependent manner compared to wild-type cells when treated for 24 hours, 48 hours, and 72 hours.
  • SW48 KRAS G12D mutant cells also showed a significant decrease in cell proliferation in the Q2a compound treatment group.
  • the number of colonies of KRAS G12D mutant cells was found to be 60% lower than that of the control group, but there was no significant difference in the wild type group.
  • NCI-H838 cells the number of colonies of KRAS G12D mutant cells was found to be less than 50% compared to the control group, and in NCI-H1975 cells, the number of colonies of KRAS G12D mutant cells was found to be 70-50% less than the control group. In contrast, there was no significant difference in the wild type group.
  • Fluorescence-activated cell sorting was performed to confirm the apoptotic effect in KRAS G12D mutant cells when treated with Q2a compound.
  • Wild-type and KRAS G12D mutant pairs of LIM1215, SW48, NCI-H1975, and NCI-H838 cells were treated with Q2a compound at different concentrations (0 ⁇ M, 1 ⁇ M, 5 ⁇ M, 10 ⁇ M) for 24, 48, and 72 h.
  • Apoptosis was measured by Annexin V and PI staining.
  • LIM1215 and SW48 compared to the control group, apoptosis in KRAS G12D mutant cells was at least 5% and at most 20% in the Q2a compound treatment group.
  • NCI-H838 KRAS G12D cells showed the greatest apoptosis effect among all cells used in the experiment.
  • an apoptosis effect of more than 60% was observed compared to the control group.
  • PARP poly(ADP-ribose) polymerase a representative marker of apoptosis, is a family of 17 proteins involved in several cellular processes, including stress response, chromatin modification, DNA repair, and apoptosis. This has been confirmed to play a role in the detection and repair of DNA breaks. The presence of cleaved PARP-1 is one of the most commonly used diagnostic tools to detect apoptosis in many cell types.
  • FIG 18 shows the Western blot results for wild-type (WT) LIM1215 cells and KRAS G12D mutant cells treated with Q2a 48.72 for 72 hours
  • Figure 19 shows Q2a 48.72 for wild-type (WT) SW48 cells and KRAS G12D mutant cells.
  • This is the Western blot result upon time processing.
  • PARP cleavage in LIM1215 and SW48 KRAS G12D mutant cells increased in both the 48-hour and 72-hour treatment groups compared to the wild type.
  • Figure 20 is a Western blot result for wild-type (WT) NCI-H1975 cells and KRAS G12D mutant cells treated with Q2a 48. 72 hours
  • Figure 21 is a Western blot result for wild-type (WT) NCI-H838 cells and KRAS G12D mutant cells.
  • Q2a 48 This is the Western blot result after 72 hours of treatment. According to this, likewise NCI-H1975. PARP cleavage also occurred in NCI-H838 KRAS G12D mutant cells and increased for both 48 and 72 hours compared to the wild type.
  • FIG 22 shows the results of Western blotting of wild type (WT) LIM1215 and SW48 cells and KRAS G12D mutant cells treated with Q2a for 24 hours.
  • WT wild type
  • colon cancer cells LIM1215 and SW48 showed decreased phosphorylation of the MAPK pathway protein C-RAF from 48 hours, and phosphorylation of AKT, a PI3K pathway protein, was also significantly decreased compared to the control group (see Figures 18 and 19).
  • Figure 23 shows the results of Western blotting of wild type (WT) NCI-H1975 and NCI-H838 cells and KRAS G12D mutant cells treated with Q2a for 24 hours. According to this, unlike at 48 and 72 hours, the phosphorylation of the MAPK pathway protein C-RAF decreased at 24 hours, and the phosphorylation of the PI3K pathway protein AKT also decreased significantly compared to the control group.
  • Figure 24 is a heatmap of three control groups and three groups treated with 5 ⁇ M of Q2a compound. According to this, differences in gene expression can be confirmed. When treated with 2a, it was confirmed that the KRAS signal in KRAS mutant cells was decreased, indicating that the Q2a compound targeted KRAS.
  • treatment with Q2a showed a decrease in the expression of MYC and NOTCH, which are oncogenic pathway proteins, in KRAS mutant cells.
  • KRAS-GTP binding decreased when treated with 5 ⁇ M for 48 hours and 10 ⁇ M for 72 hours, confirming that RAS activity was reduced.
  • C-MYC and NOTCH1 representative markers of oncogenic pathway proteins, were also found to have decreased protein expression for 24, 48, and 72 hours.
  • LIM1215 wild-type cells and KRAS G12D mutant cells were injected subcutaneously into both flanks of BALB/C nude mice. After the tumor grew to a size of 100 mm 2 , 100 ⁇ l of Q2a compound was injected intraperitoneally every two days at a concentration of 15 mg/kg and 30 mg/kg.
  • FIG. 30 A schematic diagram of the mechanism of cancer cell inhibition according to Q2a administration in the KRAS pathway is shown in Figure 30.
  • treatment with Q2a compound reduces signaling of downstream MAPK and PI3K mechanisms along with decreased KRAS-GTP activity.
  • cell death may occur and cell proliferation may be inhibited.
  • the compound of the present invention selectively inhibits cell proliferation in KRAS G12D mutant cells, it can solve the problem of low efficacy of conventional targeted therapy using epidermal growth factor receptor (EGFR), and the compound of the present invention and its salt are effective.
  • EGFR epidermal growth factor receptor
  • the pharmaceutical composition containing the composition as an ingredient can be used for targeted treatment against KRAS G12D mutant cells.

Landscapes

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

Abstract

The present invention relates to: a compound, represented by chemical formula 1, for inhibiting cell proliferation of a KRAS G12D mutation; and a pharmaceutical composition for preventing or treating cancer disease, comprising same as an active ingredient. Therefore, the present invention can exhibit significantly superior effects in targeted treatment on cancer cell lines of colon cancer, lung cancer and the like, and on KRAS G12D mutations and KRAS mutant cells thereof.

Description

KRAS G12D 돌연변이 억제용 화합물 및 이를 유효성분으로 포함하는 암질환 예방 또는 치료용 조성물Compound for inhibiting KRAS G12D mutation and composition for preventing or treating cancer disease containing the same as an active ingredient

본 발명은 KRAS G12D 돌연변이 억제용 화합물 및 이를 유효성분으로 포함하는 암질환 예방 또는 치료용 조성물에 관한 것으로, 더욱 상세하게는 KRAS G12D 돌연변이 를 표적으로 세포증식을 억제할 수 있는 화합물 및 이를 유효성분으로 포함하는 암질환 예방 또는 치료용 조성물에 관한 것이다.The present invention relates to a compound for inhibiting KRAS G12D mutation and a composition for preventing or treating cancer diseases containing the same as an active ingredient. More specifically, it relates to a compound capable of inhibiting cell proliferation by targeting the KRAS G12D mutation and a composition containing the same as an active ingredient. It relates to a composition for preventing or treating cancer disease, comprising:

암은 유전적 돌연변이를 포함하는 미성숙하고 과증식하는 세포에 의해 발생하는 질병이다. 통계청에 따르면 2021년 3대 사망원인은 암, 심장병, 폐렴으로 전체 사망자의 26%가 암이다. 이 값은 매년 0.6%씩 증가하고 있으며, 이는 암 관련 사망률 증가로 이어지고 있다(통계청, 2021). 암 발병률은 1999년에는 인구 10만명당 214.2명, 2012년에는 445.3명이었다. 암 발병 위험은 평균 수명(81세)까지 생존한 사람의 경우 37.3%이며, 남성(77세)은 37.5%, 여성(84세)은 34.9%이다.Cancer is a disease caused by immature, overproliferating cells that contain genetic mutations. According to Statistics Korea, the three leading causes of death in 2021 are cancer, heart disease, and pneumonia, with cancer accounting for 26% of all deaths. This value is increasing by 0.6% every year, leading to an increase in cancer-related mortality (Statistics Korea, 2021). The cancer incidence rate was 214.2 per 100,000 population in 1999 and 445.3 in 2012. The risk of developing cancer is 37.3% for people who survive to the average life expectancy (age 81), 37.5% for men (age 77), and 34.9% for women (age 84).

암 관련 사망이 증가하는 주요 원인은 서구화된 생활방식, 인구의 노령화, 흡연, 음주, 비만 등으로 추정된다. 암이란 세포증식 조절계의 DNA에 이상이 생기는 현상을 말하며, 이 DNA는 DNA의 이와 같은 유전적 변이와 이상 활성 유전정보에 의해 암 관련 단백질을 합성하므로 암유전자(종양유전자)라고 한다. 정상적인 조건에서 암 유전자는 암 발생을 유도하지 않고 오히려 증식을 조절한다; 이들 유전자는 유전적 변이 또는 이상에 의해 활성화되면 암을 유발한다. 암 유전자의 돌연변이로 인해 세포는 세포 성장을 억제하는 능력을 상실할 수 있다. 통제되지 않은 세포 증식은 암을 유발한다. 이들 세포는 혈관신생, 전이 등의 과정을 거쳐 신체의 다른 부위로 옮겨질 수 있다.The main reasons for the increase in cancer-related deaths are believed to be Westernized lifestyles, aging population, smoking, drinking, and obesity. Cancer refers to a phenomenon in which abnormalities occur in the DNA of the cell proliferation control system. This DNA synthesizes cancer-related proteins due to genetic mutations and abnormal active genetic information in the DNA, so it is called a cancer gene (oncogene). Under normal conditions, oncogenes do not induce cancer development but rather regulate proliferation; These genes cause cancer when activated by genetic mutation or abnormality. Mutations in cancer genes can cause cells to lose their ability to inhibit cell growth. Uncontrolled cell proliferation causes cancer. These cells can be moved to other parts of the body through processes such as angiogenesis and metastasis.

RAS 돌연변이는 대장암, 폐암, 췌장암에서 높은 비율로 발생하고 있다. RAS는 세포 성장, 분화, 증식 및 생존에 중요한 역할을 하는 소 GTPase 단백질 계열이다. RAS 유전자에는 HRAS, KRAS 및 NRAS의 세 가지 유형이 있다. RAS 유전자는 인간 암에서 가장 흔하게 발견되는 종양유전자이다. KRAS는 다양한 암 유형에서 HRAS 및 NRAS에 비해 가장 높은 돌연변이율을 나타내는 것으로 알려져 있다. KRAS는 또한 활성 상태에서 GTP에 결합하고 이를 GDP로 전환시켜 KRAS를 비활성화시키는 독특한 효소 활성을 나타내는 것으로 알려져 있다. 전환 속도는 일반적으로 느리지만 보조 GTPase 활성화 단백질(GAP)에 의해 극적으로 증가할 수 있다. KRAS는 SOS와 같은 구아닌 뉴클레오티드 교환 인자(GEF, guanine nucleotide exchange factor)를 통해 GDP를 방출한다. KRAS의 GTP 결합은 스위치 I 영역(잔기 30-40)과 스위치 II 영역(잔기 60-70)의 여러 잔기가 RAS 효과인자 단백질(effector protein)에 결합하도록 한다. 이들 스위치는 GAP 및 GEF에 의해 조절될 수 있다. KRAS는 GDP에 바인딩된 스위치 오프의 비활성 상태에서 GEF를 통해 GTP가 바인딩되는 활성 스위치 온 상태로 전환된 후 세포 분화, 증식 및 생존에 필요한 신호 전달이 진행될 수 있다. 시간이 지남에 따라 GAP의 존재 하에서 GTP는 GDP로 전환되어 ON/OFF 상태를 순환할 수 있다.RAS mutations occur at a high rate in colon cancer, lung cancer, and pancreatic cancer. RAS is a family of small GTPase proteins that play important roles in cell growth, differentiation, proliferation, and survival. There are three types of RAS genes: HRAS, KRAS, and NRAS. The RAS gene is the most commonly found oncogene in human cancer. KRAS is known to have the highest mutation rate compared to HRAS and NRAS in various cancer types. KRAS is also known to exhibit a unique enzymatic activity that binds GTP in its active state and converts it to GDP, thereby inactivating KRAS. The rate of turnover is generally slow but can be dramatically increased by auxiliary GTPase activating proteins (GAPs). KRAS releases GDP through a guanine nucleotide exchange factor (GEF), like SOS. GTP binding of KRAS causes several residues in the switch I region (residues 30-40) and switch II region (residues 60-70) to bind to the RAS effector protein. These switches can be regulated by GAP and GEF. After KRAS is converted from the inactive switch-off state bound to GDP to the active switch-on state bound to GTP via GEF, signal transduction required for cell differentiation, proliferation, and survival can proceed. Over time, in the presence of GAP, GTP can be converted to GDP and cycle through the ON/OFF state.

그러나 KRAS에 돌연변이가 발생하면 GAP 결합이 중단되고 GTP 상태로 유지되어 종양 형성 및 유지에 도움을 주는 비정상적인 신호 전달 경로가 발생하게 된다. RAS 돌연변이가 있는 암의 약 85%가 KRAS 돌연변이를 포함하며, 대장암의 45%, 췌장암의 86%, 폐암의 25% 가량이 포함된다고 알려져 있다. KRAS 돌연변이는 글리신 12(G12), 글리신 13(G13) 또는 글루타민 61에서 보고된 바 있다. G12 돌연변이는 G12D(41%), G12C(28%) 및 G12C(14%)를 포함하여 KRAS 돌연변이의 약 80%를 차지한다. 폐암(32%), 췌장암(86%), 대장암(41%)의 높은 비율은 KRAS의 코돈 12에 돌연변이를 포함한다. 폐암의 KRAS G12C 돌연변이의 경우 글리신이 시스테인으로 돌연변이가 일어난다. 췌장암의 KRAS G12D 돌연변이의 경우 글리신이 D-아스파르트산으로 돌연변이가 일어난다. KRAS 돌연변이가 있는 환자에서 표피 성장 인자 수용체(EGFR)를 이용한 표적 치료법은 이러한 돌연변이가 없는 환자에 비해 치료 효능이 낮고 생존율도 낮다고 알려져 있다.However, when KRAS is mutated, GAP binding is disrupted and remains in the GTP state, resulting in an abnormal signaling pathway that helps tumor formation and maintenance. It is known that approximately 85% of cancers with RAS mutations include KRAS mutations, including 45% of colon cancer, 86% of pancreatic cancer, and 25% of lung cancer. KRAS mutations have been reported at glycine 12 (G12), glycine 13 (G13), or glutamine 61. G12 mutations account for approximately 80% of KRAS mutations, including G12D (41%), G12C (28%), and G12C (14%). A high proportion of lung (32%), pancreatic (86%), and colorectal (41%) cancers contain mutations in codon 12 of KRAS. In the case of KRAS G12C mutation in lung cancer, glycine is mutated to cysteine. In the case of KRAS G12D mutation in pancreatic cancer, glycine is mutated to D-aspartic acid. Targeted therapy using epidermal growth factor receptor (EGFR) in patients with KRAS mutations is known to have lower treatment efficacy and lower survival rates than patients without these mutations.

수용체 티로신 키나제의 EGFR 계열은 GDP-GTP 교환을 촉진하여 RAS를 활성화한다. EGFR을 억제하면 RAS 활성화가 방지되고, SOS 또는 SHP2를 억제하면 GDP-GTP 교환율이 감소하여 RAS에 대한 GTP 결합이 감소한다. EGFR은 RAS의 업스트림에 있기 때문에 이 수용체 티로신 키나제의 불활성화는 RAS 활성을 억제할 수 있다. cetuximab 또는 panitumumab과 같은 EGFR에 대한 단일클론 항체는 야생형 KRAS 전이성 대장암 환자의 전체 생존 기간과 무진행 생존 기간을 증가시킬 수 있다. 그러나 이 항체는 KRAS 돌연변이 종양, 특히 코돈 12 또는 13 돌연변이가 있는 종양에는 효과적이지 않다. 에를로티닙(erlotinib) 및 게피티닙(gefitinib)과 같은 EGFR 티로신 키나제 억제제는 EGFR 돌연변이 비소세포폐암(non-small cell lung cancer) 치료용으로 승인되었지만 KRAS 돌연변이 비소세포폐암에서는 단일한 효과를 나타내지 않는다.The EGFR family of receptor tyrosine kinases activates RAS by promoting GDP-GTP exchange. Inhibiting EGFR prevents RAS activation, while inhibiting SOS or SHP2 reduces GDP-GTP exchange rate and thus reduces GTP binding to RAS. Because EGFR is upstream of RAS, inactivation of this receptor tyrosine kinase can inhibit RAS activity. Monoclonal antibodies against EGFR, such as cetuximab or panitumumab, can increase overall survival and progression-free survival in patients with wild-type KRAS metastatic colorectal cancer. However, this antibody is not effective against KRAS mutant tumors, especially those with codon 12 or 13 mutations. EGFR tyrosine kinase inhibitors, such as erlotinib and gefitinib, are approved for the treatment of EGFR-mutant non-small cell lung cancer, but have no single effect in KRAS-mutant non-small cell lung cancer. .

야생형 KRAS 대장암은 세툭시맙(cetuximab) 치료에 반응한다. 그러나 전이성 대장암은 이 암이 항-EGFR 치료에 내성을 갖게 만드는 KRAS 돌연변이를 획득하게 된다. 따라서 EGFR을 표적으로 하는 항암제보다는 KRAS 돌연변이를 직접적으로 표적으로 삼는 억제제가 필요하다. 또한, 폐암에서 돌연변이 KRAS G12C의 억제제가 현재 개발 및 평가되고 있지만, 대장암 및 췌장암에서 KRAS G12D 돌연변이에 대한 억제제는 광범위하게 평가되어 있지 않은 실정이다.Wild-type KRAS colorectal cancer responds to cetuximab treatment. However, metastatic colorectal cancer acquires KRAS mutations that make the cancer resistant to anti-EGFR therapy. Therefore, an inhibitor that directly targets KRAS mutations is needed rather than an anticancer drug that targets EGFR. Additionally, although inhibitors of mutant KRAS G12C in lung cancer are currently being developed and evaluated, inhibitors of KRAS G12D mutations in colon and pancreatic cancer have not been extensively evaluated.

본 발명의 KRAS G12D 돌연변이가 있는 암환자에서 표피 성장 인자 수용체(EGFR)를 이용한 표적 치료법은 이러한 돌연변이가 없는 환자에 비해 치료 효능이 낮은 문제를 해결하기 위하여 KRAS G12D 돌연변이 세포에 대해 선택적으로 세포증식을 억제하는 화합물과 이를 유효성분으로 포함하여 KRAS G12D 돌연변이 세포에 대해 표적 치료용으로 사용할 수 있는 암질환 예방 또는 치료용 약학 조성물을 제공하는 데 있다.Targeted therapy using epidermal growth factor receptor (EGFR) in cancer patients with KRAS G12D mutation of the present invention selectively proliferates KRAS G12D mutant cells to solve the problem of lower treatment efficacy compared to patients without this mutation. The aim is to provide a pharmaceutical composition for preventing or treating cancer that contains an inhibitory compound and the same as an active ingredient and can be used for targeted treatment against KRAS G12D mutant cells.

본 발명의 일 측면에 따르면,According to one aspect of the present invention,

하기 화학식 1로 표시되는 KRAS G12D 돌연변이 세포증식 억제용 화합물이 제공된다.A compound for inhibiting proliferation of KRAS G12D mutant cells represented by the following formula (1) is provided.

[화학식 1][Formula 1]

Figure PCTKR2024001239-appb-img-000001
Figure PCTKR2024001239-appb-img-000001

화학식 1에서,In Formula 1,

n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeat units, which is an integer from 1 to 10,

m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeat units that is an integer from 1 to 10,

X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom,

R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,

R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,

R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.

바람직하게는 상기 화학식 1에서,Preferably in Formula 1,

n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4,

m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4,

X1은 산소원자이고,X 1 is an oxygen atom,

R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group,

R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group,

R5

Figure PCTKR2024001239-appb-img-000002
이고, R 5 is
Figure PCTKR2024001239-appb-img-000002
ego,

X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom,

R6은 수소원자, 또는 1C 내지 10C 알킬기일 수 있다.R 6 may be a hydrogen atom or a 1C to 10C alkyl group.

더욱 바람직하게는,More preferably,

상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물일 수 있다.The compound represented by Formula 1 may be a compound represented by Formula 2 below.

[화학식 2][Formula 2]

Figure PCTKR2024001239-appb-img-000003
Figure PCTKR2024001239-appb-img-000003

..

본 발명의 다른 또 하나의 측면에 따르면,According to another aspect of the present invention,

하기 화학식 1로 표시되는 화합물 또는 이의 염을 유효성분으로 포함하는 암질환 예방 또는 치료용 약학 조성물이 제공된다.A pharmaceutical composition for preventing or treating cancer disease comprising a compound represented by the following formula (1) or a salt thereof as an active ingredient is provided.

[화학식 1][Formula 1]

Figure PCTKR2024001239-appb-img-000004
Figure PCTKR2024001239-appb-img-000004

화학식 1에서,In Formula 1,

n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeat units, which is an integer from 1 to 10,

m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeat units that is an integer from 1 to 10,

X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom,

R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,

R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,

R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.

바람직하게는 상기 화학식 1에서,Preferably in Formula 1,

n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4,

m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4,

X1은 산소원자이고,X 1 is an oxygen atom,

R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group,

R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group,

R5

Figure PCTKR2024001239-appb-img-000005
이고, R 5 is
Figure PCTKR2024001239-appb-img-000005
ego,

X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom,

R6은 수소원자, 또는 1C 내지 10C 알킬기일 수 있다.R 6 may be a hydrogen atom or a 1C to 10C alkyl group.

더욱 바람직하게는, 상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물일 수 있다.More preferably, the compound represented by Formula 1 may be a compound represented by Formula 2 below.

[화학식 2][Formula 2]

Figure PCTKR2024001239-appb-img-000006
Figure PCTKR2024001239-appb-img-000006

..

상기 암질환 예방 또는 치료용 약학 조성물은 KRAS G12D 돌연변이 세포증식 억제용일 수 있다.The pharmaceutical composition for preventing or treating cancer disease may be used to inhibit proliferation of KRAS G12D mutant cells.

상기 KRAS G12D 돌연변이 세포는 LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D 및 LS174T KRAS G12D 중에서 선택된 1종 이상일 수 있다.The KRAS G12D mutant cells may be one or more types selected from LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D, and LS174T KRAS G12D.

상기 암질환 예방 또는 치료용 조성물은 상기 KRAS G12D 돌연변이 세포에 대한 표적 치료용일 수 있다.The composition for preventing or treating cancer disease may be for targeted treatment against the KRAS G12D mutant cells.

상기 암질환 예방 또는 치료용 조성물은 MAPK 신호전달 경로 억제용일 수 있다.The composition for preventing or treating cancer disease may be for inhibiting the MAPK signaling pathway.

상기 MAPK 신호전달 경로 억제는 RAS-RAF-MEK-ERK 경로에서 C-RAF 인산화 억제에 따라 수행될 수 있다.Inhibition of the MAPK signaling pathway can be performed by inhibiting C-RAF phosphorylation in the RAS-RAF-MEK-ERK pathway.

상기 암질환 예방 또는 치료용 조성물은 PI3K 신호전달 경로 억제용일 수 있다.The composition for preventing or treating cancer disease may be for inhibiting the PI3K signaling pathway.

상기 PI3K 신호전달 경로 억제는 PI3K-AKT-mTOR 경로에서 AKT 인산화 억제에 따라 수행될 수 있다.The inhibition of the PI3K signaling pathway can be performed by inhibiting AKT phosphorylation in the PI3K-AKT-mTOR pathway.

상기 암질환은 대장암, 결장암, 직장암, 폐암, 흑색종, 갑상선암, 자궁암, 난소암, 자궁경부함, 췌장암, 위암 및 간암 중에서 선택된 어느 하나일 수 있다.The cancer disease may be any one selected from colon cancer, colon cancer, rectal cancer, lung cancer, melanoma, thyroid cancer, uterine cancer, ovarian cancer, cervix, pancreatic cancer, stomach cancer, and liver cancer.

본 발명의 화합물은 KRAS G12D 돌연변이 세포에 대해 선택적으로 세포증식을 억제하므로 종래 표피 성장 인자 수용체(EGFR)를 이용한 표적 치료법이 낮은 효능을 나타내는 문제를 해결할 수 있고, 본 발명의 화합물과 그의 염을 유효성분으로 포함하는 약학 조성물은 KRAS G12D 돌연변이 세포에 대해 표적 치료용으로 사용할 수 있다.Since the compound of the present invention selectively inhibits cell proliferation in KRAS G12D mutant cells, it can solve the problem of low efficacy of conventional targeted therapy using epidermal growth factor receptor (EGFR), and the compound of the present invention and its salt are effective. The pharmaceutical composition containing the composition as an ingredient can be used for targeted treatment against KRAS G12D mutant cells.

도 1은 실험예 1의 Q1a ~ Q11a 화합물들의 LIM1215의 야생형 세포와 KRAS 돌연변이 세포에 대한 세포성장 측정 결과이다.Figure 1 shows the cell growth measurement results of compounds Q1a to Q11a of Experimental Example 1 on wild-type cells of LIM1215 and KRAS mutant cells.

도 2는 실험예 1에서 선발된 Q2a 화합물의 분자 구조와 3차원 모델 구조를 나타낸 것이다.Figure 2 shows the molecular structure and three-dimensional model structure of the Q2a compound selected in Experimental Example 1.

도 3은 실험예 1의 Q2a 화합물이 KRAS에 결합되었는지 확인하기 위한 단백질 열 이동(PTS) 분석 결과이다.Figure 3 shows the results of protein thermal transfer (PTS) analysis to confirm whether the Q2a compound of Experimental Example 1 bound to KRAS.

도 4는 실험예 1에서 Q2a 화합물이 KRAS G12D 돌연변이에서만 특이적 활성을 갖는지 확인한 결과이다.Figure 4 shows the results of confirming whether compound Q2a has specific activity only in the KRAS G12D mutation in Experimental Example 1.

도 5는 실험예 2에 따른 대장암 세포주 LIM1215의 야생형과 KRAS G12D 돌연변이 쌍의 Q2a 화합물 처리시 세포성장율을 비교한 결과이다.Figure 5 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of colon cancer cell line LIM1215 according to Experimental Example 2 when treated with Q2a compound.

도 6은 실험예 2에 따른 대장암 세포주 SW48의 야생형과 KRAS G12D 돌연변이 쌍의 Q2a 화합물 처리시 세포성장율을 비교한 결과이다Figure 6 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of colon cancer cell line SW48 according to Experimental Example 2 when treated with Q2a compound.

도 7은 실험예 2에 따른 폐암 세포주 NCI-H838의 야생형과 KRAS G12D 돌연변이 쌍의 Q2a 화합물 처리시 세포성장율을 비교한 결과이다.Figure 7 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of lung cancer cell line NCI-H838 according to Experimental Example 2 when treated with Q2a compound.

도 8은 실험예 2에 따른 폐암 세포주 NCI-H1975의 야생형과 KRAS G12D 돌연변이 쌍의 Q2a 화합물 처리시 세포성장율을 비교한 결과이다.Figure 8 shows the results of comparing the cell growth rate of the wild type and KRAS G12D mutant pair of lung cancer cell line NCI-H1975 according to Experimental Example 2 when treated with Q2a compound.

도 9는 실험예 2의 LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과이다.Figure 9 shows the results of colony formation analysis for wild type (WT) LIM1215 cells and KRAS G12D mutant cells in Experimental Example 2.

도 10은 실험예 2의 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과이다.Figure 10 shows the results of colony formation analysis for wild type (WT) SW48 cells and KRAS G12D mutant cells in Experimental Example 2.

도 11은 실험예 2의 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과이다.Figure 11 shows the results of colony formation analysis for wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 2.

도 12는 실험예 2의 NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과이다.Figure 12 shows the results of colony formation analysis for wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 2.

도 13은 실험예 2의 LS174T KRAS G12D 돌연변이 세포의 콜로니 형성 분석 결과이다.Figure 13 shows the results of colony formation analysis of LS174T KRAS G12D mutant cells in Experimental Example 2.

도 14는 실험예 3의 LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과이다.Figure 14 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of LIM1215 cells in Experimental Example 3.

도 15는 실험예 3의 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과이다.Figure 15 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of SW48 cells in Experimental Example 3.

도 16은 실험예 3의 NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과이다.Figure 16 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 3.

도 17은 실험예 3의 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과이다.Figure 17 shows the results of analysis of the apoptosis effect on wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 3.

도 18은 실험예 4의 LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다.Figure 18 shows the results of Western blotting of wild type (WT) LIM1215 cells and KRAS G12D mutant cells in Experimental Example 4 when treated with Q2a for 48.72 hours.

도 19는 실험예 4의 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다.Figure 19 shows the results of Western blotting of wild type (WT) SW48 cells and KRAS G12D mutant cells in Experimental Example 4 when treated with Q2a for 48.72 hours.

도 20은 실험예 4의 NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다.Figure 20 shows the Western blot results of wild type (WT) and KRAS G12D mutant cells of NCI-H1975 cells in Experimental Example 4 when treated with Q2a for 48 and 72 hours.

도 21은 실험예 4의 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다.Figure 21 shows the results of Western blotting of wild type (WT) and KRAS G12D mutant cells of NCI-H838 cells in Experimental Example 4 when treated with Q2a for 48.72 hours.

도 22는 실험예 4의 LIM1215 및 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 24시간 처리시 웨스턴 블롯 결과이다.Figure 22 shows the results of Western blotting of LIM1215 and SW48 cells in Experimental Example 4 when wild type (WT) and KRAS G12D mutant cells were treated with Q2a for 24 hours.

도 23은 실험예 4의 NCI-H1975 및 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 24시간 처리시 웨스턴 블롯 결과이다.Figure 23 shows the results of Western blotting of wild type (WT) and KRAS G12D mutant cells of NCI-H1975 and NCI-H838 cells in Experimental Example 4 when treated with Q2a for 24 hours.

도 24는 실험예 5의 마이크로어레이 분석에 따른 히트맵이다.Figure 24 is a heatmap according to microarray analysis in Experimental Example 5.

도 25는 실험예 5의 웨스턴블롯 분석 결과이다.Figure 25 shows the results of Western blot analysis in Experimental Example 5.

도 26은 실험예 5의 LIM1215 야생형 세포와 KRAS G12D 돌연변이 세포 투여 마우스에서 Q2a 투여에 따른 종양 크기의 시간에 따른 변화를 나타낸 것이다.Figure 26 shows the change in tumor size over time according to Q2a administration in mice administered LIM1215 wild-type cells and KRAS G12D mutant cells in Experimental Example 5.

도 27은 실험예 5의 Q2a 화합물을 투여하면서 측정한 마우스 체중 변화 그래프이다.Figure 27 is a graph of mouse body weight change measured while administering compound Q2a of Experimental Example 5.

도 28은 실험예 5의 치료 21일 후 마우스를 희생시켜 종양을 제거하여 종양의 크기를 대비한 사진Figure 28 is a photo comparing the size of the tumor after sacrificing the mouse and removing the tumor 21 days after the treatment in Experimental Example 5.

도 29는 실험예 5에 따른 치료 전후의 종양의 중량을 측정한 결과이다.Figure 29 shows the results of measuring the weight of the tumor before and after treatment according to Experimental Example 5.

도 30은 KRAS 경로에서 Q2a 투여에 따른 암세포 억제 매커니즘이다.Figure 30 shows the mechanism of cancer cell inhibition according to Q2a administration in the KRAS pathway.

본 발명은 다양한 변환을 가할 수 있고 여러 가지 실시예를 가질 수 있는 바, 특정 실시예들을 예시하고 상세한 설명에 상세하게 설명하고자 한다. 그러나, 이는 본 발명을 특정한 실시 형태에 대해 한정하려는 것이 아니며, 본 발명의 사상 및 기술 범위에 포함되는 모든 변환, 균등물 내지 대체물을 포함하는 것으로 이해되어야 한다. 본 발명을 설명함에 있어서 관련된 공지 기술에 대한 구체적인 설명이 본 발명의 요지를 흐릴 수 있다고 판단되는 경우 그 상세한 설명을 생략한다.Since the present invention can be modified in various ways and can have various embodiments, specific embodiments will be illustrated and described in detail in the detailed description. However, this is not intended to limit the present invention to specific embodiments, and should be understood to include all transformations, equivalents, and substitutes included in the spirit and technical scope of the present invention. In describing the present invention, if it is determined that a detailed description of related known technologies may obscure the gist of the present invention, the detailed description will be omitted.

상기 "치환된"이란 적어도 하나의 수소원자가 중수소, C1 내지 C30 알킬기, C3 내지 C30 시클로알킬기, C2 내지 C30 헤테로시클로알킬기, C1 내지 C30 할로겐화알킬기, C6 내지 C30 아릴기, C1 내지 C30 헤테로아릴기, C1 내지 C30 알콕시기, C3 내지 C30 시클로알콕시기, C1 내지 C30 헤테로시클로알콕시기, C2 내지 C30 알케닐기, C2 내지 C30 알키닐기, C6 내지 C30 아릴옥시기, C1 내지 C30 헤테로아릴옥시기, 실릴옥시기(-OSiH3), -OSiR1H2(R1은 C1 내지 C30 알킬기 또는 C6 내지 C30 아릴기), -OSiR1R2H(R1 및 R2는 각각 독립적으로 C1 내지 C30 알킬기 또는 C6 내지 C30 아릴기), -OSiR1R2R3, (R1, R2, 및 R3는 각각 독립적으로 C1 내지 C30 알킬기 또는 C6 내지 C30 아릴기), C1 내지 C30 아실기, C2 내지 C30 아실옥시기, C2 내지 C30 헤테로아릴옥시기, C1 내지 C30 술포닐기, C1 내지 C30 알킬티올기, C3 내지 C30 시클로알킬티올기, C1 내지 C30 헤테로시클로알킬티올기, C6 내지 C30 아릴티올기, C1 내지 C30 헤테로아릴티올기, C1 내지 C30 인산아마이드기, 실릴기(SiR1R2R3 )(R1, R2, 및 R3는 각각 독립적으로 수소 원자, C1 내지 C30 알킬기 또는 C6 내지 C30 아릴기), 아민기(-NRR')(여기에서, R 및 R'은 각각 독립적으로, 수소 원자, C1 내지 C30 알킬기, 및 C6 내지 C30 아릴기로 이루어진 군에서 선택되는 치환기임), 카르복실기, 할로겐기, 시아노기, 니트로기, 아조기, 및 하이드록시기로 이루어진 군에서 선택되는 치환기로 치환된 것을 의미한다.The term “substituted” means that at least one hydrogen atom is deuterium, C1 to C30 alkyl group, C3 to C30 cycloalkyl group, C2 to C30 heterocycloalkyl group, C1 to C30 halogenated alkyl group, C6 to C30 aryl group, C1 to C30 heteroaryl group, C1 to C30 alkoxy group, C3 to C30 cycloalkoxy group, C1 to C30 heterocycloalkoxy group, C2 to C30 alkenyl group, C2 to C30 alkynyl group, C6 to C30 aryloxy group, C1 to C30 heteroaryloxy group, silyl oxide Group (-OSiH 3 ), -OSiR 1 H 2 (R 1 is a C1 to C30 alkyl group or C6 to C30 aryl group), -OSiR 1 R 2 H (R 1 and R 2 are each independently a C1 to C30 alkyl group or C6 to C30 aryl group), -OSiR 1 R 2 R 3 , (R 1 , R 2 , and R 3 are each independently a C1 to C30 alkyl group or a C6 to C30 aryl group), C1 to C30 acyl group, C2 to C30 acyl Oxy group, C2 to C30 heteroaryloxy group, C1 to C30 sulfonyl group, C1 to C30 alkylthiol group, C3 to C30 cycloalkylthiol group, C1 to C30 heterocycloalkylthiol group, C6 to C30 arylthiol group, C1 to C30 heteroarylthiol group, C1 to C30 phosphate amide group, silyl group (SiR 1 R 2 R 3 ) (R 1 , R 2 , and R 3 are each independently a hydrogen atom, a C1 to C30 alkyl group, or a C6 to C30 aryl group ), amine group (-NRR') (wherein R and R' are each independently a substituent selected from the group consisting of a hydrogen atom, C1 to C30 alkyl group, and C6 to C30 aryl group), carboxyl group, halogen group, It means substituted with a substituent selected from the group consisting of cyano group, nitro group, azo group, and hydroxy group.

또한 상기 치환기 중 인접한 두 개의 치환기가 융합되어 포화 또는 불포화 고리를 형성할 수도 있다.Additionally, two adjacent substituents among the above substituents may be fused to form a saturated or unsaturated ring.

또한, 상기 "치환 또는 비치환된 C1 내지 C30 알킬기" 또는 "치환 또는 비치환된 C6 내지 C30 아릴기" 등에서의 상기 알킬기 또는 아릴기의 탄소수 범위는 상기 치환기가 치환된 부분을 고려하지 않고 비치환된 것으로 보았을 때의 알킬 부분 또는 아릴 부분을 구성하는 전체 탄소수를 의미하는 것이다. 예컨대, 파라 위치에 부틸기가 치환된 페닐기는 탄소수 4의 부틸기로 치환된 탄소수 6의 아릴기에 해당하는 것을 의미한다.In addition, the carbon number range of the alkyl group or aryl group in the “substituted or unsubstituted C1 to C30 alkyl group” or “substituted or unsubstituted C6 to C30 aryl group” does not take into account the portion on which the substituent is substituted and is not substituted. It refers to the total number of carbon atoms constituting the alkyl portion or aryl portion when viewed as being formed. For example, a phenyl group substituted with a butyl group at the para position corresponds to an aryl group with 6 carbon atoms substituted with a butyl group with 4 carbon atoms.

본 명세서에서 "헤테로"란 별도의 정의가 없는 한, 하나의 작용기 내에 N, O, S 및 P로 이루어진 군에서 선택되는 헤테로 원자를 1 내지 4개 함유하고, 나머지는 탄소인 것을 의미한다.As used herein, unless otherwise defined, “hetero” means that one functional group contains 1 to 4 hetero atoms selected from the group consisting of N, O, S, and P, and the remainder is carbon.

본 명세서에서 "수소"란 별도의 정의가 없는 한, 일중수소, 이중수소, 또는 삼중수소를 의미한다.In this specification, “hydrogen” means single hydrogen, double hydrogen, or tritium hydrogen, unless otherwise defined.

본 명세서에서 "알킬(alkyl)기"란 별도의 정의가 없는 한, 지방족 탄화수소기를 의미한다. In this specification, “alkyl group” means an aliphatic hydrocarbon group, unless otherwise defined.

알킬기는 어떠한 이중결합이나 삼중결합을 포함하고 있지 않은 "포화 알킬(saturated alkyl)기" 일 수 있다.The alkyl group may be a “saturated alkyl group” that does not contain any double or triple bonds.

알킬기는 적어도 하나의 이중결합 또는 삼중결합을 포함하고 있는 "불포화 알킬(unsaturated alkyl)기"일 수도 있다.The alkyl group may be an “unsaturated alkyl group” containing at least one double or triple bond.

포화이든 불포화이든 간에 알킬기는 분쇄형, 직쇄형 또는 환형일 수 있다.Alkyl groups, whether saturated or unsaturated, may be branched, straight-chain, or cyclic.

알킬기는 C1 내지 C30 알킬기일 수 있다. 보다 구체적으로 C1 내지 C20 알킬기, C1 내지 C10 알킬기 또는 C1 내지 C4 알킬기일 수도 있다.The alkyl group may be a C1 to C30 alkyl group. More specifically, it may be a C1 to C20 alkyl group, a C1 to C10 alkyl group, or a C1 to C4 alkyl group.

예를 들어, C1 내지 C4 알킬기는 알킬쇄에 1 내지 4 개의 탄소원자, 즉, 알킬쇄는 메틸, 에틸, 프로필, 이소-프로필, n-부틸, iso-부틸, sec-부틸 및 t-부틸로 이루어진 군에서 선택됨을 나타낸다.For example, C1 to C4 alkyl groups have 1 to 4 carbon atoms in the alkyl chain, i.e., the alkyl chain is methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and t-butyl. Indicates selection from a group consisting of

구체적인 예를 들어 상기 알킬기는 메틸기, 에틸기, 프로필기, 이소프로필기, 부틸기, 이소부틸기, t-부틸기, 펜틸기, 헥실기, 에테닐기, 프로페닐기, 부테닐기, 시클로프로필기, 시클로부틸기, 시클로펜틸기, 시클로헥실기 등을 의미한다.For specific examples, the alkyl group includes methyl group, ethyl group, propyl group, isopropyl group, butyl group, isobutyl group, t-butyl group, pentyl group, hexyl group, ethenyl group, propenyl group, butenyl group, cyclopropyl group, and cyclopropyl group. It means butyl group, cyclopentyl group, cyclohexyl group, etc.

"시클로알킬(cycloalkyl)기"는 모노시클릭 또는 융합고리 폴리시클릭(즉, 탄소원자들의 인접한 쌍들을 나눠 가지는 고리) 작용기를 포함한다.“Cycloalkyl group” includes monocyclic or fused-ring polycyclic (i.e., rings splitting adjacent pairs of carbon atoms) functional groups.

"헤테로시클로알킬(heterocycloalkyl)기"는 시클로알킬기 내에 N, O, S 및 P로 이루어진 군에서 선택되는 헤테로원자를 1 내지 4개 함유하고, 나머지는 탄소인 것을 의미한다. 상기 헤테로시클로알킬기가 융합된 고리(fused ring)인 경우, 융합된 고리 중 적어도 하나의 고리가 상기 헤테로 원자를 1 내지 4개 포함할 수 있다.“Heterocycloalkyl group” means that the cycloalkyl group contains 1 to 4 heteroatoms selected from the group consisting of N, O, S, and P, and the remainder is carbon. When the heterocycloalkyl group is a fused ring, at least one ring of the fused ring may include 1 to 4 heteroatoms.

"아릴(aryl)기"는 모노시클릭 또는 융합 고리 폴리시클릭(즉, 탄소원자들의 인접한 쌍들을 나눠 가지는 고리) 작용기를 포함한다. “Aryl groups” include monocyclic or fused ring polycyclic (i.e., rings splitting adjacent pairs of carbon atoms) functional groups.

"헤테로아릴(heteroaryl)기"는 아릴기 내에 N, O, S 및 P로 이루어진 군에서 선택되는 헤테로원자를 1 내지 4개 함유하고, 나머지는 탄소인 것을 의미한다. 상기 헤테로아릴기가 융합된 고리(fused ring)인 경우, 융합된 고리 중 적어도 하나의 고리가 상기 헤테로 원자를 1 내지 4개 포함할 수 있다. “Heteroaryl group” means that the aryl group contains 1 to 4 heteroatoms selected from the group consisting of N, O, S, and P, and the remainder is carbon. When the heteroaryl group is a fused ring, at least one ring of the fused ring may include 1 to 4 heteroatoms.

아릴기 및 헤테로아릴기에서 고리의 원자수는 탄소수 및 비탄소원자수의 합이다.In aryl groups and heteroaryl groups, the number of ring atoms is the sum of the number of carbon atoms and the number of non-carbon atoms.

본 발명은 하기 화학식 1로 표시되는 KRAS G12D 돌연변이 세포증식 억제용 화합물을 제공한다.The present invention provides a compound for inhibiting proliferation of KRAS G12D mutant cells represented by the following formula (1).

[화학식 1][Formula 1]

Figure PCTKR2024001239-appb-img-000007
Figure PCTKR2024001239-appb-img-000007

화학식 1에서,In Formula 1,

n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeating units, which is an integer from 1 to 10,

m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeat units that is an integer from 1 to 10,

X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom,

R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,

R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,

R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.

바람직하게는, 상기 화학식 1에서,Preferably, in Formula 1,

n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4,

m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4,

X1은 산소원자이고,X 1 is an oxygen atom,

R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group,

R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group,

R5

Figure PCTKR2024001239-appb-img-000008
이고, R 5 is
Figure PCTKR2024001239-appb-img-000008
ego,

X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom,

R6은 수소원자, 또는 1C 내지 10C 알킬기 일 수 있다.R 6 may be a hydrogen atom or a 1C to 10C alkyl group.

더욱 바람직하게는, 상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물일 수 있다.More preferably, the compound represented by Formula 1 may be a compound represented by Formula 2 below.

[화학식 2][Formula 2]

Figure PCTKR2024001239-appb-img-000009
Figure PCTKR2024001239-appb-img-000009

또한, 본 발명은 하기 화학식 1로 표시되는 화합물 또는 이의 염을 유효성분으로 포함하는 암질환 예방 또는 치료용 약학 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating cancer disease, comprising a compound represented by the following formula (1) or a salt thereof as an active ingredient.

[화학식 1][Formula 1]

Figure PCTKR2024001239-appb-img-000010
Figure PCTKR2024001239-appb-img-000010

화학식 1에서,In Formula 1,

n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeating units, which is an integer from 1 to 10,

m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeat units that is an integer from 1 to 10,

X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom,

R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group,

R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group,

R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.

바람직하게는, 상기 화학식 1에서,Preferably, in Formula 1,

n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4,

m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4,

X1은 산소원자이고,X 1 is an oxygen atom,

R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group,

R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group,

R5

Figure PCTKR2024001239-appb-img-000011
이고, R 5 is
Figure PCTKR2024001239-appb-img-000011
ego,

X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom,

R6은 수소원자, 또는 1C 내지 10C 알킬기 일 수 있다.R 6 may be a hydrogen atom or a 1C to 10C alkyl group.

더욱 바람직하게는, 상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물일 수 있다.More preferably, the compound represented by Formula 1 may be a compound represented by Formula 2 below.

[화학식 2][Formula 2]

Figure PCTKR2024001239-appb-img-000012
Figure PCTKR2024001239-appb-img-000012

다른 예는 상기 화학식 1로 표시되는 화합물 또는 이의 염의 약학적 유효량을 이를 필요로 하는 대상에게 투여하는 단계를 포함하는 암질환의 예방 및/또는 치료 방법을 제공한다. 상기 방법은 암질환의 예방 및/또는 치료를 필요로 하는 대상을 확인하는 단계를 추가로 포함할 수 있다.Another example provides a method for preventing and/or treating cancer disease, comprising administering a pharmaceutically effective amount of the compound represented by Formula 1 or a salt thereof to a subject in need thereof. The method may further include the step of identifying a subject in need of prevention and/or treatment of a cancer disease.

본 명세서에서 사용하는 용어 "대상", "환자", "개체" 및 "숙주" 및 이들의 변이체는 호환 가능하며 본 명세서에 기재된 화합물 또는 이의 염이나 조성물이 투여되는 임의의 포유류 대상을 지칭한다. 비제한적인 예로는 진단, 처치 또는 치료가 필요한 인간, 가축(예를 들어 개, 고양이 등), 농장 동물(예를 들어 소, 양, 돼지, 말 등) 및 실험실 동물(예를 들어 원숭이, 래트, 마우스, 토끼, 기니피그 등), 특히 인간을 포함한다. 본 명세서에 기재된 방법은 인간의 예방 또는 치료 및 수의학적 용도 모두에 적용 가능하다.As used herein, the terms “subject,” “patient,” “individual,” and “host” and their variants are interchangeable and refer to any mammalian subject to which a compound or salt or composition thereof described herein is administered. Non-limiting examples include humans, livestock (e.g. dogs, cats, etc.), farm animals (e.g. cattle, sheep, pigs, horses, etc.), and laboratory animals (e.g. monkeys, rats, etc.) in need of diagnosis, treatment or treatment. , mice, rabbits, guinea pigs, etc.), especially humans. The methods described herein are applicable to both human prophylactic or therapeutic and veterinary applications.

본 명세서에서 사용하는 문구 "필요로 하는 대상"은 본 명세서에 기재된 조성물의 투여 이익이 있을 포유류 대상과 같은 대상을 포함한다.As used herein, the phrase “subject in need” includes subjects such as mammalian subjects who would benefit from administration of the compositions described herein.

다른 예는 암질환의 치료 및/또는 예방을 위한 상기 화학식 1로 표시되는 화합물 또는 이의 염의 용도를 제공한다.Another example provides the use of the compound represented by Formula 1 or a salt thereof for the treatment and/or prevention of cancer diseases.

상기 암질환 예방 또는 치료용 약학 조성물은 KRAS G12D 돌연변이 세포증식 억제용일 수 있다. The pharmaceutical composition for preventing or treating cancer disease may be used to inhibit proliferation of KRAS G12D mutant cells.

상기 KRAS G12D 돌연변이 세포는 LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D 및 LS174T KRAS G12D 중에서 선택된 1종 이상일 수 있다. 여기서, LIM1215 KRAS G12D, SW48 KRAS G12D, LS174T KRAS G12D은 대장암 세포주이고, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D은 폐암 세포주이다.The KRAS G12D mutant cells may be one or more types selected from LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D, and LS174T KRAS G12D. Here, LIM1215 KRAS G12D, SW48 KRAS G12D, and LS174T KRAS G12D are colon cancer cell lines, and NCI-H1975 KRAS G12D and NCI-H838 KRAS G12D are lung cancer cell lines.

상기 암질환 예방 또는 치료용 조성물은 상기 KRAS G12D 돌연변이 세포에 대한 표적 치료용으로 사용될 수 있다. 왜냐하면, KRAS 야생형 세포에는 세포증식 억제 효과가 거의 나타나지 않지만, KRAS G12D 돌연변이 세포에 대해서는 선택적으로 증식억제 효능을 나타낼 수 있기 때문이다.The composition for preventing or treating cancer disease can be used for targeted treatment against the KRAS G12D mutant cells. This is because, although it has little cytostatic effect on KRAS wild-type cells, it can selectively exhibit anti-proliferation effect on KRAS G12D mutant cells.

상기 암질환 예방 또는 치료용 조성물은 MAPK 신호전달 경로 억제용으로 사용될 수 있다.The composition for preventing or treating cancer diseases can be used to inhibit the MAPK signaling pathway.

상기 MAPK 신호전달 경로 억제는 RAS-RAF-MEK-ERK 경로에서 C-RAF 인산화 억제에 따라 수행될 수 있다.Inhibition of the MAPK signaling pathway can be performed by inhibiting C-RAF phosphorylation in the RAS-RAF-MEK-ERK pathway.

또한, 상기 암질환 예방 또는 치료용 조성물은 PI3K 신호전달 경로 억제용으로 사용될 수 있다.Additionally, the composition for preventing or treating cancer diseases can be used to inhibit the PI3K signaling pathway.

상기 PI3K 신호전달 경로 억제는 PI3K-AKT-mTOR 경로에서 AKT 인산화 억제에 따라 수행될 수 있다.The inhibition of the PI3K signaling pathway can be performed by inhibiting AKT phosphorylation in the PI3K-AKT-mTOR pathway.

상기 암질환은 대장암, 결장암, 직장암, 폐암, 흑색종, 갑상선암, 자궁암, 난소암, 자궁경부함, 췌장암, 위암 및 간암 중에서 선택된 어느 하나일 수 있으나, 본 발명이 범위가 여기에 한정되지 않으며 KRAS G12D 돌연변이를 일으킬 수 있는 암종은 모두 가능하다.The cancer disease may be any one selected from colon cancer, colon cancer, rectal cancer, lung cancer, melanoma, thyroid cancer, uterine cancer, ovarian cancer, cervix, pancreas cancer, stomach cancer, and liver cancer, but the scope of the present invention is not limited thereto and KRAS Any carcinoma that can cause a G12D mutation is possible.

한편, 본 명세서에서 용어 ‘유효성분으로 포함하는’이란 상기 화학식 1로 표시되는 화합물 또는 이의 염의 효능 또는 활성을 달성하는 데 충분한 양을 포함하는 것을 의미한다. 본 발명의 한 구체예에서, 본 발명의 조성물 내에서 화학식 1로 표시되는 화합물 또는 이의 염은 예를 들어, 0.001 mg/kg 이상, 바람직하게는 0.1 mg/kg 이상, 보다 바람직하게는 10 mg/kg 이상, 보다 더 바람직하게는 100 mg/kg 이상, 보다 더욱 더 바람직하게는 250 mg/kg 이상, 가장 바람직하게는 1 g/kg 이상 포함될 수 있다.Meanwhile, in this specification, the term ‘including as an active ingredient’ means containing a sufficient amount to achieve the efficacy or activity of the compound represented by Formula 1 or a salt thereof. In one embodiment of the present invention, the compound represented by Formula 1 or its salt in the composition of the present invention is, for example, 0.001 mg/kg or more, preferably 0.1 mg/kg or more, more preferably 10 mg/kg. It may contain more than kg, more preferably more than 100 mg/kg, even more preferably more than 250 mg/kg, and most preferably more than 1 g/kg.

화학식 1로 표시되는 화합물 또는 이의 염의 양적 하한 및/또는 상한은 당업자가 적절한 범위 내에서 선택하여 실시할 수 있다.The quantitative lower limit and/or upper limit of the compound represented by Formula 1 or its salt can be selected within an appropriate range by a person skilled in the art.

본 명세서에서 용어, "염", "약학적 염" 또는 "약학적으로 허용가능한 염"은, 화합물이 투여되는 유기체에 심각한 자극을 유발하지 않고 화합물의 생물학적 활성과 물성들을 손상시키지 않는, 화합물의 제형을 의미한다. 상기 약학적 염은, 본 발명의 화합물을, 염산, 브롬산, 황산, 질산, 인산 등의 무기산, 메탄술폰산, 에탄술폰산, p-톨루엔술폰산 등의 술폰산, 타타르산, 포름산, 시트르산, 아세트산, 트리클로로아세트산, 트리플루오로아세트산, 카프릭산, 이소부탄산, 말론산, 숙신산, 프탈산, 글루콘산, 벤조산, 락트산, 푸마르산, 말레인산, 살리실산 등과 같은 유기 카본산과 반응시켜 얻어질 수 있다. 또한, 본 발명의 화합물을 염기와 반응시켜, 암모니움 염, 나트륨 또는 칼륨염 등의 알칼리 금속염, 칼슘 또는 마그네슘염 등의 알칼리 토금속염 등의 염, 디시클로헥실아민, N-메틸-D-글루카민, 트리스(히드록시메틸) 메틸아민 등의 유기염기들의 염, 및 아르기닌, 리신 등의 아미노산 염을 형성함으로써 얻어질 수도 있으며, 이에 제한되지 않는다.As used herein, the term "salt", "pharmaceutical salt" or "pharmaceutically acceptable salt" refers to a compound that does not cause significant irritation to the organism to which the compound is administered and does not impair the biological activity and physical properties of the compound. It means dosage form. The pharmaceutical salts include the compounds of the present invention, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid, sulfonic acids such as methanesulfonic acid, ethanesulfonic acid, and p-toluenesulfonic acid, tartaric acid, formic acid, citric acid, acetic acid, and trichloroacid. It can be obtained by reacting with organic carboxylic acids such as loacetic acid, trifluoroacetic acid, capric acid, isobutanoic acid, malonic acid, succinic acid, phthalic acid, gluconic acid, benzoic acid, lactic acid, fumaric acid, maleic acid, salicylic acid, etc. In addition, the compound of the present invention can be reacted with a base to produce salts such as alkali metal salts such as ammonium salts, sodium or potassium salts, alkaline earth metal salts such as calcium or magnesium salts, dicyclohexylamine, and N-methyl-D-glue. It may be obtained by forming salts of organic bases such as carmine, tris(hydroxymethyl) methylamine, and amino acid salts such as arginine and lysine, but is not limited thereto.

본 발명의 약학 조성물은 상기 유효 성분 이외에 약학적으로 적합하고 생리학적으로 허용되는 보조제를 사용하여 제조될 수 있으며, 상기 보조제로는 부형제, 붕해제, 감미제, 결합제, 피복제, 팽창제, 윤활제, 활택제 또는 향미제 등을 사용할 수 있다.The pharmaceutical composition of the present invention can be prepared using pharmaceutically suitable and physiologically acceptable auxiliaries in addition to the active ingredients, and the auxiliaries include excipients, disintegrants, sweeteners, binders, coating agents, swelling agents, lubricants, and lubricants. Agents or flavoring agents can be used.

상기 약학 조성물은 투여를 위해서 상기 기재한 유효 성분 이외에 추가로 약학적으로 허용 가능한 담체를 1종 이상 포함하여 약학 조성물로 바람직하게 제제화할 수 있다.For administration, the pharmaceutical composition may be preferably formulated as a pharmaceutical composition containing one or more pharmaceutically acceptable carriers in addition to the active ingredients described above.

상기 약학 조성물의 제제 형태는 과립제, 산제, 정제, 피복정, 캡슐제, 좌제, 액제, 시럽, 즙, 현탁제, 유제, 점적제 또는 주사 가능한 액제 등이 될 수 있다. 예를 들어, 정제 또는 캡슐제의 형태로의 제제화를 위해, 유효 성분은 에탄올, 글리세롤, 물 등과 같은 경구, 무독성의 약학적으로 허용 가능한 불활성 담체와 결합될 수 있다. 또한, 원하거나 필요한 경우, 적합한 결합제, 윤활제, 붕해제 및 발색제 또한 혼합물로 포함될 수 있다. 적합한 결합제는 이에 제한되는 것은 아니나, 녹말, 젤라틴, 글루코스 또는 베타-락토오스와 같은 천연 당, 옥수수 감미제, 아카시아, 트래커캔스 또는 소듐올레이트와 같은 천연 및 합성 검, 소듐 스테아레이트, 마그네슘 스테아레이트, 소듐 벤조에이트, 소듐 아세테이트, 소듐 클로라이드 등을 포함한다. 붕해제는 이에 제한되는 것은 아니나, 녹말, 메틸 셀룰로스, 아가, 벤토니트, 잔탄 검 등을 포함한다.The pharmaceutical composition may be in the form of granules, powders, tablets, coated tablets, capsules, suppositories, solutions, syrups, juices, suspensions, emulsions, drops, or injectable solutions. For example, for formulation in the form of tablets or capsules, the active ingredient may be combined with an oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, etc. Additionally, if desired or necessary, suitable binders, lubricants, disintegrants and coloring agents may also be included in the mixture. Suitable binders include, but are not limited to, starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tracacance or sodium oleate, sodium stearate, magnesium stearate, sodium Includes benzoate, sodium acetate, sodium chloride, etc. Disintegrants include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, etc.

액상 용액으로 제제화되는 조성물에 있어서 허용 가능한 약학 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다.Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and these. One or more of the ingredients can be mixed and used, and other common additives such as antioxidants, buffers, and bacteriostatic agents can be added as needed. In addition, diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, or tablets.

본 발명의 약학 조성물은 경구 또는 비경구로 투여할 수 있고, 비경구 투여인 경우에는 정맥 내 주입, 피하 주입, 근육 주입, 복강 주입, 경피 투여 등으로 투여할 수 있으며, 바람직하게는 비경구 투여이다.The pharmaceutical composition of the present invention can be administered orally or parenterally, and in the case of parenteral administration, it can be administered by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, transdermal administration, etc., and is preferably parenteral administration. .

본 발명의 약학 조성물의 적합한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하며, 보통으로 숙련된 의사는 소망하는 치료 또는 예방에 효과적인 투여량을 용이하게 결정 및 처방할 수 있다. 본 발명의 바람직한 구현예에 따르면, 본 발명의 약학 조성물의 1일 투여량은 0.001-10 g/㎏이다.The appropriate dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration method, patient's age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity, and is usually A skilled doctor can easily determine and prescribe an effective dosage for desired treatment or prevention. According to a preferred embodiment of the present invention, the daily dosage of the pharmaceutical composition of the present invention is 0.001-10 g/kg.

본 발명의 약학 조성물은 당해 발명이 속하는 기술분야에서 통상의 지식을 가진 자가 용이하게 실시할 수 있는 방법에 따라, 약학적으로 허용되는 담체 및/또는 부형제를 이용하여 제제화 함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기 내에 내입시켜 제조될 수 있다. 이때 제형은 오일 또는 수성 매질중의 용액, 현탁액 또는 유화액 형태이거나 엑스제, 분말제, 과립제, 정제 또는 캅셀제 형태일 수도 있으며, 분산제 또는 안정화제를 추가적으로 포함할 수 있다.The pharmaceutical composition of the present invention is manufactured in unit dosage form by formulating using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person skilled in the art. Alternatively, it can be manufactured by placing it in a multi-capacity container. At this time, the formulation may be in the form of a solution, suspension, or emulsion in an oil or aqueous medium, or may be in the form of an extract, powder, granule, tablet, or capsule, and may additionally contain a dispersant or stabilizer.

이하, 본 발명이 구체적인 실시예를 들어 설명하도록 한다.Hereinafter, the present invention will be described with reference to specific examples.

[실시예][Example]

재료 준비Ingredients Preparation

소분자 퀴나졸린(Q2a) 화합물은 KIST(한국과학기술연구원)로부터 입수하였다. KRAS 돌연변이 세포는 Horizon Discovery에서 구입하였다. PARP, C-RAF, P-CRAF, AKT, P-AKT, ERK, P-ERK, HSP90, C-MYC, NOTCH1, β-actin 항체는 Cell Signaling Technology에서 구입하였다. 2차 항체 항-래빗 Ig 홀스래디쉬 퍼옥시다제(HRP) 및 항-마우스 Ig 홀스래디쉬 퍼옥시다제 (HRP)는 GeneTex에서 구입하였고, 세포증식 측정용 EZ-Cytox는 Dogen Bio에서 구입하였다. 세포사멸 측정용 ANNEXIN V-FITC 세포사멸 검출 키트는 Koma Biotech 에서 구입하였다. 활성 RAS 및 검출 키트 풀다운을 위한 Protein Thermal Shift™ 염료 키트와 Raf-1-RBD 비드는 Thermo Fisher Scientific and Cell Signaling Technology에서 구입하여 사용하였다.The small molecule quinazoline (Q2a) compound was obtained from KIST (Korea Institute of Science and Technology). KRAS mutant cells were purchased from Horizon Discovery. PARP, C-RAF, P-CRAF, AKT, P-AKT, ERK, P-ERK, HSP90, C-MYC, NOTCH1, and β-actin antibodies were purchased from Cell Signaling Technology. Secondary antibodies anti-rabbit Ig horseradish peroxidase (HRP) and anti-mouse Ig horseradish peroxidase (HRP) were purchased from GeneTex, and EZ-Cytox for cell proliferation measurement was purchased from Dogen Bio. ANNEXIN V-FITC apoptosis detection kit for measuring apoptosis was purchased from Koma Biotech. Protein Thermal Shift™ dye kit and Raf-1-RBD beads for pulling down active RAS and detection kit were purchased from Thermo Fisher Scientific and Cell Signaling Technology.

이하, 화학식 2로 표시되는 화합물 Q2a의 NMR 분석 결과를 나타내었다.Below, the NMR analysis results of compound Q2a represented by Formula 2 are shown.

[화학식 2][Formula 2]

Figure PCTKR2024001239-appb-img-000013
Figure PCTKR2024001239-appb-img-000013

1H NMR (500 MHz, MeOD) δ 8.04 (dt, J = 8.4, 1.8 Hz, 1H), 7.87 (s, 5H), 7.68 (t, J = 7.7 Hz, 1H), 7.32 (t, J = 7.8 Hz, 1H), 7.24 - 7.14 (m, 2H), 6.90 (d, J = 8.2 Hz, 1H), 6.81 (t, J = 7.4 Hz, 1H), 6.09 (d, J = 5.9 Hz, 2H), 4.77 (s, 2H), 4.72 (s, 1H), 4.70 (s, 4H), 3.78 (d, J = 1.0 Hz, 3H), 3.62 (p, J = 6.7 Hz, 2H), 3.39 - 3.33 (m, 2H), 3.16 - 3.08 (m, 2H), 2.56 (s, 1H). 1H NMR (500 MHz, MeOD) δ 8.04 (dt, J = 8.4, 1.8 Hz, 1H), 7.87 (s, 5H), 7.68 (t, J = 7.7 Hz, 1H), 7.32 (t, J = 7.8 Hz, 1H), 7.24 - 7.14 (m, 2H), 6.90 (d, J = 8.2 Hz, 1H), 6.81 (t, J = 7.4 Hz, 1H), 6.09 (d, J = 5.9 Hz, 2H), 4.77 (s, 2H), 4.72 (s, 1H), 4.70 (s, 4H), 3.78 (d, J = 1.0 Hz, 3H), 3.62 (p, J = 6.7 Hz, 2H), 3.39 - 3.33 (m , 2H), 3.16 - 3.08 (m, 2H), 2.56 (s, 1H).

13C NMR (126 MHz, MeOD) δ 167.28, 163.52, 160.39, 157.57, 153.40, 139.03, 135.09, 130.50, 129.11, 128.72, 125.67, 124.84, 124.61, 123.42, 120.11, 116.47, 110.30, 54.48, 42.43, 35.59, 30.27. 13 C NMR (126 MHz, MeOD) δ 167.28, 163.52, 160.39, 157.57, 153.40, 139.03, 135.09, 130.50, 129.11, 128.72, 125.67, 124.84, 124.61, 123.42, 120.11, 116.47, 110.30, 54.48, 42.43, 35.59, 30.27.

세포주 및 세포배양Cell lines and cell culture

야생형 암세포와 KRAS G12D 돌연변이 암세포주는 Horizon Discovery에서 구입한 것을 사용하였다. 대장 세포주 LIM1215, SW48, 및 NSCLC(비소세포폐암) 세포주 NCI-H1975, NCI-H838을 10% FBS 및 1% 페니실린이 보충된 RPMI-1640 배지에서 배양하였다. 또한, COLO205, LOVO, HCT8, HCT15, SW480, SW620도 10% FBS와 1% 페니실린이 첨가된 RPMI-1640 배지에서 배양하였다. 모든 세포는 37℃의 5% CO2 인큐베이터에서 배양되었다.Wild-type cancer cells and KRAS G12D mutant cancer cell lines purchased from Horizon Discovery were used. Colon cell lines LIM1215 and SW48, and non-small cell lung cancer (NSCLC) cell lines NCI-H1975 and NCI-H838 were cultured in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin. Additionally, COLO205, LOVO, HCT8, HCT15, SW480, and SW620 were cultured in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin. All cells were cultured in a 5% CO 2 incubator at 37°C.

웨스턴 블롯에 의한 단백질 분석Protein analysis by Western blot

세포를 37℃의 5% CO2 인큐베이터에서 밤새 60 mm2 세포배양접시에 분주하였다. 화합물은 0μM, 1μM, 5μM, 10μM로 농도로 처리되었다. 트립신 EDTA를 사용하여 세포를 수득한 후 세포를 1500rpm에서 5분간 원심분리하여 상층액을 제거하고, 차가운 PBS로 1500rpm에서 5분간 세척하였다. 상층액을 제거한 후, 2x 샘플 완충액(100㎕)을 사용하여 세포를 용해시켰다. 히팅 블록을 사용하여 5~7분간 가열한 후, BCA 단백질 분석 키트(Thermal Fisher Scientific)를 사용하여 정량하였다. 동일한 양의 단백질 샘플을 SDS-PAGE로 분리하고 전기 블로팅 장치(Bio-Rad)를 사용하여 PVDF 막으로 전이시켰다. 막을 블로킹 완충액(TBS-T + 5% 탈지유)에서 1시간 동안 블로킹하고 1차 항체와 함께 밤새 4℃에서 배양하였다. 막을 TBS-T로 세척하고 홀스래디쉬 퍼옥시다제가 결합된 2차 항체와 함께 실온에서 1시간 동안 배양하였다. ECL 키트(Amersham Biosciences)를 사용하여 단백질 발현을 시각화하였다.Cells were seeded in a 60 mm 2 cell culture dish overnight in a 5% CO 2 incubator at 37°C. Compounds were treated at concentrations of 0 μM, 1 μM, 5 μM, and 10 μM. After obtaining the cells using trypsin-EDTA, the cells were centrifuged at 1500 rpm for 5 minutes to remove the supernatant, and washed with cold PBS for 5 minutes at 1500 rpm. After removing the supernatant, cells were lysed using 2x sample buffer (100 μl). After heating for 5 to 7 minutes using a heating block, it was quantified using a BCA protein analysis kit (Thermal Fisher Scientific). Equal amounts of protein samples were separated by SDS-PAGE and transferred to PVDF membranes using an electroblotting device (Bio-Rad). Membranes were blocked in blocking buffer (TBS-T + 5% skim milk) for 1 h and incubated with primary antibodies overnight at 4°C. The membrane was washed with TBS-T and incubated with horseradish peroxidase-conjugated secondary antibody for 1 hour at room temperature. Protein expression was visualized using the ECL kit (Amersham Biosciences).

세포증식 분석Cell proliferation analysis

KRAS G12D 돌연변이 암세포와 야생형 암세포를 96웰 플레이트에 웰당 5 Х 103 내지 1 Х 104 개 세포로 분주하고 밤새 성장시켰다. 이후, 세포에 퀴나졸린 유도체 Q2a 화합물을 0μM, 1μM, 5μM, 10μM 농도로 처리하였다. 화합물 처리 후 Ez-cytox를 이용한 WST 분석을 수행하여 세포 증식을 측정하였다. 10% Ez-cytox 처리 후, 37℃, 5% CO2 인큐베이터에서 2시간 동안 반응시켰다. 마이크로플레이트 리더를 사용하여 490-500 nm에서 흡광도를 측정하여 광학 밀도(OD) 값을 측정하였다.KRAS G12D mutant cancer cells and wild-type cancer cells were seeded in a 96-well plate at 5 Х 10 3 to 1 Х 10 4 cells per well and grown overnight. Afterwards, the cells were treated with quinazoline derivative Q2a compound at concentrations of 0μM, 1μM, 5μM, and 10μM. After compound treatment, WST analysis using Ez-cytox was performed to measure cell proliferation. After treatment with 10% Ez-cytox, the reaction was performed in a 5% CO 2 incubator at 37°C for 2 hours. Optical density (OD) values were determined by measuring absorbance at 490-500 nm using a microplate reader.

클론생성 분석Cloning analysis

세포를 6웰 플레이트에 웰당 1 Х 103개 세포로 분주하고 밤새 성장시켰다. 세포에 Q2a 화합물을 0μM, 1μM, 5μM, 10μM 농도로 처리하였고, 배지는 3일마다 Q2a 화합물을 함유한 새로운 배지로 교체하였다. Q2a 화합물 처리 21일 후, 배지를 제거하고 차가운 PBS로 세척하였다. 세포 콜로니를 메탄올로 10분 동안 고정하고 크리스탈 바이올렛(20% 메탄올 중 0.1%)으로 20분 동안 염색하였다. 염색이 끝나면 증류수로 조심스럽게 세척하였다. 적어도 3회 이상의 독립적인 실험 후에 생존한 콜로니의 수를 계산하고, 생존한 콜로니의 수(SF)를 하기 식 1에 따라 계산하였다.Cells were seeded in 6-well plates at 1 Х 10 3 cells per well and grown overnight. Cells were treated with Q2a compound at concentrations of 0 μM, 1 μM, 5 μM, and 10 μM, and the medium was replaced with new medium containing Q2a compound every 3 days. After 21 days of Q2a compound treatment, the medium was removed and washed with cold PBS. Cell colonies were fixed with methanol for 10 min and stained with crystal violet (0.1% in 20% methanol) for 20 min. After staining, it was carefully washed with distilled water. After at least three independent experiments, the number of surviving colonies was calculated, and the number of surviving colonies (SF) was calculated according to Equation 1 below.

[식 1][Equation 1]

SF = 처리된 콜로니 수/대조군 콜로니의 수SF = number of colonies treated/number of control colonies

세포사멸 분석Apoptosis analysis

KRAS G12D 돌연변이 세포에서 Q2a 화합물 처리시 세포사멸을 측정하기 위해 형광 활성화 세포 분류(FACS)를 수행하였다. KRAS G12D 돌연변이 암세포와 야생형 암세포를 6웰 플레이트에 웰당 1 Х 105 내지 1.5 Х 105개 세포를 분주하고 밤새 성장시켰다. 이후, 세포에 Q2a 화합물을 0μM, 1μM, 5μM, 10μM 농도로 처리하고, 키트 프로토콜에 따라 수행하였다. 트립신 EDTA를 사용하여 세포를 수득하고 세포를 1000 x g에서 5분간 원심분리하여 상층액을 제거하고, 차가운 PBS로 1000 x g에서 5분간 세척하였다. 상층액을 제거한 후 1x 결합 완충액 500㎕를 첨가하고 현탁시켰다. 세포에 Annexin V 1.25㎕(5㎍/㎖)을 첨가하고 실온 암실에서 15분간 염색하였다. 15분 후 1000 x g로 5분간 원심분리하고 상층액을 제거하였다. 1 x 결합 완충액 500㎕를 첨가하고 PI 10㎕를 첨가한 후, 즉시 유세포 분석기(Beckman cytoflex)를 사용하여 세포 사멸을 측정하였다. 생존 세포 또는 사멸 세포의 백분율은 형광-활성화 세포 분류기(FACS) 분석에 의해 분석하였다.Fluorescence-activated cell sorting (FACS) was performed to measure apoptosis in KRAS G12D mutant cells upon treatment with Q2a compound. KRAS G12D mutant cancer cells and wild-type cancer cells were seeded at 1 Х 10 5 to 1.5 Х 10 5 cells per well in a 6-well plate and grown overnight. Afterwards, the cells were treated with Q2a compound at concentrations of 0 μM, 1 μM, 5 μM, and 10 μM, and performed according to the kit protocol. Cells were obtained using trypsin EDTA, centrifuged at 1000 xg for 5 minutes to remove the supernatant, and washed with cold PBS at 1000 xg for 5 minutes. After removing the supernatant, 500 μl of 1x binding buffer was added and suspended. 1.25㎕ (5㎍/㎖) of Annexin V was added to the cells and stained for 15 minutes at room temperature in the dark. After 15 minutes, it was centrifuged at 1000 xg for 5 minutes and the supernatant was removed. After adding 500 μl of 1×binding buffer and 10 μl of PI, cell death was immediately measured using a flow cytometer (Beckman cytoflex). The percentage of viable or dead cells was analyzed by fluorescence-activated cell sorter (FACS) analysis.

세포 내 KRAS 활성 측정Measurement of intracellular KRAS activity

LIM1215 KRAS G12D 돌연변이 세포주를 37℃의 5% CO2 인큐베이터에서 밤새 60mm2 세포배양접시에 분주하였다. Q2a 화합물은 0μM, 1μM, 5μM, 10μM 농도로 처리하였다. KRAS 단백질의 세포 활성은 활성 RAS pull-down and detection kit를 사용하여 제조업체의 지침(Cell Signaling Technology)에 따라 측정하였다. GST-Raf-1 RBD 비드를 사용하여 GTP-결합 형태의 KRAS를 세포 용해물로부터 추출하였다. 세척 후 시료에 로딩 완충액을 첨가하고 5분간 끓인 후 SDS-PAGE와 KRAS 항체를 이용한 웨스턴 블롯팅을 수행하였다.The LIM1215 KRAS G12D mutant cell line was seeded in a 60 mm 2 cell culture dish overnight in a 5% CO 2 incubator at 37°C. Q2a compound was treated at concentrations of 0μM, 1μM, 5μM, and 10μM. Cellular activity of KRAS protein was measured using the active RAS pull-down and detection kit according to the manufacturer's instructions (Cell Signaling Technology). GTP-bound form of KRAS was extracted from cell lysates using GST-Raf-1 RBD beads. After washing, loading buffer was added to the sample, boiled for 5 minutes, and then SDS-PAGE and Western blotting using KRAS antibody were performed.

대장암 이종이식(xenograft) 및 치료Colon cancer xenograft and treatment

6주령 암컷 BALB/C 누드마우스를 나라바이오텍에서 구입하였다. 3×106 LIM1215 야생형 및 KRAS G12D 돌연변이 세포를 누드 마우스의 양쪽 옆구리 부위의 피하 조직에 투여하였다. 치료를 시작하기 전에 종양이 약 100mm3의 평균 부피로 성장하도록 하고, 마우스를 복강 내 투여(i.p.)에 따라 Q2a 화합물로 치료하였다. 치료 기간 동안, 종양 부피(V)를 2일 마다 한 번씩 캘리퍼로 측정하고 다음 식 2에 따라 계산하였다.Six-week-old female BALB/C nude mice were purchased from Nara Biotech. 3×10 6 LIM1215 wild type and KRAS G12D mutant cells were administered to the subcutaneous tissue of both flanks of nude mice. Tumors were allowed to grow to an average volume of approximately 100 mm 3 before starting treatment, and mice were treated with compound Q2a by intraperitoneal administration (ip). During the treatment period, tumor volume (V) was measured with calipers once every 2 days and calculated according to the following equation 2.

[식 2][Equation 2]

종양 부피(V) = ( L × S2 ) / 2 (L은 장축, S는 단축)Tumor volume (V) = ( L × S 2 ) / 2 (L is long axis, S is short axis)

열이동 분석을 통한 Q2a 화합물 KRAS 결합 측정Measurement of KRAS binding of Q2a compound through thermal shift analysis

열 이동 분석(Thermal shift analysis)은 Protein Thermal Shift 염료 키트(Thermo Fisher Scientific)를 사용하여 수행하였다. 대장암 세포인 LIM1215 세포 야생형과 KRAS G12D 돌연변이 세포에 Q2a 화합물을 72시간 처리한 후 1x 리파 완충액(lipa buffer)을 이용하여 단백질 추출을 수행하였다. 추출된 단백질은 BCA 분석을 통해 1㎍으로 정량하고 Thermal Shift 염료가 포함된 Protein Thermal Shift 완충액을 사용하여 단백질과 반응시켰다. 이후, 실시간 PCR을 이용하여 Tm(녹는점)을 측정하였다.Thermal shift analysis was performed using the Protein Thermal Shift dye kit (Thermo Fisher Scientific). Colon cancer cells, LIM1215 wild type cells and KRAS G12D mutant cells, were treated with Q2a compound for 72 hours, and then protein extraction was performed using 1x lipa buffer. The extracted protein was quantified at 1㎍ through BCA analysis and reacted with the protein using Protein Thermal Shift buffer containing thermal shift dye. Afterwards, Tm (melting point) was measured using real-time PCR.

통계 분석statistical analysis

모든 데이터는 최소 세 번의 독립적인 실험에서 평균 ± 표준 편차(SD)로 표시하였다. 통계분석은 GraphPad InStat 6 소프트웨어를 사용하여 수행하였다. 그룹 간 비교를 위해 일원분산분석(one-way ANOVA)과 tukey's post-hoc test 방법을 사용하였다. 또한, 그룹 간 유의성 평가로는 t-test 방법을 사용하였다. p-값의 유의성은 *p < 0.05, **p < 0.01, ***p < 0.001로 평가하였다.All data are expressed as mean ± standard deviation (SD) from at least three independent experiments. Statistical analysis was performed using GraphPad InStat 6 software. For comparison between groups, one-way ANOVA and tukey's post-hoc test methods were used. Additionally, the t-test method was used to evaluate significance between groups. The significance of the p-value was evaluated as *p < 0.05, **p < 0.01, and ***p < 0.001.

[실험예][Experimental example]

실험예 1: KRAS G12D를 표적으로 하는 소분자의 스크리닝Experimental Example 1: Screening of small molecules targeting KRAS G12D

KRAS 돌연변이 세포에 반응하는 화합물을 찾기 위해 Q1a ~ Q11a까지 총 11개의 화합물을 대장암 세포주인 LIM1215의 야생형 세포와 KRAS 돌연변이 세포에 각각 5 μM의 농도로 처리하였고, 세포증식은 72시간 후에 측정하였다. 세포 성장을 측정하는 방법은 WST-1 분석으로 하였다. 화합물 처리 72시간 후 Ez-cytox 시약을 첨가하고 2시간 반응 후 마이크로플레이트 리더를 이용하여 세포 성장을 측정하였고, 그 결과를 도 1에 나타내었다. 이에 따르면, 대부분의 화합물은 야생형보다 KRAS 돌연변이에서 15% 이상의 평균 세포 증식 억제를 나타냈다. To find compounds that react to KRAS mutant cells, a total of 11 compounds from Q1a to Q11a were treated at a concentration of 5 μM each in wild-type cells and KRAS mutant cells of the colon cancer cell line LIM1215, and cell proliferation was measured after 72 hours. The method for measuring cell growth was WST-1 analysis. After 72 hours of compound treatment, Ez-cytox reagent was added, and after 2 hours of reaction, cell growth was measured using a microplate reader, and the results are shown in Figure 1. According to this, most compounds showed an average inhibition of cell proliferation of more than 15% in KRAS mutants compared to the wild type.

세포성장 억제 효과가 우수한 일부 화합물을 선택하여 형광 활성화 세포 분류(FACS)로 세포사멸 정도를 다시 측정하였다. FACS에서는 WST-1에서와 같이 5μM의 화합물을 처리한 후 72시간 후에 Annexin V와 PI(Propidium Iodide) 염색을 사용하여 세포사멸을 측정하였다. 이들 중 Q2a의 세포사멸 효과는 다른 화합물에 비해 약 3% 더 높았으며, 세포사멸의 지표인 PARP의 단백질 발현도 다른 화합물에 비해 증가하는 것으로 나타났다. 따라서 최종적으로 11개 화합물 중에서 Q2a 화합물을 선택하여 실험을 진행하였다. Q2a 화합물의 분자 구조와 3차원 모델 구조를 도 2에 나타내었다. Some compounds with excellent cell growth inhibition effects were selected and the degree of cell death was measured again using fluorescence-activated cell sorting (FACS). In FACS, apoptosis was measured using Annexin V and PI (Propidium Iodide) staining 72 hours after treatment with 5 μM of the compound as in WST-1. Among these, the apoptosis effect of Q2a was about 3% higher than that of other compounds, and the protein expression of PARP, an indicator of apoptosis, was also found to increase compared to other compounds. Therefore, compound Q2a was finally selected among 11 compounds and the experiment was conducted. The molecular structure and three-dimensional model structure of the Q2a compound are shown in Figure 2.

Q2a 화합물이 KRAS에 결합되었는지 확인하기 위해 단백질 열 이동(PTS) 분석을 수행하였다. 대장암 세포주인 LIM1215 야생형 세포와 KRAS 돌연변이 세포에 각각 5 μM의 Q2a 화합물을 처리한 후 72시간 후에 Protein Thermal Shift 키트를 이용하여 Tm(녹는점)을 측정하고, 그 결과를 도 3에 나타내었다. 이에 따르면, 야생형(WT) 대조군의 Tm 값은 60.81℃, 야생형의 Q2a 화합물 처리군의 Tm 값은 58.31℃였으나, KRAS G12D 돌연변이군의 Tm 값은 55.41℃, KRAS G12D 돌연변이의 Q2a 화합물 처리군의 Tm 값은 약 3.51℃ 증가하는 것으로 나타났다. 이와 같은 결과에 따라 Q2a 화합물이 KRAS에 결합한다는 것을 확인할 수 있었다.Protein thermal shift (PTS) analysis was performed to confirm that the Q2a compound bound to KRAS. 72 hours after treating colon cancer cell lines LIM1215 wild-type cells and KRAS mutant cells each with 5 μM of Q2a compound, Tm (melting point) was measured using a Protein Thermal Shift kit, and the results are shown in Figure 3. According to this, the Tm value of the wild type (WT) control group was 60.81℃, the Tm value of the wild type Q2a compound treatment group was 58.31℃, the Tm value of the KRAS G12D mutant group was 55.41℃, and the Tm value of the KRAS G12D mutant Q2a compound treatment group was 58.31℃. The value was found to increase by about 3.51℃. According to these results, it was confirmed that compound Q2a binds to KRAS.

이후, Q2a 화합물이 KRAS G12D 돌연변이에서만 특이적 활성을 갖는지 확인하기 위해 KRAS G12D 이외의 다른 돌연변이 세포에 Q2a 화합물을 처리한 후 세포 성장을 측정하였고, 그 결과를 도 4에 나타내었다. 구체적으로, 대장암 세포주로 야생형 COLO205, KRAS-G12D 돌연변이인 LS174T 및 SNUC2A, KRAS-G12V 돌연변이인 SW620 및 SW480, KRAS-G13D 돌연변이인 LOVO 및 HCT15 세포를 대상으로 실험을 수행하였다. 각 세포에 5μM의 Q2a 화합물을 처리한 후 48시간 후와 72시간 후 Ez-cytox를 이용하여 세포 성장을 측정하였다. 그 결과, 야생형인 KRAS G12V와 KRAS G13D 돌연변이체는 별 다른 효과가 나타나지 않았으나, KRAS G12D 돌연변이체 세포인 LS174T와 LOVO에서는 세포 성장 억제 효과가 나타났다. 즉, Q2a 화합물이 KRAS G12D 돌연변이에서만 특이적 활성을 나타냄을 확인할 수 있다.Thereafter, in order to confirm whether the Q2a compound had specific activity only in the KRAS G12D mutant, mutant cells other than KRAS G12D were treated with the Q2a compound and cell growth was measured, and the results are shown in Figure 4. Specifically, experiments were performed on colon cancer cell lines such as wild-type COLO205, KRAS-G12D mutant LS174T and SNUC2A, KRAS-G12V mutant SW620 and SW480, and KRAS-G13D mutant LOVO and HCT15 cells. After treating each cell with 5 μM of Q2a compound, cell growth was measured using Ez-cytox 48 and 72 hours later. As a result, the wild type KRAS G12V and KRAS G13D mutants did not show any significant effect, but the KRAS G12D mutant cells LS174T and LOVO showed a cell growth inhibitory effect. In other words, it can be confirmed that the Q2a compound exhibits specific activity only in the KRAS G12D mutant.

실험예 2: KRAS G12D 돌연변이 세포와 대장암 및 폐암 야생형 세포의 성장억제 효과 분석Experimental Example 2: Analysis of growth inhibition effect of KRAS G12D mutant cells and wild-type colon cancer and lung cancer cells

선별된 Q2a 화합물에 대하여, 동일한 세포주에서 한 쌍의 야생형 및 KRAS G12D 돌연변이 세포의 Q2a 처리에 따른 세포성장 억제 정도를 비교하기 위해 WST-1 분석을 수행하였다.For the selected Q2a compound, WST-1 analysis was performed to compare the degree of cell growth inhibition following Q2a treatment of a pair of wild-type and KRAS G12D mutant cells in the same cell line.

대장암 세포주인 LIM1215 및 SW48, 폐암 세포주인 NCI-H838 및 NCI-H1975의 야생형 및 KRAS G12D 돌연변이 쌍에 대하여 Q2a 화합물을 다양한 농도(0μM, 1μM, 5μM, 10μM)로 24시간, 48시간, 72시간 동안 각각 처리하고 세포성장 억제율을 측정하고, 그 결과를 도 5 내지 8에 나타내었다. Ez-cytox와 2시간 동안 반응시킨 후 마이크로플레이트 리더기를 이용하여 세포 증식률을 측정한 결과, 대장암 세포주 LIM1214와 SW48 모두 야생형에 비해 KRAS G12D 돌연변이체에서 세포 성장 억제 경향을 보였다.Q2a compound was administered at various concentrations (0 μM, 1 μM, 5 μM, 10 μM) to wild-type and KRAS G12D mutant pairs of colon cancer cell lines LIM1215 and SW48 and lung cancer cell lines NCI-H838 and NCI-H1975 for 24, 48, and 72 hours. After each treatment, the cell growth inhibition rate was measured, and the results are shown in Figures 5 to 8. After reacting with Ez-cytox for 2 hours, the cell proliferation rate was measured using a microplate reader. As a result, both colon cancer cell lines LIM1214 and SW48 showed a tendency to inhibit cell growth in the KRAS G12D mutant compared to the wild type.

도 5에 따르면, LIM1215 세포는 24시간과 48시간 처리시 5μM과 10μM 처리군에서 야생형과 비교했을 때 세포증식 억제가 나타났으며, 72시간 처리시에는 1μM, 5μM, 10μM 모든 처리 농도에서 세포증식 억제가 나타났다.According to Figure 5, LIM1215 cells showed inhibition of cell proliferation compared to the wild type in the 5μM and 10μM treatment groups when treated for 24 and 48 hours, and cell proliferation was observed at all treatment concentrations of 1μM, 5μM, and 10μM when treated for 72 hours. Inhibition appeared.

도 6에 따르면, SW48 세포에서는 24시간, 48시간, 72시간 처리시 KRAS G12D 돌연변이 세포에서만 1μM, 5μM, 10μM 농도별 세포 성장이 억제되는 것을 확인할 수 있다.According to Figure 6, in SW48 cells, when treated for 24 hours, 48 hours, and 72 hours, cell growth was inhibited only in KRAS G12D mutant cells at concentrations of 1 μM, 5 μM, and 10 μM.

폐암 세포주는 대장암 세포주에 비해 더 우수한 세포 증식 억제 효과를 나타냈다. 도 7에 따르면, NCI-H838 세포에서는 야생형 세포와 비교하여 KRAS G12D 돌연변이 세포는 48시간 처리시 1 μM 농도 처리군에서 약 30% 정도의 세포 증식 억제를 나타냈으며, 24시간, 72시간 처리시에도 농도 의존적으로 세포 증식이 억제되는 것을 확인하였다.Lung cancer cell lines showed a better cell proliferation inhibition effect than colon cancer cell lines. According to Figure 7, in NCI-H838 cells, compared to wild-type cells, KRAS G12D mutant cells showed about 30% inhibition of cell proliferation in the 1 μM concentration group when treated for 48 hours, even when treated for 24 hours and 72 hours. It was confirmed that cell proliferation was inhibited in a concentration-dependent manner.

도 8에 따르면, NCI-H1975 세포에서도 KRAS G12D 돌연변이 세포에서 24시간, 48시간, 72시간 처리시 야생형 세포에 비해 농도 의존적으로 세포 성장이 억제되는 것으로 나타났다.According to Figure 8, in NCI-H1975 cells, KRAS G12D mutant cells were shown to be inhibited in cell growth in a concentration-dependent manner compared to wild-type cells when treated for 24 hours, 48 hours, and 72 hours.

한편, Q2a의 세포 증식 억제 효과를 콜로니 형성 분석(Colony Formation Assay)를 통해 확인하였다. LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과를 도 9에 나타내었다. 이에 따르면, LIM1215 세포에서는 KRAS G12D 돌연변이 세포의 콜로니 수가 대조군에 비해 50% 적은 것으로 나타났다. Meanwhile, the cell proliferation inhibitory effect of Q2a was confirmed through colony formation assay. The results of colony formation analysis for wild type (WT) LIM1215 cells and KRAS G12D mutant cells are shown in Figure 9. According to this, in LIM1215 cells, the number of colonies of KRAS G12D mutant cells was 50% lower than that of the control group.

SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과를 도 10에 나타내었다. 이에 따르면, SW48 KRAS G12D 돌연변이 세포도 Q2a 화합물 처리군에서 세포 증식이 현저하게 감소한 것으로 나타났다. KRAS G12D 돌연변이 세포의 콜로니 수가 대조군에 비해 60% 적은 것으로 나타났으나, 야생형 그룹에서는 큰 차이가 나타나지 않았다.The results of colony formation analysis for wild type (WT) SW48 cells and KRAS G12D mutant cells are shown in Figure 10. According to this, SW48 KRAS G12D mutant cells also showed a significant decrease in cell proliferation in the Q2a compound treatment group. The number of colonies of KRAS G12D mutant cells was found to be 60% lower than that of the control group, but there was no significant difference in the wild type group.

한편, 대장암 외에도 폐암 세포주에서도 콜로니 수가 현저히 감소하는 것으로 나타났다. NCI-H838 및 NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 콜로니 형성 분석 결과를 각각 도 11 및 도 12에 나타내었다. 이에 따르면, NCI-H838와 NCI-H1975 세포에서도 KRAS G12D 돌연변이군은 대조군에 비해 Q2a 화합물 처리군에서 콜로니가 유의하게 감소한 것으로 나타났다. NCI-H838 세포에서는 KRAS G12D 돌연변이 세포의 콜로니 수가 대조군에 비해 50% 이하인 것으로 나타났고, NCI-H1975 세포에서도 KRAS G12D 돌연변이 세포의 콜로니 수가 대조군에 비해 70~50% 적은 것을 확인할 수 있었다. 이에 반해, 야생형 그룹에서는 큰 차이가 없었다.Meanwhile, in addition to colon cancer, the number of colonies was also found to be significantly reduced in lung cancer cell lines. The results of colony formation analysis for wild type (WT) and KRAS G12D mutant cells of NCI-H838 and NCI-H1975 cells are shown in Figures 11 and 12, respectively. According to this, in NCI-H838 and NCI-H1975 cells, the KRAS G12D mutant group showed a significant decrease in colonies in the Q2a compound treatment group compared to the control group. In NCI-H838 cells, the number of colonies of KRAS G12D mutant cells was found to be less than 50% compared to the control group, and in NCI-H1975 cells, the number of colonies of KRAS G12D mutant cells was found to be 70-50% less than the control group. In contrast, there was no significant difference in the wild type group.

또한, 대장암 세포주인 LS174T KRAS G12D 돌연변이 세포의 콜로니 형성 분석 결과를 도 13에 나타내었다. 이에 따르면, 콜로니 수는 대조군에 비해 Q2a 화합물군에서 유의하게 적은 것으로 나타났다.Additionally, the results of colony formation analysis of LS174T KRAS G12D mutant cells, a colon cancer cell line, are shown in Figure 13. According to this, the number of colonies was significantly lower in the Q2a compound group compared to the control group.

실험예 3: KRAS G12D 돌연변이 세포와 대장암 및 폐암 야생형 세포의 세포사멸 효과 분석Experimental Example 3: Analysis of apoptosis effect of KRAS G12D mutant cells and wild-type colon cancer and lung cancer cells

Q2a 화합물을 처리했을 때 KRAS G12D 돌연변이 세포에서 세포사멸 효과를 확인하기 위해 형광 활성화 세포 분류(FACS)를 수행하였다. LIM1215, SW48, NCI-H1975, NCI-H838 세포의 야생형 및 KRAS G12D 돌연변이 쌍에 대해 서로 다른 농도(0μM, 1μM, 5μM, 10μM)로 24시간, 48시간 및 72시간 동안 Q2a 화합물로 처리하였다. 세포사멸은 Annexin V와 PI 염색으로 측정하였다. 대장암 세포주 LIM1215와 SW48에서는 대조군과 비교하여 KRAS G12D 돌연변이 세포에서 세포사멸이 Q2a 화합물 처리군에서 최소 5%, 최대 20%로 나타났다. 그러나 야생형에서는 대조군에 비해 Q2a 화합물 처리군에서 유의한 효과를 나타내지 않았다.Fluorescence-activated cell sorting (FACS) was performed to confirm the apoptotic effect in KRAS G12D mutant cells when treated with Q2a compound. Wild-type and KRAS G12D mutant pairs of LIM1215, SW48, NCI-H1975, and NCI-H838 cells were treated with Q2a compound at different concentrations (0 μM, 1 μM, 5 μM, 10 μM) for 24, 48, and 72 h. Apoptosis was measured by Annexin V and PI staining. In colon cancer cell lines LIM1215 and SW48, compared to the control group, apoptosis in KRAS G12D mutant cells was at least 5% and at most 20% in the Q2a compound treatment group. However, in the wild type, there was no significant effect in the Q2a compound treatment group compared to the control group.

LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과를 도 14에 나타내었다. 이에 따르면, LIM1215 KRAS G12D 돌연변이 세포에서는 24시간, 48시간, 72시간 동안 농도 의존적으로 세포사멸율이 높아졌으며, 72시간에서는 10μM 농도 처리군에서 20%의 세포사멸이 나타났다..The results of analysis of the apoptosis effect on wild type (WT) LIM1215 cells and KRAS G12D mutant cells are shown in Figure 14. According to this, in LIM1215 KRAS G12D mutant cells, the apoptosis rate increased in a concentration-dependent manner for 24, 48, and 72 hours, and at 72 hours, 20% of apoptosis was observed in the 10μM concentration treatment group.

SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과를 도 15에 나타내었다. 이에 따르면, SW48 KRAS G12D 돌연변이 세포에서도 대조군에 비해 Q2a 화합물 처리군에서 24시간, 48시간, 72시간 처리시 세포사멸이 일어나는 것을 확인하였다.The results of analysis of the apoptosis effect on wild type (WT) SW48 cells and KRAS G12D mutant cells are shown in Figure 15. According to this, it was confirmed that cell death occurred in SW48 KRAS G12D mutant cells when treated for 24 hours, 48 hours, and 72 hours in the Q2a compound treatment group compared to the control group.

폐암 세포주에서는 세포 성장 억제 효과와 마찬가지로 대장암 세포주에 비해 세포사멸 효과가 더 좋은 것을 확인할 수 있었다. NCI-H1975, NCI-H838 세포에서 KRAS G12D 돌연변이 세포는 야생형에 비해 더 현저한 세포사멸 효과가 나타났으며, 돌연변이 세포에서는 대조군과 비교했을 때 10% 미만, 최대 60%까지 세포사멸 효과가 나타난 것을 확인할 수 있었다.In lung cancer cell lines, it was confirmed that the cell death effect was better than that in colon cancer cell lines, as was the cell growth inhibition effect. In NCI-H1975 and NCI-H838 cells, KRAS G12D mutant cells showed a more significant apoptotic effect compared to the wild type, and the apoptotic effect was confirmed to be less than 10% and up to 60% in mutant cells compared to the control group. I was able to.

NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과를 도 16에 나타내었다. 이에 따르면, NCI-H1975 KRAS G12D 세포에서는 Q2a 화합물 처리군의 세포사멸 효과가 대조군에 비해 24시간, 48시간, 72시간에 걸쳐 유의하게 증가하는 것으로 나타났다. NCI-H1975 KRAS G12D 세포에서는 Q2a 화합물 처리군의 세포사멸 효과가 대조군에 비해 24시간, 48시간, 72시간에 걸쳐 유의하게 증가하였으며, 24시간 동안 5μM, 10μM 처리군과 48시간 동안 1μM 처리군에서 약 20% 정도의 세포사멸 효과가 나타났다.The results of analysis of the apoptosis effect on wild type (WT) NCI-H1975 cells and KRAS G12D mutant cells are shown in Figure 16. According to this, in NCI-H1975 KRAS G12D cells, the apoptosis effect of the Q2a compound treatment group was found to significantly increase over 24 hours, 48 hours, and 72 hours compared to the control group. In NCI-H1975 KRAS G12D cells, the apoptosis effect of the Q2a compound treatment group was significantly increased over 24, 48, and 72 hours compared to the control group, in the 5μM and 10μM treatment groups for 24 hours and the 1μM treatment group for 48 hours. An apoptosis effect of approximately 20% was observed.

NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 세포사멸효과 분석 결과를 도 17에 나타내었다. 이에 따르면, NCI-H838 KRAS G12D 세포는 실험에 사용된 모든 세포 중에서 가장 많은 세포사멸 효과를 나타냈다. 24시간 동안 1μM 농도 처리시 대조군에 비해 60% 이상의 세포사멸 효과가 나타났다.The results of analysis of the apoptosis effect on wild type (WT) NCI-H838 cells and KRAS G12D mutant cells are shown in Figure 17. According to this, NCI-H838 KRAS G12D cells showed the greatest apoptosis effect among all cells used in the experiment. When treated at a concentration of 1 μM for 24 hours, an apoptosis effect of more than 60% was observed compared to the control group.

실험예 4: 세포사멸, KRAS 다운스트림 관련 단백질 발현 변화 분석Experimental Example 4: Analysis of changes in apoptosis and KRAS downstream-related protein expression

(1) KRAS G12D 돌연변이 세포의 세포사멸 관련 단백질 발현 감소 효과 분석(1) Analysis of the effect of reducing apoptosis-related protein expression in KRAS G12D mutant cells

Q2a 화합물의 처리에 의한 세포사멸과 관련된 기전을 확인하기 위해 웨스턴블롯을 실시하였다. LIM1215, SW48, NCI-H1975 및 NCI-H838 세포의 야생형 및 KRAS G12D 돌연변이 쌍에 Q2a 화합물을 농도별로(0μM, 1μM, 5μM, 10μM) 처리하고, 48시간 및 72시간 후 단백질 추출을 하였다. 10% SDS-Page에 로딩한 후 PVDF 멤브레인으로 옮긴 뒤 1차 항체와 2차 항체를 부착시킨 후 ECL 키트로 검출하였다.Western blot was performed to confirm the mechanism related to cell death caused by treatment with Q2a compound. Wild type and KRAS G12D mutant pairs of LIM1215, SW48, NCI-H1975, and NCI-H838 cells were treated with Q2a compound at different concentrations (0 μM, 1 μM, 5 μM, 10 μM), and protein extraction was performed after 48 and 72 hours. After loading on 10% SDS-Page, it was transferred to a PVDF membrane, primary and secondary antibodies were attached, and it was detected with an ECL kit.

세포사멸의 대표적인 마커인 PARP 폴리(ADP-리보스) 폴리머라제는 스트레스 반응, 염색질 변형, DNA 복구 및 세포사멸을 포함한 여러 세포 과정에 관여하는 17개 단백질 계열이다. 이는 DNA 절단의 검출 및 복구 역할로 확인된 바 있다. 절단된 PARP-1의 존재는 많은 세포 유형에서 세포사멸을 감지하기 위해 가장 일반적으로 사용되는 진단 도구 중 하나이다.PARP poly(ADP-ribose) polymerase, a representative marker of apoptosis, is a family of 17 proteins involved in several cellular processes, including stress response, chromatin modification, DNA repair, and apoptosis. This has been confirmed to play a role in the detection and repair of DNA breaks. The presence of cleaved PARP-1 is one of the most commonly used diagnostic tools to detect apoptosis in many cell types.

세포사멸의 대표적인 마커인 PARP의 절단은 LIM1215, SW48, NCI-H1975, NCI-H838 세포의 KRAS G12D 돌연변이 세포 모두에서 일어났다. 단백질 발현에서의 세포사멸을 확인한 결과, KRAS G12D 돌연변이체에서 Q2a 화합물이 세포사멸을 유도하는 것을 확인하였다. Cleavage of PARP, a representative marker of apoptosis, occurred in all KRAS G12D mutant cells of LIM1215, SW48, NCI-H1975, and NCI-H838 cells. As a result of confirming apoptosis in protein expression, it was confirmed that Q2a compound induces apoptosis in KRAS G12D mutant.

도 18은 LIM1215 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이고, 도 19는 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다. 이에 따르면, LIM1215, SW48 KRAS G12D 돌연변이 세포에서 PARP 절단이 야생형에 비해 48시간 처리군 및 72시간 처리군에서 모두 증가한 것으로 나타났다.Figure 18 shows the Western blot results for wild-type (WT) LIM1215 cells and KRAS G12D mutant cells treated with Q2a 48.72 for 72 hours, and Figure 19 shows Q2a 48.72 for wild-type (WT) SW48 cells and KRAS G12D mutant cells. This is the Western blot result upon time processing. According to this, PARP cleavage in LIM1215 and SW48 KRAS G12D mutant cells increased in both the 48-hour and 72-hour treatment groups compared to the wild type.

도 20은 NCI-H1975 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이고, 도 21은 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 48. 72시간 처리시 웨스턴 블롯 결과이다. 이에 따르면, 마찬가지로 NCI-H1975. NCI-H838 KRAS G12D 돌연변이 세포에서도 PARP 절단이 일어나고 야생형에 비해 48시간과 72시간 모두 증가하는 것으로 나타났다.Figure 20 is a Western blot result for wild-type (WT) NCI-H1975 cells and KRAS G12D mutant cells treated with Q2a 48. 72 hours, and Figure 21 is a Western blot result for wild-type (WT) NCI-H838 cells and KRAS G12D mutant cells. Q2a 48. This is the Western blot result after 72 hours of treatment. According to this, likewise NCI-H1975. PARP cleavage also occurred in NCI-H838 KRAS G12D mutant cells and increased for both 48 and 72 hours compared to the wild type.

(2) KRAS G12D 돌연변이 세포에서 KRAS 다운스트림 관련 단백질의 발현 감소 효과(2) Effect of reducing expression of KRAS downstream-related proteins in KRAS G12D mutant cells

Q2a 화합물이 KRAS 다운스트림 활성 억제에도 영향을 미치는지 확인하였다. KRAS의 다운스트림에는 MAPK 경로 단백질 C-RAF 및 ERK와 PI3K 경로 단백질 AKT가 있다.It was confirmed whether compound Q2a also affects the inhibition of KRAS downstream activity. Downstream of KRAS are the MAPK pathway proteins C-RAF and ERK and the PI3K pathway protein AKT.

도 22는 LIM1215 및 SW48 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 24시간 처리시 웨스턴 블롯 결과이다. 이에 따르면, 대장암 세포 LIM1215 및 SW48은 48시간부터 MAPK 경로 단백질 C-RAF의 인산화를 감소시켰고 PI3K 경로 단백질인 AKT의 인산화도 대조군에 비해 유의하게 감소한 것으로 나타나났다(도 18, 도 19 참조).Figure 22 shows the results of Western blotting of wild type (WT) LIM1215 and SW48 cells and KRAS G12D mutant cells treated with Q2a for 24 hours. According to this, colon cancer cells LIM1215 and SW48 showed decreased phosphorylation of the MAPK pathway protein C-RAF from 48 hours, and phosphorylation of AKT, a PI3K pathway protein, was also significantly decreased compared to the control group (see Figures 18 and 19).

폐암세포주 NCI-H1975와 NCI-H838의 경우 48시간과 72시간에 KRAS G12D 돌연변이에서 KRAS 다운스트림 단백질의 변화는 NCI-H1975 세포의 72시간 처리시 AKT의 인산화와 다르지 않았다(도 20 및 도 21 참조).For lung cancer cell lines NCI-H1975 and NCI-H838, changes in KRAS downstream proteins in the KRAS G12D mutant at 48 and 72 hours did not differ from phosphorylation of AKT upon 72-hour treatment of NCI-H1975 cells (see Figures 20 and 21 ).

도 23는 NCI-H1975 및 NCI-H838 세포의 야생형(WT)과 KRAS G12D 돌연변이 세포에 대한 Q2a 24시간 처리시 웨스턴 블롯 결과이다. 이에 따르면, 48시간, 72시간과 달리 24시간째에는 MAPK 경로 단백질 C-RAF의 인산화가 감소하였고, PI3K 경로 단백질 AKT의 인산화도 대조군에 비해 유의하게 감소하는 것으로 나타났다.Figure 23 shows the results of Western blotting of wild type (WT) NCI-H1975 and NCI-H838 cells and KRAS G12D mutant cells treated with Q2a for 24 hours. According to this, unlike at 48 and 72 hours, the phosphorylation of the MAPK pathway protein C-RAF decreased at 24 hours, and the phosphorylation of the PI3K pathway protein AKT also decreased significantly compared to the control group.

NCI-H838 세포에서는 P-CRAF와 P-AKT가 유의하게 감소했으며 P-ERK는 1μM 및 5μM 농도에서 대조군에 비해 감소한 것으로 나타났다.In NCI-H838 cells, P-CRAF and P-AKT were significantly decreased, and P-ERK was decreased compared to the control group at 1 μM and 5 μM concentrations.

실험예 5: LIM 1215 KRAS G12D 세포에서 Q2a 처리군과 대조군의 유전자 발현 차이 분석Experimental Example 5: Analysis of gene expression differences between Q2a treatment group and control group in LIM 1215 KRAS G12D cells

절단 PARP의 단백질 발현이 증가하고 RAS 다운스트림 관련 단백질 PC-RAF, P-AKT, P-ERK이 웨스턴 블롯에서 단백질 발현이 감소한 LIM1215 KRAS G12D 세포를 사용하여 대조군과 Q2a 화합물 처리군의 유전자 발현 차이를 확인하기 위해 마이크로어레이(Microarray)를 수행하였다. 마이크로어레이는 Macrogene에서 수행되었다.Gene expression differences between control and Q2a compound treatment groups were examined using LIM1215 KRAS G12D cells, which showed increased protein expression of cleaved PARP and decreased protein expression of RAS downstream-related proteins PC-RAF, P-AKT, and P-ERK in Western blot. Microarray was performed to confirm. Microarrays were performed at Macrogene.

도 24는 3개 대조군과 Q2a 화합물 5μM을 처리한 3개 그룹의 히트맵(heatmap)이다. 이에 따르면, 유전자 발현의 차이를 확인할 수 있다. 2a를 처리한 경우 KRAS 돌연변이 세포의 KRAS 신호가 감소하는 것을 확인할 수 있었으며, 이는 Q2a 화합물이 KRAS를 타겟으로 했음을 나타낸다. Figure 24 is a heatmap of three control groups and three groups treated with 5 μM of Q2a compound. According to this, differences in gene expression can be confirmed. When treated with 2a, it was confirmed that the KRAS signal in KRAS mutant cells was decreased, indicating that the Q2a compound targeted KRAS.

또한, Q2a를 처리한 경우 KRAS 돌연변이 세포에서 발암 경로 단백질인 MYC 및 NOTCH의 발현이 감소하는 것으로 나타났다.Additionally, treatment with Q2a showed a decrease in the expression of MYC and NOTCH, which are oncogenic pathway proteins, in KRAS mutant cells.

마이크로어레이의 결과를 바탕으로 풀다운 분석(Pull down assay)을 수행하였다. Raf-1-RBD를 사용하여 RAS 활성을 측정하고 NOTCH1 및 C-MYC 단백질의 발현을 웨스턴 블롯으로 확인하여 그 결과를 도 25에 나타내었다.Pull down analysis was performed based on the microarray results. RAS activity was measured using Raf-1-RBD, and expression of NOTCH1 and C-MYC proteins was confirmed by Western blot, and the results are shown in Figure 25.

이에 따르면, KRAS-GTP 결합은 48시간 5μM 처리시, 72시간 10μM 처리시 감소되어 RAS 활성이 감소되었음을 확인하였다. 유사하게, 발암 경로 단백질의 대표적인 마커인 C-MYC와 NOTCH1도 24시간, 48시간, 72시간 동안 단백질 발현이 감소된 것으로 확인되었다.According to this, KRAS-GTP binding decreased when treated with 5 μM for 48 hours and 10 μM for 72 hours, confirming that RAS activity was reduced. Similarly, C-MYC and NOTCH1, representative markers of oncogenic pathway proteins, were also found to have decreased protein expression for 24, 48, and 72 hours.

실험예 6: 이종이식 모델에서 돌연변이 KRAS G12D 대장암을 억제 효과 분석Experimental Example 6: Analysis of the inhibitory effect on mutant KRAS G12D colon cancer in a xenograft model

in vitro에서 세포 성장 및 세포 사멸 억제 효과를 확인한 후, in vivo에서 Q2a 화합물의 종양 성장 억제 효과를 알아보기 위해 마우스 모델을 이용한 동물 실험을 수행하였다. LIM1215 야생형 세포와 KRAS G12D 돌연변이 세포를 BALB/C 누드 마우스의 양쪽 옆구리에 피하 주사하였다. 종양이 100 mm2 크기로 성장한 후, 이틀에 한 번씩 Q2a 화합물 100㎕를 15mg/kg과 30mg/kg의 농도로 복강 주사하였다.After confirming the inhibitory effect on cell growth and apoptosis in vitro, an animal experiment using a mouse model was performed to determine the tumor growth inhibitory effect of Q2a compound in vivo. LIM1215 wild-type cells and KRAS G12D mutant cells were injected subcutaneously into both flanks of BALB/C nude mice. After the tumor grew to a size of 100 mm 2 , 100 μl of Q2a compound was injected intraperitoneally every two days at a concentration of 15 mg/kg and 30 mg/kg.

LIM1215 야생형 세포와 KRAS G12D 돌연변이 세포 투여 마우스에서 Q2a 투여에 따른 종양 크기의 시간에 따른 변화를 도 26에 나타내었다. 이에 따르면, 대조군에는 생리식염수 100㎕를 복강 내 주사하였다. 야생형군의 종양 크기는 대조군과 Q2a 화합물 처리군 모두에서 증가를 보였으나, KRAS G12D 돌연변이군의 종양 크기는 대조군에서만 시간이 지남에 따라 증가하는 것으로 나타났고, 그룹은 종양 크기는 농도 의존적으로 감소하는 것으로 나타났다.Changes in tumor size over time according to Q2a administration in mice administered LIM1215 wild-type cells and KRAS G12D mutant cells are shown in Figure 26. According to this, 100㎕ of physiological saline was injected intraperitoneally in the control group. The tumor size of the wild-type group showed an increase in both the control and Q2a compound treatment groups, but the tumor size of the KRAS G12D mutant group was found to increase over time only in the control group, and the tumor size of the group decreased in a concentration-dependent manner. It was found that

한편, 21일 동안 Q2a 화합물을 투여하면서 측정한 마우스 체중 변화를 도 27에 나타내었다. 이에 따르면, 대조군에 비해 약물 투여군에서 유의한 체중 감소가 나타나지 않았다. 즉, Q2a에 독성이 없는 것을 확인할 수 있다.Meanwhile, the change in mouse body weight measured while administering the Q2a compound for 21 days is shown in Figure 27. According to this, there was no significant weight loss in the drug administration group compared to the control group. In other words, it can be confirmed that Q2a is not toxic.

치료 21일 후 마우스를 희생시켜 종양을 제거하여 종양의 크기를 대비한 사진을 도 28에 나타내었다. 이에 따르면, 야생형군에서는 Q2a 화합물 치료군과 대조군 사이에 종양 크기에 차이가 없었으나, KRAS G12D 돌연변이군의 Q2a 화합물 치료군에서는 유의한 감소를 나타내었다. After 21 days of treatment, the mouse was sacrificed and the tumor was removed. A photo comparing the size of the tumor is shown in Figure 28. According to this, there was no difference in tumor size between the Q2a compound treatment group and the control group in the wild type group, but there was a significant decrease in the Q2a compound treatment group in the KRAS G12D mutant group.

치료 전후의 종양의 중량을 측정한 결과를 도 29에 나타내었다. 이에 따르면, 종양의 무게도 야생형군에서 대조군에 비해 평균 15mg/kg에서 더 컸으며, KRAS G12D 돌연변이군에서는 농도 의존적 종양의 중량이 감소하였다.The results of measuring the weight of the tumor before and after treatment are shown in Figure 29. According to this, the weight of the tumor was larger in the wild-type group compared to the control group at an average of 15 mg/kg, and the weight of the tumor decreased in a concentration-dependent manner in the KRAS G12D mutant group.

KRAS 경로에서 Q2a 투여에 따른 암세포 억제 매커니즘의 개략도를 도 30에 나타내었다. 이에 따르면, Q2a 화합물이 처리되면 다운스트림 MAPK 및 PI3K 메커니즘의 신호 감소가 KRAS-GTP 활성 감소와 함께 감소한다. 이에 따라 세포사멸이 일어남으로써 세포 증식이 억제될 수 있다.A schematic diagram of the mechanism of cancer cell inhibition according to Q2a administration in the KRAS pathway is shown in Figure 30. According to this, treatment with Q2a compound reduces signaling of downstream MAPK and PI3K mechanisms along with decreased KRAS-GTP activity. As a result, cell death may occur and cell proliferation may be inhibited.

이상, 본 발명의 실시예들에 대하여 설명하였으나, 해당 기술 분야에서 통상의 지식을 가진 자라면 특허청구범위에 기재된 본 발명의 사상으로부터 벗어나지 않는 범위 내에서, 구성 요소의 부가, 변경, 삭제 또는 추가 등에 의해 본 발명을 다양하게 수정 및 변경시킬 수 있을 것이며, 이 또한 본 발명의 권리범위 내에 포함된다고 할 것이다.Although the embodiments of the present invention have been described above, those skilled in the art can add, change, delete or add components without departing from the spirit of the present invention as set forth in the patent claims. The present invention may be modified and changed in various ways, and this will also be included within the scope of rights of the present invention.

본 발명의 화합물은 KRAS G12D 돌연변이 세포에 대해 선택적으로 세포증식을 억제하므로 종래 표피 성장 인자 수용체(EGFR)를 이용한 표적 치료법이 낮은 효능을 나타내는 문제를 해결할 수 있고, 본 발명의 화합물과 그의 염을 유효성분으로 포함하는 약학 조성물은 KRAS G12D 돌연변이 세포에 대해 표적 치료용으로 사용할 수 있다.Since the compound of the present invention selectively inhibits cell proliferation in KRAS G12D mutant cells, it can solve the problem of low efficacy of conventional targeted therapy using epidermal growth factor receptor (EGFR), and the compound of the present invention and its salt are effective. The pharmaceutical composition containing the composition as an ingredient can be used for targeted treatment against KRAS G12D mutant cells.

Claims (14)

하기 화학식 1로 표시되는 KRAS G12D 돌연변이 세포증식 억제용 화합물;A compound for inhibiting proliferation of KRAS G12D mutant cells represented by the following formula (1); [화학식 1][Formula 1]
Figure PCTKR2024001239-appb-img-000014
Figure PCTKR2024001239-appb-img-000014
화학식 1에서,In Formula 1, n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeat units, which is an integer from 1 to 10, m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeat units that is an integer from 1 to 10, X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom, R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group, R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group, R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
제1항에 있어서,According to paragraph 1, 상기 화학식 1에서,In Formula 1, n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4, m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4, X1은 산소원자이고,X 1 is an oxygen atom, R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group, R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group, R5
Figure PCTKR2024001239-appb-img-000015
이고,
R 5 is
Figure PCTKR2024001239-appb-img-000015
ego,
X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom, R6은 수소원자, 또는 1C 내지 10C 알킬기인 것을 특징으로 하는, KRAS G12D 돌연변이 세포증식 억제용 화합물.A compound for inhibiting proliferation of KRAS G12D mutant cells, wherein R 6 is a hydrogen atom or a 1C to 10C alkyl group.
제2항에 있어서,According to paragraph 2, 상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물인 것을 특징으로 하는, KRAS G12D 돌연변이 세포증식 억제용 화합물;The compound represented by Formula 1 is a compound for inhibiting proliferation of KRAS G12D mutant cells, characterized in that it is a compound represented by Formula 2 below; [화학식 2][Formula 2]
Figure PCTKR2024001239-appb-img-000016
Figure PCTKR2024001239-appb-img-000016
..
하기 화학식 1로 표시되는 화합물 또는 이의 염을 유효성분으로 포함하는 암질환 예방 또는 치료용 약학 조성물;A pharmaceutical composition for preventing or treating cancer disease comprising a compound represented by the following formula (1) or a salt thereof as an active ingredient; [화학식 1][Formula 1]
Figure PCTKR2024001239-appb-img-000017
Figure PCTKR2024001239-appb-img-000017
화학식 1에서,In Formula 1, n은 1 내지 10의 정수인 반복단위수이고,n is the number of repeating units, which is an integer from 1 to 10, m은 1 내지 10의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 10, X1은 산소원자 또는 황원자이고,X 1 is an oxygen atom or a sulfur atom, R1은 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 1 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group, R2 내지 R4는 각각 독립적으로 수소원자, 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이고,R 2 to R 4 are each independently a hydrogen atom, a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, or a substituted or unsubstituted A C6 to C30 aryl group, or a substituted or unsubstituted C1 to C30 heteroaryl group, R5는 치환 또는 비치환된 1C 내지 30C 알킬기, 치환 또는 비치환된 C3 내지 C30 시클로알킬기, 치환 또는 비치환된 C1 내지 C30 헤테로시클로알킬기, 치환 또는 비치환된 C6 내지 C30 아릴기, 또는 치환 또는 비치환된 C1 내지 C30 헤테로아릴기이다.R 5 is a substituted or unsubstituted 1C to 30C alkyl group, a substituted or unsubstituted C3 to C30 cycloalkyl group, a substituted or unsubstituted C1 to C30 heterocycloalkyl group, a substituted or unsubstituted C6 to C30 aryl group, or a substituted or It is an unsubstituted C1 to C30 heteroaryl group.
제4항에 있어서,According to clause 4, 상기 화학식 1에서,In Formula 1, n은 1 내지 4의 정수인 반복단위수이고,n is the number of repeat units that is an integer from 1 to 4, m은 1 내지 4의 정수인 반복단위수이고,m is the number of repeating units that is an integer from 1 to 4, X1은 산소원자이고,X 1 is an oxygen atom, R1은 1C 내지 10C 알킬기이고,R 1 is a 1C to 10C alkyl group, R2 내지 R4는 각각 독립적으로 수소원자, 또는 1C 내지 10C 알킬기이고,R 2 to R 4 are each independently a hydrogen atom or a 1C to 10C alkyl group, R5
Figure PCTKR2024001239-appb-img-000018
이고,
R 5 is
Figure PCTKR2024001239-appb-img-000018
ego,
X2는 산소원자 또는 황원자이고,X 2 is an oxygen atom or a sulfur atom, R6은 수소원자, 또는 1C 내지 10C 알킬기인 것을 특징으로 하는, KRAS G12D 돌연변이 세포증식 억제용 화합물.A compound for inhibiting proliferation of KRAS G12D mutant cells, wherein R 6 is a hydrogen atom or a 1C to 10C alkyl group.
제5항에 있어서,According to clause 5, 상기 화학식 1로 표시되는 화합물은 하기 화학식 2로 표시되는 화합물인 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물;A pharmaceutical composition for preventing or treating cancer disease, wherein the compound represented by Formula 1 is a compound represented by Formula 2 below; [화학식 2][Formula 2]
Figure PCTKR2024001239-appb-img-000019
Figure PCTKR2024001239-appb-img-000019
..
제4항에 있어서,According to paragraph 4, 상기 암질환 예방 또는 치료용 약학 조성물은 KRAS G12D 돌연변이 세포증식 억제용인 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.The pharmaceutical composition for preventing or treating cancer diseases is characterized in that it is used to inhibit proliferation of KRAS G12D mutant cells. 제7항에 있어서,In clause 7, 상기 KRAS G12D 돌연변이 세포는 LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D 및 LS174T KRAS G12D 중에서 선택된 1종 이상인 것을 특징으로 하는, 암질환 예방 또는 치료용 조성물.The KRAS G12D mutant cells are one or more selected from LIM1215 KRAS G12D, SW48 KRAS G12D, NCI-H1975 KRAS G12D, NCI-H838 KRAS G12D, and LS174T KRAS G12D. A composition for preventing or treating cancer disease. 제8항에 있어서,According to clause 8, 상기 암질환 예방 또는 치료용 조성물은 상기 KRAS G12D 돌연변이 세포에 대한 표적 치료용인 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer diseases, characterized in that the composition for preventing or treating cancer diseases is for targeted treatment of the KRAS G12D mutant cells. 제7항에 있어서,In clause 7, 상기 암질환 예방 또는 치료용 조성물은 MAPK 신호전달 경로 억제용인 것을 특징으로 암질환 예방 또는 치료용 약학 조성물.The composition for preventing or treating cancer diseases is a pharmaceutical composition for preventing or treating cancer diseases, wherein the composition is for inhibiting the MAPK signaling pathway. 제10항에 있어서,According to clause 10, 상기 MAPK 신호전달 경로 억제는 RAS-RAF-MEK-ERK 경로에서 C-RAF 인산화 억제에 따라 수행되는 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer diseases, wherein the inhibition of the MAPK signaling pathway is carried out by inhibiting C-RAF phosphorylation in the RAS-RAF-MEK-ERK pathway. 제7항에 있어서,In clause 7, 상기 암질환 예방 또는 치료용 조성물은 PI3K 신호전달 경로 억제용인 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer diseases, characterized in that the composition for preventing or treating cancer diseases is for inhibiting the PI3K signaling pathway. 제12항에 있어서,According to clause 12, 상기 PI3K 신호전달 경로 억제는 PI3K-AKT-mTOR 경로에서 AKT 인산화 억제에 따라 수행되는 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer diseases, wherein the inhibition of the PI3K signaling pathway is carried out by inhibiting AKT phosphorylation in the PI3K-AKT-mTOR pathway. 제4항에 있어서,According to paragraph 4, 상기 암질환은 대장암, 결장암, 직장암, 폐암, 흑색종, 갑상선암, 자궁암, 난소암, 자궁경부함, 췌장암, 위암 및 간암 중에서 선택된 어느 하나인 것을 특징으로 하는, 암질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer diseases, wherein the cancer disease is any one selected from colon cancer, colon cancer, rectal cancer, lung cancer, melanoma, thyroid cancer, uterine cancer, ovarian cancer, cervix cancer, pancreatic cancer, stomach cancer, and liver cancer.
PCT/KR2024/001239 2023-01-25 2024-01-25 Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient Ceased WO2024158242A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20230009711 2023-01-25
KR10-2023-0009711 2023-01-25
KR10-2024-0011679 2024-01-25
KR1020240011679A KR20240117500A (en) 2023-01-25 2024-01-25 Compound for inhibiting KRAS G12D mutation and composition for preventing or treating cancer disease comprising the same as an active ingredient

Publications (1)

Publication Number Publication Date
WO2024158242A1 true WO2024158242A1 (en) 2024-08-02

Family

ID=91970940

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2024/001239 Ceased WO2024158242A1 (en) 2023-01-25 2024-01-25 Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient

Country Status (1)

Country Link
WO (1) WO2024158242A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2025080946A2 (en) 2023-10-12 2025-04-17 Revolution Medicines, Inc. Ras inhibitors
WO2025171296A1 (en) 2024-02-09 2025-08-14 Revolution Medicines, Inc. Ras inhibitors
US12448400B2 (en) 2023-09-08 2025-10-21 Gilead Sciences, Inc. KRAS G12D modulating compounds
WO2025240847A1 (en) 2024-05-17 2025-11-20 Revolution Medicines, Inc. Ras inhibitors
WO2025255438A1 (en) 2024-06-07 2025-12-11 Revolution Medicines, Inc. Methods of treating a ras protein-related disease or disorder

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020140001A1 (en) * 2018-12-28 2020-07-02 Riboscience Llc Quinazoline derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
WO2022002102A1 (en) * 2020-06-30 2022-01-06 InventisBio Co., Ltd. Quinazoline compounds, preparation methods and uses thereof
KR20220071193A (en) * 2019-08-29 2022-05-31 미라티 테라퓨틱스, 인크. KRAS G12D inhibitor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020140001A1 (en) * 2018-12-28 2020-07-02 Riboscience Llc Quinazoline derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
KR20220071193A (en) * 2019-08-29 2022-05-31 미라티 테라퓨틱스, 인크. KRAS G12D inhibitor
WO2022002102A1 (en) * 2020-06-30 2022-01-06 InventisBio Co., Ltd. Quinazoline compounds, preparation methods and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"M.S. thesis", 1 February 2023, WELLNESS BIO INDUSTRY DEPARTMENT, GANGNEUNG-WONJU NATIONAL UNIVERSITY, Korea, article LEE, JIYUN: "Inhibition of KRAS G12D mutant with Small Molecules", pages: 1 - 67, XP009557408 *
WANG YUTING, ZHANG HAI, LI JINDONG, NIU MIAO-MIAO, ZHOU YANG, QU YUANQIAN: "Discovery of potent and noncovalent KRASG12D inhibitors: Structure-based virtual screening and biological evaluation", FRONTIERS IN PHARMACOLOGY, FRONTIERS RESEARCH FOUNDATION, CH, vol. 13, 22 December 2022 (2022-12-22), CH , pages 1094887, XP093194742, ISSN: 1663-9812, DOI: 10.3389/fphar.2022.1094887 *
ZHU CHUNXIAO, GUAN XIAOQING, ZHANG XINUO, LUAN XIN, SONG ZHENGBO, CHENG XIANGDONG, ZHANG WEIDONG, QIN JIANG-JIANG: "Targeting KRAS mutant cancers: from druggable therapy to drug resistance", MOLECULAR CANCER, vol. 21, no. 1, 4 August 2022 (2022-08-04), XP055979853, DOI: 10.1186/s12943-022-01629-2 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12448400B2 (en) 2023-09-08 2025-10-21 Gilead Sciences, Inc. KRAS G12D modulating compounds
WO2025080946A2 (en) 2023-10-12 2025-04-17 Revolution Medicines, Inc. Ras inhibitors
WO2025171296A1 (en) 2024-02-09 2025-08-14 Revolution Medicines, Inc. Ras inhibitors
WO2025240847A1 (en) 2024-05-17 2025-11-20 Revolution Medicines, Inc. Ras inhibitors
WO2025255438A1 (en) 2024-06-07 2025-12-11 Revolution Medicines, Inc. Methods of treating a ras protein-related disease or disorder

Similar Documents

Publication Publication Date Title
WO2024158242A1 (en) Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient
WO2023003417A1 (en) Kras mutation-specific inhibitor and composition for preventing or treating cancer comprising same
WO2021158071A1 (en) Pharmaceutical composition for prevention or treatment of cancers associated with kras mutation
WO2019112344A1 (en) Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
WO2011159137A2 (en) Novel thiourea or urea derivative, preparation method thereof, and pharmaceutical composition for preventing or treating aids, containing same as active ingredient
WO2012053768A2 (en) Aryloxyphenoxyacetyl-based compound having hif-1 inhibition activity, preparation method thereof and pharmaceutical composition containing the same as an active ingredient
WO2012128521A2 (en) Pharmaceutical composition for treating aging-associated diseases, containing progerin expression inhibitor as active ingredient, and screening method of said progerin expression inhibitor
WO2021149900A1 (en) Disubstituted adamantyl derivative or pharmaceutically acceptable salt thereof, and pharmaceutical composition for suppressing cancer growth comprising same as active ingredient
WO2022197103A1 (en) Novel salts of heterocyclic compound as protein kinase inhibitor and uses thereof
WO2019035522A1 (en) Composition for preventing or treating cancer containing triazolopyridine-based derivative as active ingredient
WO2021235811A1 (en) Pharmaceutical composition for preventing or treating small-cell lung cancer associated with ron mutants and method using same
WO2021235812A1 (en) Pharmaceutical composition for preventing or treating non-small cell lung cancer associated with ron mutation, and method using same
WO2024158237A1 (en) Compound for inhibiting kras mutation, and composition for preventing or treating cancer, comprising same as active ingredient
WO2017048069A1 (en) Novel pyrazoline derivative and use thereof
WO2021085888A1 (en) Novel heterocyclic-substituted pyrimidine derivative exhibiting cancer cell growth inhibitory effect, and pharmaceutical composition containing same
WO2020040343A1 (en) Isoxazole derivatives and preparation process thereof
WO2023249228A1 (en) Novel compound and use thereof for treatment of autoimmune diseases
WO2014157965A1 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
WO2021235813A1 (en) Pharmaceutical composition for preventing or treating pancreatic cancer associated with ron mutation and method using same
WO2023063751A1 (en) Compounds inhibiting alk and/or egfr mutation kinases and medical use thereof
WO2025071172A1 (en) Novel compound, and pharmaceutical composition for prevention or treatment of cancer or tumors comprising same
WO2022065968A1 (en) Pharmaceutical composition comprising evodiamine as active ingredient for prevention or treatment of non-small cell lung cancer
WO2016144087A1 (en) N-phenyl-n&#39;-phenoxycarbonyl-phenylsulfonhydrazide derivative and pharmaceutical composition comprising same
WO2022014975A1 (en) Bi-1 antagonist and use thereof
WO2021100897A1 (en) Pharmaceutical composition for preventing or treating cancer, comprising biguanide-based compound and ferrocene or ferrocene derivative as active ingredients

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: 24747475

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE