CN117440954A - Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors - Google Patents

Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors Download PDF

Info

Publication number
CN117440954A
CN117440954A CN202280038735.6A CN202280038735A CN117440954A CN 117440954 A CN117440954 A CN 117440954A CN 202280038735 A CN202280038735 A CN 202280038735A CN 117440954 A CN117440954 A CN 117440954A
Authority
CN
China
Prior art keywords
alkyl
cancer
haloalkyl
cyano
hydroxy
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.)
Pending
Application number
CN202280038735.6A
Other languages
Chinese (zh)
Inventor
S·班纳吉
S·邓
李伟
K·A·F·穆哈默德
D·D·米勒
Z·吴
R·王
T·西格罗夫斯
R·克鲁蒂里娜
K·哈特曼
S·波尚帕利
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.)
University of Tennessee Research Foundation
Original Assignee
University of Tennessee Research Foundation
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 University of Tennessee Research Foundation filed Critical University of Tennessee Research Foundation
Priority claimed from PCT/US2022/025637 external-priority patent/WO2022226118A1/en
Publication of CN117440954A publication Critical patent/CN117440954A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Abstract

The present invention encompasses novel dihydroquinoxalinone compounds having significantly improved water solubility and reduced toxicity to achieve higher therapeutic indices, and the use of the compounds for the treatment of cancer, viral infection and inflammation.

Description

Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors
Cross Reference to Related Applications
The present application claims priority from U.S. provisional application Ser. No. 63/177,183, filed on month 4 of 2021, and U.S. provisional application Ser. No. 63/317,931, filed on month 3 of 2022, which are hereby incorporated by reference.
Statement of government interest
The present invention proceeds in whole or in part with government support under grant numbers R01CA148706, 1S10OD010678-01, RR-026377-01 and 1S10OD016226 awarded by the national institutes of health (National Institutes of Health). The government may have certain rights in this invention.
Background
Dynamic Microtubules (MT) are key elements of the cytoskeleton and are known to have a significant contribution in cell proliferation, migration and mitosis. The assembly and disassembly of MT depends on the polymerization and depolymerization of tubulin. Thus, given the deep participation of MT in mitosis, disruption of MT kinetics has become an effective strategy in anticancer therapies.
Three major classes of tubulin inhibitors that have been widely used for clinical intervention of cancer are taxanes, vinca alkaloids and epothilones. However, the generation of multi-drug resistance (MDR), peripheral neuropathy, and the narrow therapeutic index often limit the clinical efficacy of these drugs. Colchicine binds at the interface of the alpha beta-tubulin dimer. Colchicine and other small molecules that bind to the colchicine site have been observed to inhibit the ability of tubulin dimers to polymerize and form functional microtubules. Thus, colchicine Binding Site Inhibitors (CBSI) have shown significant cytotoxicity in many studies. Although colchicine itself is susceptible to efflux transporters as well as to β3-tubulin (class III β -tubulin) -mediated MDR, small molecule CBSI is significantly less susceptible to these MDR mechanisms, which is why currently FDA-approved tubulin inhibitors have limited clinical efficacy. However, the clinical use of small molecule CBSI is limited by its significantly undesirable toxicity to normal cells, low solubility, and low oral bioavailability.
In recent years, a new class of small molecule CBSI, in particular vascular blockers (VDAs), has been extensively studied that target the colchicine binding site. VDA has significant advantages over Angiogenesis Inhibitors (AI) in that it can disrupt the established vascular network in tumors, thereby introducing extensive necrosis and apoptosis via vascular collapse. Notably, VDAs are known to block blood flow primarily in solid tumors, leaving blood vessels intact in normal tissues. Currently, a large number of VDA agents targeting the colchicine site are in the stage of a post clinical trial for the treatment of different cancer types.
For example, phosphate analogs of combretastatin A-4 (CA-4P) are in phase III trials targeting undifferentiated thyroid cancer and in phase II trials for the treatment of non-small cell lung cancer (NSCLC). Vitamin Lu Bulin (verubulin) (MPC-6827) (Azixa) is a highly potent tubulin polymerization inhibitor that was introduced several years ago and has a powerful ability to initiate vascular rupture by rapidly collapsing tumor blood flow and inhibiting tumor growth. Vitamin Lu Bulin has shown low nanomolar efficacy against various cancers including melanoma, brain cancer, prostate cancer and breast cancer. Vitamin Lu Bulin entered phase I and phase II clinical trials, however, was subsequently exited due to potential cardiovascular toxicity.
These successful experiences have prompted intensive research into VDA, leading to the recent advent of numerous reports on small molecule VDA drug candidates with significantly high antiproliferative activity. Most of the recent colchicine-binding VDAs have attracted considerable attention due to their dual mechanism of action as antimitotic agents as well as vascular damaging agents, and are therefore being postulated to be successful in cancer chemotherapy.
Disclosure of Invention
One embodiment of the present invention encompasses compounds having the structure of formula I:
Wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Halogenated compoundsAlkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, or wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
Another embodiment of the invention encompasses compounds having the structure of formula IA:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocycle having at least one N, O or S atom, wherein said cycloalkyl ring or heterocycle can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocycle can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
Another embodiment of the invention encompasses compounds having the structure of formula II:
wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of formula II and a pharmaceutically acceptable excipient.
An embodiment of the present invention encompasses compounds of formula I represented by any one of the following compounds 5j-5r, 5t-5v or 12a-12m and 12o-12 q:
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of any one of formulas 5j-5r, 5t-5v or 12a-12m and 12o-12q, and a pharmaceutically acceptable excipient.
One embodiment of the present invention encompasses compounds represented by 5 s:
another embodiment of the invention encompasses a method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula I wherein the structure of formula I is
Wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein said substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
Or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof. In another embodiment of the method, the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer. In one embodiment of the method, the cancer is at least one of the following: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
Another embodiment of the invention encompasses a method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula I wherein the structure of formula IA is
Wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein the cycloalkyl ringOr a heterocycle can optionally have at least one unsaturation, wherein the cycloalkyl ring or heterocycle can be optionally substituted, wherein the substituents of the cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof. In another embodiment of the method, the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer. In one embodiment of the method, the cancer is at least one of the following: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
One embodiment of the invention encompasses a method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula II, wherein the structure of formula II is
Wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof. In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of formula II and a pharmaceutically acceptable excipient. In one embodiment of the method, the cancer is at least one of the following: a drug resistant tumor; metastatic cancer; or drug resistant cancer. In another embodiment of the method, the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
One embodiment of the invention encompasses a method of treating cancer in a subject in need thereof by administering a therapeutically effective amount of: a compound of formula I represented by any one of the following compounds 5j-5r, 5t-5v or 12a-12m and 12o-12 q:
/>
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses a pharmaceutical composition for treating cancer comprising a compound of any one of formulas 5j-5r, 5t-5v or 12a-12m and 12o-12q, or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof, and a pharmaceutically acceptable excipient.
One embodiment of the invention encompasses the treatment of cancer with a compound represented by 5 s:
in one embodiment of the method, the cancer is at least one of the following: a drug resistant tumor; metastatic cancer; or drug resistant cancer. In another embodiment of the method, the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
One embodiment of the invention encompasses a method of treating cancer in a subject in need thereof by administering a therapeutically effective amount of compound 5 s:
/>
in one embodiment of the method, the cancer is at least one of the following: a drug resistant tumor; metastatic cancer; or drug resistant cancer. In another embodiment of the method, the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
Drawings
The subject matter regarded as the invention is particularly pointed out and distinctly claimed in the concluding portion of the specification. The invention, however, both as to organization and method of operation, together with objects, features, and advantages thereof, may best be understood by reference to the following detailed description when read with the accompanying drawings in which:
FIG. 1 shows the x-ray crystal structure of heterocyclic-pyridopyrimidine 1a and dihydroquinoxalinone 2a, and 2a showing binding to colchicine sites in tubulin.
FIG. 2 shows examples of compounds of the invention, for example 5j, 5l, 5m, 5r and 5t.
FIG. 3 shows the synthesis of modified A-ring and B-ring dihydroquinoxalinone analogs.
FIG. 4 shows the synthesis of ethylamine-substituted B-ring dihydroquinoxalinone analogs 5t-5 u.
FIGS. 5A and 5B show the binding patterns of compounds 5m and 5t to tubulin and their localization within cells. FIG. 5A shows 5M (10. Mu.M) and 5t (10. Mu.M) tubulin polymerization assays using tubulin from bovine brain sources. Colchicine and paclitaxel were used at the same concentration, colchicine was used as positive control and paclitaxel was used as negative control. FIG. 5B shows a comparison between morphology and α -tubulin distribution of A375/TxR melanoma cells at the interphase (top) and mitotic phase (bottom) after in vitro treatment with 2nM colchicine, paclitaxel, 5m and 5t.
FIGS. 6A to 6H show the X-ray co-crystal structures of tubulin-RB 3-SLD-TTL proteins complexed with 2a, 5j, 5k, 5l, 5m, 5t and colchicine. FIG. 6A shows the position of the device inComplex with 2a at resolution. Fig. 6B shows at +.>Complex with 5j at resolution. Fig. 6C shows at +.>Complex with 5k at resolution. Fig. 6D shows at +.>Complex with 5l at resolution. Fig. 6E shows at +.>Complex with 5m at resolution. Fig. 6F shows at +.>Complex with 5t at resolution. Fig. 6G shows the overlap with complexes of 5m and 5 t. Fig. 6H shows complexes of tubulin complexes (PDB 5 XIW) bound to colchicine. For fig. 6A-6H, tubulin a-monomers are shown in cyan and β -monomers are shown in gold.
FIGS. 7A and 7B show the effect of compounds 5m and 5t on the clonogenic assay. FIG. 7A shows representative colony formation images of A375/TxR cells after 5m one week treatment with various concentrations. FIG. 7B shows representative colony formation images of A375/TxR cells after 5t of treatment with various concentrations for one week. Colony formation rate was expressed as colony area density% ± SEM. * P=0.0004, p <0.0001.
FIGS. 8A and 8B show the effect of compounds 5m and 5t on A375/TxR cell migration. Scarification is caused by the trauma manufacturer. Fig. 8A shows representative images of wound healing captured by IncuCyte after 12 hours or 24 hours of treatment with 5m, and blue lines show wound edges of cell monolayers. Fig. 8B shows representative images of wound healing captured by IncuCyte after 12 hours or 24 hours of treatment with 5t, and blue lines show wound edges of cell monolayers. Wound closure is shown as a percentage of the relative wound density at each time point.
FIGS. 9A and 9B show that compounds 5M and 5t induce G2/M phase cell cycle arrest and apoptosis in A375/TxR cells. FIG. 9A shows cell cycle analysis of A375/TxR cells by compounds 5m and 5 t. A375/TxR cells were incubated with 1nM, 2nM and 5nM 5m or 5t for 24 hours without serum starvation. Cells were harvested, stained with Propidium Iodide (PI), and then analyzed by flow cytometry. Quantification of cell cycle distribution was analyzed by GraphPad based on two independent experiments in triplicate. Fig. 9B shows that compounds 5m and 5t induce apoptosis. A375/TxR cells were treated with 5m or 5t for 24 hours at the concentration ranges shown in panel A. Cells were collected and stained with FITC-labeled annexin V and PI and analyzed by flow cytometry. The bar graph on the right represents the percentage of the sum of early and late apoptotic cells in triplicate of two independent experiments. P=0.0001, p <0.0001 compared to control.
FIGS. 10A-10D show inhibition of tumor growth by dihydroquinoxalinone pyrimidine analog 5m in A375/TxR xenograft studies. In this study, 10mg/kg paclitaxel treatment was included as a reference control. Intravenous (IV) administration was carried out at two different doses (2 mg/kg and 4 mg/kg) every 2 times per week for 5m. Similarly, paclitaxel was administered at a dose of 10mg/kg (IV) at the same frequency as 5m. Fig. 10A shows tumor growth of the vaccinated tumor, and fig. 10B shows mouse body weight as measured during treatment. Fig. 10C shows the final weight of resected tumor after 21 days of treatment. Fig. 10D shows images of all tumors as captured. Significant differences between groups were determined by one-way ANOVA followed by Dunnett multiple comparison test. (p <0.05, p <0.0001 compared to control). Data = mean ± SD.
FIGS. 11A-11D show that dihydroquinoxalinone pyrimidine analog 5t inhibits tumor growth of A375/TxR xenografts in NSG mice. Mice were treated twice weekly with 2.5mg/kg or 5mg/kg for 5t intravenous injection. Paclitaxel treatment group (10 mg/kg) was used as positive control. FIG. 11A shows the tumor growth curve of the vaccinated A375/TxR xenograft. Fig. 11B illustrates changes in mouse body weight. Fig. 11C illustrates final tumor weight at the end of the study. Fig. 11D shows representative tumor images in this study. Data are presented as mean ± SD. P <0.05, p <0.0001, as analyzed by one-way ANOVA followed by Dunnett's multiple comparison test, compared to control.
FIG. 12 shows necrosis in A375/TxR tumors caused by 5m or 5t treatment. Tumors were harvested, fixed, embedded, sectioned and stained with hematoxylin and eosin (H & E), slides from 5m xenograft model (5 m model, upper panel) and 5t xenograft model (lower panel) were scanned by panoramic FLASH III system and representative images were captured using CaseViewer.
Fig. 13A-13D show the anti-lung and liver spontaneous metastasis effect of compound 5m in a375/TxR subcutaneous xenograft model. After 21 days of treatment, mice bearing the a375/TxR tumor depicted in fig. 10A-10D were sacrificed and lung and liver tissue were harvested, fixed and stained with H & E or anti-human mitochondrial antibodies. Fig. 13A shows the number of lung metastases counted in all mice by averaging the percentage of the transfer area of 3 to 4 representative H & E images per mouse (p <0.0001 compared to control). Fig. 13B shows the number of liver metastases counted in all mice by averaging the percentage of the transfer area of 3 to 4 representative H & E images per mouse (p <0.0001 compared to control). Fig. 13C shows anti-human mitochondrial Immunohistochemical (IHC) staining of lung tissue, which demonstrates the presence of melanoma metastasis in vehicle-or paclitaxel-treated mice, and treatment with 5m reduced metastasis. Fig. 13D shows anti-human mitochondrial IHC staining of liver tissue, which confirms the presence of melanoma metastasis in vehicle-or paclitaxel-treated mice, and treatment with 5m reduced metastasis. Images were obtained by Keyence BZ-X700 microscope and brown staining indicated transfer.
Fig. 14A-14D show in vivo efficacy of 5t in spontaneous lung or liver metastasis. Fig. 14A shows a scatter plot of mean ± SEM showing quantification of metastasis present in the lungs by counting the mean of the percentage of the area of metastasis in 3 to 4 representative H & E images per mouse (p=0.016, p <0.0001 compared to control). Fig. 14B shows a scatter plot of mean ± SEM showing quantification of metastasis present in liver by counting the mean of the percentage of the area of metastasis in 3 to 4 representative H & E images per mouse (p=0.016, p <0.0001 compared to control). Fig. 14C shows IHC staining of anti-human specific mitochondria to detect metastasis in lung sections. Fig. 14D shows IHC staining of anti-human specific mitochondria to detect metastasis in liver sections. Images were obtained by Keyence BZ-X700 microscope and brown staining indicated transfer.
Fig. 15A and 15B show histopathological evaluation of tumor nodules in lung and liver tissue of a 5m subcutaneous xenograft model. FIG. 15A shows representative H & E staining images of lungs in mice treated with vehicle, 10mg/kg paclitaxel, 2mg/kg 5m, and 4mg/kg 5m for 21 days. Fig. 15B shows representative H & E stained images of liver harvested in the same treatment group as in fig. 15A, wherein in both figures, examples of transfer are indicated by yellow arrows.
Figure 16 shows that compound 5m showed potent anti-metastatic effects in anti-human mitochondrial IHC staining with representative images of whole lung and liver treated with vehicle, 10mg/kg paclitaxel, 2mg/kg 5m and 4mg/kg5m 21 days after anti-human mitochondrial antibody staining. Brown staining was reduced in the 5m treated lungs compared to vehicle or paclitaxel group and liver was clearer in the 5m treated group than in vehicle or paclitaxel group.
Fig. 17A and 17B show histopathological evaluation of melanoma metastasis of tissues in a 5t subcutaneous xenograft model. FIG. 17A shows representative H & E staining images of lungs in mice treated with vehicle, 10mg/kg paclitaxel, 2.5mg/kg 5t, and 5mg/kg 5t for 24 days. Fig. 17B shows representative H & E stained images of liver in the same treatment group as in fig. 17A.
Figure 18 shows that compound 5t treatment reduced melanoma metastasis in anti-human mitochondrial IHC staining. Fig. 18 shows representative images of whole lung and liver treated with vehicle, 10mg/kg paclitaxel, 2.5mg/kg 5t and 5mg/kg 5t for 24 days after staining with anti-human mitochondrial antibody, wherein brown staining was reduced in compound 5t treated lung or liver compared to vehicle or paclitaxel group.
Fig. 19A-19F show that compound 5m treatment is effective to overcome taxane resistance and/or compound 17ya resistance or castration resistance in xenografts derived from a variety of cancers including taxane-resistant melanoma (a 375/TxR), compound 17 ya-resistant prostate cancer (DU-145/VxR), taxane and compound 17 ya-resistant breast cancer (MDA-MB-231/VxR), castration-resistant prostate cancer (22 RV 1) and taxane-resistant ovarian cancer (a 2780/TxR). FIG. 19A shows tumor growth inhibition of A375/TxR xenografts in NSG mice by treatment with vehicle, 10mg/kg paclitaxel IV or 2mg/kg or 4mg/kg5 mIV. FIG. 19B shows tumor growth inhibition of DU-145/VxR xenografts in NSG mice by treatment with vehicle (control), 20mg/kg compound 17ya PO or 1mg/kg5 mIV. FIG. 19C shows tumor growth inhibition of MDA-MB-231/VxR xenografts in NSG mice by treatment with vehicle, 10mg/kg paclitaxel IP, 20mg/kg compound 17ya PO or 2mg/kg 5 mIV. FIG. 19D shows tumor growth inhibition of 22RV1 xenografts in NSG mice by treatment with vehicle (control) or 1mg/kg5 mIV. Figure 19E shows tumor growth inhibition as shown by images of A2780/TxR ovarian cancer xenografts grown in mouse left ovaries (right ovaries as control remained unchanged, as shown) by treatment with vehicle (control), 5mg/kg paclitaxel IV, 1mg/kg5m IV, or 20mg/kg compound 17ya PO. Fig. 19F shows the tumor weights of these a2780/TxR ovarian cancer xenografts shown in fig. 19E. The data presented in fig. 19B, 19C, 19E and 19F show that 5m has unexpectedly superior activity compared to 17 ya. Data are expressed as mean ± SEM and analyzed by two-way ANOVA followed by Dunnett multiple comparison test using GraphPad Prism 9 software (San Diego, CA). Statistical significance was presented as p <0.05, p <0.01, p <0.001, and p <0.0001.
FIG. 20 illustrates that 5m shows comparable cytotoxic efficacy in Pancreatic Ductal Adenocarcinoma (PDAC) cells as Paclitaxel (PTX) in Mia PaCa-2 and PANC-1 cell lines.
Fig. 21A and 21B show 5m shows more effective dose-responsive inhibition of colony formation and cell migration than Paclitaxel (PTX) in Pancreatic Ductal Adenocarcinoma (PDAC) cell lines Mia PaCa-2 and PANC-1. Fig. 21A shows the effect of 5m and PTX on PDAC cell growth in a clonogenic assay compared to vehicle-treated controls (controls). Colony formation in Mia PaCa-2 and PANC-1 cell lines treated with 5m or Paclitaxel (PTX) (1 nM, 2.5nM and 5nM for both compounds) were compared in the representative colony formation images shown. The bar graph shows that for Mia PaCa-2, colony formation was completely inhibited by 5m at the lowest dose (1 nM), whereas for PTX, complete inhibition was observed at only 5 nM. Whereas for PANC-1 cells the inhibition efficacy of colony formation was comparable for 5nM and PTX, with only 5nM doses showing almost complete inhibition of colony formation. * P <0.0001. Fig. 21B shows a representative image of wound healing captured by IncuCyte. Cells were monitored live with incuCyte and photographs were taken every 2 hours. Wound closure is shown as the wound width in micrometers (μm) at each time point compared to the control (vehicle), as summarized in the bar graph. Both 5m (2 nM) and PTX (4 nM) inhibited cell migration in Mia PaCa-2 cell culture for more than 48 hours compared to control. As shown by the bar graph, 5m (2 nM) inhibited wound healing more effectively than PTX (4 nM) at each time point. * P <0.001, and p <0.0001.
Fig. 22A and 22B show that compound 5M induced cell cycle arrest in G2/M phase and induced apoptosis in a dose-dependent manner. Figure 22A shows the ability of compound 5M to dose-dependently increase the proportion of cells in G2/M phase in PANC-1 and Mia PaCa-2 cell lines, indicating mitotic arrest in PDAC cell lines. Figure 22B shows that compound 5m and PTX induced apoptosis in Mia PaCa-2 cells as measured by increased cleaved PARP to PARP ratio. In the Mia PaCa-2 cell line, induction of apoptosis by 5m is dose dependent and more efficient compared to PTX. For example, this ratio of 5m of 10nM is comparable to 20nM PTX.
FIGS. 23A-23E show that 5m inhibited PDAC tumor growth with minimal signs of toxicity in the Mia PaCa-2-Luc subcutaneous xenograft model. Figure 23A shows the effect of compound 5m (2 mg/kg) on tumor volume compared to control (vehicle), as measured over 42 days. It can be seen that 5m (2 mg/kg) significantly reduced tumor growth compared to the control. Figure 23B shows the effect of compound 5m on body weight over 42 days compared to control. Body weight is presented as% weight change. It can be seen that limited overall toxicity was observed with 5m, as body weight tended to decrease slightly compared to the control. FIG. 23C illustrates the effect of compound 5m (2 mg/kg) on tumor volume ex vivo compared to control, as measured over 42 days. FIG. 23D illustrates the effect of compound 5m (2 mg/kg) on tumor weight ex vivo over 42 days as compared to control. Figure 23E shows a comparison of resected tumor sizes after treatment with compound 5m compared to control. As seen in fig. 23, tumor volume and tumor weight were significantly reduced by treatment with 5m (2 mg/kg) for 42 days as compared to the control, which can also be understood in photographs of resected tumors. Data are presented as mean ± sign of mean Quasi-error (SEM). Significant differences associated with the control group were measured by P-value<0.05(*p<0.05,**p<0.01,***p<0.001,****p<0.001 Indicated, e.g., by a two-tailed unpaired Welch t-test or two-way ANOVA, followed byOr Dunnett multiple comparisons. Calculation of IC by nonlinear regression 50 . All data were analyzed using GraphPad Prism 9.
Fig. 24A-24E show that 5m inhibited PDAC tumor growth in PANC-1-Luc subcutaneous xenograft models with no signs of toxicity. 5m (1 mg/kg or 2mg/kg;1 dose/week for 7 weeks) or vehicle (control) was administered by i.v. injection to the right flank of each NSG mouse (male, 6 weeks to 8 weeks). Tumor volumes and body weights were measured twice weekly. Figure 24A shows the effect of compound 5m on tumor volume compared to control. The 5m dose-dependent inhibition of xenograft tumor growth compared to the control. Figure 24B shows the effect of compound 5m on body weight (weight change%) compared to control. After 49 days of treatment with two doses of 5m, no differences in body weight were observed in the treated animals compared to the control, indicating a lack of significant overall toxicity at 5 m. FIG. 24C illustrates compound 5m versus control versus tumor volume ex vivo (mm 3 ) Is a function of (a) and (b). Figure 24D illustrates the effect of compound 5m on tumor weight ex vivo (g) compared to control. Consistent with the results for tumor volumes, 5m dose-dependently inhibited xenograft tumor growth compared to control, as measured by ex vivo tumor volume and ex vivo tumor weight. Fig. 24E photo shows a comparison of resected tumor size after treatment with compound 5m compared to control. Tumor volume was calculated by the following formula: volume (mm) 3 ) =0.5× (length×width 2 ). All animals were euthanized at the end of the study. Tumors were resected, ex vivo weights and sizes were recorded and imaged. Data are presented as mean ± Standard Error of Mean (SEM). Significant differences associated with the control group were measured by P-value<0.05(*p<0.05,**p<0.01,***p<0.001,****p<0.001 For example, a two-tailed unpaired Welch test, or one-way ANOVA, followed by Dunnett multiple comparisons, or two-way ANOVA, thenOr Dunnett multiple comparisons. Calculation of IC by nonlinear regression 50 . All data were analyzed using GraphPad Prism 9 (GraphPad Software inc.).
FIG. 25 shows a scheme for the synthesis of compounds 12 a-n. a Reagents and conditions: (i) t-BuOK/t-BuOH; (ii) POCl (Point of care testing) 3 90 ℃; (iii) IPA/HCl, room temperature, 5 hours to 6 hours; (iv) Zn/AcOH, CH 2 Cl 2 The method comprises the steps of carrying out a first treatment on the surface of the (v) Chloroacetyl chloride/K 2 CO 3 Acetone, 0 ℃; (vi) NaH, THF,0 ℃ to room temperature; (vii) potassium hydrogen persulfate, methanol/water, room temperature.
Fig. 26A to 26D show the crystal structure of tubulin-rb3_sld (sT 2R) complex with compounds 5m (not labeled), 12e, 12j, 12k and 5 v. FIG. 26A shows the position of the first part in the following mannerThe original complex (PDB ID: PDB 6X 1F) with compound 5m (unlabeled) at resolution. Fig. 26B shows at +.>Has the crystal structure of compound 12e at resolution. Fig. 26C shows at +. >Has the crystal structure of compound 12j at resolution. Fig. 26D shows at +.>Has the crystal structure of compound 12k at resolution. Fig. 26E shows at +.>Has the crystal structure of compound 5v at resolution. Tubulin a-and β -monomers are shown in cyan and gold, respectively.
Fig. 27 shows plasma concentration-time curves (mean ± SEM) of 12k in mice (n=3) after intravenous administration of 4 mg/kg.
Fig. 28A to 28E show the anti-tumor efficacy of compound 12k on PC3/TxR xenograft tumor growth in NSG male mice. PC3/TxR cells (3X 10) 6 Individual cells/mice) were inoculated subcutaneously into the right flank of NSG mice (n=8). FIG. 28A is a graph comparing tumor growth curves over time with control (vehicle), 10mg/kg (1X/week) of paclitaxel and compound 2.5mg/kg (2X/week) 12 k. Statistical significance was determined using two-way ANOVA followed by multiple comparison tests. Fig. 28B graphically depicts percent change in body weight (weight%) during the treatment day compared with control, paclitaxel and compound 12 k. Fig. 28C illustrates comparing the volume of isolated tumors treated with control, paclitaxel and compound 12 k. Figure 28D illustrates the weight of the tumor ex vivo after treatment with control, paclitaxel and compound 12 k. Fig. 28E shows photographs of isolated tumors in 35mm dishes after treatment with control, paclitaxel and compound 12 k. Data are presented as mean +/-SEM. Significant differences (×p) between groups were determined by one-way ANOVA followed by Dunnett multiple comparison test <0.005,***p<0.0005,****p<0.0001)。
Figure 29 shows that 5v induces tubulin depolymerization and disruption of the microtubule network. (A) Tubulin depolymerization was induced by 5v at 10 μm in a tubulin mixture containing porcine brain tubulin (3 mg/mL) and GTP (1 mM) at 37 ℃. 10. Mu.M colchicine and paclitaxel were used as positive and negative controls. (B) Representative immunofluorescence images of A375/TxR cells at interphase or mitotic stage treated with colchicine (2 nM), paclitaxel (2 nM) or 5v (1 nM or 2 nM) for 24 hours. Tubulin (red) was stained with an alpha-tubulin antibody. Nuclei were stained with DAPI (blue).
FIG. 30 shows that 5v exhibited growth inhibitory effects on a panel of paclitaxel-sensitive and paclitaxel-resistant cancer cells. (A) Five paclitaxel sensitive cancer cell lines (A375, M14, MDA-MB-231, PC3 and DU 145) were treated with 0.1nM to 3. Mu.M colchicine, paclitaxel (paclitaxel), azixa and 5v for 72 hours. Cell viability was expressed relative to DMSO control. (B) Anti-proliferative Activity of 5v against five paclitaxel resistant cancer cell lines (A375/TxR, M14/LCC6MDR1, MDA-MB-231/TxR, PC3/TxR and DU 145/TxR). (C) Representative colony images of A375/TxR cells treated with 5v (0.5 nM, 1nM and 2 nM) for 7 days and quantification of colony formation density in the treated and control groups. Data are presented as total mean ± SEM, P <0.0001.
FIG. 31 shows 5v induction of inhibition of cell migration, apoptosis and mitotic arrest in A375/TxR cells. (A) Migration ability of A375/TxR cells after treatment with 5v (0.5 nM, 1nM, 2nM or 5 nM) was studied by scratch wound assay. Representative wound photographs were obtained by intycyte after 5v exposures of 0 hours, 24 hours and 48 hours. The percentage of wound closure relative to cells at 0 hours ± SEM for each group was calculated at each time point by the IncuCyte scratch wound module. (B) Apoptotic A375/TxR cells treated with 5V (1 nM, 2nM or 5 nM) were measured by annexin V/PI staining. Bar graphs represent the% ± SEM of apoptotic cells for each group. (C) Cell cycle distribution of A375/TxR cells treated identically in B. The percentage of cells in the G1, S or G2/M phase of each treatment group was plotted. * P <0.01; * P <0.001; * P <0.0001.
FIG. 32 shows that 5v shows anti-tumor efficacy in A375/TxR xenograft models. (A) Tumor volume of A375/TxR melanoma xenograft.+ -. SD. Mice were given intravenous vehicle, 4mg/kg paclitaxel, 2mg/kg 5v or 4mg/kg 5v twice weekly. (B) mouse weight change of tumor-bearing mice.+ -. SD. (C) Representative images of resected tumors in vehicle and treatment groups. (D) final tumor wet weight.+ -. SD for each group. Assessing statistical significance by Dunnett multiple comparison test, P <0.001; * P <0.0001 compared to vehicle.
FIG. 33 shows the confirmed H & E and IHC staining of A375/TxR tumors showing the effect of 5v on tumor cell apoptosis, proliferation and angiogenesis in vivo. (A) Representative images of tumor sections stained with & E, ki67, CD31 and cleaved caspase-3. Keyence microscope magnification, 20×. Scale bar, 50 μm. Yellow arrows in H & E stained tumor sections indicate necrotic tumor cells. (B) Quantification of mean Ki67, CD31 and cleaved caspase-3 expression ± SD in tumor sections relative to vehicle control. Assessing statistical significance by Dunnett multiple comparison test, < P0.05, < P0.001; * P <0.0001 compared to vehicle.
FIG. 34 shows that 5v inhibits spontaneous metastasis of A375/TxR subcutaneous tumors. (A) Representative images of axillary lymph nodes collected from each group in a375/TxR xenograft model. (B-C) transfer was detected in lung and liver sections of each group of mice using anti-human mitochondrial IHC staining. The histogram represents the area of metastasis present in the lungs (B) and liver (C) in each group. (D) Representative images of lung (top) or liver (bottom) metastases against human mitochondrial staining in each group. Lung and liver metastases are indicated with red arrows. Keyence microscope magnification, 20×. Scale bar, 200 μm. * P <0.01; * P <0.0001, relative to vehicle.
Fig. 35 shows that 5v shows no acute toxicity. H & E staining of kidney, heart and spleen in each group of mice. After 3 weeks of treatment, the major organs (kidney, heart and spleen) were harvested from mice and stained with H & E. Keyence microscope magnification, 20×. Scale bar, 50 μm.
Figure 36 shows the tolerability assessment of 5v in healthy NSG mice. For example, 5v is tolerated at 5mg/kg IP but not at 10mg/kg IP. 5mg/kg 5v (A) or 10mg/kg 5v (B) was administered to 3 NSG mice by Intraperitoneal (IP) injection at a dosing frequency of 5 times per week. 5mg/kg 5v (C) or 10mg/kg 5v (D) was administered to 4 NSG mice by Intravenous (IV) injection at a dosing frequency of 2 times per week.
FIG. 37 shows that 5v inhibited spontaneous metastasis of A375/TxR tumors. Representative images of whole lung (top), H & E stained lung (middle) and H & E stained liver (bottom) treated with vehicle, 4mg/kg paclitaxel, 2mg/kg 5v or 4mg/kg5v for 23 days. Lung or liver metastasis in H & E stained slides is indicated by yellow arrows.
FIG. 38 shows that 5v inhibits spontaneous metastasis of A375/TxR tumors to lung and liver. Representative images of the whole lung (left) and the whole liver (right) of mice with axillary lymph nodes were not observed in vehicle group (zero).
Figures 39A to 39C show that 5m, 12k and 5v HCl have low nM to pM potent cytotoxicity in two different cell lines a-253 and Detroit 562 of head and neck cancer. FIG. 39A shows compounds 5m, 12k and 5v HCl in IC in two different cell lines A-253 and Detroit 562 for head and neck cancer 50 Values, expressed in nM. FIG. 39B is a graph of cell viability (%) versus concentration (nM) of the A-253 cell line. FIG. 39C is a graph of cell viability (%) versus concentration (nM) of Detroit-562 cell line. Computing ICs as described elsewhere herein 50 Values. Similarly, cytotoxicity assays were performed as described elsewhere herein.
Fig. 40A to 40D show dose-responsiveness decrease with time (hours) of proliferation (confluency%) of cervical cancer cells. FIG. 40A shows a plot of% confluence versus elapsed time (hours) for 12k in the A-253 cell line. Fig. 40B shows a plot of% confluence versus elapsed time (hours) for 12k in the Detroit 562 cell line. FIG. 40C shows a plot of% confluence of compound 17ya versus elapsed time (hours) in the A-253 cell line. FIG. 40D shows a plot of% confluence of compound 17ya versus elapsed time (hours) in the A-253 cell line. Compound 12k or 17ya was added at increasing concentrations to wells of the cell line seeded with the indicated number of indicated cell lines and% confluence was monitored over a period of 62 hours. Both compounds showed reduced dose responsiveness to cell proliferation in both a-253 and Detroit 562 cells, however 12k was more effective in both cell lines.
FIGS. 41A and 41B show that colony formation of A-253 and Detroit 562 cells was also effectively inhibited by 12 k. FIG. 41A shows the effect of compound 12k on colony formation in the A-253 cell line. Figure 41B shows the effect of compound 12k on colony formation in the Detroit 562 cell line. Colony formation experiments were performed as described elsewhere herein.
Fig. 42A and 42B show that 12k induced apoptosis in head and neck cancer cell lines a-253 and Detroit 562, as demonstrated by the elevation of cleaved PARP (c-PARP) and cleaved cas 3 (c-cas 3) of the apoptosis markers revealed by western blot analysis (performed as described elsewhere herein). FIG. 42A shows that compound 12k induced apoptosis in the A-253 cell line. Figure 42B shows that compound 12k induced apoptosis in the Detroit 562 cell line.
Fig. 43 shows that 5v can be synthesized according to the scheme in the figure.
Fig. 44 shows plasma and brain concentrations of 5m and 12k in mice treated as described in example 13.
Fig. 45 shows that 5m is an effective, stable and brain permeable compound, with the same characteristics as Azixa and 17ya, remains able to overcome taxane resistance and thus can be used to treat advanced breast cancer, even when the subject was previously treated with taxane or 17ya and subsequently transferred to the brain. This is an unmet clinical need.
Fig. 46A and 46B show that 5m suppresses BrnMets and increases OS. FIG. 46A shows the effect of compound 5m on (IC) injection of 231-BrM2 cells (200,000) in 8-to 9-week-old NSG female centers, and drug treatment was initiated 2 days after IC injection; animals were dosed twice weekly by the IV route. All mice were euthanized on day 24 of treatment and brain signals were displayed at endpoint after ex vivo bioimaging. Fig. 46A shows representative images of whole brains from vehicle (top) and 5m treated (bottom) mice. Fig. 46B shows that in a separate experiment measuring total survival (OS), 231-BrM2 cells (100,000) IC were injected into NSG females at 8 to 9 weeks of age and treatment was started after 24 hours. Animals were dosed twice weekly by the IV route. Mice were bioimaged once a week and once moribund animals were removed from the study by local IACUC standard. Total photon flux data of living mice (head only) are shown up to day 28. Note that 2 out of 6 vehicle mice moribund/deleted after imaging on day 24, resulting in a decrease in balance average flux on day 28. The Y-axis is plotted on a log10 scale. Survival data is shown in figure 51 below.
Figure 47 shows that 5m treatment allowed mice to maintain body weight throughout day 24, while vehicle-treated mice steadily decreased in body weight from day 15 to day 24. Weight loss is common when mice are ill due to metastatic load. In addition, weight loss is associated with signs and symptoms of brain metastases, including sleepiness, difficulty walking, head tilting, etc.
Figure 48 shows excised brains of vehicle-treated (top) and 5 m-treated (bottom) mice harvested on day 24 after ex vivo bioimaging. Quantitative analysis showed that the 5m treatment averaged total photon flux from 6.4 compared to vehicle treatment×10 7 The p/s is reduced to 2.2X10 7 p/s, and this difference was statistically significant (p=0.044).
Fig. 49A and 49B show a comparison of in vivo head imaging (right panel of each treatment) and later ex vivo head imaging (left panel of each treatment) for vehicle and 5m treated animals showing intact mice of the same animals. Figure 49A shows vehicle treated mice. Fig. 49B shows mice treated with compound 5 m. The reduction in photon flux in the ex vivo brain can be further appreciated using the same capture time (1 minute); ex vivo brain treated at 5m (see rightmost image; 1.51X10) 7 p/s) with the flux of photons observed in vehicle-treated brains (second plot from left; 6.01X10 7 p/s) is reduced. Animals were imaged using Perkin Elmer XMRS instrument.
Fig. 50A-50C show BrM cells will transfer to bone, lung and spleen. Extracranial metastases were observed in both treatment groups, but treatment with 5m reduced or delayed metastatic growth of MBC not only to the brain (as described above), but also statistically reduced metastasis to bone, lung and spleen, as determined by reduced total photon flux measured ex vivo in these organs in the same experiment. Fig. 50A illustrates the ex vivo effect on bone. Fig. 50B shows the ex vivo effect on the lung. Fig. 50A shows the ex vivo effect on the spleen.
Figure 51 shows that 5m significantly improved Overall Survival (OS) of mice carrying 231BrM2 BrnMets. Survival data was generated only in experiment #2 using 5 m. 100,000 cells were infused, treatment was initiated 24 hours later, and mice were deleted once dying. Brains were imaged in vivo until the harvest day, while also imaged ex vivo. The last data including all vehicle mice was on day 24, and the last in vivo imaging day including all 5m treatments was on day 28.
Fig. 52 shows that mice treated with 5m survived longer, which is also evident by the% change in body weight over time (see survival study (experiment # 2)). In addition, the 5m cohort increased body weight on day 21 of dosing, while vehicle-treated mice began to lose body weight on day 11. The% change in body weight in both cohorts appeared to change in the latter phase of the experiment, as the average reflects animals that remain alive only on those days, and as extreme weight loss (15-20%) was the primary euthanasia criterion. Arrows indicate days of administration of 5m or vehicle.
Figure 53 shows that the 5m treatment delayed the progression of metastasis in the brain as observed by the average total photon flux decreasing at each time point greater than 14 days (the signal tracked in the figure was from the brain alone). Despite the high variability of the data, the effect of the 5m treatment was statistically significant at day 28 (p-value 0.0141).
Fig. 54A and 54B show that in vivo imaging (n=1, hence no error bars) of a single representative mouse from each cohort reveals the same trend as ex vivo imaging. FIG. 54A shows a graphical representation of the effect of compound 5m and vehicle. Figure 54B shows ex vivo imaging of vehicle-treated mice and mice treated with compound 5 m. Individual representative mice from each cohort (using the same biological imaging capture time) were tracked over time. Again, by day 14, vehicle-treated mice began to show differences in total luminous flux compared to the 5 m-treated mice, again indicating that 5m delayed the transfer progression, and that on day 28 the transfer was increased 6.7-fold, although each mouse showed similar BrnMets luminous flux onset values (7.5 x 10 5 Compared with 6.3X10 5 ). In addition, representative vehicle-treated mice died at day 28, while 5 m-treated mice survived until day 35. Animals were imaged using Perkin Elmer XMRS instrument.
Fig. 55 (including the table) shows 5m increased survival. The survival of each queue was tracked over time and plotted on a kaplan-Meier survival curve. The table shows the day of euthanasia for each of the six mice in each cohort. All vehicle-treated mice died at 30 days, while all 5 m-treated mice survived for more than 30 days. The median survival of 5m treated mice was 36.5 days, while vehicle treated mice were 25 days. Finally, the kaplan-Meier curve shows a statistically significant increase in survival up to the 5m incidence treatment cohort, with a risk ratio of 5.13, and a p-value of 0.018.
It will be appreciated that for simplicity and clarity of illustration, elements shown in the figures have not necessarily been drawn to scale. For example, the dimensions of some of the elements may be exaggerated relative to other elements for clarity. In addition, where considered appropriate, reference numerals may be repeated among the figures to indicate corresponding or analogous elements.
Detailed Description
Heterocycle-pyridopyrimidine 1a (see fig. 1) and hydroquinoxalinone 2a (see fig. 1) were found to be potent tubulin polymerization inhibitors with significant vascular damaging capabilities. The X-ray co-crystal structure shows that 1a and 2a bind to colchicine sites in tubulin (figure 1). The crystal structure suggests that the pyrimidine moiety of these molecules forms water-mediated hydrogen bonds with β -C239 and β -V236, making these analogs one of the most potent inhibitors of tubulin polymerization. In addition, in compound 2a and its derivatives, the dihydroquinoxalinone moiety forms hydrogen bonds with α -T179. Thus, compound 2a is a strong inhibitor of microtubule polymerization and is highly active in humans (t 1/2 =5.5 hours), mice (t 1/2 =1 hour) and rats (t 1/2 =5.07 hours) liver microsomes have improved metabolic stability. Compound 2a was found to have nanomolar potency on a single order of magnitude for a variety of melanoma, prostate, lung and breast cancer cell lines. However, compound 2a has poor water solubility, making it very difficult to work in intraperitoneal or intravenous in vivo experiments. Furthermore, compound 2a in the Maximum Tolerated Dose (MTD) study showed toxicity above the 1mg/kg dose, resulting in weight loss and death of mice, a highly undesirable property.
To address these shortcomings, the present invention encompasses novel dihydroquinoxalinone compounds having significantly improved water solubility and reduced toxicity to achieve higher therapeutic indices. The present invention encompasses a novel class of pyrimidine analogs having different a-rings (see figure 1). The compounds of the invention include the a-ring, such as a) fused heterocycle-pyrimidine; b) Fused saturated cycloalkane-pyrimidine; c) Ring-opened substituted pyridine-pyrimidine analogs having a dihydroquinoxalinone head group, and the like, as shown in fig. 2. However, without being limited by theory, the present invention is based on the insight that the conversion of a fused pyridopyrimidine tail group to other fused heterocyclic-pyrimidines and fused saturated hydrocarbon-pyrimidine tail groups will provide two advantages: a) Improved water solubility; and b) reduced toxicity while retaining critical hydrogen bonding interactions, one with the T-5 loop of the alpha-tubulin monomer and the other with the H-7 helix of the beta-monomer mediated by water. By incorporating these different structures within the molecule, the present invention seeks to overcome the solubility and toxicity drawbacks found in other compounds.
Without being limited by theory, it is believed that embodiments involving a fused hydrocarbon ring in the tail group should make the compound more capable of forming hydrophobic interactions with the predominantly hydrophobic pocket in β -tubulin, thereby resulting in stronger tubulin binding and improved anticancer efficacy.
The present invention encompasses compounds having the structure of formula I:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
R 4 And R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, orIf R is 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine;
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
The present invention encompasses compounds having the structure of formula IA:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
R 4 And R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocycle having at least one N, O or S atom, wherein said cycloalkyl ring or heterocycle can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocycle can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine;
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
The present invention encompasses compounds having the structure of formula II:
wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
R 4 And R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein said cycloalkyl ring or heterocyclic ringThe substituents of (C) include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine;
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof. In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of formula II and a pharmaceutically acceptable excipient.
The present invention encompasses compounds having the structure of formula IIA:
wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 NaphtheneRadical), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
R 4 And R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocycle having at least one N, O or S atom, wherein said cycloalkyl ring or heterocycle can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocycle can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine;
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of formula II and a pharmaceutically acceptable excipient.
An embodiment of the invention encompasses compounds of formula I, IA, II or IIA represented by any of the following compounds 5j-5r, 5t-5v or 12a-12m and 12o-12 q:
/>
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of any one of formulas 5j-5r, 5t-5v or 12a-12m and 12o-12q, and a pharmaceutically acceptable excipient.
One embodiment of the present invention encompasses compounds represented by 5 s:
the present invention encompasses compounds having the structure of formula III:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
The present invention encompasses compounds having the structure of formula IIIA:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
The present invention encompasses compounds of the structure of formula IIIB:
wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
An embodiment of the present invention encompasses compounds of formula III represented by any one of the following compounds 5l-5n, 5v, 12a-12m and 12o-12 q:
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
In another embodiment, the invention encompasses pharmaceutical compositions comprising a compound of formula III represented by any one of 5l-5n, 5v, 12a-12m, and 12o-12q, and a pharmaceutically acceptable excipient.
One embodiment of the present invention encompasses compounds represented by 5 s:
the present invention includes "pharmaceutically acceptable salts" of the compounds of the invention as described above, which salts may be produced by the reaction of a compound of the invention with an acid or base. Certain compounds, particularly those having an acid or basic group, may also be in the form of salts, preferably pharmaceutically acceptable salts. As used herein, the term "pharmaceutically acceptable salts" refers to those salts that retain the biological effectiveness and properties of the free base or free acid, which are not biologically or otherwise undesirable. Salts are formed from inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like) and organic acids (e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxy acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcysteine, and the like). Other salts are known to those skilled in the art and may be readily adapted for use in accordance with the present invention.
Suitable pharmaceutically acceptable salts of amines of the compounds used in the methods of the invention may be prepared from inorganic or organic acids. In one embodiment, examples of inorganic salts of amines are bisulfate, borate, bromide, chloride, hemisulfate, hydrobromide, hydrochloride, 2-isethionate (hydroxyethane sulfonate), iodate, iodide, isoparaffinate, nitrate, persulfate, phosphate, sulfate, sulfamate, sulfanilate, sulfonic acid (alkyl sulfonate, aryl sulfonate, halogen substituted alkyl sulfonate, halogen substituted aryl sulfonate), sulfonate, and thiocyanate.
Organic salts of amines include, but are not limited to, organic acids of aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic acids, examples thereof are acetic acid, arginine, aspartic acid, ascorbate, adipate, anthranilate, alginate, alkane carboxylate, substituted alkane carboxylate, alginate, benzenesulfonate, benzoate, bisulfate, butyrate, bicarbonate, bitartrate, citrate, camphorate, camphorsulfonate, cyclohexylsulfamate, cyclopentanepropionate, calcium ethylenediamine tetraacetate, dextromethorsulfonate, carbonate, clavulanate (clavulanate), cinnamate, dicarboxylate, digluconate, dodecylsulfonate, dihydrochloride, decanoate, heptanoate, ethane sulfonate, ethylenediamine tetraacetate, ethane disulfonate, propionate dodecylsulfate, ethane sulfonate, fumaric diacid salt formate, fluoride, galacturonate, gluconate, glutamate, glycolate, glucarate, glucoheptonate, glycerophosphate, glucoheptonate, para-hydroxyacetylaminobarate, glutarate, glutamate, heptanoate, caproate, hydroxymaleate, hydroxycarboxylic acid, hexylresorcinol salt, hydroxybenzoate, hydroxynaphthoate, hydrofluoric acid salt, lactate, lactobionate, laurate, malate, maleate, methylenebis (beta-oxynaphthoate), malonate, mandelate, methanesulfonate, methyl bromide, methyl nitrate, methanesulfonate, monopotassium maleate, muciate, monocarboxylate, naphthalenesulfonate, 2-naphthalenesulfonate, nicotinate, and pharmaceutical compositions containing the same, nitrate, naphthalene sulfonate, N-methyl glucamine, oxalate, caprylate, oleate, pamoate, phenylacetate, picrate, phenylbenzoate, trimethylacetate, propionate, phthalate, phenylacetate, pectate, phenylpropionate, palmitate, pantothenate, polygalacturonate, pyruvate, quiniate, salicylate, succinate, stearate, sulfanilate, basic acetate, tartrate, theophyllinate, p-toluene sulfonate (tosylate), trifluoroacetate, terephthalate, tannate, theachlorate, trihaloacetate, triethyliodide, tricarboxylate, undecanoate and valerate.
Examples of inorganic salts of carboxylic acids or hydroxyl groups may be selected from ammonium, alkali metals including lithium, sodium, potassium, cesium; alkaline earth metals including calcium, magnesium, aluminum; zinc, barium, choline, quaternary ammonium.
Examples of organic salts of carboxylic acids or hydroxyl groups may be selected from arginine, organic amines, including aliphatic organic amines, alicyclic organic amines, aromatic organic amines, benzathine, t-butylamine, phenethylamine (N-benzyl phenethylamine), dicyclohexylamine, dimethylamine, diethanolamine, ethanolamine, ethylenediamine, sea Zhuo An (hydrabamine), imidazole, lysine, methylamine, meglumine, N-methyl-D-glucamine, N' -dibenzylethylenediamine, nicotinamide, organic amines, ornithine, pyridine, picoline, piperazine, procaine (procain), tris (hydroxymethyl) methylamine, triethylamine, triethanolamine, trimethylamine, bradykinin and urea.
Typical salts include, but are not limited to, hydrofluoric acid, hydrochloric acid, hydrobromic acid, hydroiodic acid, boric acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, acetic acid, citric acid, maleic acid, malic acid, or methanesulfonic acid. Preferred salts include hydrofluoric acid, hydrochloric acid, hydrobromic acid, hydroiodic acid, acetate, citrate, maleate or mesylate salts. More preferred salts include hydrochloric acid, acetic acid or maleic acid salts.
Salts may be formed in a conventional manner, such as by reacting the free base or free acid form of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble or in a solvent which may be removed in vacuo or by freeze drying, such as water, or by exchanging ions of an existing salt with another ion or a suitable ion exchange resin.
Pharmaceutical composition
The invention also encompasses pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds described above. Generally, the pharmaceutical composition may comprise at least one of the above compounds or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. The term "pharmaceutically acceptable excipient" refers to any suitable adjuvant, carrier, excipient, flavoring or stabilizer, and may be used in a pharmaceutical formulation in solid or liquid form. Such forms include, but are not limited to, lozenges, capsules, powders, solutions, suspensions, or emulsions.
When the condition of the subject is improved, a maintenance dose of the compound, composition or formulation may be administered, if desired. Subsequently, the dose or frequency of administration, or both, can be reduced to a level, depending on the symptoms, at which the improved condition is maintained when the symptoms have been alleviated to a desired level. However, once disease symptoms recur, the subject may require long-term intermittent treatment.
The solid unit dosage form may be of conventional type. The solid form may be capsules and the like, such as the usual gelatin type containing the compound and carrier. Carriers include, but are not limited to, lubricants and inert fillers such as lactose, sucrose, or corn starch. The formulation may be combined with conventional lozenge bases such as lactose, sucrose or corn starch; and binders, such as acacia, corn starch or gelatin; disintegrants, such as corn starch, potato starch or alginic acid; and lubricants such as stearic acid or magnesium stearate.
Lozenges, capsules and the like may also contain binders such as tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; disintegrants such as corn starch, potato starch, alginic acid; lubricants such as magnesium stearate; and sweeteners such as sucrose, lactose or saccharin. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a fatty oil.
Various other materials may be present in a coated form or used to adjust the physical form of the dosage unit. For example, the troches may be coated with shellac, sugar or both. In addition to the active ingredient, syrups may contain sucrose as a sweetener, methyl and propyl parabens as preservatives, dyes and flavors (e.g., cherry or orange flavor).
For oral therapeutic administration, the formulations may include excipients and be used in the form of lozenges, capsules, elixirs, suspensions, syrups and the like. Such compositions and formulations should contain at least 0.1% active compound. Of course, the percentage of compounds in these compositions may vary, and may suitably be between about 2% and about 60% by weight of the unit. The amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained. Preferred compositions according to the invention are prepared such that the oral dosage unit contains from about 0.1mg to 80mg of the active compound, or alternatively from about 1mg to 800mg, or alternatively from about 2mg to 108mg.
The formulation may be administered orally with an inert diluent or with an absorbable edible carrier, or it may be enclosed in hard or soft shell capsules, or it may be compressed into lozenges, or it may be incorporated directly into the diet of the diet. The preparation can be oral preparation, intraperitoneal preparation, intravenous preparation, etc.
Pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form should be sterile and should be fluid to the extent that easy injection is possible. It should be stable under the conditions of manufacture and storage and should be preserved from the contaminating action of microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
The compounds or pharmaceutical compositions used in the methods of the invention may also be administered in injectable dosages as solutions or suspensions of these materials in physiologically acceptable diluents and pharmaceutical adjuvants, carriers or excipients. Such adjuvants, carriers, and/or excipients include, with or without the addition of surfactants and other pharmaceutically and physiologically acceptable components, sterile liquids, such as water and oils. Illustrative oils are those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil or mineral oil. In general, water, physiological saline, dextrose and related aqueous sugar solutions, and glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
The formulation may also be administered parenterally. Solutions or suspensions of these formulations can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in oils containing glycerol, liquid polyethylene glycols, and mixtures thereof. Illustrative oils are those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil or mineral oil. In general, water, physiological saline, dextrose and related aqueous sugar solutions, and glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these formulations contain preservatives to prevent microbial growth.
When the formulation is administered in the methods of the invention, the formulation may be administered systemically or sequentially. Administration may be accomplished in any manner effective to deliver the compound or pharmaceutical composition to the desired site. Exemplary modes of administration include, but are not limited to, oral, topical, transdermal, parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, by intranasal instillation, by intracavity or intravesical instillation, intraocular, intraarterial, intralesional, or by mucosal administration to mucous membranes such as the nose, throat, and bronchi.
The present invention encompasses methods of treating cancer by providing to a subject in need thereof a therapeutically effective amount of at least one compound of the present invention or as at least one composition sufficient to treat cancer in the subject. Drug resistance is the primary cause of cancer chemotherapy failure. Thus, the invention may also encompass the treatment of a subject who has been previously treated with hormone, chemotherapy, radiation therapy or biological therapy; the method comprises administering to a subject in need thereof at least one compound of the invention.
The invention also encompasses a method of treating at least one of the following: a drug resistant tumor; metastatic cancer; or drug resistant cancer. The invention also encompasses a method of treating at least one of the following: prostate cancer, breast cancer, ovarian cancer, skin cancer (e.g., melanoma), lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer (e.g., glioma, glioblastoma).
The invention also encompasses the use of the medicaments of the invention for the treatment of cancer, wherein the cancer is adrenocortical carcinoma, anal carcinoma, bladder carcinoma, brain tumor, brain stem tumor, breast carcinoma, glioma, cerebellar astrocytoma, cerebral astrocytoma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumor, pineal tumor, hypothalamic glioma, breast carcinoma, carcinoid tumor, carcinoma, cervical carcinoma, colon carcinoma, central Nervous System (CNS) cancer, endometrial cancer, and/or peripheral tumor esophageal cancer, extrahepatic cholangiocarcinoma, ewing's family of tumors (Pnet), extracranial germ cell tumor, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer, germ cell tumor, extragonadal gestational trophoblastic tumor, head and neck cancer, hypopharyngeal carcinoma, islet cell carcinoma, laryngeal carcinoma, leukemia, acute lymphoblastic leukemia, oral cancer, liver cancer, lung cancer, non-small cell lung cancer, cervical cancer, lung cancer, malignant tumor, hemoptysis, and malignant tumor small cell lymphoma, AIDS-related lymphoma, central nervous system (primary) lymphoma, cutaneous T-cell lymphoma, hodgkin's disease, non-hodgkin's disease, malignant mesothelioma, melanoma, mekel's cell carcinoma, metastatic squamous carcinoma, multiple myeloma, plasmacytoma, mycosis fungoides, myelodysplastic syndrome, myeloproliferative disorders, nasopharyngeal carcinoma, neuroblastoma, oropharyngeal carcinoma, osteosarcoma, ovarian carcinoma, ovarian epithelial carcinoma, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic carcinoma, exocrine pancreatic carcinoma, islet cell carcinoma, paranasal and nasal cavity carcinoma, parathyroid adenocarcinoma, penile carcinoma, pheochromocytoma carcinoma, pituitary carcinoma, plasmacytoma, prostate carcinoma, rhabdomyosarcoma, rectal carcinoma, renal cell carcinoma, salivary gland cancer, szezary syndrome (Sezary syndrome), skin cancer, cutaneous T cell lymphoma, skin cancer, kaposi's sarcoma, skin cancer, melanoma, small intestine cancer, soft tissue sarcoma, testicular cancer, thymoma, malignant thyroid cancer, urinary tract cancer, uterine cancer, sarcoma, childhood rare cancer, vaginal cancer, vulvar cancer, wilms' tumor, or any combination thereof. In another embodiment, the subject has been previously treated with a hormone, chemotherapy, radiation therapy, or biological therapy. Preferably, the cancer is melanoma, breast cancer, ovarian cancer, prostate cancer or pancreatic cancer.
Despite significant advances in breast cancer treatment, effective treatment of Metastatic Breast Cancer (MBC) remains challenging. The major metastatic sites in MBC generally include bone (41%), lung (22%), liver (8%) and brain (7%). While the precise distribution in these primary metastatic sites depends on MBC subtype, the most common site is bone and the most refractory site is brain. Patients with bone destruction lesions (osteolytic) are particularly susceptible to fractures and chronic pain.
In a preferred embodiment, the invention encompasses methods of treating cancers with compounds of formula III (including formulas IIIA and IIIB), wherein the cancers comprise melanoma, breast cancer, pancreatic cancer, prostate cancer, metastatic bone cancer, or metastatic brain cancer. In another preferred embodiment, the invention encompasses a method of treating breast cancer with a compound of formula III (including formula IIIA and formula IIIB), wherein the breast cancer comprises at least one of: progressive breast cancer; metastatic breast cancer; AR positive breast cancer; ER positive breast cancer; AR positive breast cancer with or without ER, PR and/or HER2 expression; triple positive breast cancer (ER, PR and HER2 positive), AR positive breast cancer with or without ER expression; ER positive breast cancer with or without AR expression; AR-positive and ER-positive breast cancers; refractory breast cancer; AR positive refractory breast cancer; ER positive refractory breast cancer; AR positive metastatic breast cancer; ER positive metastatic breast cancer.
In another preferred embodiment, the invention encompasses methods of treating triple negative breast cancer with a compound of formula III (including formula IIIA and formula IIIB).
As used herein, the term "metastatic cancer" refers to cancer that spreads (metastasizes) from its original location to another area of the body. Almost all cancers have the potential to spread. Whether metastasis occurs depends on a complex interaction of many tumor cell factors, including the type of cancer, the degree of maturation (differentiation) of the tumor cells, the location and time of existence of the cancer, and other factors that are not fully understood. Metastasis spreads in three ways-through local extension from the tumor to surrounding tissue, through blood flow to distant sites, or through the lymphatic system to adjacent or distant lymph nodes. Each cancer may have a typical diffusion pathway. Tumors are referred to as primary sites (e.g., breast cancer that spreads to the brain is referred to as metastatic breast cancer to the brain). In one embodiment of the invention, the compounds of the invention are useful for treating breast cancer that metastasizes to bone. In one embodiment of the invention, the compounds of the invention are useful for preventing metastatic breast cancer to bone development in patients with breast cancer. In another embodiment of the invention, the compounds of the invention are useful for treating breast cancer that metastasizes to the brain. In one embodiment of the invention, the compounds of the invention are useful for preventing the development of metastatic breast cancer to the brain in patients with breast cancer.
As used herein, the term "drug resistant cancer" refers to cancer cells that are resistant to hormone or chemotherapy. Cancer cells can acquire resistance to hormones or chemotherapy through a variety of mechanisms, including mutation or overexpression of drug targets, drug inactivation, or elimination of drugs from cells. Tumors that recur after initial response to hormone or chemotherapy may be resistant to multiple drugs (which are multi-resistant). In conventional resistance views, one or several cells in a tumor population acquire genetic changes that confer resistance. The cause of resistance is therefore, inter alia: a) Some cells that are not killed by hormone or chemotherapy are mutated and resistant to drugs. Once they proliferate, drug resistant cells may be more than cells sensitive to chemotherapy; b) Cancer cells can produce gene amplification of hundreds of copies of a particular gene. This gene triggers the overproduction of proteins that invalidate anticancer drugs; c) Cancer cells can use molecules called P-glycoproteins to pump drugs out of the cell as quickly as possible; d) Cancer cells can stop taking drugs because proteins that transport drugs across the cell wall stop working; e) Cancer cells can learn how to repair DNA breaks caused by some anticancer drugs; f) Cancer cells can develop mechanisms that inactivate drugs. One major contributor to multi-drug resistance is the overexpression of P-glycoprotein (P-gp). This protein is a clinically important transporter that belongs to the ATP-binding cassette family of cell membrane transporters. Which can pump substrates including anticancer drugs out of tumor cells through ATP-dependent mechanisms. Therefore, resistance to anticancer agents used in chemotherapy is a major cause of failure in treatment of malignant tumor, which makes tumors resistant. Drug resistance is the primary cause of cancer hormone or chemotherapy failure.
The invention also encompasses methods of treating viral infections caused by Flaviviridae (flavoviridae) or Herpesviridae (Herpesviridae) (PMID: 31861082) viruses by administering the compounds of the invention. One embodiment of the invention encompasses methods of treating a viral infection by administering a compound of the invention, wherein the infection is caused by SARS-CoV, MERS-CoV, COVID-19 or SARS-CoV-2. Another embodiment of the invention encompasses a method of treating a viral infection, wherein the infection is caused by COVID-19. Another embodiment of the invention encompasses a method of treating a herpes virus infection or potential infection, wherein the virus infection is caused by HSV, VZV, CMV, EBV or PRV. Another embodiment of the invention encompasses a method of treating a viral infection, wherein the infection is caused by a flavivirus. Another embodiment of the invention encompasses methods wherein the flavivirus infection is caused by Dengue fever (Dengue), west Nile, hepatitis C or Zika. Another embodiment of the invention encompasses a method of treating a viral infection, wherein the viral infection is caused by an influenza virus. In another embodiment, the influenza virus is influenza a. In another embodiment, the influenza virus is influenza b. In another embodiment, the influenza virus is influenza delta. In another embodiment, the influenza virus is influenza c.
One embodiment of the invention encompasses methods of treating a viral infection by administering a compound of the invention, wherein the infection is caused by a coronavirus. Another embodiment of the invention encompasses methods of treating a viral infection by administering a compound of the invention, wherein the viral infection is caused by SARS-CoV, MERS-CoV or SARS-CoV-2. A preferred embodiment of the invention encompasses methods of treating a subject for SARS-CoV-2 infection by administering a compound of the invention. Another embodiment of the invention encompasses a method of treating a subject having a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for infection with SARS-CoV-2. Another embodiment of the invention encompasses methods of reducing morbidity in a subject treated for SARS-CoV-2 infection. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces mortality. Another embodiment of the invention encompasses methods of treating a subject with SARS-CoV-2 infection to reduce the incidence of the infection. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces the incidence. Another embodiment of the invention encompasses methods wherein treating a subject with SARS-CoV-2 infection reduces respiratory failure, days in the ICU, days of use of mechanical ventilators, or improves the clinically improved WHO order scale. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces respiratory failure, days in the ICU, days using mechanical ventilators, or improves the clinically improved WHO order scale. Another embodiment of the invention encompasses a method wherein treating a subject with SARS-CoV-2 infection reduces mortality or respiratory failure in subjects older than 60 years. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for a SARS-CoV-2 infection reduces mortality or respiratory failure in subjects >60 years of age. Another embodiment of the invention encompasses a method wherein the co-2 infection or respiratory failure is reduced in a subject when administered with adefovir (remdesivir) and/or dexamethasone (desmethavisone) and/or moneupiravir (mosiupiravir) and/or sotovimab Wei Shankang (sotrovimab) and/or bitrotuzumab (bebtelovisab) and/or tolizumab (tocilizumab) and/or baroretinib (Baricitinib) and/or convalescence plasma and/or bamanib/etfumagma Wei Shankang (etesevisab) and/or karivizumab (casivivisab)/idevisomab) and/or enrouzumab Bei Pu (enstrolivir)/rituximab (ritonavir). Another embodiment of the invention encompasses a method wherein treating a subject having a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces mortality or respiratory failure when administered in combination with adefovir and/or dexamethasone and/or knoop-pivir and/or sotopwei mab and/or bitorubin and/or tolbutamide and/or pamphlet-tinib and/or emmravisuzumab and/or karivizumab/idelizumab and/or enrolment Bei Pu and/or nemorpivir/ritonavir. As used herein, a decrease in mortality, morbidity or respiratory failure, ICU days, ventilator days, etc., refers to a decrease compared to subjects (or a population of subjects) treated with placebo. Likewise, any improvement (such as in the WHO ordinal scale of clinical improvement) refers to an improvement compared to a subject (or population of subjects) treated with placebo.
Another embodiment of the invention encompasses a method of treating a viral infection by administering a compound of the invention, which method further comprises at least one additional therapy. One embodiment of the method of treating a viral infection further comprises a second antiviral therapy, such as a neuraminidase inhibitor, adefovir, hydroxychloroquine, azithromycin (azithromycin) or a hemagglutinin inhibitor. One embodiment of the method of treating a virus further comprises a drug that modulates an immune system or host cytokine, such as dexamethasone or another corticosteroid; IL-6 inhibitors, such as tolizumab, interferon, IL-1 inhibitors; or a kinase inhibitor such as baroretinib. In another embodiment of the invention, the method further comprises antibody therapy, such as high titer covd-19 convalescence plasma, IVIG; monoclonal antibody therapies, such as carlizumab galectin, pamphleb or pamphleb Shan Kangjia angstrom bevacizumab. One embodiment of the method further comprises additional therapies such as adefovir and/or dexamethasone or other corticosteroids. One embodiment of the method further comprises additional therapies, such as tolizumab. One embodiment of the method further comprises additional therapies, such as baroretinib. One embodiment of the method further comprises additional therapies, such as high titer covd-19 convalescence plasma. One embodiment of the method further comprises additional therapies, such as IVIG. One embodiment of the method further comprises additional therapies, such as karivizumab plus idevezumab. One embodiment of the method further comprises additional therapies, such as bamla mab. One embodiment of the method further comprises additional therapies, such as bamla ney Shan Kangjia angstrom bevacizumab. Another embodiment of the method includes a second antiviral therapy that is at least one of: favirapivir (favipiravir), lopinavir (lopinavir), ritonavir, radevivir, janus kinase inhibitors, hydroxychloroquine, azithromycin (azithromycin), amantadine (amantadine), rimantadine (rimantadine), ribavirin (ribavirin), herpesmine (idoxidine), trifluridine, vidarabine, acyclovir (acyclovir), ganciclovir (ganciclovir), sodium foscarnet (foscarnet), zidovudine (zidovudine), didanosine (peraminosine), peramivir (zalcitabine), stavudine (stavudine), famciclovir (famciclovir), oseltamivir (zaelvir), zafimi (zanamivir) or acyclovir (acyclovir). Another embodiment of the method includes a second therapy that is at least one of: vitamin C or D, zinc, famotidine, ivermectin or Angiotensin Converting Enzyme Inhibitor (ACEI) or angiotensin receptor binding Agent (ARB).
One embodiment of the invention encompasses methods of treating viral infections by administering a compound of the invention, wherein the compound of the invention is administered in an amount of about 0.1mg to about 100 mg. Another embodiment of the invention for treating viral infections by administering a compound of the invention encompasses methods wherein the compound of the invention is administered in an amount of about 1mg to about 90 mg. Another embodiment of the invention for treating viral infections by administering a compound of the invention encompasses methods wherein the compound of the invention is administered in an amount of about 3mg to about 30 mg. Another embodiment of the invention encompasses methods wherein the compounds of the invention are administered in an amount of about 9mg to about 18 mg. Another embodiment of the invention encompasses methods wherein the compounds of the invention are administered in an amount of about 4mg to about 45 mg. In another embodiment of the method, at least one pharmaceutically acceptable excipient is contemplated.
Methods of treating viral infections by administering the compounds of the present invention may be administered in combination with other antiviral therapies to treat the infection or diseases associated with viral infections, e.g., combination therapies. Suitable antiviral agents contemplated for use in combination with the methods of the invention can include nucleoside and Nucleotide Reverse Transcriptase Inhibitor (NRTI) non-nucleoside reverse transcriptase inhibitor (NNRTI), protease inhibitors, and other antiviral agents. Examples of suitable NRTIs include zidovudine (AZT); didanosine (ddI); zalcitabine (ddC); stavudine (d 4T); lamivudine (lamivudine) (3 TC); abacavir (abacavir) (1592U 89); adefovir dipivoxil (adefovir dipivoxil) [ bis (POM) -PMEA ]; lobucavir (BMS-180194); BCH-I0652; emtricitabine [ ((-) -FTC ]; beta-L-FD 4 (called beta-L-D4C and named beta-L-2 ',3' -dichloro-5-fluoro-cytidine); DAPD ((-) - β -D-2, 6-diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delavirdine (BHAP, U-90152); efavirenz (efavirenz) (DMP-266); PNU-142721; AG-1549; typical suitable protease inhibitors include saquinavir (Ro 31-8959), ritonavir (ABT-538), indinavir (indinavir) (MK-639), nelfinavir (nelfinavir) (AG-1343), amprenavir (141W 94), rascinavir (BMS-234475), DMP-450, BMS-2623, ABT-378, and AG-1549 other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuvirucide and YIsu project No. 11607.
Other antiviral agents include, but are not limited to, neuraminidase inhibitors, hemagglutinin inhibitors, hydroxychloroquine, azithromycin, or agents that modulate the immune system or host cytokines, such as dexamethasone. Examples include, but are not limited to, at least one of the following: fapisoravir, lopinavir, ritonavir, redexivir, janus kinase inhibitors, hydroxychloroquine, azithromycin, amantadine, rimantadine, ribavirin, herpeszin, trifluouridine, vidarabine, acyclovir, ganciclovir, sodium foscarnet, zidovudine, didanosine, peramivir, zalcitabine, stavudine, famciclovir, oseltamivir, zanamivir and valacyclovir. One embodiment of the method further comprises additional therapies, such as adefovir and/or dexamethasone. One embodiment of the method further comprises additional therapies, such as karivizumab plus idevezumab. One embodiment of the method further comprises additional therapies, such as bamla mab.
Methods of treating viral infections may also include other therapies. For example, the method may include a second antiviral therapy, such as a neuraminidase inhibitor, adefovir, hydroxychloroquine, azithromycin, or a hemagglutinin inhibitor. Other therapies included in these methods are drugs that modulate the immune system or host cytokines, such as dexamethasone; corticosteroids; IL-6 inhibitors, such as tolizumab; an interferon; an IL-1 inhibitor; or a kinase inhibitor such as baroretinib. The method may further comprise antibody therapy, such as high titer covd-19 convalescence plasma, IVIG, monoclonal antibody therapy, such as karivizumab plus idevezumab, bamla niumab, or bamla niumab Shan Kangjia angstrom niumab. These methods may also include tolizumab or baroretinib. These methods may also include additional therapies, such as high titer covd-19 convalescence plasma; IVIG; carduzumab plus idevezumab; bamanimumab; or pamphleb Shan Kangjia angstrom bevacizumab. The methods may include a second antiviral therapy that is at least one of: famprivir, lopinavir, ritonavir, radevir, janus kinase inhibitors, hydroxychloroquine, azithromycin, amantadine, rimantadine, ribavirin, herpeszin, trifluoracetin, vidarabine, acyclovir, ganciclovir, sodium foscarnet, zidovudine, didanosine, peramivir, zalcitabine, stavudine, famciclovir, oseltamivir, zanamivir or valacyclovir. The methods may include a second therapy that is at least one of vitamin C or D, zinc, famotidine, ivermectin, or an Angiotensin Converting Enzyme Inhibitor (ACEI) or an angiotensin receptor binding Agent (ARB).
The invention also relates to methods of treating inflammation using the above compounds and formulations. These compounds and their formulations have utility in the treatment of inflammation by disrupting tubulin polymerization. The formulations may optionally include additional active ingredients whose activity may be useful in the treatment of diseases associated with inflammation, in the treatment of side effects associated with the dosage of the compound or particular formulation, and/or in delaying or prolonging the release of these ingredients.
Another embodiment of the invention encompasses methods of treating unwanted inflammation by administering a compound of the invention, wherein the inflammation is caused by a viral infection caused by SARS-CoV, MERS-CoV, covd-19 or SARS-CoV-2 virus.
Embodiments of the present invention encompass methods of treating inflammation wherein a compound of the present invention is administered in an amount of about 0.1mg to about 100 mg. Another embodiment of the invention encompasses a method of treating inflammation wherein a compound of the invention is administered in an amount of about 1mg to about 90 mg. Another embodiment of the invention for treating inflammation by administering a compound of the invention encompasses methods wherein the compound of the invention is administered in an amount of about 3mg to about 30 mg. Another embodiment of the invention encompasses a method of treating inflammation wherein a compound of the invention is administered in an amount of about 9mg to about 18 mg. Another embodiment of the invention encompasses a method of treating inflammation wherein a compound of the invention is administered in an amount of about 4mg to about 45 mg. In another embodiment of the method of treating inflammation, at least one pharmaceutically acceptable excipient is contemplated.
The methods of the invention are useful for treating inflammation caused by diseases including, but not limited to, chronic inflammatory diseases and autoimmune diseases. Examples include virus-induced inflammation, arthritis, gout, acute Respiratory Distress Syndrome (ARDS), systemic Acute Respiratory Syndrome (SARS), allergy, alzheimer's disease, asthma, autoimmune diseases, cardiovascular diseases, cancer, chronic obstructive pulmonary disease, celiac disease, crohn's disease, type I diabetes, type II diabetes, endometriosis, fatty liver disease, glomerulonephritis, hepatitis, inflammatory bowel disease, multiple sclerosis, muscular dystrophy (such as duchenne muscular dystrophy (Duchenne muscular dystrophy)), obesity, parkinson's disease, periodontitis, psoriasis, rheumatoid arthritis, sinusitis, pulmonary tuberculosis, ulcerative colitis. a) Preventing, treating or reversing arthritis; b) Preventing, treating or reversing arthritic conditions such as Behcet's disease (autoimmune vasculitis), bursitis, calcium pyrophosphate dihydrate crystals (calcium pyrophosphate dihydrate crystal; CPPD), depositional disease (or pseudogout), carpal tunnel syndrome, connective tissue disease, crohn's disease, elley-swerve syndrome (Ehlers-Danlos syndrome; EDS), fibromyalgia, gout, infectious arthritis, inflammatory bowel disease (inflammatory bowel disease; IBD), juvenile arthritis, systemic lupus erythematosus (systemic lupus erythematosus; SLE), lyme's disease (Lyme's disease), ma Fanzeng syndrome (Marfan syndrome), myositis, osteoarthritis, polyarteritis nodosa, polymyalgia rheumatica, psoriasis, psoriatic arthritis, raynaud's phenomenon (Raynaud's phenomenon), reflex sympathetic dystrophic syndrome, reiter's syndrome (Reiter's syndrome), rheumatoid arthritis, scleroderma, sjogrens 'syndrome (Sjogrens' syndrome), tendinitis or ulcerative colitis; c) Preventing, treating or reversing autoimmune diseases.
The methods of the invention are useful for treating inflammation caused by viruses including Coronaviridae, and possibly viruses of the flaviviridae and herpesviridae superfamily. In addition, the methods of the invention are useful for treating inflammatory responses caused by viruses including, but not limited to RSV, KSHV, CMV, DENV, CHIKV, TBEV, VSV, ZIKV, HCV, SARS, MERS-CoV and COVID-19. Preferably, the method of the invention treats inflammation caused by SARS-CoV, MERS-CoV or COVID-19. The methods of the invention may also be used to treat inflammatory responses caused by herpes viruses, such as herpes simplex virus (HSV-1, HSV-2), varicella Zoster Virus (VZV), cytomegalovirus (CMV) or Epstein-Barr virus (EBV).
The methods of the invention are useful for treating inflammation caused by SARS-CoV, MERS-CoV or SARS-CoV-2, and in particular SARS-CoV-2 infection. The methods of the invention are useful for treating a subject having a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for infection with SARS-CoV-2. The subject may have a SARS-CoV-2 infection that reduces mortality. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces mortality. Another embodiment of the invention encompasses methods of treating a subject with SARS-CoV-2 infection to reduce the incidence of the infection. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces the incidence. Another embodiment of the invention encompasses methods wherein treating a subject with SARS-CoV-2 infection reduces respiratory failure, days in the ICU, days of use of mechanical ventilators, or improves the clinically improved WHO order scale. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces respiratory failure, days in the ICU, days using mechanical ventilators, or improves the clinically improved WHO order scale. Another embodiment of the invention encompasses a method wherein treating a subject with SARS-CoV-2 infection reduces mortality or respiratory failure in subjects older than 60 years. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for a SARS-CoV-2 infection reduces mortality or respiratory failure in subjects >60 years of age. Another embodiment of the invention encompasses methods wherein treating a subject with SARS-CoV-2 infection reduces mortality or respiratory failure when administered in combination with adefovir and/or dexamethasone. Another embodiment of the invention encompasses a method wherein treating a subject with a high risk of Acute Respiratory Distress Syndrome (ARDS) or Severe Acute Respiratory Syndrome (SARS) for SARS-CoV-2 infection reduces mortality or respiratory failure when administered in combination with adefovir and/or dexamethasone.
Biological activity
Fourteen compounds were synthesized and tested for in vitro anticancer efficacy against a panel of melanoma and breast cancer cell lines. The results are shown in table 1 below.
Ten new designed dihydroquinoxalinone analogues are highly potent, in particular 5l, 5m, 5r, 5t, 5u and 5v show promising cytotoxic activity towards all tested cell lines, their IC 50 The values were 0.4nM to 26nM.
Compound 5p also has moderate activity, IC 50 The values are in the range of 19 nanomole to 37 nanomole. Two compounds 5q and 5r with a ring-opened pyrimidine analogs were prepared as shown in fig. 3. Compound 5r was found to be very effective, IC 50 Values were 4 nanomolar to 18 nanomolar (see table 1). Compound 5q, having a chloro-substituted B ring, was found to be inactive. Thus, the a ring is flexible and can be modified to obtain improved potency as well as water solubility. Pyridopyrimidine analogs 5s were prepared because the nitrogen atom at position 1 of 1a and 2a was involved in water-mediated hydrogen bonding with tubulin beta-C239 and beta-V236. Compound 5s has reduced potency compared to pyrimidine counterpart 2a, IC thereof 50 The values were 13nM to 24nM. In addition, the chlorine atom attached to the B ring is located in the pocket between the beta-H7 and beta-T7 rings. The bag is hydrophobic in nature and has more free space allowing further synthetic modifications. Thus, three ethylamine-substituted B-ring analogues 5t, 5u and 5v were synthesized. See fig. 4. The ethylamine moiety is capable of forming hydrogen bonds with the β -H7 backbone and the β -T7 backbone and improving the water solubility of the analog for better in vivo efficacy. Compound 5v showed the highest potency with IC50 values in the pM range of 0.4nM to 0.8nM.
Since the major clinical limitation of existing tubulin inhibitors such as taxanes is their high sensitivity to efflux pumps, compounds 5M, 5t and 5v were evaluated in other cancer cell lines (A375/TxR, M14/LCC6MDR1, MB-231/TxR and A549/TxR) that are highly resistant to taxanes (Table 2). Paclitaxel, colchicine, vitamin Lu Bulin and compound 17ya [ (2- (1H-indol-3-yl) -1H-imidazol-4-yl) (3, 4, 5-trimethoxyphenyl)]Have been used as controls for side-by-side comparisons. Unlike paclitaxel, which lost potency significantly in these cell lines, 5m, 5t, 5v and dimension Lu Bulin retained their potency (table 2). Compounds 5m, 5t and 5v showed one of the most potent cytotoxic activities against melanoma and breast cancer (table1). IC of Compounds 5m, 5t and 5v against taxane-resistant melanoma, breast and lung cancer cells 50 Values ranged from 0.3 nanomolar to 5 nanomolar (table 2). From table 2, compound 5v has an equivalent potency to the paclitaxel resistant cell line as vitamin Lu Bulin, and 5m and 5t are considered to be almost equivalent. Compounds 5m, 5t and 5v differ from compound 17ya in that they retain low nM (1.2 nM to 2.9 nM) activity in compound 17ya resistant triple negative breast cancer cells (MDA-MD-231/VxR; grown in the presence of 100nM compound 17 ya), which is comparable to their activity in the MDA-MB-231 parental line (1.0 nM to 2.2 nM). In contrast, compound 17ya showed at least 100-fold resistance (903.4 nM versus 6.1 nM) activity.
The main obstacle to drug discovery is insufficient in vivo stability of the compounds, which leads to rapid loss of pharmacological activity and adverse effects due to the formation of potentially toxic metabolites. Thus, half-life and clearance of compound 5m in human and mouse liver microsomes were measured.
Table 3 summarizes the data.
The results indicated that compound 5m had acceptable microsomal stability, which was significantly higher in humans than rodents.
In vivo pharmacokinetic studies of compound 5m in rats after intravenous and oral administration showed low cumulative urine output in unchanged form, indicating a high degree of metabolic conversion. The in vivo terminal half-life was 14.7 hours long, probably due to the large volume distribution. Systemic exposure, quantified as area under the concentration-time curve, was almost twice as high per milligram dose as that of vitamin Lu Bulin observed in mice [ data not shown ]. The oral bioavailability of 5m was 3.2%. Table 4 summarizes the data.
Evaluation of antitumor Activity of Compound 5m in pancreatic cancer
Pancreatic Ductal Adenocarcinoma (PDAC) is a lethal malignancy with high mortality. Compound 5m is one of many dihydroquinoxalinones reported herein that acts as a tubulin inhibitor targeting the colchicine binding site, supporting their use in PDACs and various other cancer types. The in vitro effect of compound 5m on PDAC cell lines was surprising compared to one of the first line treatment options (paclitaxel). This supported in vivo test of compound 5m was effective in the luciferase-labeled Mia PaCa-2 cell line Mia PaCa-2-luc for live monitoring of tumor progression and evaluation of anti-tumor effects in a subcutaneous mouse model. 5m showed significant tumor growth inhibition and limited overall toxicity in Mia PaCa-2-luc xenografts (fig. 23). Similarly, PANC-1-luc xenografts treated with 5m also showed potent and dose-dependent anti-cancer activity of compound 5m in PDAC. The results of example 7 show that compound 5m is dose-dependently effective in inhibiting cell proliferation, colony formation and cell migration at low nanomolar concentrations. Immunoblots also demonstrated that compound 5m induced apoptosis in a dose-dependent manner. Cell cycle arrest assay demonstrated that compound 5M arrested cells in G2/M phase. In vivo studies showed that compound 5m significantly inhibited tumor growth of PDAC tumors with little or no low toxicity in the subcutaneous xenograft model. Preclinical data shows that compound 5m inhibits proliferation, cell migration and induces apoptosis in PDAC cells, indicating a chemotherapeutic agent to treat PDAC.
Inhibition of tubulin polymerization
To determine that these compounds exhibit potent antiproliferative activity due to binding to microtubules, a cell-free tubulin polymerization assay was performed using both compounds 5m and 5t and the reference compounds colchicine and paclitaxel. As shown in fig. 5, compounds 5m and 5t significantly inhibited microtubule polymerization, similar to colchicine as a positive control. The negative control paclitaxel showed the expected acceleration of microtubule nucleation and growth, thus causing enhanced polymerization. Consistent with the tubulin polymerization assay, compounds 5m and 5t showed soluble cytoplasmic tubulin associated with microtubule fragmentation, resulting in severe disruption of microtubule dynamics in interphase a375/TxR cells (fig. 5B). Whereas during the interphase, a regular network of microtubules was observed in cells in the vehicle control group, which was packed towards the nucleus with unconcentrated chromosomes. Colchicine or paclitaxel treated cells also showed intact tubulin networks with normal filamentous arrangements at a concentration of 2 nM. In comparison to mitotic cells with a normal functioning mitotic spindle in the control, colchicine or paclitaxel groups, 2nM of 5m treated cells were defective in the assembly of the mitotic spindle, forming multipolar spindles and misaligned chromosomes. Similar results were observed in mitotic cells treated with 2nM 5 t.
X-ray crystallography of compounds 5j, 5k, 5l, 5m and 5t complexed with tubulin
The molecular interactions of compounds with the colchicine binding site were studied by the tubulin crystal structure complexed with: 5j (PDB: 6X1C,resolution), 5k (PDB: 6X1E, < >>Resolution), 5l (PDB: 6X1E, < >>Resolution), 5m (PDB: 6X1F,)>Resolution) and 5t (PDB: 7LZ8, < >>Resolution) (fig. 6B to 6G). These crystal structures show that the engineered analogs bind to the colchicine binding site as expected.
All five designed analogues had a binding orientation in which the a-and B-loops go deep into the β -tubulin pocket and the C-and D-loops are in the interface with α -tubulin (fig. 6B-6G). Most molecular interactions of co-crystallizing ligands with tubulin are hydrophobic in nature, including α0-A352 from sheet S9, α1-A314 and α2-I316 from sheet S8, α6-I368 from sheet S10, α7-L253 and α9-M257 from helix H8, βL246 and β1-A248 from loop T7, β3-L240, β -C239 and β -V236 from helix H7, as observed for compounds 1a and 2 a. The NH group from the amide portion of the C-ring acts as a hydrogen bond donor from the backbone carbonyl α3-T179 of ring T5. The carbonyl group of the amide moiety in the C-ring forms additional water mediated hydrogen bonds with α4-T179 and α5-N101, as observed in FIG. 2 a. Both compounds 5j and 5m also retain the N atom in the B-ring and water-mediated H-bonding between β -C239 and β -V236 from helix H7. As with compounds 1a and 2a, all co-crystalline structures present new H-bonds between α8-S178 from ring T5 and β -K350 from sheet S9. Thus, these findings indicate that the new analogues with hydrophobic a-rings as well as amide C-rings have a stronger interaction with the β0-tubulin monomers and the β -tubulin monomers and thus bring them closer together into a tighter conformation, leading to the possibility of H-bonding between residues from the β2-monomers and the β -monomers. The 5m eutectic structure binds better to the colchicine binding site than other compounds. The strongest binding of 5m to tubulin depends on low single digit nanomolar IC against different conventional and paclitaxel resistant melanoma, breast and lung cancer cell lines 50 Experimental findings of values (1 nM to 5 nM). Dihydroquinoxalinone analogues (5 l, 5 m) with saturated hydrophobic a-rings have a stronger binding than previously disclosed 1a and 2a, resulting in potentially improved antitumor efficacy.
Inhibition of colony formation and migration of paclitaxel-resistant melanoma cells.Based on sensitivity (parent) of dihydroquinoxalinone analogues to different taxanesResults of cytotoxic effects of paclitaxel resistant cancer cell lines (tables 1 and 2), two analogs 5m and 5t were selected for further in vitro and in vivo experiments to investigate the anti-cancer mechanism against a375/TxR cells. Since cancer cells proliferate by forming colonies, inhibition of colony formation is considered as a key attribute of good anticancer drugs. Clonogenic assays were performed to investigate the efficacy of 5m and 5t in inhibiting colony formation.
FIGS. 7A and 7B show that the newly synthesized compounds inhibited colony formation in A375/TxR cells in a concentration-dependent manner. Compounds 5m and 5t showed a decrease in colony number and size compared to the control group and inhibited cell growth over a long exposure time (7 days) even at very low concentrations (1 nM). Compounds 5m and 5t completely inhibited colony formation at high doses (i.e., 2 nM) (fig. 7A).
Since cancer cell migration is very important in tumor progression and metastasis, and metastasis is the most common contributing factor leading to high mortality in cancer patients, compounds 5m and 5t were tested to inhibit migration of a375/TxR cells by using a wound healing assay. Figure 8A shows that 24 hours after wound, the cell monolayer DMSO-treated control cells filled the wound area, while the wound in 2nM or 5nM 5 m-treated cells healed slower than the control group. 5nM 5m significantly inhibited migration of A375/TxR cells, even after 12 hours of incubation. FIG. 8B shows that 5t treatment has a strong ability to inhibit migration of A375/TxR cells and its efficacy is the same as 5 m. In summary, the results indicate that 5m and 5t are effective in inhibiting colony formation and migration of cancer cells.
Compound 5M and 5t treatment caused G2/M cell cycle arrest and apoptosis in a375/TxR cells.As shown in fig. 5B, a flow cytometry-based cell cycle analysis was performed to evaluate the effect of 5m and 5t on cell mitosis, considering the critical role of inhibiting microtubule polymerization in disrupting cell cycle progression and mitotic spindle defects observed in 5m and 5t treated a375/TxR cells. A375/TxR cells were treated with compounds at different concentrations (1 nM, 2nM and 5 nM) for 5m or 5t for 24 hours without serum starvation. FIG. 9A shows the normal cell cycle distribution (G1: 48%; S33) in comparison to control cells The%; G2/M: 19%) of the comparison, 5M or 5t treatment induced significant cell cycle arrest in the G2/M phase concentration dependence. At a concentration of 2nM, 5M and 5t arrested A375/TxR cells at G2/M phase at percentages of 60% and 64%, respectively. The results show that 5M and 5t can effectively induce cell cycle arrest in the G2/M phase against a375/TxR cells and affect cell mitosis even without serum starvation.
Using ModFit LT TM The software analyzed the cell cycle distribution and a significant peak of apoptosis in the sub G1 region was observed after 24 hours of treatment with 5M or 5t (we gated this cell population to quantify the percentage of correct cells in G2/M phase in fig. 9A). The annexin V-FITC/PI staining assay demonstrated the effect of 5m and 5t on apoptosis induction in A375/TxR cells using the same treatments as in the cell cycle assay. The total percentage of apoptotic cells is the sum of early and late apoptotic cells relative to the total number of cells. As can be seen from fig. 9B, the percentage of apoptotic cells after 24 hours treatment was only 9% in the control group, whereas after treatment with 5m and 5t of 2nM, apoptotic cells increased to 30% and 33%, respectively. In addition, compounds 5m and 5t induced apoptosis of A375/TxR in a concentration-dependent manner. In conclusion, compounds 5m and 5t significantly caused apoptosis of tumor cells, consistent with their activity in antiproliferative and cell cycle arrest.
In vivo experiments of Compounds 5m and 5t against A375/TxR xenograft models.Test compounds 5m and 5t treat tumors in a375/TxR xenograft model to determine the potential of dihydroquinoxalinone pyrimidine analogs for in vivo treatment of cancer. A375/TxR melanoma cells were developed to be resistant to paclitaxel treatment. This tumor model can be used to study the therapeutic benefit of newer agents over existing therapies where acquired resistance is a major issue. Treatment with compound 5m inhibited a375/TxR melanoma tumor growth in a dose-dependent manner (fig. 10A). At a dose of 4mg/kg, 5m significantly inhibited tumor growth throughout the study period (p= 0.0452). Treatment with 2mg/kg 5m also resulted in 70.45% tumor reduction compared to control (88.18% compared to control) compared to 4 mg/kg. Purple (purple)The paclitaxel therapy was ineffective and showed a similar tumor growth trend as the vehicle-treated group, indicating drug resistance. Individual mice body weight was monitored during the study. The results showed consistent weight gain without any significant adverse effects caused by the test compounds (fig. 10B). At the end of the study, tumors were collected and weighed. Tumor weights of mice treated with 2mg/kg and 4mg/kg 5m were reduced by 76.7% and 91.8%, respectively, compared to vehicle group (FIGS. 10C and 10D). Relatively high doses of paclitaxel (10 mg/kg) did not show the benefit of tumor weight reduction. The in vivo antitumor efficacy of compound 5t was evaluated in the same a375/TxR melanoma model, using paclitaxel treatment as a positive control. Since in A375/TxR cells, 5t of IC 50 (1.3 nM) is slightly higher than 5m (1.1 nM), so the dose of 5t is increased to 2.5mg/kg and 5mg/kg in mice bearing A375/TxR tumors during treatment. Both 2.5mg/kg and 5mg/kg 5t treatments showed excellent antitumor efficacy compared to the control and paclitaxel-treated groups (FIG. 11A). Treatment with 2.5mg/kg and 5mg/kg 5t reduced the final a375/TxR tumor volume in mice at a rate of 64.63% and 78.38%, respectively (p=0.0374) compared to vehicle-treated groups. There was no significant difference in body weight between the different treatment groups compared to the control (fig. 11B). Furthermore, compound 5t dose-dependently reduced tumor weight in mice (67.38% lower in the 2.5mg/kg treated group and 78.65% lower in the 5mg/kg treated group compared to vehicle group), and the results were remarkable (p<0.0001 (fig. 11C). Photographs of all tumors in this study further confirm the in vivo efficacy of 5t (fig. 11D). In conclusion, compound 5t showed enhanced antitumor efficacy against the paclitaxel resistant melanoma model without significant side effects.
Compound 5m and 5t treatment induced tumor necrosis in vivo.Because of the strong role of compounds 5m and 5t in apoptosis induction observed in vitro, in vivo destructive effects of 5m and 5t were studied using tumors in a375/TxR xenograft model, as shown in fig. 10 and 11. After measurement and imaging, tumors were fixed in 10% buffered formalin, embedded in paraffin and sectioned for H &E staining. As shown in fig. 12, in the vehicle-treated group, the tumor cells appeared to have circlesShape and normal shape of intact nuclei, and tumor cells are closely and constantly arranged. Paclitaxel-treated tumor cells showed similar cell morphology and distribution. In contrast, after 5m or 5t treatment, tumor cells were loosely and unevenly arranged, and a broad necrotic area with several necrotic cells was clearly observed in the tumor, and the tumor necrotic area increased in a dose-dependent manner. These results confirm the potent antitumor capacity of 5m and 5 t.
Compound 5m and 5t treatments inhibited spontaneous lung and liver metastasis.Malignant melanoma is a dangerous disease with invasive metastatic potential, a process in which tumor cells spread from a primary site to internal organs. One of the main steps of metastasis is the formation of a tumor microenvironment in the internal organs that is suitable for the survival and growth of tumor cells. Many studies report that spontaneous metastasis xenograft models are widely used to study the critical steps of such metastasis, such as the occurrence of lung or brain metastasis caused by melanoma. The effect of compounds 5m and 5t in inhibiting spontaneous melanoma metastasis in vivo was studied using lung and liver tissue of a375/TxR subcutaneous xenograft model (see above) in which spontaneous lung metastasis would occur. First, H of lung tissue of 5m xenograft model was performed &E staining.
Fig. 15A shows a number of large metastases (yellow arrows) observed in the control group, indicating severe melanoma lung metastases. Whereas the 5m treatment group showed a limited number of smaller size metastases, and the 5m treatment dose dependently inhibited lung metastases. The paclitaxel-treated group served as a positive control, which had no effect on spontaneous lung metastasis compared to vehicle treatment. FIG. 13A is a graphical representation of the quantitative analysis of tumor burden in the lung further demonstrating the inhibition of metastatic tumor cell migration by 5 m. Since liver is another major organ for seeding and growth of tumor cells, H & E staining of liver tissue was performed. The decrease in liver surface metastasis in the 5m treated group demonstrated the efficacy of 5m in inhibiting spontaneous migration of melanoma, as shown in fig. 15B and 13B. The livers of 4mg/kg 5m treated mice were all clean, demonstrating the strong inhibitory effect of 5m on liver metastasis. Significant inhibition of tumor migration by 5m treatment was further demonstrated by a decrease in the density of anti-human mitochondrial immunostaining (brown stained tissue) in lung or liver sections, consistent with the results obtained by H & E staining (fig. 13C, 13D and 16). Considering the promising data obtained from the 5m xenograft model, H & E staining of lung and liver tissue in the 5t xenograft model was performed. Depending on the final tumor weight, the number of lung and liver metastases was increased in the control or paclitaxel group from the 5t xenograft model relative to the control or paclitaxel group from the 5m xenograft model, confirming the difference in tumor progression between the two xenograft models, as shown in fig. 17 and 14A-14B. H & E staining results showed a strong effect of 5t treatment on inhibition of lung and liver metastasis in a dose-dependent manner. In addition, anti-human mitochondrial IHC staining also showed sparse and smaller metastasis in 5t treated mice, and as the dose of 5t increased, the number and size of metastases decreased, as shown in fig. 14C and 14D and fig. 18. These data support the role of 5m and 5t as potent tubulin destabilizers in inhibiting spontaneous metastasis of melanoma to mouse lung and liver.
Compound 5m in melanoma (A375/TxR), prostate cancer (DU-145/VxR and 22RV 1), breast cancer (MDA-MD- Overcoming P-taxane and/or Compound 17ya or in vivo xenograft models of 231/Vx) and ovarian cancer (A2780/TxR) Resistance to castration.
To determine whether dihydroquinoxalinone pyrimidine analogs have potential for in vivo treatment of cancer, compound 5m was selected as a representative compound for treatment of tumors in the a375/TxR xenograft model. A375/TxR melanoma cells were developed to be resistant to paclitaxel treatment. Thus, this tumor model may be very helpful in studying the therapeutic benefit of new agents over existing therapies where acquired resistance is a major issue. We found that 5m treatment strongly inhibited a375/TxR melanoma tumor growth in a dose-dependent manner (fig. 19A). At a dose of 4mg/kg, 5m significantly inhibited tumor growth throughout the study period (p= 0.0452). Treatment with 2mg/kg 5m also resulted in a 70.5% tumor reduction compared to the control, which was slightly less than a 4mg/kg tumor reduction (88.2% compared to the control). As expected, 10mg/kg paclitaxel therapy was ineffective and showed similar tumor growth trend as vehicle-treated group, indicating taxane drug resistance.
To assess the effect of 5m on the resistance of compound 17ya (VxR) [ Compound 17ya is (2- (1H-indol-3-yl) -1H-imidazol-4-yl) (3, 4, 5-trimethoxyphenyl)]Compound 17ya resistant DU-145/VxR cells and MDA-MB-231/VxR cells were generated by continuous incubation of the cells with compound 17 ya. When the cells were resistant to compound 17ya at 100nM, both cell lines were confirmed to be resistant to compound 17ya and expanded for in vivo studies. Interestingly, DU-145/VxR cells and MDA-MB-231/VxR cells grew slowly in tumor bearing mice. We wait 16 days until the average tumor volume reaches about 80mm 3 And treatment is started. During treatment, we found a significant tumor suppression effect of 5m on DU-145/VxR xenografts (fig. 19B). The mean tumor volume at the end point of the group IV 5m (2 x/wk) at 1mg/kg was almost the same as that at the start point and was 80mm 3 Whereas the control group had an end point average tumor volume of about 340mm 3 . Surprisingly, a high dose of 20mg/kg of the PO compound 17ya (3 x/wk) was still effective in this compound 17ya resistant prostate cancer model. Unlike 5m, compound 17ya has high oral bioavailability.]The mean tumor volume at the end point was about 190mm between the control group and the 5m group 3 . Nevertheless, the liquid is not allowed to flow through the device,>compound 17ya at 20-fold dose had a worse efficacy than 5 m.
We also used the in situ MDA-MB-231/VxR xenograft model to determine the effect of 5m on Triple Negative Breast Cancer (TNBC) that is resistant to compound 17 ya. This time we included 10mg/kg paclitaxel and 20mg/kg compound 17ya as controls. To alleviate the discomfort of IV injected mice we increased the dose of 5m to 2mg/kg and reduced the dose frequency from 2 times per week to 1 time per week. Similar to the DU-145/VxR xenograft model, 20mg/kg of compound 17ya (3 x/wk) effectively inhibited the growth of MDA-MB-231/VxR xenografts, although its antitumor efficacy was weaker than that of paclitaxel (10 mg/kg IV 3 x/wk) or 5m (1 mg/kg IV 1 x/wk) treatment, thus indicating that compound 17ya may have antitumor activity even when compound 17ya resistance developed (FIG. 19C). As reflected by the flat tumor growth curve shown in fig. 19C, tumor growth inhibition was still evident in the 5m treated group. And its antitumor effect is greater than that of paclitaxel or compound 17ya, even at very low doses and with a lower frequency of administration. Both compounds 17ya and 5m act through the colchicine binding site of tubulin, and thus unexpectedly, 5m will be able to overcome the resistance to compound 17ya even at much lower doses than compound 17 ya. Furthermore, other compounds of the invention having a different structure and thus a different binding pattern than 17ya are expected to also be able to overcome 17ya resistant tumors.
Additional animal studies were performed using 22RV1 cells to determine the effect of 5m on castration resistant prostate cancer. We divided mice into untreated control groups and 1mg/kg 5m treated groups based on tumor volume and mouse body weight. This is the first time we evaluate the efficacy of 5m to overcome castration resistance of the 22RV1 prostate cancer xenograft model, so once mice are ready for treatment we keep the original dose and dose frequency of 5m (1 mg/kg, IV,2 times/week). As shown in fig. 19D, 1mg/kg 5m significantly inhibited the growth of 22RV1 xenografts (p < 0.0001), demonstrating the strong anti-tumor capacity of 5m for castration resistant prostate cancer models.
Furthermore, in addition to melanoma, prostate and breast cancer models, we tested 5m for effect on invasive orthotopic ovarian cancer models. We selected paclitaxel resistant A2780/TxR cells for this study. Similar to the other models, we treated mice for 3 weeks and collected all tumors at the study endpoint. As shown in FIG. 19E, we demonstrate that paclitaxel has limited effect on this invasive A2780/TxR model, even though the IV injection dose is 5mg/kg. While at a dose of 1mg/kg 5m, tumor growth was significantly inhibited, with only 1 out of 5 mice having visible tumors. And of 20mg/kg compound 17ya, 2 mice had visible tumors and tumor sizes were greater than those in the 5m treatment group. Furthermore, the tumor weight of each group demonstrated a strong antitumor efficacy of 5m against a2780/TxR ovarian cancer model, and its efficacy was greater than that of paclitaxel and compound 17ya (fig. 19F). While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those skilled in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.
Analogs of compounds 10 and 12a-12q were tested for cytotoxic activity against a panel of cancer cell lines such as melanoma (A375, M14), breast cancer (MDA-MB-231, MDA-MB-453), pancreatic cancer (Mia PaCa-2, PANC-1) and prostate cancer (PC 3, PC 3/TxR). The half maximal inhibitory concentration values (IC) for inhibition of cell growth are summarized in example 9 50 ). This study reveals that the size of the heteroatoms has a significant effect on the cytotoxic potency, and reducing the size of the heteroatoms tends to increase potency. For example, thioether 10 (IC 50 Approximately 3.4 nM.+ -. 0.5nM, A375 cell line), ether 12b (IC 50 Approximately 3.2 nM.+ -. 0.5 nM) and secondary amine 12k (IC 50 Approximately 1.2nM ± 0.2 nM) is relatively small and has a single digit nM potency. However, other than morpholine derivatives exhibiting moderate to high potency, such as N-methylpiperazine 12d (IC 50 About 542.8 nM.+ -. 111.0 nM), morpholine 12e (IC 50 About 13.6 nM.+ -. 2.0 nM), piperidine 12f (IC 50 Approximately 436.1 nM.+ -. 76.2 nM) and pyrrolidine 12g (IC 50 Substitutions of cyclic derivatives of +.82.1 nM.+ -. 12.9 nM) have proven to be relatively low in potency. Aromatic heterocycles (i.e. imidazole 12h (IC 50 Approximately 5.7nM ± 0.9 nM)). Tertiary amine 12I (IC) 50 The derivative of +.22.6 nM.+ -. 4.5 nM) shows moderate potency. The results obtained with compound 12k were a study of secondary amines (such as N-ethyl 5v (IC 50 Approximately 1.6 nM.+ -. 0.3 nM) and N-cyclopropyl 12j (IC 50 The pharmacological potency of 1.4 nM.+ -. 0.3 nM) paves the way, and these secondary amines are highly potent. Of the molecules studied, 12k is the best of the three secondary amines. Adding additional hydrogen bonding donors, such as 12m (IC 50 the-OH group in the ethanolamine moiety of +.8.6nM.+ -. 0.2nM is slightly less potent than 5v (ethylamine form) and isothiocyanate derivative 12l (IC 50 Approximately 3.3 nM.+ -. 0.5 nM) also shows very good potency. In pyrimidine (2-Py) ring 12a (IC 50 An unprotected phenolic OH at C2 position on 646.5 nM.+ -. 124.2 nM) as otherElectron withdrawing groups (such as sulfone derivative 11 (IC) 50 And +.84.9 nM.+ -.17 nM)) as well as significantly reducing potency. On the other hand, the free amine 12c at the same position yields improved potency (IC 50 2.01 nM.+ -. 0.4 nM). Compound 12o-12p (i.e., OCF) as a surrogate for OMe group 3 OBn and OH) are not well tolerated, resulting in reduced efficacy. The general trend for aryl substituents is that 4-OMe compounds have the highest affinity, with 4-OCF 3 (12o,IC 50 ≈43.1±6.9)、OH(12q,IC 50 The compounds of ≡19.0±2.9) are moderate in potency and the compounds with an OBn (12 p) substitution are the lowest in potency.
Examples
While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those skilled in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.
Chemical: general procedure. All nonaqueous reactions were carried out in oven-dried glassware under an inert atmosphere of dry nitrogen. All reagents and solvents were purchased from Aldrich (St. Louis, mo.), alfa-Aesar (Ward Hill, mass.), combi-Blocks (San Diego, calif.), ark Pharm (Libertyville, ill.) and used without further purification. Analytical thin layer chromatography was performed on silica gel GHLF 10cm×20cm Analtech TLC Uniplates (Analtech, newark, DE) and visualized by fluorescence quenching under UV light. The compounds were purified using a Biotage SP1 flash chromatography purification system (Charlotte, NC) (Biotage SNAP cartridge, silica, 50g and 100 g). Recording on a Varian Inova-500 spectrometer (500 MHz) (Agilent Technologies, santa Clara, calif.) or a Bruker Assend 400 (400 MHz) (Billerica, mass.) spectrometer 1 H NMR 13 C NMR spectrum. Chemical shifts are reported in ppm on delta scale and referenced to the appropriate solvent residual peak (CDCl) 3 For the following 1 H is 7.26ppm and for 13 C is 77.23ppm; DMSO-d 6 For the following 1 H is 2.50ppm and for 13 C is 39.51 ppm). In BrukerMass spectra were collected in positive and negative modes on an ESQUIRE electrospray/ion trap instrument. High Resolution Mass Spectrometer (HRMS) data were obtained on a Waters Xevo G2-S qTOF (Milford, MA) system equipped with an Acquity I type UPLC system. Porcine brain tubulin (catalog number T-238P) was purchased from Cytoskeleton, inc. By passing through 1 The purity of all test compounds was not less than 95% as determined by H NMR and HPLC. The HPLC method for determining purity is as follows: compound purity was analyzed using an Agilent 1100HPLC system (Santa Clara, CA) with a Zorbax SB-C18 column (particle size 3.5 μm,4.6mm x 150 mm) from Agilent. The mobile phase consisted of water (a) with 0.1% formic acid and acetonitrile (B) with 0.1% formic acid. A flow rate of 1mL/min was used. Gradient elution was started from 50% B. It reaches 100% B from 0 to 9 minutes, remains from 9 to 12 minutes, then decreases from 12 to 15 minutes to 50% B and stops. Compound purity was monitored with a DAD detector set at 254 nm.
Example 1:2- ((5-fluoro-4-methoxy-2-nitrophenyl) amino) acetic acid ethyl ester (7 a) or 2- ((4-methoxy-) 2-nitrophenyl) amino) Synthesis of ethyl acetate (7 b) (FIG. 3)
Synthesis of ethyl 2- ((5-fluoro-4-methoxy-2-nitrophenyl) amino) acetate (7 a) or ethyl 2- ((4-methoxy-2-nitrophenyl) amino) acetate (7 b). A1000 mL three-necked flask was charged with 25g of commercially available 5-fluoro-4-methoxy-2-nitroaniline (148.7 mmol) (compound 6 a) or 25g of 4-methoxy-2-nitroaniline (compound 6 b). A100 mL volume of ethyl bromoacetate (901.8 mmol) was slowly poured into the flask under an argon atmosphere. K in an amount of 102.7g 2 CO 3 (743.5 mmol) was added to the solution. The mixture was heated to reflux for 12 hours. The mixture was cooled to room temperature and diluted with EtOAc (250 mL). The organic layer was extracted with water, over MgSO 4 Drying and evaporating to dryness gave the crude product. Then by column chromatography at 20% Et 2 The crude product was purified in O/hexane to give compound 7a or 7b (20 g, 53-55%) as a red solid powder. 1 H NMR(7a)(CDCl 3 ,400MHz)δ8.31(bs,1H),7.87(d,1H,J=7.19Hz),6.45(d,1H,J=7.12Hz),4.30(q,2H,J=7.28Hz),4.10(m,2H),3.82(s,3H),1.33(t,3H,J=7.19Hz)。 1 HNMR(7b)(CDCl 3 ,400MHz)δ8.30(bs,1H),7.68(m,1H),7.18(m,1H),6.69(d,1H,J=8.27Hz),4.30(q,2H,J=7.28Hz),4.10(s,2H),3.82(s,3H),1.33(t,3H,J=7.19Hz)。
Example 2: 6-fluoro-7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (8 a) or 7-methoxy-3, 4-dihydro Synthesis of quinoxalin-2 (1H) -one (8 b)
Synthesis of 6-fluoro-7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (8 a) or 7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (8 b): an amount of 19g of Compound 7a (69.7 mmol) or 7b (74.7 mmol) was dissolved in 150mL of a solution containing 3% CH 2 Cl 2 Is contained in MeOH. 2g of 10% Pd-C were carefully added to the solution. Then at H 2 The reaction was continued for 5 hours under an atmosphere, at which point the reaction was complete as inferred by TLC. Passing the reactants throughThe bed was filtered and the filtrate was evaporated to dryness to give pure compound 8a (8.88 gm,45.3mmol, 65%) or 8b (12.47 gm,71.7mmol, 96%) as a light brown solid. 1 H NMR(8a)(DMSO-d 6 ,400MHz)δ10.16(bs,1H),6.60(m,2H),5.8(s,1H),3.63(m,5H)。 1 H NMR(8b)(DMSO-d 6 400 MHz) delta 10.16 (bs, 1H), 6.60 (d, 1H, j=8.0 Hz), 6.38 (m, 2H), 3.63 (s, 6H). Compounds 8a and 8b were then used in the final coupling step without further purification.
Example 3: general procedure for preparation of the dihydroquinazolinone-pyrimidine/pyridine analogues (5 i-5 s) (FIG. 3).
Compounds 5i-5s were prepared according to either procedure A or procedure B.
Procedure AA solution (1 eq) of a commercially available 2, 4-dichloropyrimidine/pyridine analog 3b-3l was added to 10mL of anhydrous isopropanol followed by the addition of solid head groups 8a or 8b (1 eq). To the solution was added a catalytic amount of concentrated HCl (3 drops to 4 drops) and the solution was stirred under an argon atmosphere for 12 hours. The reaction was diluted with water and extracted with dichloromethane (3X 30 mL). The organic layer was neutralized with saturated sodium bicarbonate solution and then with MgSO 4 And (5) drying. Concentrating the organic layerThen a silica column (20% -30% EtOAc/CH was used 2 Cl 2 ) The crude material obtained was purified to give the pure product as a solid.
Procedure B: commercial pyrimidine/pyridine analogues 3b-3l (1 eq) were dissolved in 10mL absolute ethanol, then solid head groups 8a or 8b (1 eq) were added. Sodium carbonate (2 eq) was added to the solution and the solution was refluxed under argon for 12 hours. The reaction was diluted with water, extracted with dichloromethane (3X 30 mL) and then over MgSO 4 And (5) drying. The organic layer was concentrated and then purified using a silica column (20% -30% EtOAc/CH 2 Cl 2 ) The crude product obtained was purified to give the pure product as a solid.
4- (2-chloropyridine [3, 2-d)]Synthesis of pyrimidin-4-yl) -6-fluoro-7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 i) (FIG. 3): compound 5i was prepared according to procedure a, a general procedure used for the preparation of 5i-5 s. To a solution of 216mg of compound 8a (1.1 mmol) in 20mL of anhydrous isopropanol was added 3b (1 mmol) in an amount of 200mg followed by a catalytic amount of HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as pale yellow solid (252 mg,0.7mmol, 70%). 1 H NMR(400MHz,DMSO-d 6 )δ10.72(s,1H),8.81(d,J=3.2Hz,1H),8.21(d,J=8.3Hz,1H),7.90(dd,J=8.6,4.2Hz,1H),7.34(d,J=12.6Hz,1H),6.77(d,J=8.3Hz,1H),5.03(s,2H),3.85(s,3H)。 13 C NMR(101MHz,DMSO)δ167.13,160.52,155.64,149.36,149.02,145.80,145.68,145.30,135.77,132.86,129.42,128.59,120.97,112.44,112.21,56.52,51.84。HRMS[C 16 H 12 ClFN 5 O 2 + ]Calculated 360.0664, found 360.0650.HPLC purity 96.20% (t) R =2.60 minutes).
4- (2-Chlorofurano [3, 2-d) ]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 j) (FIG. 3): compound 5j was prepared according to procedure a, a general procedure used for the preparation of 5i-5 s. To a solution of 196mg of compound 8b (1.1 mmol) in 20mL of anhydrous isopropanol was added 3c (1 mmol) in an amount of 188mg followed by a catalytic amount of HCl (3 drops to 4 drops). Then by flash silica (30% EtOAc)/CH 2 Cl 2 ) The crude product was purified to give the pure product as a pale yellow solid (235 mg,0.71mmol, 71%). 1 H NMR(400MHz,DMSO-d 6 )δ10.75(s,1H),8.30(d,J=2.0Hz,1H),7.34(d,J=8.8Hz,1H),7.06(d,J=2.0Hz,1H),6.72–6.49(m,2H),4.67(s,2H),3.77(s,3H)。 13 C NMR(101MHz,DMSO)δ167.28,157.98,155.11,153.16,152.50,146.50,133.85,133.26,124.46,119.66,107.81,107.52,101.89,55.84,31.17。HRMS[C 15 H 12 ClN 4 O 3 + ]Calculated 331.0598, found 331.0584.HPLC purity 96.27% (t) R =2.53 min).
4- (3, 6-dimethylisoxazolo [5, 4-d)]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 k) (FIG. 3; r is H): compound 5k was prepared according to procedure B, which was used to prepare general procedure 5i-5 s. To a solution of 196mg of compound 8b (1.1 mmol) in 20mL of anhydrous EtOH was added 3d (1 mmol) in an amount of 184mg followed by K 2 CO 3 (276 mg,2 mmol). By flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a white solid (221 mg,0.68mmol, 68%). 1 HNMR(400MHz,DMSO-d 6 )δ10.79(s,1H),7.21(d,J=8.8Hz,1H),6.80–6.44(m,2H),4.51(s,2H),3.75(s,3H),2.60(s,3H),1.65(s,3H)。 13 C NMR(101MHz,DMSO)δ177.03,168.19,167.80,157.86,156.97,154.52,133.63,122.77,121.17,108.55,102.80,97.97,55.91,50.35,26.16,12.69。HRMS[C 16 H 16 N 5 O 3 + ]Calculated 326.1253, found 326.1294.HPLC purity of 99.6% (t) R =2.37 min).
4- (2-chloro-6, 7-dihydro-5H-cyclopenta [ d ] ]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 l): compound 5l was prepared following procedure A, which was used to prepare 5i-5 s. To a solution of 196mg of compound 8b (1.1 mmol) in 20mL of anhydrous isopropanol was added 3e (1 mmol) in an amount of 188mg followed by a catalytic amount of HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a white solid (205 mg,0.62mmol, 62%). 1 H NMR(400MHz,DMSO-d 6 )δ10.72(s,1H),6.93(d,J=8.5Hz,1H),6.60(d,J=9.5Hz,2H),4.44(s,2H),3.74(s,3H),2.78(t,J=7.6Hz,2H),2.18(t,J=7.1Hz,2H),1.97–1.73(m,2H)。 13 C NMR(101MHz,DMSO)177.90,167.34,157.61,157.28,156.94,132.64,122.80,119.87,118.46,106.80,101.63,55.32,49.25,33.33,30.32,21.98。HRMS[C 16 H 16 ClN 4 O 2 + ]Calculated 331.0962, found 331.0974.HPLC purity 99.4% (t) R =2.59 minutes).
7-methoxy-4- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (5 m): compound 5m was prepared according to procedure a, a general procedure used for the preparation of 5i-5 s. To a solution of 256mg of compound 8b (1.4 mmol) in 20mL of anhydrous isopropanol was added an amount of 220mg of 3f (1.3 mmol) followed by a catalytic amount of HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give the pure product as a reddish solid (323 mg,1.04mmol, 80%). 1 H NMR(400MHz,DMSO-d 6 )δ10.66(s,1H),6.79(d,J=9.2Hz,1H),6.58(s,2H),4.43(s,2H),3.73(s,3H),2.74(t,J=7.3Hz,2H),2.47(s,3H),2.17(t,J=6.8Hz,2H),1.92–1.74(m,2H)。 13 C NMR(101MHz,DMSO)δ175.16,167.81,165.09,156.33,156.22,132.23,121.84,121.07,116.34,106.79,101.64,55.27,49.09,33.51,30.42,25.30,21.88。HRMS[C 17 H 19 N 4 O 2 + ]Calculated 311.1508, found 331.1525.HPLC purity 99.9% (t) R =1.94 min).
4- (6, 7-dihydro-5H-cyclopenta [ d ] ]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 n): compound 5n was prepared according to procedure a, a general procedure used for the preparation of 5i-5 s. To a solution of 249mg of compound 8b (1.4 mmol) in 20mL of anhydrous isopropanol was added 3g (1.3 mmol) in an amount of 200mg followed by a catalytic amount of HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as an off-white solid (300 mg,1.01mmol, 78%). 1 H NMR(400MHz,DMSO-d 6 )δ10.68(s,1H),8.59(s,1H),6.95–6.71(m,1H),6.66–6.35(m,2H),4.44(s,2H),3.74(s,3H),2.80(t,J=7.6Hz,2H),2.24(t,J=7.2Hz,2H),1.94–1.69(m,2H)。 13 C NMR(101MHz,DMSO)δ174.69,167.69,156.48,156.37,156.35,132.31,121.88,120.91,119.69,106.78,101.67,55.28,49.21,33.59,30.73,21.80。HRMS[C 16 H 17 N 4 O 2 + ]Calculated 297.1352, found 297.1346.HPLC purity of 97.9% (t) R =1.66 min).
Synthesis of 4- (2-chloro-5, 6,7, 8-tetrahydroquinazolin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 o): compound 5o was prepared according to procedure a, which was used to prepare general procedure 5i-5 s. To a solution of 196mg of compound 8b (1.1 mmol) in 20mL of anhydrous isopropanol was added an amount of 203mg for 3h (1 mmol) followed by a catalytic amount of HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a white solid (224 mg,0.65mmol, 65%). 1 H NMR(400MHz,DMSO-d 6 )δ10.59(s,1H),7.60(d,J=9.0Hz,1H),6.88–6.20(m,2H),4.57(s,2H),3.75(s,3H),2.83(s,1H),2.68(d,J=3.7Hz,2H),2.57(s,1H),1.79(dd,J=10.1,6.9Hz,4H)。 13 C NMR(101MHz,DMSO)δ169.38,167.81,160.56,157.03,132.55,125.40,120.96,119.07,107.75,101.84,55.52,47.76,32.70,24.98,22.27,21.92。HRMS[C 17 H 18 ClN 4 O 2 + ]Calculated 345.1118, found 345.1133.HPLC purity 95.40% (t) R =3.33 minutes).
4- (2-chloro-5, 7-dihydrofuro [3, 4-d) ]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 p): compound 5p was prepared according to procedure B, which was used to prepare general procedure 5i-5 s. To a solution of 205mg of Compound 8b (1.15 mmol) in 20mL of anhydrous EtOH was added 3i (1.04 mmol) in an amount of 200mg followed by Na 2 CO 3 (244 mg,2.3 mmol). Then through flash silica (25% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as an off-white solid (201 mg,0.6mmol, 58%). 1 HNMR(400MHz,DMSO-d 6 )δ10.75(s,1H),7.14(d,J=8.7Hz,1H),6.65(d,J=8.7Hz,1H),6.59(s,1H),4.79(s,2H),4.53(s,2H),4.34(s,2H),3.76(s,3H)。 13 C NMR(101MHz,DMSO)δ207.03,173.02,167.65,158.36,156.93,134.16,123.91,119.19,113.73,107.30,102.22,71.68,55.88,49.23,31.17。HRMS[C 15 H 14 ClN 4 O 3 + ]Calculated 333.0754, found 333.0754.HPLC purity 99.08% (t) R =2.37 min).
Synthesis of 4- (2-chloro-5, 6-dimethylpyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 q): compound 5q was prepared according to procedure a, a general procedure used for the preparation of 5i-5 s. To a solution of 221mg of compound 8b (1.2 mmol) in 20mL of anhydrous isopropanol was added 3j (1.1 mmol) in an amount of 200mg followed by a catalytic amount of concentrated HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a white solid (217 mg,0.68mmol, 62%). 1 H NMR(400MHz,DMSO-d 6 )δ10.60(s,1H),7.60(d,J=9.0Hz,1H),6.63(d,J=8.9Hz,1H),6.55(d,J=2.4Hz,1H),4.56(s,2H),3.73(s,3H),2.38(s,3H),2.17(s,3H)。 13 C NMR(101MHz,DMSO)δ169.08,167.82,159.95,156.95,156.85,132.78,125.55,120.85,117.97,107.78,101.79,55.75,47.77,23.55,14.30。HRMS[C 15 H 16 ClN 4 O 2 + ]Calculated 319.0962, found 319.0962.HPLC purity 95.9% (t) R =3.00 min).
Synthesis of 7-methoxy-4- (2, 5, 6-trimethylpyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (5 r): compound 5r was prepared according to procedure B, which was used to prepare general procedure 5i-5 s. To a solution of 250mg of Compound 8b (1.4 mmol) in 20mL of anhydrous EtOH was added 3k (1.3 mmol) followed by Na in an amount of 200mg 2 CO 3 . Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a white solid (280 mg,0.94mmol, 72%). 1 H NMR(400MHz,DMSO-d 6 )δ10.65(s,1H),6.89–6.22(m,3H),4.18(s,2H),3.70(s,3H),2.36(s,3H),1.72(s,3H)。 13 C NMR(101MHz,DMSO)δ167.72,166.24,163.53,158.90,155.04,130.42,123.65,117.65,115.26,107.04,102.07,55.20,50.79,25.16,22.08,14.15。HRMS[C 16 H 19 N 4 O 2 + ]Calculated 299.1508, found 299.1517.HPLC purity of 99.6% (t) R =1.74 min).
Synthesis of 7-methoxy-4- (2-methyl-1, 5-naphthyridin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (5 s) (FIG. 3): compound 5s was prepared according to procedure a, a general procedure for the preparation of 5i-5 s. To a solution of 219mg of compound 8b (1.2 mmol) in 20mL of anhydrous isopropanol was added 3l (1.1 mmol) in an amount of 200mg followed by a catalytic amount of concentrated HCl (3 drops to 4 drops). Then through flash silica (30% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a yellow-red solid (247 mg,0.77mmol, 70%). 1 H NMR(400MHz,DMSO-d 6 )δ10.65(s,1H),8.75(dd,J=4.0,1.6Hz,1H),8.24(dd,J=8.5,1.6Hz,1H),7.69(dd,J=8.5,4.1Hz,1H),7.01(s,1H),6.71(d,J=8.8Hz,1H),6.61(s,1H),6.48(dd,J=8.8,2.8Hz,1H),4.61(s,2H),3.72(s,3H),2.52(s,3H)。 13 C NMR(101MHz,DMSO)δ167.60,160.28,156.33,150.47,147.88,144.75,137.20,137.12,132.49,125.12,125.06,122.13,114.51,108.55,102.37,55.73,54.10,25.36。HRMS[C 18 H 17 N 4 O 2 + ]Calculated 321.1352, found 321.1353.HPLC purity 98.6% (t) R =1.70 min).
Example 4: the procedure used to prepare 5t-5v (FIG. 4).
Compound 2a or 2b (1 equivalent) was dissolved in 3ml of isopropanol in a microwave tube, to which was added ethylamine (5 equivalents). The reaction was allowed to proceed at 80℃under microwave conditions (150 watts) for 30 minutes. The reaction mixture was adjusted to pH 7 using 10% hcl. The precipitate was filtered and dried under air to give the pure product as a solid. Flash chromatography was then used with 50% -60% EtOAc/CH 2 Cl 2 The crude product was purified to give the pure product as a tan solid.
4- (2- (ethylamino) pyrido [3, 2-d)]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 t): compound 5t was prepared following the general procedure used for the preparation of 5t-5u starting with 345mg of 2a (1 mmol) and 331mL of ethylamine (5 mmol). By means of fast speedSilica (55% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give pure product as a tan solid (263 mg,0.75mmol, 75%). 1 H NMR(400MHz,CDCl 3 )δ8.83(s,1H),8.36(dd,J=4.1,1.6Hz,1H),7.81(d,J=8.3Hz,1H),7.46(dd,J=8.6,4.1Hz,1H),6.96(d,J=8.9Hz,1H),6.49(dd,J=8.9,2.7Hz,1H),6.39(d,J=2.7Hz,1H),4.97(s,2H),3.76(s,3H),3.57–3.36(m,2H),1.24(t,J=7.2Hz,3H)。 13 C NMR(101MHz,CDCl 3 )δ168.51,159.98,158.60,157.03,150.17,143.07,133.60,130.84,127.60,124.24,123.32,108.48,101.64,55.58,51.63,36.45,15.08。HRMS[C 18 H 19 N 6 O 2 + ]Calculated 351.1569, found 351.1568.HPLC purity 96.74% (t) R =2.41 minutes).
4- (2- (ethylamino) pyrido [2, 3-d)]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 u): compound 5u was prepared following the general procedure used for the preparation of 5t-5u starting with 345mg of 2b (1 mmol) and 331mL of ethylamine (5 mmol). By flash silica (55% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give the pure product as a pale yellow solid (284 mg,0.81mmol, 81%). 1 H NMR(400MHz,DMSO-d 6 )δ10.79(s,1H),8.65(s,1H),7.66–7.25(m,2H),6.88(s,1H),6.78–6.53(m,2H),6.43(d,J=8.9Hz,1H),4.38(s,2H),3.72(s,3H),3.40(dt,J=13.7,7.0Hz,2H),1.18(t,J=7.1Hz,3H)。 13 C NMR(101MHz,DMSO)δ168.18,162.41,161.01,156.76,156.00,135.04,132.00,124.10,121.23,116.33,108.09,102.57,55.75,51.32,35.88,15.05,0.56。HRMS[C 18 H 19 N 6 O 2 + ]Calculated 351.1569, found 351.1572.HPLC purity 95.01% (t) R =2.41 minutes).
4- (2- (ethylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 v). Compound 5v was prepared according to the general procedure for preparation of 5t starting from 345mg of 5l (1 mmol) and 331mL of ethylamine (5 mmol). By flash silica (55% EtOAc/CH) 2 Cl 2 ) The crude product was purified to give the pure product (284) as a pale yellow solidmg,0.81mmol,81%)。 1 H NMR(400MHz,DMSO-d 6 )δ10.60(s,1H),6.77(d,J=8.4Hz,1H),6.71(t,J=4.9Hz,1H),6.57(d,J=9.6Hz,2H),4.35(s,1H),3.72(s,3H),3.72(s,2H),3.31–3.20(m,2H),2.59(t,J=7.4Hz,2H),2.12–2.00(m,2H),1.83–1.68(m,2H),1.11(t,J=7.0Hz,2H)。HRMS[C 18 H 22 N 5 O 2 ] + Accurate mass 340.1773, a value of 340.1768 is obtained.
Example 5: biological science
Cell culture and reagents. Human melanoma cell line A375, RPMI-7951, human breast cancer cell line MDA-MB-231, MDA-MB-453, MDA-MB-468 and human lung cancer cell line A549 were purchased from the American type culture Collection (ATCC, manassas, va.). M14 and M14 Multidrug resistance lines M14/LCC6MDR1 were given by Robert Clarke doctor at university Qiao Zhidui (Georgetown University). Melanoma cells and breast cancer cells were cultured in Du Erbei grams of modified Eagle's medium (DMEM) (Corning, manassas, va.) supplemented with 10% fetal bovine serum (FBS, atlanta Biologicals, lawrenceville, GA) and 1% antibiotic/antifungal solution (Sigma-Aldrich, st. Louis, mo.). A549 cells were cultured with RPMI 1640 medium (Gibco, carlsbad, CA) supplemented with 10% fetal bovine serum and 1% antibiotic/antifungal mixture. Paclitaxel resistant A375/TxR, MDA-MB-231/TxR and A549/TxR cells were formed by sequential treatment with paclitaxel and contained 5% CO 2 Is maintained in a medium with 100nM paclitaxel at 37 ℃. Taxane was removed from the medium one week prior to the actual experiment. Compound 17ya resistant MDA-MB-231/VxR cell lines were formed by continuous treatment with compound 17ya and maintained in a cell culture medium with 100nM compound 17ya in a cell culture incubator. Compound 17ya was removed from the medium two weeks prior to the actual experiment. For biological studies, dihydroquinoxalinone pyrimidine analogs were prepared in DMSO (ATCC) at a stock solution concentration of 20mM and stored in a-20 ℃ freezer. The stock solution was diluted with a suitable medium prior to the experiment.
Cytotoxicity assays (e.g., table 1 and table 2).
Cancer cells were seeded in 96-well plates at concentrations of 3,500 to 5,000 cells/well, depending on their growth rate. The next day, the medium was replaced in quadruplicate with fresh medium containing test compound at a concentration ranging from 0.1nM to 3. Mu.M. After 72 hours of incubation, MTS reagent (Promega, madison, wis.) was added to each well in the dark and incubated for 1 to 2 hours at 37℃depending on the cell type. Absorbance at 490nm was recorded using a microplate reader (BioTek Instruments inc., winooski, VT). IC was calculated by GraphPad Prism software (San Diego, calif.) 50 Values.
Microsomal stability assay (table 3).
Compound 5m or other compounds of the invention and verapamil (1 μg/mL) were evaluated for liver microsomal temperature (1 mg microsomal protein/mL) incubation with human (Corning Life Sciences, oneonta, NY), rat and mouse microsomes (Sekisui XenoTech, kansas City, KS) in the presence of NADPH (Acros Organics, fair, NJ) (1 mM). At predetermined times (0 min, 5 min, 10 min, 30 min, 45 min and 60 min), an aliquot (50 μl) was removed and the reaction quenched by adding 200 μl ice-cold methanol containing an internal standard. The sample was briefly vortexed and centrifuged at 3,200xg for 5 minutes at 4 ℃. The supernatant was collected and analyzed by LC-MS/MS. In vitro half-life and intrinsic clearance were assessed according to standard procedures. See Obach, R.S. "Cytochrome P450-catalyzed metabolism of ezlopitant alkene (CJ-12,458), a pharmacologically active metabolite of ezlopitant: enzyme kinetics and mechanism of an alkene hydration reaction," drug. Metab. Dispos.,2001,29 (7), 1057-67.
In vivo pharmacokinetics in rats (table 4).
All animal studies were conducted following NIH experimental animal care guidelines (Principles of Laboratory Animal Care) and only started after pre-approval by institutional animal care and use committee (Institutional Animal Care and Use Committee) of the university of tennessee health science center (University of Tennessee health Science Center). Intubated male and female Sprague-Dawley rats (225 g to 250g;Harlan Bioscience,Indianapolis,IN) were kept under 12 hours of light/card circulation, with food and water ad libitum. Groups of 4 rats (2 males and 2 females) received a single Intravenous (IV) dose of 2mg/kg of compound 5m by injection via femoral vein catheter or a single oral dose of 5mg/kg of compound 5m by oral gavage. The compounds were formulated in PEG300 (40%) and water (60%). After drug administration, blood samples (200 μl) were collected via jugular vein catheter at up to 10 predetermined time points over 24 hours. Plasma was immediately separated by centrifugation (6,000Xg, 10 min, 4 ℃) and stored at-70 ℃ until analysis. For urine excretion, urine was collected cumulatively after intravenous administration, the volume was recorded and the samples were stored at-70 ℃ until analysis.
Quantification of compound concentration
To quantify the 5m concentration of compound in plasma and urine, samples were treated with 4 volumes of methanol by protein precipitation and analyzed by LC-MS/MS. Chromatographic separation was performed on a Phenomenex C18,2.6 μm, 100X 4.6mm column (Phenomenex, torrance CA) using Nexera XR liquid chromatography (Shimadzu Corp., columbia, md.). The mobile phase consisted of: a) 95% water and 5% acetonitrile with 2mM ammonium formate and 0.1% formic acid, and b) 95% acetonitrile and 5% water with 2mM ammonium formate and 0.1% formic acid. Compound 5m and internal standard [ (2- (1H-indol-3-yl) -1H-imidazol-4-yl) (3, 4, 5-trimethoxyphenyl) were eluted with a gradient of 0.5mL/min]A ketone. The eluate was directed into an API 4500 triple quadrupole mass spectrometer (Applied Biosystem, foster City, CA) equipped with a turbo spray ion source, operating in positive ion mode at a source temperature of 500 ℃ and a capillary voltage of 4500 kV. Nitrogen was used as the source gas, curtain gas and collision gas. The characteristic mass transfer monitored was m/x 311.1/296.0 for compound 5m and m/z 378.4/210.1 for the internal standard. Using a 5m calibration standard ranging from 2.93ng/mL to 3000ng/mL and a mass control at concentrations of 20,200ng/mL and 2000ng/mL, the calibration standard was analyzed by weighted least squares regression (1/x 2 ) The concentration was calculated. The resulting plasma concentration-time curves were analyzed by standard non-atrioventricular pharmacokinetic analysis using the software package WinNonlin 8.0 (cetra, princeton, NJ).
Tubulin polymerization assay (fig. 5A).
According to the manufacturer's protocol, a test compound-containing universal tubulin buffer (80 mM PIPES, 2.0mM MgCl) was prepared by adding 100. Mu.L of tubulin from bovine brain source (3 mg/ml, cytoskeleton, denver, CO) to 10. Mu.M 2 0.5mM EGTA and 1mM GTP) to initiate tubulin polymerization. The reaction kinetics are recorded every thirty seconds at 37℃for 1 hour, for which purpose a microplate reader equipped with an absorbance setting at a wavelength of 340nm is used. This experiment was performed in duplicate.
Clonogenic assay (FIG. 7).
In the case of clonogenic assays, a375/TxR cells were seeded in 6-well plates at very low concentrations (1000 cells/well). When each individual cell was split into 4 cells in each well, the cells were treated with different concentrations (0.5 nM, 1nM and 2 nM) of 5m or 5t or medium alone and incubated for 7 days. The medium was replaced once with fresh drug during treatment. Cells were then fixed with cold methanol and stained with 0.5% crystal violet. Colony area density was quantified using a Keyence mix cytometry module.
Wound healing assay (fig. 8).
Scratch assays were performed with 5m or 5t treatment (1 nM, 2nM and 5 nM) using an IncuCyte S3 living cell imager. Briefly, A375/TxR cells (50000 cells/well) were seeded in 96-well ImageLock plates (Essen Bioscience) and allowed to attach overnight. Then use WoundMaker TM (Essen Bioscience) produced uniform scratches in all wells and washed cell debris three times with growth medium. Growth medium or medium containing 5m or 5t was added to each well and plates were monitored every two hours for up to 2 days by IncuCyte. Using an IncuCyte TM The scratch wound software module processes the representative image and the relative wound density calculation.
Cell cycle and apoptosis analysis (fig. 9).
A375/TxR cells were seeded into 10mm dishes (2X 10) 6 Individual/well) and with concentrations of 1nM, 2nM and5nM 5m or 5t treatment for 24 hours. Cells were digested with trypsin, washed and fixed in ice-cold 70% ethanol overnight. Next, the cells were incubated with 100. Mu.g/ml RNase A for 1 hour, followed by propidium iodide staining. After 5 minutes incubation, samples were analyzed by Bio-Rad ZE5 instrument at the University of Tennex Health Science Center (UTHSC) flow cytometry and cell sorting core. By ModFit LT TM Software (erity Software House, topsham, ME) processes the data. For apoptosis analysis, 10 was collected after the same treatment as in cell cycle analysis 5 Individual cells, washed, resuspended in annexin-V-FITC binding buffer (eBioscience, grand Island, NY) and stained with annexin-V-FITC (eBioscience) and propidium iodide based on kit instructions. The samples were then incubated in the dark for 10 minutes and analyzed using Bio-Rad ZE 5.
Immunofluorescent staining (fig. 5B).
A375/TxR cells (100000 cells) were seeded into six well plates with glass coverslips per well and incubated overnight. 2nM colchicine, paclitaxel, 5m or 5t was added to the cells and treated for 24 hours. Immunofluorescent staining was performed as follows: alpha-tubulin antibodies (Thermo Scientific, rockford, il.) were incubated overnight at 4℃followed by Alexa Fluor 647 conjugated goat anti-mouse IgG (Molecular Probes, eugene, OR) as secondary antibodies for 1 hour at room temperature. Coverslips were washed three times with PBS and mounted in DAPI slides containing Prolong Diamond Antifade mounting agent (Invitrogen, eugene, OR). Photographs were taken and processed using a Keyence BZ-X700 microscope (Itasca, IL).
In vivo A375/TxR melanoma xenograft models (FIGS. 10 and 11).
All animal experiments were conducted according to the guidelines of the Institutional Animal Care and Use Committee (IACUC) of the National Institutes of Health (NIH) and the university of tennessee health science center (UTHSC, memphis, TN). An equal number of male and female pathogen-free Nod-skip-Gamma (NSG) mice (n=8 mice/group) 6 to 8 weeks old were maintained in an animal facility with a controlled environmental condition of 12:12 hours light-dark cycleAnd (3) downwards. A375/TxR melanoma cells were suspended in FBS and phenol red free medium and diluted with matrigel solution prior to inoculation. An insulin syringe will be used to contain a total of 2x 10 6 100. Mu.L of a solution of A375/TxR cells was inoculated into the right flank of each mouse. Anesthetized mice were inhaled with 2% to 4% isoflurane prior to tumor cell inoculation. Tumor growth was carefully monitored and tumor volume was calculated as a×b using calipers 2 X 0.5, wherein a and b represent the larger diameter and the smaller diameter, respectively. When the tumor reaches about 100mm 3 Vehicle or drug treatment is started at this time. Paclitaxel was dissolved in ethanol and diluted in ethanol to Cremophor EL to PBS at a ratio of 1:1:18. 5m was formulated in PEG300 solution and further diluted with isotonic saline (ratio 1:4). Paclitaxel (10 mg/kg) and 5m (2 mg/kg and 4 mg/kg) were Intravenously (IV) administered to mice twice a week (2 x/Wk) over a total of 3 weeks by tail vein injection. In another separate study, a375/TxR tumor xenograft models were also established in NSG mice (50% male and 50% female) following the protocol described above (n=7 to 8 mice per group). Compound 5t was dissolved in PEG300 solution using water bath ultrasound and further diluted with sterile saline (PEG 300: saline = 3:7 ratio). Tumor bearing mice (average 90 mm) were treated with two different doses of compound 5t by intravenous injection over 24 days 3 To 100mm 3 Tumor volume), total 7 doses. At the end of the study, mice were euthanized by excessive use of isoflurane and tumor samples were obtained from the mice to analyze tumor size as well as various histological factors.
Histological and anti-mitochondrial IHC staining (fig. 12-18).
Fixed lung, liver and tumor tissues were embedded in paraffin and cut into 4 μm thick sections. By dewaxing with hematoxylin and eosin (H&E) Staining, rehydration and sealing for attachment to a slide for histological processing of specimens. Anti-mitochondrial IHC staining was performed according to the previously disclosed protocol to visualize metastasis. See Deng et al, "An orally Available Tubulin Inhibitor, compound 17ya,Suppresses Triple-Negative Breast Cancer Tumor Growth and Metastasis and Bypasses Taxane Resistance,"mol. Cancer Ther.," 2020,19 (2), 16146-16154. Briefly, after blocking with 10% horse serum, lung and liver slides were stained overnight with anti-human mitochondrial antibodies (AbCAM, cambridge, MA, cat# ab 92824) diluted 1:1000. The next day, the slides were incubated with secondary anti-mouse antibody, visualized with DAB reagent (Sigma-Aldrich, catalog number D5637), counterstained in Gill hematoxylin and with Permount TM And (5) sealing by a sealing agent. Representative tissue images were captured by Keyence BZ-X700 microscope. Representative whole lung or liver images were digitally scanned by a panoramic FLASH III system (3D Histech). Measurement of each H by using Keyence mix cytometry Module&The percentage of transfer area in 3 to 4 representative fields of E stained tissue was used to quantify the lung or liver transfer burden of each mouse.
Taxane-resistant and/or compound 17 ya-resistant in vivo xenograft models (fig. 19A to 19C, 19E And FIG. 19F)
FIG. 19A (A375/TxR): all animal experiments were conducted according to institutional animal care and use committee guidelines of the National Institutes of Health (NIH) and UTHSC (Memphis, TN). An equal number of male and female pathogen-free NSG mice (n=8 mice/group) of 6 to 8 weeks of age were maintained under controlled environmental conditions with 12:12 hours light-dark cycle in animal facilities. A375/TxR melanoma cells were suspended in FBS and phenol red free medium and diluted with matrigel solution prior to inoculation. An insulin syringe will be used to contain a total of 2 x 10 6 100. Mu.L of a solution of A375/TxR cells was inoculated into the right flank of each mouse. Anesthetized mice were inhaled with 2% to 4% isoflurane prior to tumor cell inoculation. Tumor growth was carefully monitored and tumor volume was calculated as a×b using calipers 2 X 0.5, wherein a and b represent the larger diameter and the smaller diameter, respectively. When the tumor reaches about 100mm 3 Vehicle or drug treatment is started at this time. Paclitaxel was dissolved in ethanol and diluted in ethanol to Cremophor EL to PBS at a ratio of 1:1:18. 5m was formulated in PEG300 solution and further diluted with isotonic saline (1:4 ratio). Paclitaxel (10 mg/kg) and 5m (2 mg/kg and 4 mg/kg) were intravenous by tail vein twice weekly (2 x/Wk) for a total of 3 weeksMice were administered intravenously and tumor volumes of each mouse were measured during treatment until the study endpoint was reached.
Fig. 19B: for the DU-145/VxR (Compound 17ya resistance; compound 17ya is (2- (1H-indol-3-yl) imidazol-4-yl) (3, 4, 5-trimethoxyphenyl) methanone) xenograft model, 21 male NSG mice aged 5 to 6 weeks were used. Then suspended in 100. Mu.L of a solution consisting of 50% FBS and phenol red-free medium and 50% matrigel in 2.5X10 6 The right flank of each NSG mouse was inoculated with DU-145/VxR cells. After about 16 days, mice were randomly divided into 3 groups [ untreated controls; 20mg/kg compound 17ya (PO, 3 times/week); 1mg/kg 5m (IV, 2 times/week)]And treatment is started. During treatment, we monitored tumor growth by measuring tumor volume three times per week. After 22 days of drug treatment, we terminated the study.
Fig. 19C: for the MDA-MB-231/VxR (Compound 17ya resistant) xenograft model, we will suspend 2.5X10 s in 10. Mu.L HBSS solution 5 The MDA-MB-231/VxR cells were injected in situ into the left and right sites of the mammary fat pad. Tumor growth was monitored externally using calipers. When the average tumor volume of each mouse reached 100mm 3 At this time, mice were randomly divided into 4 groups including vehicle (ethanol: cremophor EL: PBS solution=1:1:18, ip,3 times/week), 10mg/kg paclitaxel (ethanol: cremophor EL: PBS solution=1:1:18, ip,3 times/week), 20mg/kg compound 17ya (PEG 300: water=3:7, po,3 times/week), and 2mg/kg 5m (PEG 300: saline=1:4, iv,1 times/week). Mice were dosed for 20 days before the end of the study. Fig. 19E and 19F: by mixing 5X 10 5 Individual a2780/TxR cells were intraperitoneally injected into the bursa of left ovarian to construct ovarian cancer xenograft models with the right unchanged ovary as a control. Anesthesia and analgesia are used to minimize pain in animals. After one week, all 20 female mice were randomly divided into 4 groups based on body weight. Group 4 included untreated control, 5mg/kg paclitaxel (IV, 2 times/week), 20mg/kg compound 17ya (PO, 2 times/week), and 1mg/kg 5m (IV, 2 times/week). Tumor progression was monitored by intraperitoneal injection of D-luciferin into mice using bioluminescence imaging. After 3 weeks of administration to mice, the mice were treated with Mice were euthanized and ovaries were collected, weighed and imaged.
Castration resistant in vivo xenograft model (22 RV)
Fig. 19D: similarly, for a 22RV1 prostate cancer xenograft model we will be 2.5×10 6 Each 22RV1 cell was inoculated subcutaneously into the right flank of each mouse. Cells were prepared in a mixture of FBS and DMEM medium without phenol red and matrigel (1:1 ratio). When the average tumor volume reaches about 100mm 3 At (11 days of use), all 14 male mice were randomly divided into 2 groups, i.e., untreated control group and 1mg/kg 5m group. 1mg/kg 5m was administered by IV injection at a dose rate of 2 times per week. We measured tumor volumes 2 times per week until the study endpoint was reached.
Statistical analysis
All quantitative data were analyzed using GraphPad Prism 7 (San Diego, CA). One-way ANOVA followed by Dunnett multiple comparison test was applied to all in vitro experiments and quantification of lung and liver metastasis. Significant levels are defined as p <0.05, p <0.01, p <0.001, p <0.0001.
Example 6: x-ray crystallography (fig. 6).
Specific reagents. Porcine brain tubulin (catalog number T-238P) was obtained from Cytoskeleton. Bis-Tris propane, tyrosine, DTT, MES and AMPPCP were purchased from Sigma. Glycerol and anti-protease mixtures were obtained from Sangon Biotech. Beta-mercaptoethanol was obtained from XiYa Reagent.
Protein expression and purification.The microtubule-labile protein (stathmin) -like domain of RB3 (RB 3-SLD) is composed ofCloning by the group of Gigant doctor (university of Paris-Saclay, france, university of Paris, france). Purification follows the protocol disclosed in Charbaut et al, "Family Proteins Display Specific Molecular and Tubulin Binding Properties," J.biol.chem.,2001,276 (19), 16146-16154; dorleans et al, "Variations in the colchicine-binding domain provideinsight into the structural switch of tubulin, "proc.Natl.Acad.Sci.,2009,106 (33), 13775-13779. Briefly, genes were transformed into E.coli (E.coli) and overexpressed, and bacterial cells were harvested by centrifugation and resuspended with lysis buffer. The supernatant was collected by centrifugation, and RB3-SLD was purified by anion exchange chromatography and gel filtration chromatography. The peak fractions of the target protein were finally concentrated to 10mg/mL and stored at-80 ℃. The TTL plasmid was donated by Michel O.Steinmetz doctor (Paul Scherrer Institut, PSI, switzerland) and expressed and purified as described above. Prota et al, "Molecular Mechanism of Action of Microtubule-Stabilizing Anticancer Agents," Science,203,339 (6119), 587-590. Briefly, transformed E.coli was induced in LB medium with IPTG overnight at 25 ℃. Cells were then collected and lysed by sonication in lysis buffer. The lysate was then clarified by centrifugation and TLL purified by Ni-NTA affinity chromatography and gel filtration chromatography for purification. Finally the purified protein was concentrated to 20mg/ml and stored at-80 ℃ until use. Purity of RB3 and TTL was checked by SDS-PAGE. Pig brain tubulin (catalog number T-238P, cytoskeleton, inc.) at 10mg/ml in G-PEM (Universal tubulin buffer: 80mM PIPES pH 6.9, 2mM MgCl) 2 0.5mM EGTA and 1mM GTP) was supplied as a frozen liquid and stored at-80 ℃.
Crystallization and crystal soaking.Crystals were grown by sitting-drop vapor diffusion. The detailed procedure for the T2R-TTL crystal is as described above. See Wang et al, "Mechanism of microtubule stabilization by taccalonolide AJ," nat. Commun.,2017,8,15787. Briefly, protein mixtures containing tubulin (10 mg/ml), TTL (20 mg/ml) and RB3 (10 mg/ml) in a molar ratio of 2:1.3:1.2 (tubulin: RB3: TTL) were incubated on ice supplemented with 1mM AMPPCP, 5mM tyrosine and 10mM DTT. It was then concentrated to 20mg/ml at 4℃and 1.0. Mu.L of protein was used in combination with 1.0. Mu.L of crystallization buffer (4% -8% PEG4K, 5% glycerol, 0.1M MES, 30mM CaCl) 2 、30mM MgCl 2 pH 6.7) to allow crystal growth. After two days, the initial crystals were observed, and then the crystals could reach a length of about 200 μm to 300 μm in 3 days to 5 daysFinal dimensions. Thereafter, the compounds 5j, 5k, 5l, 5m and 5t were dissolved in DMSO at a concentration of 10mM, and then immersed in the crystals at 20℃for 12 hours. The soaked crystals were rapidly transferred to cryoprotectant (30 mM MgCl) 2 、30mM CaCl 2 0.1M MES, pH 6.7, containing 20% glycerol) and flash frozen at 100K for synchrotron X-ray data collection.
X-ray data collection and structural determination. Crystals of the T2R-TTL-compound complex were mounted in nylon loops and rapidly cooled at 100K in a cold nitrogen stream. Diffraction data were collected on beam line BL19U1 of the Shanghai Synchrotron Radiation Facility (SSRF) in Shanghai, china. The dataset was initially processed by the HKL2000 package. The previously disclosed T2R-TTL structure (PDB ID:4I 55) was used as a starting model to determine the structure by molecular substitution. The structures were constructed, optimized and refined using Coot and phix. See, emsley et al, "Coot: model-building tools for molecular graphics," Acta Cryst.,2004,60 (12 part 1), 2126-2132; tervilliger et al, "PHENIX: building new software for automated crystallographic structure determination," Acta Cryst.,2002,58 (11), 1948-1954. The refined structure was verified using molprobit. The data parameters and refinement statistics are summarized in table 5.
TABLE 5 data collection and refinement statistics
a The values in brackets refer to the highest resolution shell. b R merge =∑|(I-<I>) I/Σ (I), where I is the observed intensity.
Example 7: pancreatic cancer treatment.
Cell culture:
human pancreatic cancer cell lines Mia PaCa-2 and PANC-1 cell lines were obtained from ATCC and were supplemented with 10% fetal bovine serum (Atlanta Biologicals) and 1% antibiotic/antimycotic solution (Sigma-Aldrich) in DMEM at 37℃with 5% CO 2 Is routinely cultivated in a humid atmosphere. Additional 2.5% horse serum was supplemented when the Mia PaCa-2 cell line was cultured. Mia PaCa-2-Luc cell line was transfected in the laboratory with lentiviruses donated by Junming Yue doctor laboratories, cultured with DMEM supplemented with 10% FBS, 2.5% horse serum and 0.5. Mu.g/mL puromycin (Sigma). The medium was refreshed 2 times per week and the cells were maintained to 80% -90% confluence. 20mmol/L of the compound stock was dissolved in dimethyl sulfoxide (DMSO, ATCC) and further diluted to the indicated concentration in fresh cell culture medium before use.
Cytotoxicity assay (fig. 20):
mia PaCa-2 and PANC-1 cells were seeded in 96-well plates at 4000 and 5000 cells/well, respectively. After overnight incubation, cells were treated in quadruplicate with test compounds at increasing concentrations ranging from 1nmol/L to 1.25. Mu. Mol/L for 72 hours. MTS reagent (Promega) was added and incubated at 37℃for 1.5 hours, then measured in a plate reader (BioTek Instruments Inc.) at 490nM absorbance. IC was calculated by nonlinear regression analysis using GraphPad Prism 9 based on normalized values on log scale for vehicle control 50 Values.
Colony formation assay (fig. 21A):
cells were seeded in triplicate in 6-well plates at 1000 cells/well and treated with vehicle control or 1nmol/L to 5nmol/L of test compound for 11 days to 14 days. Colonies were fixed with 100% methanol and stained with 0.5% crystal violet. Colony area was quantified using Image J software.
Scratch migration assay (fig. 21B):
mia PaCa-2 cells (25,000 cells/well) were seeded in 96-well plates and allowed to adhere overnight. At 80% -90% confluence, scraping with a wound marker. After washing out the residue, the medium is replaced and the test compound is added. Cells were monitored live with incuCyte and photographs were taken every 2 hours. The wound width of cells under different treatment conditions was quantified according to the end-point width at the start point compared to the control.
Cell cycle analysis (fig. 22A):
cells were plated in 1-2X 10 100mm dishes 6 The concentration of individual cells/dish was plated in serum-free DMEM. After 24 hours of serum starvation, the medium was replaced with complete medium containing the test compound. For cell cycle analysis, cells were trypsinized and fixed overnight at-20℃with 70% ethanol. The immobilized cells were incubated with 100. Mu.g/mL RNase A and 50. Mu.g/mL propidium iodide for 30 minutes at room temperature. Stained cells were analyzed using Bio-Rad ZE5 and ModFit LT 5.0.9 software at the University of Tennessee Health Science Center (UTHSC) flow cytometry and cell sorting core and the ratio of G1 phase, S phase, G2/M phase was determined.
Western blot analysis (fig. 22B):
Cells (1X 10) 6 Individual cells/well) were seeded overnight in 6-well plates and treated with increasing concentrations of 5m and PTX for 24 hours. The total cell lysates were collected in ice-cold RIPA buffer containing protease inhibitor cocktail for 30 min and centrifuged at 13000rpm for 10 min at 4 ℃. The total protein concentration was quantified by BCA protein assay kit (Thermo Fisher Scientific). A20. Mu.g sample of protein was loaded and separated by TGX 4% -15% gradient gel (Bio-Rad) and transferred onto activated PVDF membrane. Membranes were blocked with 5% skim milk for 1 hour, then primary antibody (parp#9542 (1:500)); beta-actin #4970 (1:1000), cell Signaling Technology) was incubated overnight at 4 ℃ and blotted with secondary antibody for 1 hour at room temperature. Proteins were detected by a Bio-Rad Chemidoc imager and analyzed by Image J software.
Sub-endothelial Mia PaCa-2-Luc and PANC-1-Luc xenograft models (FIGS. 23 and 24)
All animal procedures were performed according to Institutional Animal Care and Use Committee (IACUC) approved protocols for UTHSC. Two pancreatic cancer cell lines Mia PaCa-2-Luc and PANC-1-Luc were used in this study.mu.L of Mia PaCa-2-Luc (5X 10) 6 Individual cells in HBSS) or PANC-1-Luc (3X 10) 6 Individual cells in HBSS) cell suspension was mixed with the same volume of matrigel (1:1 mixture). The cell mixture was subcutaneously injected into the right flank of each NSG mouse (male, 6 weeks to 8 weeks). 5m was dissolved in PEG 300:brine (1:4 v/v). The control group received no treatment. When the average tumor size reached 70mm 3 To 100mm 3 Treatment was started at that time. For mice bearing Mia PaCa-2-Luc tumors, we administered 5m,1 dose/week for 6 weeks by intravenous (i.v.) injection at a dose of 2 mg/kg. For mice bearing PANC-1-Luc tumors, we also administered 5m,1 dose/week for 7 weeks by i.v. injection at a dose of 1mg/kg or 2 mg/kg. Tumor volumes and body weights were measured twice weekly. Tumor volume was calculated by the following formula: volume (mm) 3 ) =0.5× (length×width 2 ). All animals were euthanized at the end of the study. Tumors were resected, ex vivo weights and sizes were recorded and imaged.
Results:
as shown in fig. 20, compound 5m showed comparable cytotoxic efficacy in Pancreatic Ductal Adenocarcinoma (PDAC) cell lines Mia PaCa-2 and PANC-1 compared to Paclitaxel (PTX). Calculated IC 50 The values indicate that in Mia PaCa-2 cells, compound 5m (1.4 nM) is slightly more potent than PTX (2.5 nM), while in PANC-1 cells, PTX (1.1 nM) is slightly more potent than compound 5m (2.7 nM). Compound 5m inhibited colony formation and cell migration in vivo as shown in fig. 21A and 21B. FIG. 21A shows the effect of compound 5m on colony formation and cell migration in Mia PaCa-2 and Panc-1 cell lines, wherein compound 5m was compared to Paclitaxel (PTX) at 1nM, 2.5nM and 5 nM. Representative colony forming images shown show that compound 5m was shown to inhibit colony formation more effectively than Paclitaxel (PTX) in the two PDAC cell lines Mia PaCa-2 and PANC-1. The bar graph shows that for Mia PaCa-2, the lowest dose of compound 5m (1 nM) completely inhibited colony formation, whereas for PTX, complete inhibition was observed at only 5 nM. Whereas for PANC-1 cells, the inhibition efficacy of colony formation was comparable for 5nM and PTX, with only 5nM doses showing colony formation Almost complete inhibition. * P<0.0001. FIG. 21B illustrates the effect of compound 5m on cell migration in Mia PaCa-2 cells, wherein compound 5m (2 nM) was compared to PTX (4 nM). Representative images show wound healing as captured by intycyte. Cells were monitored live with incuCyte and photographs were taken every 2 hours. Wound closure is shown as the wound width in micrometers (μm) at each time point compared to the control, as summarized in the bar graph. Both 5m (2 nM) and PTX (4 nM) inhibited cell migration in Mia PaCa-2 cell culture for more than 48 hours compared to control. As shown by the bar graph, 5m (2 nM) inhibited wound healing more effectively than PTX (4 nM) at each time point. * P<0.001, and p<0.0001。
Compound 5M induced cell cycle arrest in G2/M phase and apoptosis in PDAC cell lines Mia PaCa-2 and PANC-1 in a dose-dependent manner, as shown in fig. 22A and 22B. Figure 22A shows the ability of compound 5M to dose-dependently increase the proportion of cells in the G2/M phase (relative to G1 or S phase) in PANC-1 and Mia PaCa-2 cell lines, indicating mitotic arrest in these PDAC cell lines. Figure 22B shows that compounds 5m and PTX induced apoptosis in Mia PaCa-2 cells, as measured by western blot, demonstrating an increased cleaved PARP to PARP ratio. Beta-actin was used as an internal standard to correct for total protein loading. As shown in the bar graph (fig. 22B), in the Mia PaCa-2 cell line, the induction of apoptosis by compound 5m was dose dependent and more effective compared to PTX. For example, this ratio of 5m of 10nM is comparable to 20nM PTX.
Compound 5m inhibited PDAC tumor growth with minimal signs of toxicity in the Mia PaCa-2-Luc subcutaneous xenograft model, as shown in fig. 23A-23E. Compound 5m inhibited PDAC tumor growth with minimal signs of toxicity in the Mia PaCa-2-Luc subcutaneous xenograft model, as shown in fig. 23A-23E. It can be seen that compound 5m (2 mg/kg) significantly reduced tumor growth by about 70% -80% compared to the control (untreated). Figure 23B shows the effect of compound 5m on body weight over 42 days compared to control. Body weight is presented as% weight change. It can be seen that limited overall toxicity was observed with compound 5mAs body weight tends to be a slightly reduced value compared to the control. FIG. 23C illustrates the effect of compound 5m (2 mg/kg) on tumor volume ex vivo compared to control, as measured over 42 days. FIG. 23D illustrates the effect of compound 5m (2 mg/kg) on tumor weight ex vivo over 42 days as compared to control. The image captured in fig. 23E shows that the size of the resected tumor after treatment with compound 5m is smaller compared to the control treated tumor. In agreement with fig. 23A and 23E, tumor volume (fig. 23C) and tumor weight (fig. 23D) were significantly reduced by 42 days of compound 5m (2 mg/kg) treatment as compared to the control, which can also be understood in photographs of resected tumors. Data are presented as mean ± Standard Error of Mean (SEM). Significant differences associated with the control group were measured by P-value <0.05(*p<0.05,**p<0.01,***p<0.001,****p<0.001 Indicated, e.g., by a two-tailed unpaired Welch t-test or two-way ANOVA, followed byOr Dunnett multiple comparisons. Calculation of IC by nonlinear regression 50 . All data were analyzed using GraphPad Prism 9.
Fig. 24A-24E show that compound 5m inhibited PDAC tumor growth in PANC-1-Luc subcutaneous xenograft models with no signs of toxicity. Compound 5m (1 mg/kg or 2mg/kg;1 dose/week for 7 weeks) was administered by i.v. injection to the right flank of each NSG mouse (male, 6 weeks to 8 weeks). Tumor volumes and body weights were measured twice weekly. Figure 24A shows the effect of compound 5m on tumor volume compared to control (untreated). Compound 5m showed dose-dependent and significant inhibition of xenograft tumor growth compared to the control. Figure 24B shows the effect of compound 5m on body weight (weight change%) compared to control. After 49 days of treatment with two doses of compound 5m, no differences in body weight were observed in the treated animals compared to the control, indicating that compound 5m did not show significant overall toxicity. FIGS. 24C and 24D illustrate the compound 5m versus control, respectively, in vitro tumor volume (mm 3 ) And the effect of ex vivo tumor weight (g). Consistent with the results of tumor volume, compound 5m dose-dependently inhibited xenograft swelling compared to control Tumor growth as measured by ex vivo tumor volume and ex vivo tumor weight. Fig. 24E photo shows a comparison of resected tumor size after treatment with compound 5m compared to control. Tumor volume was calculated by the following formula: volume (mm) 3 ) =0.5× (length×width 2 ). All animals were euthanized at the end of the study. Tumors were resected, ex vivo weights and sizes were recorded and imaged. Data are presented as mean ± Standard Error of Mean (SEM). Significant differences associated with the control group were measured by P-value<0.05(*p<0.05,**p<0.01,***p<0.001,****p<0.001 For example, a two-tailed unpaired Welch test, or one-way ANOVA, followed by Dunnett multiple comparisons, or two-way ANOVA, followed byOr Dunnett multiple comparisons. Calculation of IC by nonlinear regression 50 . All data were analyzed using GraphPad Prism 9 (GraphPad Software inc.).
Example 8: for the preparation of dihydro-quinazolinone-pyrimidine/pyridine analogues (12 a-12m and 5 v) and for the preparation of 12o- General procedure of 12 q.
General procedure
All nonaqueous reactions were carried out in oven-dried glassware under an inert atmosphere of dry nitrogen. All reagents and solvents were purchased from Aldrich (St. Louis, mo.), alfa-Aesar (Ward Hill, mass.), combi-Blocks (San Diego, calif.), ark Pharm (Libertyville, ill.) and used without further purification. Analytical thin layer chromatography was performed on silica gel GHLF 10cm×20cm Analtech TLC Uniplates (Analtech, newark, DE) and visualized by fluorescence quenching under UV light. The compound was purified using silica gel (60 mesh-120 mesh or 100 mesh-200 mesh). Recording on a Varian Inova-500 spectrometer (400 MHz) (Agilent Technologies, santa Clara, calif.) or a Bruker ascase 400 (400 MHz) (Billerica, mass.) spectrometer 1 H NMR 13 C NMR spectrum. Chemical shifts are reported in ppm on delta scale and referenced to the appropriate solvent residual peak (CDCl) 3 For the following 1 H is 7.27ppm and for 13 C is77.23ppm, and DMSO-d 6 For the following 1 H is 2.50ppm and for 13 C is 39.51 ppm), all coupling constants (J) are given in hertz (Hz). Mass spectra were collected in positive and negative modes on a Bruker amazon SL electrospray/ion trap instrument. High Resolution Mass Spectrometer (HRMS) data were obtained on a Waters Xevo G2-S qTOF (Milford, MA) system equipped with an Acquity I type UPLC system. Porcine brain tubulin (catalog number T-238P) was purchased from Cytoskeleton, inc. By passing through 1 The purity of all test compounds was not less than 95% as determined by H NMR and HPLC. The HPLC method for determining purity is as follows: compound purity was analyzed using an Agilent 1100HPLC system (Santa Clara, CA) with a Zorbax SB-C18 column (particle size 3.5 μm,4.6mm x 150 mm) from Agilent. The mobile phase consisted of water (a) with 0.1% formic acid and acetonitrile (B) with 0.1% formic acid. A flow rate of 1mL/min was used. Gradient elution was started from 50% B. It reaches 100% B from 0 to 9 minutes, remains from 9 to 12 minutes, then decreases from 12 to 15 minutes to 50% B and stops. Compound purity was monitored with a DAD detector set at 254 nm. FIG. 25 shows a synthetic scheme for the following compounds.
N- (4-methoxy-2-nitrophenyl) -2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-amine (7). A mixture of compound 5 (10 g,0.05 mol) and 4-methoxy-2-nitroaniline 6 (9.2 g,0.055 mol) in anhydrous IPA (50 mL) was stirred with a catalytic amount of HCl (concentrated, 10 drops) at 50deg.C for 8 hours and monitored by TLC until the reaction was complete. The reaction mass was taken up with saturated NaHCO 3 Aqueous solution (ph=7), diluted, filtered, washed with water and dried to obtain 7, 15.0g,90.3% yield as orange solid; 1 H NMR(400MHz,CDCl 3 )δ9.91(s,1H),8.99(d,J=7.4Hz,1H),7.70(d,J=6.3Hz,1H),7.26(s,1H),3.87(s,3H),2.96(t,J=7.8Hz,2H),2.88(t,J=7.1Hz,2H),2.57(s,3H),2.22(dd,J=16.0,9.1Hz,2H); 13 C NMR(100MHz,CDCl 3 ) Delta 155.65,154.30,136.94,128.59,124.06,123.68,121.97,115.43,109.62,108.23,55.95,33.81,26.45,21.70,14.45; LCMS [ m+h was measured]333.2。
2-chloro-N- (5-methoxy-2- ((2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d)]Synthesis of pyrimidin-4-yl) amino) phenyl) acetamide (9).7 (15.0 g,0.045 mol) and zinc powder (6.0 g,0.09 mol) were combined in the presence of 1.5mL of AcOH in 100mL of CH 2 Cl 2 The mixture was stirred at 0℃for 0.5 h. After the addition of the raw materials, it is passed throughThe bed was filtered and the filtrate was concentrated to obtain aniline derivative (8). It was immediately dissolved in acetone (100 mL) and powder K was added 2 CO 3 (20.5 g,0.15 mol) and the mixture was cooled to 0 ℃. Chloroacetyl chloride (5 mL, excess) was slowly added dropwise to the mixture, which was stirred at 0 ℃ for an additional 2 hours. The mixture was then diluted with water, with CH 2 Cl 2 Extraction and washing with brine solution, washing with Na 2 SO 4 Drying and concentrating. The crude product was purified by column chromatography to give 9 (12 g,63.8% yield) as a bright pink solid; 1 H NMR(400MHz,DMSO-d 6 )δ9.64(s,1H),8.25(s,1H),7.34–7.31(m,2H),6.80–6.77(m,1H),4.30(s,2H),3.74(s,3H),2.73(t,J=7.4Hz,2H),2.64(t,J=7.6Hz,2H),2.28(s,3H),2.01–1.97(m,2H); 13 C NMR(100MHz,CDCl 3 ) Delta 165.23,158.20,157.30,132.71,127.67,122.83,113.21,112.02,108.91,55.63,43.03,33.32,26.85,21.70,14.05; LCMS [ m+h was measured]379.81。
7-methoxy-4- (2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (10). 60% sodium hydride (1.258 g,0.039 mol) was added in portions to anhydrous THF (100 mL) containing compound 9 (10.0 g,0.026 mol) at 0deg.C and allowed to slowly reach room temperature, which was stirred until judged complete by TLC monitoring. The mixture was poured into ice water and the solid product 10 was removed by filtration, washed with water and dried to give 6.0g of a grey solid with a yield of 66.6%; 1 H NMR(400MHz,CDCl 3 )δ9.62(s,1H),7.34(d,J=9.1Hz,2H),6.78(dd,J=8.8,2.8Hz,1H),4.25(s,2H),3.83(s,3H),2.94(t,J=7.8Hz,2H),2.73(s,2H),2.39(s,3H),2.15(dd,J=17.0,9.6Hz,2H); 13 C NMR(100MHz,CDCl 3 )δ168.40,156.73,131.17,122.96,115.74,108.16,102.36,55.78,49.64,30.99,22.60,14.21;HRMS[C 17 H 18 N 4 O 2 S + ]calculated 343.1229, measured 343.1233; HPLC purity 99.7%; mp=170 ℃ to 171 ℃.
7-methoxy-4- (2- (methylsulfonyl) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) one (11). A mixture of 10 (5 g,0.014 mol) and potassium peroxymonosulfate (also known as potassium hydrogen persulfate) (11.1 g,0.073 mol) in water/MeOH (1:1 vol) was stirred at room temperature for 5 hours, then the reaction mixture was diluted with water, filtered and dried under vacuum to give 5.0g of 92.6% of the title compound 11 without further purification; 1 H NMR(400MHz,DMSO-d 6 )δ10.74(s,1H),6.98(d,J=9.3Hz,1H),6.62–6.59(m,2H),4.53(s,2H),3.74(s,3H),2.86(d,J=13.5Hz,2H),2.25(d,J=7.1Hz,2H),1.93–1.91(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ176.79,167.87,163.91,157.57,157.10,133.16,123.25,122.89,120.25,107.32,102.15,55.82,49.78,33.97,31.44,22.46;HRMS[C 17 H 19 N 4 O 4 S + ]Calculated 375.1127, measured 375.1130; HPLC purity 95.6%. Mp=156 ℃ to 157 ℃.
4- (2-hydroxy-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 a). A mixture of 11 (100 mg,0.26 mmol) and 1N NaOH (5 mL) in 1, 4-dioxane was heated to 90℃for 6 hours. After the conversion was complete, the mixture was poured into ice water and the solid was collected, washed with water and dried. The crude product was purified by column chromatography to give pure 12a (80 mg,96.3% yield) as a pale brown solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.67(s,1H),6.90(d,J=8.5Hz,1H),6.60–6.57(m,2H),4.40(s,2H),3.72(s,3H),2.61(t,J=7.5Hz,2H),1.99(t,J=6.6Hz,2H),1.80(dd,J=14.3,7.1Hz,2H); 13 C NMR(100MHz,DMSO-d 6 )δ168.44,163.52,160.99,157.42,133.18,124.18,120.80,107.44,105.49,102.04,72.63,66.77,60.66,55.79,49.30,30.83,22.44;HRMS[C 16 H 17 N 4 O 3 + ]calculated 313.1310, measured 313.1306; HPLC purity 98.7%; mp=198 ℃ to 199 ℃.
7-methoxy-4- (2-methoxy-6, 7-)dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 b). 11 (100 mg,0.29 mmol) and 0.5M CH 3 A mixture of ONa in MeOH (5 mL) was heated to 90℃in a sealed tube for 6 hours. The mixture was poured into ice water, the solid was isolated, washed with water and dried. The crude product was purified by column chromatography to give pure 12b (79 mg,90.7% yield) as a haematochrome solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.66(s,8H),6.86(d,J=8.3Hz,7H),6.58(t,J=5.6Hz,2H),4.44(s,2H),3.86(s,3H),3.73(s,3H),2.71(t,J=7.6Hz,2H),2.14(t,J=7.1Hz,2H),1.86(dd,J=14.6,7.2Hz,2H); 13 C NMR(100MHz,DMSO-d 6 )δ177.04,168.20,164.33,158.09,157.07,132.88,123.14,121.00,113.21,107.41,102.09,55.78,54.77,49.57,33.79,30.51,22.68;HRMS[C 17 H 19 N 4 O 3 + ]calculated 327.1457, measured 327.1459; HPLC purity 95.02%; mp=163 ℃ to 164 ℃.
4- (2-amino-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 c). A mixture of 11 (100 mg,0.26 mmol) and 0.5M ammonia in 1, 4-dioxane (5 mL) was heated in a sealed tube to 90℃for 8 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12c (70 mg,84.2% yield) as a pale brown solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.61(s,1H),6.76(d,J=8.4Hz,1H),6.59–6.54(m,2H),6.23(s,2H),4.34(s,2H),3.72(s,3H),2.58(t,J=7.6Hz,2H),2.08–2.04(m,2H),1.79-1.75(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ167.78,157.83,156.73,156.30,155.58,150.40,133.05,122.97,120.44,119.08,107.36,102.30,55.85,50.51,32.35,29.74,23.09,21.22,14.54;HRMS[C 16 H 18 N 5 O 2 + ]calculated 312.1460, measured 312.1461; HPLC purity 95.26%; mp=208 ℃ to 209 ℃.
7-methoxy-4- (2- (4-methylpiperazin-1-yl) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 d). 11 (100 mg,0.29 mmol) and methylpiperazine (80 mg)0.8 mmol) of the mixture in 1, 4-dioxane was heated in a sealed tube to 110℃for 10 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12d (68 mg,64.5% yield) as a pale yellow solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.61(s,1H),6.78(d,J=8.2Hz,1H),6.58(d,J=7.7Hz,2H),4.37(s,2H),3.72(s,3H),3.68(s,4H),2.63(t,J=7.6Hz,2H),2.35-2.32(m,4H),2.20(s,3H),2.10(t,J=7.1Hz,2H),1.87-1.71(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ179.49,176.51,169.60,168.51,161.53,157.13,156.42,156.29,152.28,151.73,132.41,130.50,124.22,122.49,121.87,112.39,108.47,107.39,107.24,102.06,96.24,55.96,55.73,54.92,49.40,46.28,34.40,34.30,30.41,29.22,22.52,22.36;HRMS[C 21 H 27 N 6 O 2 + ]calculated 395.2195, measured 395.2209; mp=187 ℃ to 188 ℃.
7-methoxy-4- (2-morpholino-6, 7-dihydro-5H-cyclopenta [ d ] ]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 e). A mixture of 11 (100 mg,0.26 mmol) and morpholine (69.8 mg,0.8 mmol) in 1, 4-dioxane was heated in a sealed tube to 110℃for 10 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12e (101 mg,79.2% yield) as a pale yellow solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.60(s,1H),6.77(d,J=8.2Hz,1H),6.60-6.53(m,2H),4.36(s,2H),3.71(s,3H),3.64(s,8H),2.63(dd,J=16.6,9.1Hz,2H),2.10(dd,J=18.6,11.6Hz,2H),1.84-1.71(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ175.49,167.39,160.54,156.08,155.40,131.42,121.41,120.76,107.81,106.28,101.00,65.47,54.67,48.35,43.64,33.26,29.37,21.58;HRMS[C 20 H 24 N 5 O 3 + ]calculated 382.1879, measured 382.1883; decomposition at=266℃.
7-methoxy-4- (2- (piperidin-1-yl) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 f). 11 (100 mg,0.26 mmol) and piperidine (68 mg,0.8 mmol) were combined inThe mixture in 1, 4-dioxane was heated to 110 ℃ in a sealed tube for 9 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12f (80 mg,78.9% yield) as an off-white solid; 1 H NMR(400MHz,CDCl 3 )δ8.12(s,1H),6.75(d,J=8.8Hz,1H),6.58(dd,J=8.8,2.3Hz,1H),6.44(d,J=2.0Hz,1H),4.56(s,2H),3.80(s,3H),3.77(t,J=5.3Hz,4H),2.75(t,J=7.6Hz,2H),2.18(t,J=7.1Hz,2H),1.94-1.83(m,2H),1.63(bs,6H); 13 C NMR(100MHz,DMSO-d 6 )δ176.49,168.53,161.43,157.17,156.35,122.46,121.97,107.77,107.39,102.05,66.78,55.72,49.40,44.89,34.34,30.37,25.75,24.90,22.52;HRMS[C 21 H 26 N 5 O 2 + ]calculated 380.2087, measured 380.2096; HPLC purity 95.88%; mp=248 ℃ to 249 ℃.
7-methoxy-4- (2- (pyrrolidin-1-yl) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 g). A mixture of 11 (100 mg,0.26 mmol) and pyrrolidine (57 mg,0.8 mmol) in 1, 4-dioxane was heated in a sealed tube to 110℃for 10 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12g (82 mg,84% yield) as a pale yellow solid; 1 H NMR(400MHz,CDCl 3 )δ8.20(s,1H),6.76(d,J=8.8Hz,1H),6.58(dd,J=8.8,2.6Hz,1H),6.44(d,J=2.6Hz,1H),4.58(s,2H),3.80(s,3H),3.59(bs,4H),2.77-2.71(m,2H),2.18(t,J=3.5Hz,2H),1.98-1.95(m,4H),1.89-1.83(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ175.22,167.48,159.32,156.04,155.22,141.67,131.35,122.53,121.13,106.31100.99,54.65,54.39,48.33,45.67,33.24,29.31,24.43,21.52;HRMS[C 20 H 24 N 5 O 2 + ]Calculated 366.1930, measured 366.1930; mp=144 ℃ to 145 ℃.
4- (2- (1H-imidazol-1-yl) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12H). 11 (100 mg,0.29 mmol), imidazole (54 mg,0.8 mmol) and DIPA (71 mg,0.5 mmol) were combined in 1,4The mixture in dioxane was heated to 110 ℃ in a sealed tube for 12 hours. The mixture was poured into ice water, the solid was collected, washed with water and dried. The crude product was purified by column chromatography to give pure 12h (78 mg,80.6% yield) as a pale yellow solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.69(s,1H),8.54(s,1H),7.92(s,1H),7.08(s,1H),6.95(d,J=8.3Hz,1H),6.61(d,J=9.1Hz,2H),4.55(s,2H),3.74(s,3H),2.83(t,J=7.6Hz,2H),2.23(t,J=7.1Hz,2H),1.92-1.88(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ177.38(s),168.13,157.62,157.28,153.13,136.08,133.09,130.34,123.25,120.74,117.57,117.22,107.30,102.11,66.79,55.79,49.74,34.12,31.00,22.52;HRMS[C 19 H 19 N 6 O 2 + ]calculated 363.1569, measured 363.1577; mp=242 ℃ to 243 ℃.
4- (2- (dimethylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 i). A mixture of 11 (100 mg,0.26 mmol), dimethylamine salt (65 mg,0.8 mmol) and DIPA (2 mL) in 1, 4-dioxane was heated in a sealed tube to 110℃for 10 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12i (70 mg,77.2% yield) as a pale yellow solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.59(s,1H),6.77(d,J=8.2Hz,1H),6.59-6.56(m,2H),4.38(s,2H),3.72(s,3H),3.08(s,6H),2.63(t,J=7.4Hz,2H),2.09(t,J=6.7Hz,2H),1.80-1.77(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ176.35,168.51,162.22,157.07,156.32,132.43,122.36,122.04,107.44,107.31,102.05,55.71,49.41,37.20,34.36,30.34,22.56;HRMS[C 18 H 22 N 5 O 2 + ]calculated 340.1773, measured 340.1787; HPLC purity 96.92%; mp=198 ℃ to 199 ℃.
4- (2- (cyclopropylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 j). A mixture of 11 (100 mg,0.26 mmol) and cyclopropylamine (45 mg,0.8 mmol) in 1, 4-dioxane was heated in a sealed tube to 80℃for 10 hours. Will be mixedThe compound was poured into ice water, the solid was removed by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12j (62 mg,61% yield) as a pale yellow solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.60(bs,1H),6.95(d,J=3.3Hz,1H),6.79(d,J=8.5Hz,1H),6.59-6.55(m,2H),4.38(s,2H),3.75(s,3H),2.69(dq,J=10.6,3.6Hz,1H),2.61(t,J=7.5Hz,2H),2.08(t,J=7.1Hz,2H),1.79(dd,J=14.5,7.3Hz,2H),0.62(dt,J=6.5,3.1Hz,2H),0.44-0.43(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ176.31,168.54,163.27,157.23,156.35,132.49,122.50,122.08,107.31,102.03,66.79,55.72,49.43,34.12,30.45,24.38,22.55,6.89;HRMS[C 19 H 22 N 5 O 2 + ]calculated 352.1773, measured 352.1782; HPLC purity 95.9%; mp=171 ℃ to 172 ℃.
7-methoxy-4- (2- (methylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 k). A mixture of 11 (100 mg,0.26 mmol) and 2M methylamine in 1, 4-dioxane containing THF (0.4 mL,8 mmol) was heated in a sealed tube to 110℃for 7 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure 12k (70 mg,80.6% yield) as an off-white solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.58(s,1H),6.75(d,J=8.4Hz,1H),6.65(q,J=4.7Hz,1H),6.56(dd,J=12.2,2.7Hz,2H),4.36(s,2H),3.71(s,3H),2.77(d,J=4.8Hz,3H),2.59(t,J=7.4Hz,2H),2.06(t,J=7.1Hz,2H),1.78–1.75(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ176.36,168.53,162.94,157.29,156.30,132.43,122.39,122.11,107.32,102.05,55.72,49.40,30.40,28.49,22.54;HRMS[C 17 H 20 N 5 O 2 + ]calculated 326.1617, measured 326.1624; HPLC purity 96.7%; mp=201 ℃ to 202 ℃.
4- (2-isothiocyanato-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 l). Thiophosgene (0.55 mL,0.4 mmol) was added to anhydrous CH 2 Cl 2 Is cooled to 0 c under argon. Adding12c (100 mg,0.3 mmol) in dry CH 2 Cl 2 And DIPA (0.7 mL). The resulting solution was allowed to warm to ambient temperature over 8 hours. The reaction mass was quenched with 1N HCl (5 mL) and quenched with CH 2 Cl 2 (20 mL. Times.2) extraction. Combining organic layers via Na 2 SO 4 Drying and concentrating. The crude product was purified by column chromatography to give isothiocyanate as a pale yellow solid (50 mg,44.2% yield); 1 H NMR(400MHz,DMSO-d 6 )δ10.72(s,1H),6.91(d,J=8.5Hz,1H),6.60(d,J=7.8Hz,2H),4.43(s,2H),3.74(s,3H),2.78(t,J=7.6Hz,2H),2.20(t,J=7.2Hz,2H),1.90-1.86(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ182.66,166.12,156.44,154.36,153.29,153.26,127.11,126.48,119.80,116.90,109.81,55.90,55.54,34.45,26.89,22.32;HRMS[C 17 H 16 N 5 O 2 S + ]calculated 354.1025, measured 354.1031; HPLC purity 96.4%; decomposing at 139 ℃.
4- (2- ((2-hydroxyethyl) amino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (12 m). A mixture of 11 (100 mg,0.26 mmol), 2-aminoethanol (500 mg,0.80 mmol) and DIPA (2 mL) in 1, 4-dioxane was heated in a sealed tube to 110℃for 12 hours. The mixture was poured into ice water and then with CH 2 Cl 2 (30 mL. Times.2) extraction. The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 Dried, concentrated, and then purified by silica gel column chromatography to give pure 12m (75 mg,79% yield) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ10.60(s,1H),6.77(d,J=8.4Hz,1H),6.60(s,1H),6.59(dt,J=11.6,4.1Hz,2H),4.68(bs,1H),4.35(s,2H),3.72(s,3H),3.51-3.48(m,2H),3.35-3.30(m,2H),2.50(t,J=7.5Hz,2H),2.07(t,J=6.8Hz,2H),1.79 1.76(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ168.51,162.39,157.31,156.34,132.45,122.43,122.04,107.32,102.05,60.56,55.72,55.37,49.41,34.13,30.43,22.53;HRMS[C 18 H 22 N 5 O 3 + ]Calculated 340.1773, measured 340.1789; HPLC purity 99.73%; mp=143 ℃ to 144 ℃.
4- (2- (ethylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 v) (alternative procedure outlined in example 4). A mixture of 11 (100 mg,0.26 mmol), ethylamine (36 mg,0.80 mmol) in 1, 4-dioxane was heated in a sealed tube to 110℃for 6 hours. The mixture was poured into ice water, the solid was collected by filtration, washed with water and dried. The crude product was purified by column chromatography to give pure salt-free 5v (50 mg,50% yield) as an off-white solid; 1 HNMR(400MHz,DMSO-d 6 )δ10.84(s,1H),8.04(bs,2H),7.08(d,J=8.1Hz,1H),6.65-6.63(m,2H),3.88(bs,1H),3.76-3.72(m,4H),3.44-3.41(m,2H),2.83(bs,1H),2.08(bs,1H),1.88(bs,2H),1.18(t,J=6Hz,2H); 13 CNMR (100MHz,DMSO-d 6 ) δ167.56,158.48,133.92,125.18,119.29,107.38,102.11,55.90,49.70,36.50,31.18,22.44,14.75; HRMS[C 18 H 22 N 5 O 2 + ]calculated 340.1773, measured 340.1789; HPLC purity 97.3%; decomposing at 210 ℃ to 211 ℃.
4- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7- (trifluoromethoxy) -3, 4-dihydroquinoxalin-2 (1H) -one (12 o). Sodium hydride (15 mg,0.3 mmol) was added at 0deg.C to a solution containing 2-chloro-N- (2- ((2-methyl-6, 7-dihydro-5H-cyclopenta [ d ])]Pyrimidin-4-yl) amine) -2- ((4-trifluoromethoxy) phenyl) acetamide (100 mg,0.25 mmol) in anhydrous THF (5 mL) and the mixture was stirred at room temperature until complete as determined by TLC monitoring. The mixture was poured into ice water and the solids were removed by filtration, washed with water and dried. The crude product was purified by column chromatography to give 12o off-white solid (50 mg,55% yield); 1 H NMR(400MHz,DMSO-d 6 )δ10.87(s,1H),6.96(d,J=7.6Hz,1H),6.86(d,J=9.1Hz,2H),4.44(s,2H),2.79(t,J=7.6Hz,2H),2.49(s,3H),2.21(t,J=7.1Hz,2H),1.89-1.86(m,4H); 13 C NMR(100MHz,DMSO-d 6 )δ176.30,167.83,165.81,156.41,144.28,132.31,127.44,121.44,118.13,114.62,108.88,49.36,34.06,30.73,25.77,22.48;HRMS[C 17 H 15 F 3 N 4 O 2 + ]Calculated 365.1225, measured 365.1236; HPLC purity 98.7%;mp=188 ℃ to 189 ℃.
7- (benzyloxy) -4- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 p). Sodium hydride (71 mg,1.7 mmol) was added in portions to a solution containing N- (5- (benzyloxy) -2- ((2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]) at 0deg.C]Pyrimidin-4-yl) amino) phenyl) -2-chloroacetamide (500 mg,1.1 mmol) in anhydrous THF (20 mL) and the mixture was stirred at room temperature until complete as mediated by TLC monitoring. The mixture was poured into ice water and the solids were removed by filtration, washed with water and dried. The crude product was purified by column chromatography to give 12p brown solid (300 mg,65.6% yield); 1 H NMR(400MHz,DMSO-d 6 )δ10.69(s,1H),7.46-7.34(m,5H),6.79(d,J=8.4Hz,1H),6.67-6.65(m,2H),5.06(s,2H),4.43(s,2H),2.75(t,J=7.6Hz,2H),2.47(s,3H),2.17(t,J=7.1Hz,2H),1.85-1.82(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ175.58,168.26,165.53,156.81,155.79,137.33,132.71,128.90,128.36,128.25,122.27,121.75,116.88,108.09,103.10,69.98,49.57,33.98,30.92,25.76,22.37;HRMS[C 23 H 23 N 4 O 2 + ]calculated 387.1821, measured 387.1833; HPLC purity 99.4%; mp=150 ℃ to 151 ℃.
7-hydroxy-4- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -3, 4-dihydroquinoxalin-2 (1H) -one (12 q). A solution of 12p (200 mg,0.5 mmol) was prepared under nitrogen in a mixture of anhydrous MeOH/EtOAc (1:1 vol) and 5% Pd/C (10 wt%) was added. Then the nitrogen atmosphere was removed under vacuum and the material was subjected to a vacuum of 1atm H 2 Stir at room temperature for 2 hours (hydrogen balloon) until complete consumption of starting material by TLC. The hydrogen atmosphere was then removed under vacuum and the reaction mixture was thoroughly flushed with nitrogen. By passing through The suspended Pd/C was removed by filtration, and the solvent was evaporated and purified by column chromatography to give 12q of a white solid (100 mg,65.3% yield); 1 H NMR(400MHz,DMSO-d 6 )δ10.58(s,1H),9.47(s,1H),6.68(d,J=8.5Hz,1H),6.47(d,J=2.6Hz,1H),6.40(dd,J=8.6,2.6Hz,1H),4.42(s,2H),2.73(d,J=7.7Hz,2H),2.45(s,3H),2.16(t,J=7.1Hz,2H),1.84-1.80(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ175.45,168.38,165.48,156.95,155.04,132.80,122.65,120.06,116.49,108.93,103.25,49.63,33.97,30.91,25.78,22.33;HRMS[C 16 H 17 N 4 O 2 + ]calculated 297.1352, measured 297.1353; HPLC purity 96.9%; mp=161 ℃ to 162 ℃.
2- ((5-methoxy-2- ((2- (methylsulfanyl) -6, 7-dihydro-5H-cyclopenta [ d)]Pyrimidin-4-yl) amino) phenyl) amino) -2-oxoacetic acid ethyl ester (13). Aniline derivative 8(3 g,9 mmol) was dissolved in acetone (40 mL) and powdered K was added 2 CO 3 (5.4 g,4 mmol) and the mixture was cooled to 0 ℃. Ethyl oxalyl chloride (2 mL, excess) was slowly added to the mixture, which was stirred at 0 ℃ for an additional 1 hour. The mixture was then diluted with water, with CH 2 Cl 2 Extraction and washing with brine solution, washing with Na 2 SO 4 Drying and concentrating. The crude product was purified by column chromatography to give a pure bright brown solid (2.50 g,62.6% yield); 1 H NMR(400MHz,DMSO-d 6 )δ9.99(s,1H),8.53(s,1H),7.39(d,J=2.8Hz,1H),7.33(d,J=8.8Hz,1H),6.85(dd,J=8.8,2.8Hz,1H),4.23(q,J=7.1Hz,2H),3.76(s,3H),2.75(t,J=7.7Hz,2H),2.68(t,J=7.1Hz,2H),2.07-1.96(m,2H),1.23(t,J=7.1Hz,3H); 13 C NMR(100MHz,CDCl 3 ) Delta 177.41,173.72,165.18,162.20,162.02,159.86,137.43,133.13,128.90,117.71,116.20,114.19,67.86,60.56,38.77,32.01,26.35,18.86,18.47; LCMS [ m+h was measured]403。
6-methoxy-1- (2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) quinoxaline-2, 3 (1H, 4H) -dione (14). To a stirred solution of compound 13 (1 g,2.4 mmol) in THF (20 mL) at 0deg.C was added 60% sodium hydride (0.86 g,5.0 mmol). The mixture was stirred at room temperature for 12 hours and monitored by TLC. After the reaction was completed, the mixture was diluted with water, and washed with CH 2 Cl 2 (30 mL. Times.2) extraction, andthe combined organic layers were washed with 0.5N HCl (2X 5 mL) and dried over Na 2 SO 4 Drying and concentrating. The crude product was purified by column chromatography to give pure bright brown solid 14 (0.75 g, 79% yield): 1 H NMR(400MHz,DMSO-d 6 )δ12.18(s,1H),6.80(d,J=2.4Hz,1H),6.65(dd,J=9.1,2.5Hz,1H),6.54(d,J=9.1Hz,1H),3.74(s,3H),3.05(d,J=7.0Hz,2H),2.68-2.66(m,2H),2.49(s,3H),2.08-2.05(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ180.80,171.27,156.46,154.33,153.35,152.57,128.95,127.16,119.80,116.87,109.78,01.17,55.91,34.39,27.32,21.99,14.22;HRMS[C 17 H 17 N 4 O 3 S + ]calculated 357.1021, measured 357.1034; HPLC purity 97.6%; mp=210 ℃ to 211 ℃.
6-methoxy-1- (2- (methylsulfone) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) quinoxaline-2, 3 (1H, 4H) -dione (15). A mixture of 14 (500 mg,1.4 mmol) and potassium peroxymonosulfate (0.65 g,4.2 mmol) in water/MeOH (1:1 volumes) was stirred at room temperature for 12 hours then the reaction mixture was diluted with water, filtered and dried under vacuum to give (0.5 g, 92.5%) of the title compound 15 without further purification; 1 H NMR(400MHz,DMSO-d 6 )δ12.19(s,1H),6.81(d,J=2.0Hz,1H),6.65-6.63(m,2H),3.75(s,3H),3.40(s,3H),3.22(s,2H),2.84(s,2H),2.17(s,2H); 13 C NMR(100MHz,DMSO-d 6 )δ182.85,165.17,156.65,154.15,153.55,152.80,138.21,127.36,119.81,117.21,109.57,101.23,55.94,34.54,28.31,22.25;HRMS[C 17 H 17 N 4 O 5 S + ]calculated 389.0920, measured 389.0927; mp=178 ℃ to 179 ℃,
6-methoxy-1- (2-methoxy-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) quinoxaline-2, 3 (1H, 4H) -dione (16). A mixture of 15 (100 mg,0.25 mmol) and 5N sodium methoxide in methanol (2.2 mL) was stirred in a closed tube at room temperature for 48 hours. After completion of the reaction, the mixture was diluted with water, filtered and dried in vacuo to give 16 (50 mg, 57.4%) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ12.16(s,1H),6.79(d,J=2.4Hz,1H),6.63(dd,J=9.1,2.4Hz,1H),6.57(d,J=9.1Hz,1H),3.89(s,3H),3.73(s,3H),3.02(dd,J=13.6,6.8Hz,2H),2.63-2.57(m,2H),2.07-2.03(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ182.87,166.12,156.44,154.36,153.29,153.27,127.11,126.48,119.80,116.98,109.81,101.11,55.90,55.54,34.44,26.89,22.31;HRMS[C 17 H 17 N 4 O 4 + ]Calculated 341.1250, measured 341.1257; HPLC purity 98.3%; mp=126 ℃ to 127 ℃.
5-methoxy-1- (2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Pyrimidin-4-yl) -1H-benzo [ d ]]Synthesis of imidazol-2 (3H) -one (18). Compound 8(200 mg,0.06 mmol) was dissolved in acetone (25 mL) and powdered K was added 2 CO 3 (0.45 mg,3.3 mmol) and the mixture was cooled to 0 ℃. Ethyl chloroformate (0.4 mL, excess) was slowly dropped into the mixture, which was stirred at 0 ℃ for an additional 2 hours. The mixture was then diluted with water, with CH 2 Cl 2 (20 mL. Times.2) and washed with brine solution, dried over Na 2 SO 4 Dried, concentrated, and crude 17 was dissolved in anhydrous THF (5 mL), cooled to 0 ℃ and sodium hydride (41 mg,1.06 mmol) was added, allowed to warm to room temperature until completion. The mixture was poured into ice water and the solid product was removed by filtration, washed with water and dried to give off-white solid 18 (98 mg, 56%); 1 H NMR(400MHz,CDCl 3 )δ8.96(bs,1H),7.40(d,J=8.6Hz,1H),6.69-6.66(m,2H),3.82(s,3H),3.06(td,J=7.5,4.1Hz,4H),2.58(s,3H),2.21-2.09(m,2H); 13 C NMR(100MHz,CDCl 3 )δ178.41,169.49,156.71,153.04,151.84,129.49,124.51,122.25,112.67,107.79,96.42,55.91,34.23,29.50,22.57,14.34;HRMS[C 16 H 17 N 4 O 2 S + ]calculated 329.1072, measured 329.1076; HPLC purity 95%; mp=167 ℃ to 168 ℃.
1- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ]]Pyrimidin-4-yl) -5- (trifluoromethoxy) -1H-benzo [ d ]]Synthesis of imidazol-2-one (18 a). The compound N1- (2-methyl-6, 7-dihydro-5H-cyclopenta [ d ] ]Pyrimidin-4-yl) -4- (trisFluoromethoxy) benzene-1, 2-diamine (500 mg,1.5 mmol) was dissolved in acetone (20 mL), and K was added as a powder 2 CO 3 (850 mg,6.1 mmol) and the mixture was cooled to 0deg.C. Ethyl chloroformate (1.0 mL, excess) was slowly dropped into the mixture, which was stirred at 0 ℃ for an additional 2 hours. The mixture was then diluted with water, with CH 2 Cl 2 (20 mL. Times.2) extraction and washing with brine solution, na 2 SO 4 Dried, concentrated, and the crude product was dissolved in anhydrous THF (5 mL), cooled to 0 ℃ and 60% sodium hydride (71 mg,1.7 mmol) was added and allowed to warm to room temperature until completion. The mixture was poured into ice water and the solid product was removed by filtration, washed with water and dried to give off-white solid 18a (180 mg, 43.6%); 1 H NMR(400MHz,DMSO-d 6 )δ11.55(s,1H),7.44(d,J=8.6Hz,1H),7.15-6.97(m,2H),3.03-2.89(m,4H),2.61(s,3H),2.09-1.98(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ179.19,166.25,152.26,151.00,144.15,130.35,127.77,126.19,121.93,119.39,114.37,112.32,103.53,34.41,29.15,25.54,22.32;HRMS[C 16 H 14 F 3 N 4 O 2 + ]calculated 351.1069, measured 351.1080; HPLC purity 95.2%; mp=136 ℃ to 137 ℃.
5-methoxy-1- (2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Pyrimidin-4-yl) -2- (2, 3, 4-trimethoxyphenyl) -1H-benzo [ d ]]Synthesis of imidazole (19). Compound 8A mixture of (200 mg,0.6 mmol) and the corresponding 2,3, 4-trimethoxybenzaldehyde (142 mg,0.7 mmol) in EtOH (6 mL) was refluxed for 1 hour. The solvent was then evaporated under reduced pressure to give the crude imine which was redissolved in CH 2 Cl 2 (6 mL) followed by the sequential addition of iodine (75 mg,0.45 mmol) and K 2 CO 3 (115 mg,0.8 mmol). After the reaction was completed, the reaction mixture was stirred at room temperature, which was quenched with 5% na 2 S 2 O 3 (15 mL) quenching followed by CH 2 Cl 2 (30 mL. Times.2) extraction. The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 Drying and concentratingContracted, and then purified by silica gel column chromatography to give 19 (99 mg, 49.7%) as an off-white solid; 1 H NMR(400MHz,CDCl 3 )δ7.47(d,J=8.6Hz,1H),7.36(d,J=1.9Hz,1H),7.32(d,J=8.9Hz,1H),6.96(dd,J=8.9,2.1Hz,1H),6.79(d,J=8.7Hz,1H),3.91(s,3H),3.90(s,3H),3.68(s,3H),3.49(s,3H),2.95(t,J=7.4Hz,2H),2.32(s,3H),2.09-1.97(m,2H); 13 C NMR(100MHz,CDCl 3 )δ179.88,171.57,151.42,141.66,127.14,123.45,112.67,107.85,77.35,77.04,76.72,61.19,61.11,56.22,56.06,34.46,28.45,23.01,14.16;HRMS[C 25 H 27 N 4 O 4 S + ]calculated 479.1753, measured 479.1755; HPLC purity 95.8%; mp=127 ℃ to 128 ℃.
4-methoxy-N1- (2-methoxy-6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) benzene-1, 2-diamine (20). Will be 15A mixture of (100 mg,0.25 mmol) and 5N sodium methoxide in methanol (3.0 mL) was stirred in a closed tube at 70℃for 2 hours. After completion of the reaction, the mixture was diluted with water, filtered and dried in vacuo to give 20 (60 mg, 81%) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ7.98(bs,1H),6.90(d,J=2.8Hz,1H),6.30(d,J=9.1Hz,1H),6.12(dd,J=9.2,2.5Hz,1H),4.85(bs,2H),3.68(s,3H),3.67(s,3H),2.66(t,J=7.1Hz,2H),2.55-2.50(bs,2H),1.97-1.93(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ165.29,160.13,158.61,146.06,129.37,117.54,110.31,101.93,100.59,55.24,53.97,33.96,27.06,21.92;HRMS[C 15 H 19 N 4 O 2 + ]calculated 287.1508, measured 287.1519; HPLC purity 98.2%; mp=189 ℃ to 190 ℃.
N1- (5-methoxy-2- ((2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d)]Pyrimidin-4-yl) amino) phenyl) -N2-methyl oxamide (21). To Compound 17 at 0deg.C(0.2 g,0.49 mmol) methylamine in THF (2 mL, excess) was added. The mixture was stirred in a sealed tube at room temperature for 12 hours and monitored by TLC. After completion of the reaction, the mixture was diluted with water, filtered and dried in vacuo to give 21 (100 mg, 52%) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ9.96(s,1H),9.02(d,J=2.6Hz,1H),8.59(s,1H),7.55(d,J=9.3Hz,1H),7.27(d,J=9.1Hz,1H),6.81(dd,J=8.9,2.1Hz,1H),3.77(s,3H),2.77-2.73(m,2H),2.69-2.66(m,5H),2.27(s,3H),2.03–1.99(m,2H); 13 C NMR(100MHz,DMSO-d 6 )δ172.75,169.0,160.44,158.11,157.77,157.56,133.51,128.77,123.46,112.74,110.72,108.31,55.77,33.98,27.28,26.59,21.61,13.70;HRMS[C 18 H 22 N 5 O 3 S + ]calculated 388.1443, measured 388.1457; HPLC purity 99.2%; mp=165 ℃ to 166 ℃.
Example 9: biological characterization of cyclopenta-pyrimidine dihydroquinoxalinones (12 a-12m, 12o-12q, 5v, etc.).
Biological science. Cell culture and reagents. Human melanoma cell lines a375 and M14; human breast cancer cell lines MDA-MB-231 and MDA-MB-453; human pancreatic cancer cell lines MIA Paca-2 and PANC-1; the prostate cancer cell line PC-3 was purchased from American type culture Collection (ATCC, manassas, va.). A375, M14, MDA-MB-231, MDA-MB-453 and MIA Paca-2 were cultured in Du Erbei g of modified eagle medium (Corning, manassas, va.) supplemented with 10% fetal bovine serum (FBS, atlanta Biologicals, lawrenceville, GA) and 1% antibiotic/antifungal solution (Sigma-Aldrich, st. Louis, mo.). MIA Paca-2 pancreatic cancer cells were cultured in medium also supplemented with 2.5% horse serum. PANC-1 and PC-3 were cultured in RPMI 1640 medium (Gibco, carlsbad, calif.) supplemented with 10% FBS and 1% antibiotic/antifungal mixture. Paclitaxel resistant PC-3 cells (PC 3/TxR) cells are formed by sequential treatment with paclitaxel and contain 5% CO 2 Is maintained in a medium with 10nM paclitaxel at 37 ℃. For biological experiments, pyrimidine dihydroquinoxalinone derivatives were dissolved in DMSO to generate 20mM stock solutions and stored at-20 ℃ until use.
The compounds synthesized in example 8 were tested for cytotoxic activity against a panel of cancer cell lines such as melanoma (A375, M14), breast cancer (MDA-MB-231, MDA-MB-453), pancreatic cancer (Mia PaCa-2, PANC-1) and prostate cancer (PC 3, PC 3/TxR).
The half maximal inhibitory concentration values (IC) for inhibition of cell growth are summarized in table 6 50 )。
Studies have revealed that the size of heteroatoms has a significant impact on cytotoxic potency, and reducing the size of heteroatoms tends to increase potency. For example, thioether 10 (IC 50 Approximately 3.4 nM.+ -. 0.5nM, A375 cell line, table 6), ether 12b (IC 50 Approximately 3.2 nM.+ -. 0.5 nM) and secondary amine 12k (IC 50 Approximately 1.2nM ± 0.2 nM) is relatively small and has single digit nM potency. Except morpholine derivatives such as N-methylpiperazine 12d (IC) which show moderate to high potency 50 About 542.8 nM.+ -. 111.0 nM), morpholine 12e (IC 50 About 13.6 nM.+ -. 2.0nM, table 6), piperidine 12f (IC 50 Approximately 436.1 nM.+ -. 76.2 nM) and pyrrolidine 12g (IC 50 The substitution of the cyclic derivative of 82.1 nM.+ -. 12.9 nM) has relatively low potency. Aromatic heterocycles (i.e. imidazole 12h (IC 50 Approximately 5.7nM ± 0.9 nM)). Tertiary amine 12I (IC) 50 The derivative of +.22.6 nM.+ -. 4.5 nM) shows moderate potency. The results obtained with compound 12k were a study of secondary amines (such as N-ethyl 5v (IC 50 Approximately 1.6 nM.+ -. 0.3 nM) and N-cyclopropyl 12j (IC 50 The pharmacological potency of 1.4 nM.+ -. 0.3 nM) paves the way, and these secondary amines are highly potent. Adding additional hydrogen bonding donors, such as 12m (IC 50 the-OH group in the ethanolamine moiety of +.8.6nM.+ -. 0.2nM is slightly less potent than 5v (ethylamine form) and isothiocyanate derivative 12l (IC 50 Approximately 3.3 nM.+ -. 0.5 nM) also shows very good potency. In pyrimidine (2-Py) ring 12a (IC 50 An unprotected phenol O at the C2 position on 646.5 nM.+ -. 124.2 nM)H is as other electron withdrawing groups (such as sulfone derivative 11 (IC 50 And +.84.9 nM.+ -.17 nM)) as well as significantly reducing potency. On the other hand, the free amine 12c at the same position yields improved potency (IC 50 ≈2.01nM±0.4nM)。
Compound 12o-p (i.e., OCF) as a substitute for OMe group 3 OBn and OH) have reduced efficacy. The general trend for aryl substituents is that 4-OMe compounds have the highest affinity with 4-OCF 3 (12o,IC 50 ≈43.1±6.9)、OH(12q,IC 50 Those compounds of table 6) were moderate in potency, and the compounds with OBn (12 p) substitution had the lowest potency. Furthermore, modification of the C-ring structure did not lead to efficacy improvement. Closed loop systems (such as benzimidazole 19 (IC, respectively 50 > 3. Mu.M) and 2-imidazolone derivatives 18a-b (IC 50 Approximately 1182 nM+ -339 nM, 238.1 nM+ -126.5 nM)), various quinoxalinedione derivatives 14, 15, 16 (IC) 50 Both > 3 μm) and open systems, such as amine 20 (table 6) and amide 21, were negatively affected, resulting in little or no cytotoxic activity.
Pharmacokinetic assessment.
Female NSG mice 7 to 9 weeks old were used in pharmacokinetic studies. Mice received a single dose of 4mg/kg 12k (50% PEG300:50% saline) intravenously or 10mg/kg (90% PEG300:10% saline) orally. Blood from animals (n=3) was collected in heparinized tubes by terminal intracardiac blood collection at predetermined time points (0.08 hours, 0.25 hours, 0.5 hours, 1 hour, 3 hours, 6 hours, 12 hours, 24 hours) and plasma was immediately isolated by centrifugation (10,000 rpm,10 minutes, 4 ℃) and stored at-80 ℃ until analysis. The concentration-time curve of this experiment is shown in fig. 27.
Metabolic stability of compounds 12j, 12k and 5v in human and mouse liver microsomes.Compounds 12j, 12k, 5v and verapamil (1 μg/mL) were evaluated for liver microsomes (1 mg microsomal protein/mL) in the presence of NADPH (Acros Organics, fair down, NJ) (1 mM) with human (Corning Life Sciences, oneonta, NY) and mouse microsomes (Sekisui XenoTech, kansas City, KS). At a predetermined time Between (0 min, 5 min, 15 min and 60 min), an aliquot (50 μl) was removed and the reaction quenched by the addition of 200 μl ice-cold methanol containing internal standard. The sample was briefly vortexed and centrifuged at 3200 Xg for 5 minutes at 4 ℃. The supernatant was collected and analyzed by LC-MS/MS. In vitro half-life was assessed according to standard procedures (t 1/2 ) And intrinsic Clearance (CL) int )。Obach,R.S.,Cytochrome P450-catalyzed metabolism of ezlopitant alkene(CJ-12,458),a pharmacologically active metabolite of ezlopitant:enzyme kinetics and mechanism of an alkene hydration reaction.Drug metabolism and disposition,2001,29(7),1057-1067。
In vitro microsomal stability of compounds 12j, 12k and 5v in human and mouse liver microsomes was determined and the results are summarized in table 7. Compounds 12j and 5v exhibited limited stability and in the mouse microsomal formulation, compound 12k exhibited good stability in both species with half-lives exceeding 300 minutes.
a Verapamil was used as an assay control in this study. Data are presented as mean (% CV).
* Based on t 1/2 And CL int Is a detection limit of (2).
Compound 12k was carried into an in vivo study, which is graphically depicted in fig. 28. In vivo pharmacokinetic studies of compound 12k in NSG mice after intravenous 4mg/kg (FIG. 27) or oral administration of 10 mg/kg. The in vivo half-life was 238 minutes (3.97 hours). As shown in table 8, oral bioavailability remained limited to 2.02%.
Data are presented as mean (% CV).
Cytotoxicity assay.Cancer cells are divided into 3,500 to 5,000 cellsCell/well concentrations were seeded in 96-well plates. After 24 hours, the medium was replaced with test compound in fresh medium ranging in concentration from 0.1nmol/L to 3. Mu. Mol/L in A375, M14, MDA-MB-231 and MDA-MB-453 cancer cells. A concentration range of 1nmol/L to 1.25. Mu. Mol/L was used for Mia Paca-2, PANC-1, PC3 and PC3/TxR cancer cells. Each experiment consisted of four replicates. Cancer cells were treated for 72 hours, then MTS reagent (Promega, madison, wis.) was added to each well and incubated in the dark at 37℃for 1 to 2 hours depending on the cell type. Absorbance at 490nm was recorded using a microplate reader (BioTek Instruments inc., winooski, VT). IC was calculated by GraphPad Prism software (San Diego, calif.) 50 Values.
Subendothelial PC-3/TxR xenograft model.Compound 12k in vivo antitumor efficacy in a subcutaneous human prostate cancer drug resistant cell line PC-3/TxR xenograft mouse model was evaluated. All animal procedures were performed according to protocol (protocol # 20-0166) approved by the Institutional Animal Care and Use Committee (IACUC) of UTHSC. Male Nod-Skid-Gamma (NSG) mice (n=8 mice/group) 7 to 10 weeks old were kept in an animal facility under 12/12 light. PC-3/TxR prostate cancer tumors were tested as mycoplasma free and were validated for in vivo resistance to paclitaxel prior to study. Will be 3X 10 6 Individual cells were suspended in 75 μl of HBSS and matrigel mixture (2:1) and injected subcutaneously into the right flank of each mouse using a 28g 1/2 insulin syringe. Mice were anesthetized with 5% isoflurane and maintained at 2% isoflurane while cell injection was performed. Average tumor size reached 70mm when inoculated for approximately two weeks 3 To 100mm 3 At this time, mice were randomly divided into control, paclitaxel-treated, and 12 k-treated groups. Paclitaxel was first dissolved in ethanol and diluted with Cremophor EL/saline solution (1:1:18 ratio). Compound 12k was dissolved in PEG 300 and diluted with saline solution (1:4 ratio) prior to use. Paclitaxel (10 mg/kg,1 dose/week) and compound 12k (2.5 mg/kg,2 doses/week) were administered intravenously (i.v.) until the endpoint. Tumor volumes were measured twice weekly with calipers and calculated by the following formula: volume=0.5× (length×width 2). All animals were euthanized at the endpoint after 2 weeks of treatment. Tumors were resected and weights and sizes were recorded ex vivo and imaged in petri dishes as a size reference.
As shown in fig. 28A to 28E, compound 12k significantly inhibited tumor growth in the treated group compared to the paclitaxel-treated group and the control group. Figure 28A shows tumor growth curves (two-way ANOVA followed by multiple comparison test) for control, paclitaxel and compound 12k treated groups. All mice were stable and had no significant weight loss in compound 12k treatment, indicating that the 2.5mg/kg treatment dose twice weekly was well tolerated, while both the control and paclitaxel-treated groups experienced stable weight loss until the end of the study, as shown in fig. 28B. At the end of the study, all mice were euthanized and tumors and major organs were harvested. Tumors were weighed ex vivo and tumor volumes were measured. Treatment with compound 12k inhibited tumor volume growth by about 85.6% (fig. 28C) and tumor weight by 84.5% (fig. 28D), respectively, compared to the control. Higher doses of paclitaxel showed no significant tumor weight reduction. The results show that compound 12k is able to attenuate the progression of prostate cancer tumors and overcome taxane resistance in vivo at low and safe but effective doses. FIG. 28E is a comparison of tumors isolated in 35mm dishes for the control, paclitaxel and compound 12k treated groups. Data are presented as mean +/-SEM. Significant differences between the groups (fig. 28C-28D) were determined by one-way ANOVA followed by Dunnett multiple comparison test (< p <0.005, < p <0.0005, < p < 0.0001).
Example 10:5v inhibited tubulin polymerization, paclitaxel resistant melanoma growth and spontaneous metastasis.
X-ray crystal structure and tubulin polymerization assays confirm that 5v is a Colchicine Binding Site Inhibitor (CBSI) that disrupts microtubule dynamics and interferes with microtubule assembly. In vitro studies have shown that 5v has sub-nanomolar antiproliferative activity against a panel of cancer cell lines and some of their paclitaxel resistant cell lines (TxR). 5v inhibited colony formation and migration of A375/TxR cells and induced apoptosis and G2/M phase arrest of A375/TxR cells. The following in vivo studies demonstrate that 5v strongly inhibits tumor growth and induces tumor necrosis, anti-angiogenesis and apoptosis in a375/TxR melanoma xenografts. Furthermore, 5v treatment significantly inhibited spontaneous axillary lymph node, lung and liver metastasis from subcutaneous tumors and had no apparent toxicity to the major organs of mice, demonstrating the therapeutic potential of 5v as a novel anticancer agent for cancer treatment.
Microtubules are the key elements of polymeric alpha-tubulin and beta-tubulin heterodimers arranged in a head-to-tail fashion, with the alpha-subunit in one dimer being bound to the beta-subunit in the next dimer. Non-covalent binding of these subunits forms a filament. The filaments are assembled longitudinally into a cylindrical structure, typically 13 pieces, constituting a 22nm microtube. Microtubules undergo an alternation of two phases (growth and contraction), a behavior known as dynamic instability. The continuous change in slow polymerization and fast depolymerization kinetics allows microtubules to play a fundamental and essential role in many cellular processes including cell division, maintenance of cellular architecture, intracellular trafficking and regulation of movement.
Microtubule Targeting Agents (MTAs) affect microtubule dynamics by binding to microtubules via different mechanisms. In the intercellular phase, MTA has a great influence on microtubules involved in intracellular transport of proteins, vesicles and organelles, and the interphase cytoskeleton. In mitosis, the structure required to isolate the replicated chromosomes (i.e., the mitotic spindle consisting of the cytoskeleton of rearranged microtubules) is also greatly affected by MTA. Disruption of microtubule dynamics results in cell cycle arrest in the G2/M phase, leading to mitotic arrest. Given the important role of microtubules in cell growth, microtubules have become attractive targets for the development of anticancer drugs.
Colchicine Binding Site Inhibitors (CBSI) target the binding domain of microtubules located at the interface between the alpha and beta subunits of tubulin heterodimers. CBSI has several advantages over other MTAs or colchicine itself, including overcoming ABC transporter mediated multidrug resistance and β3-tubulin overexpression as well as vascular damaging activity. Several CBSIs are currently in clinical trials, such as CA-4P, OXi4503, ABT-751, and 17ya, however, no FDA approved CBSIs is currently available for cancer treatment, mainly due to undesirable adverse events (e.g., hematological toxicity, neurotoxicity), lack of bioavailability, or low water solubility. Thus, there is still a need for much effort to find more CBSI that can avoid multi-drug resistance (MDR) or that is a non-MDR substrate, and these promising CBSIs should have a broader therapeutic window, excellent pharmacokinetic/pharmacodynamic properties and better efficacy.
All CBSIs we produce have different chemical structures, but they are efficient and able to overcome MDR, and some of them even have good bioavailability. Among them, the CBSI of the present invention having pyridopyrimidine and dihydroquinoxalinone structures represented by 5m and 5t shows the most potent antiproliferative activity against a group of human cancer cell lines, including melanoma, lung cancer and breast cancer, IC 50 The value is one digit (example 9). And in vivo studies demonstrated the effective efficacy of 5m (4 mg/kg) and 5t (5 mg/kg) on tumor growth and spontaneous metastasis of subcutaneous melanoma to lung and liver using the paclitaxel-resistant a375/TxR xenograft model (e.g., fig. 10 and 11). We hypothesize that by combining the structures of 5m and 5t we can obtain new dihydroquinoxalinone analogues, which are more efficient than 5m or 5 t. In this context, in this report we designed and synthesized the new analogue 5v and evaluated its efficacy in vitro and in vivo. Through X-ray crystal structure studies and a set of in vitro techniques, we demonstrate that 5v is an effective CBSI that can inhibit cancer cell growth at sub-nanomolar concentration ranges. We also demonstrated the in vivo efficacy of 5v in inhibiting tumor growth and spontaneous metastasis using a375/TxR xenograft model without causing toxicity to major organs in mice. In summary, the results we report here show that 5v is a promising CBSI with anti-proliferative activity comparable to paclitaxel and can be used to treat paclitaxel sensitive or paclitaxel resistant cancers. In addition, 5v has the same biological characteristics as the other CBSI of the present invention, which is able to overcome 17ya resistance. Thus, it is reasonably expected that 5v may also overcome 17ya resistance.
Cell culture:colchicine, paclitaxel and AzixaPurchased from Sigma-Aldrich, LC Laboratories and APExBIO Technology LLC, respectively. Human melanoma cell lines A375 and RPMI-7951, and human breast cancer cell lines MDA-MB-231, MDA-MB-453 and MDA-MB-468 were purchased from the American Type Culture Collection (ATCC). Melanoma cell lines M14 and M14 multidrug resistance sub-line M14/LCC6MDR1 were given by Robert Clarke doctor at the university of George. The prostate cancer cell lines PC-3, PC-3/TxR, DU-145 and DU145/TxR were donated by Evan Keller doctor from Michigan University (University of Michigan.). Melanoma and breast cancer cells were cultured in Du Erbei grams of modified eagle medium (Corning) supplemented with 10% fetal bovine serum (FBS, atlanta Biologicals) and 1% antibiotic-antifungal solution (Sigma-Aldrich). Paclitaxel resistant A375/TxR and MDA-MB-231/TxR cells were generated by gradual and continuous culture in a medium containing paclitaxel. At 37℃with 5% CO 2 All cells were cultured in the atmosphere. A375/TxR and MDA-MB-231/TxR cells were maintained in medium containing 100nM paclitaxel. PC-3/TxR and DU-145/TxR cells were stored in medium containing 10nM paclitaxel or docetaxel, respectively. Taxane was removed from the medium one week prior to the experiment.
Cell proliferation inhibition assay:the cell proliferation inhibition of the test compounds was first determined by the MTS assay as described previously. That is, 3000 to 7500 cells were added to 96-well cell culture plates, followed by treatment of test compounds (0.1 nM to 3 μm) in quadruplicate after inoculation and overnight incubation. After 72 hours of treatment, MTS (Promega) was added to the wells and absorbance was measured at 590nm after incubation for 1.5 to 2 hours. Calculation of IC by GraphPad Prism 8 (GraphPad Software) 50 Values. Inhibition of 5v colony formation was determined on A375/TxR cells as described previously. Briefly, A375/TxR cells (1000 cells/well) were treated with growth medium containing 0.5nM, 1nM or 2nM 5v and incubated for 7 days or more (incubation medium was changed every 3 days). After fixation and staining with 0.5% crystal violet, cell colonies were quantified by the mix count module of Keyence BZ-X700 microscope. Assays were performed in triplicate.
Tubulin polymerization inhibition assay:according to the manufacturerIn vitro tubulin polymerization assays were performed according to the instructions of (Cytoskeleton). Briefly, 10. Mu.M of 5v was added to tubulin (3 mg/mL, purity>99%) and the mixture was transferred to a microplate reader and incubated at 37 ℃. The absorbance of the mixture was recorded at 350nm for 1 hour every 30 seconds. Colchicine and paclitaxel were used as positive controls. Immunofluorescent staining of alpha-tubulin in a375/TxR cells treated with 5v as previously described. Briefly, A375/TxR cells seeded on glass coverslips were treated with 2nM colchicine, 2nM paclitaxel, 1nM 5v or 2nM 5v for 24 hours. The cells were then incubated with alpha-tubulin antibodies (Thermo Scientific, # 62240) followed by incubation with Alexa Fluor 647 conjugated goat anti-mouse IgG (Molecular Probes) after fixation and permeabilization. The stained microtubules were observed with a Keyence BZ-X700 microscope and imaged. Assays were performed in duplicate.
X-ray crystallographyPerformed as described in example 5 to determine the crystal structures of 5m, 12e, 12j, 12k and 5v, as shown in fig. 26.
Scratch wound assay:as previously reported, 5v anti-migration effect was determined by scratch wound assay using an intycyte S3 living cell imager. After overnight incubation, monolayers of A375/TxR cells were scraped with WoundMaker (Essen BioScience) and cells were treated with 5v (0.5 nM, 1nM, 2nM and 5 nM) for 48 hours. Wounds were imaged every 2 hours by intycyte and relative wound densities were calculated by the intycyte scratch wound software module. The assays were performed in quadruplicates.
Flow cytometry analysis of cell cycle distribution and apoptosis:after overnight fixation in ice-cold 70% ethanol and incubation with 100. Mu.g/mL RNase A for 1 hour, the cell cycle distribution of A375/TxR treated with 1nM, 2nM and 5nM 5v was determined by propidium iodide staining. The cell cycle distribution was then analyzed by a ZE5 cell analyzer (Bio-Rad) at the University of Tennessee Health Science Center (UTHSC) flow cytometry and cell sorting core, and the results were processed by ModFit LT software (erity Software House). As previously described, the same cell cycle analysis was determined by FITC annexin V apoptosis detection kit (eBioscience) Apoptosis of treated A375/TxR cells. The data were analyzed by FlowJo software (Becton, dickinson, and co.).
In vivo anti-tumor study:all animal experiments were approved by the UTHSC Animal Care and Use Committee (ACUC) and performed according to protocol (protocol # 17-056) according to rules of the NIH guidelines for laboratory animal care. NOD Scid Gamma (NSG) mice (5 to 6 weeks old) purchased from Jackson Laboratories were kept in a controlled animal facility with 12:12 hours light-dark cycle. 5v tolerability was tested by using 5mg/kg or 10mg/kg Intraperitoneal (IP) or Intravenous (IV) injections per day for at least 5 consecutive days in NSG mice. For the IP group, 3 NSG mice were used in each group. For group IV, 4 NSG mice were included in each group. Mice were observed daily for physical activity, respiration, feeding, fur status and body weight to monitor for possible signs of toxicity. For xenograft models, 2×10 suspended in FBS and phenol red free medium and matrigel (50%/50%) 6 Individual a375/TxR cells were inoculated subcutaneously into the right flank of NSG mice. When the tumor grows to 100mm 3 At this time, the mice were randomly divided into 4 groups (6 mice per group): vehicle (DMSO: PEG300: tween 80: saline = 2:20:5:73), 5v (2 mg/kg), 5v (4 mg/kg), and paclitaxel (4 mg/kg). Mice were given 5v and paclitaxel intravenously twice weekly for 3 consecutive weeks. Volume = (width) by formula 2 measurements per week 2 X length)/2 calculated tumor volume and mouse body weight. When the tumor volume in the vehicle group exceeds 1500mm 3 At that time, the study was terminated. Mice were euthanized and tumors and major organs were rapidly dissected and fixed in 10% buffered formalin for further experiments.
Histological and Immunohistochemical (IHC) analysis:fixed tumor tissues and major organs (lung, liver, kidney, heart and spleen) were embedded in paraffin and histological cores studied by UTHSC were sectioned at a thickness of 4 μm. Hematoxylin-eosin (H) of major organs was performed on all tumors and major organs&E) Staining for histological examination. Representative images were obtained by a Keyence BZ-X700 microscope. As previously described, anti-Ki 67 (1:400, #9027,Cell Signaling Technology), anti-CD 31 (1:100, #77699,CST), rabbit anti-lytic caspase-3 (1:200, #9661, CST) and mouse anti-human specific mitochondria (1:1000, # ab92824, abcam) were IHC stained and anti-human mitochondrial stained. IHC slides were imaged by Keyence BZ-X700 microscope and quantification of Ki67, CD31, cleaved caspase-3 and human mitochondrial staining area was performed by 7 or 8 representative fields per group via IHC Profiler module in imageJ.
Statistical analysis: we analyzed independent groups for in vitro assays using one-way ANOVA followed by paired two-tailed Student t-test (Student t-test) or Dunnett multiple comparison test. The treatment group of the in vivo xenograft model was compared to the control group using two-way ANOVA followed by the Dunnett multiple comparison test. Significance level is expressed as:, P<0.05;**,P<0.01;***,P<0.001;****,P<0.0001。
5v targeting the colchicine binding site and inhibiting tubulin polymerization (synthesized as described in example 4): based on the X-ray crystal structures of 5m and 5t (fig. 6E to 6G), we found that the large pocket near the pyrimidine B ring can accommodate both the ethylamine moiety and saturated cycloalkanes. Thus, for 5m and 5t, 5v was synthesized following the procedure of example 4, which proved to be effective tubulin polymerization inhibitors targeting the colchicine binding site. To determine the effect of 5v on microtubules, we first characterized the molecular interactions between 5v and the colchicine binding site (fig. 26E).
To confirm the inhibition of tubulin polymerization by 5v on the microtubule network, a tubulin polymerization assay was performed in vitro. After DMSO was added to the tubulin mixture at 37 ℃, the control showed an increase in absorbance at 340nm over 40 minutes due to tubulin assembly and stabilized at a340 of 0.3 (fig. 29A). As expected, paclitaxel induced rapid tubulin polymerization within 20 minutes and stabilized tubulin assembly at a340 of about 0.4. Similar to the action of colchicine, a known tubulin polymerization inhibitor, 5v inhibited and stabilized tubulin assembly at a340 of about 0.05 in vitro, indicating that 5v is a tubulin polymerization inhibitor. We also characterized the effect of 5v on the microtubule system using A375/TxR cells. Because the A375/TxR cells were the taxol-resistant subfamily of A375 cells and were reported to have over-expression of P-glycoprotein (P-gp), and colchicine and taxol were substrates for P-gp, 2nM colchicine or taxol-treated A375/TxR cells showed a complete microtubule network similar to the untreated control group during the interphase or mitosis (FIG. 29B). However, at nanomolar concentrations of the unit order, 5v destroyed the tissue of intact microtubules and induced unorganized and diffuse microtubules. Even at a concentration of 1nM, 5v induced multipolar spindle formation (FIG. 29B). Although 2nM 5v appeared to inhibit all A375/TxR cells entering the mitotic phase, we could not find mitotic cells under the microscope (FIG. 29B).
Antiproliferative effect of 5v on paclitaxel sensitive and paclitaxel resistant cancer cell lines:as shown in FIG. 30A, 5v growth inhibition was first assessed on a panel of human cancer cell lines including melanoma cell lines A375, M14 and RPMI-7951, breast cancer cell lines MDA-MB-231, MDA-MB-453 and MDA-MB-468, and prostate cancer cell lines PC3 and DU145. We used colchicine, paclitaxel and Azixa (N- (4-methoxyphenyl) -N, 2-dimethylquinazolin-4-amine), which is currently in clinical trials, as controls. 5v showed very high antiproliferative efficacy on all cancer cell lines tested, average IC 50 The value was 0.5nM. The cytotoxicity is stronger than colchicine and Azixa and is equivalent to paclitaxel, which is a cytotoxic anticancer drug for the treatment of solid tumors. We then further determined the efficacy of 5v on paclitaxel resistant cancer cell lines (A375/TxR, M14/LCC6MDR1, MDA-MB-231/TxR, PC3/TxR, and DU 145/TxR). Despite the limited efficacy of colchicine or paclitaxel, 5v remained highly potent cytotoxicity in inhibiting the growth of all paclitaxel resistant cancer cells tested, with IC 50 The values are in the sub-nanomolar order and they are more efficient than Azixa (fig. 30B). In summary, our results show that 5v can inhibit the growth of all the paclitaxel-sensitive and paclitaxel-resistant cancer cells tested, and that its cytotoxicity to paclitaxel-sensitive cells is similar to that of paclitaxel. We also used a paclitaxel resistant cell line a375/TxR assay 5v anti-colony formation. As shown in fig. 30C, the number and size of colonies in the 5v treated a375/TxR cells were significantly smaller than the control cells. And 5v can inhibit proliferation of a375/TxR cell colonies in a concentration-dependent manner.
5v results in inhibition of cell migration, cell cycle arrest and eventual cell death of A375/TxR cells: since cell motility is dependent on microtubule structure and microtubule targeting agents are reported to always lead to G2/M cycle arrest and ultimately cell death in cancer cells, and 5v shows very strong cytotoxicity in inhibiting colony formation of a375/TxR cells, we continued to evaluate the effect of 5v on cell migration, cell cycle distribution and apoptosis induction using a375/TxR cells. As shown in fig. 31A, 5v showed significant efficacy in inhibiting cell migration of a375/TxR cells in the single digit nanomolar range. And it slowed wound healing of a375/TxR cells in a concentration-dependent manner and showed maximum inhibition after 48 hours of 5v treatment. Furthermore, flow cytometry analysis using annexin V-FITC/PI staining showed a significant increase in the number of percent apoptosis of a375/TxR cells in the 5V incubation group compared to the untreated control group, from 28% to 42% in a dose-dependent manner starting from a concentration of 2nM (fig. 31B). Furthermore, cell cycle analysis by single PI staining showed that even without serum starvation, 5v treatment induced a significant increase in the number of a375/TxR cells arrested in G2/M phase with maximum intensity at a concentration of 5nM (fig. 31C). Together, 5v showed potent anti-migration, pro-apoptotic and G2/M phase arrest effects against a375/TxR cells at single digit nanomolar concentrations.
5v strongly inhibits melanoma tumor growth in vivo: before assessing the efficacy of 5v in xenograft models, we performed a simple tolerability study to find a safe dose of 5v for in vivo studies. We first treated healthy NSG mice intraperitoneally with doses of 5mg/kg and 10mg/kg at a dose frequency of five times per week (fig. 36A-36B). During the treatment period, mice in the 5mg/kg 5v treated group were still healthy, but those mice in the 10mg/kg 5v treated group had villi and weight loss. In the first placeFive days, mice in the 10mg/kg 5v treatment group died or euthanized due to adverse conditions, so the 10mg/kg 5v treatment injected five times per week IP was toxic to the mice. We then selected 5mg/kg and 10mg/kg 5v as doses to determine potential toxicity for IV injections and 2 times per week as dose frequency. The results showed that both 5mg/kg and 10mg/kg were safe by IV injection 2 times per week (fig. 36C-36D). Our previous studies showed that the parent compounds of 5v, 5m and 5t were very effective in inhibiting the growth of paclitaxel resistant a375/TxR xenografts. To compare the effects of 5v with 5m and 5t, we also used a375/TxR xenograft model to evaluate the anti-tumor effects of 5 v. When the average tumor volume reaches 100mm 3 At this time, mice were randomly grouped according to tumor volume and body weight and given vehicle, 2mg/kg 5v, 4mg/kg 5v or reference compound paclitaxel (4 mg/kg) intravenously twice weekly until three weeks of treatment. As shown in fig. 32A, the paclitaxel-treated group had no significant tumor suppression, whereas the 5v treatment significantly reduced tumor volume, particularly at a dose of 4 mg/kg. Tumor growth inhibition values of 2Mg/kg and 4Mg/kg 5v treatment reached 62.3% and 76.6%, respectively. And no weight loss was observed in all treatment groups, indicating that mice were tolerised to two doses of 5v in this a275/TxR xenograft model (figure 32B). Furthermore, the tumor weight of mice treated with 2mg/kg 5v was reduced by 47.4% and the tumor weight of mice treated with 4mg/kg 5v was reduced by 59.9% compared to vehicle group, indicating that high dose 5v had better inhibition of tumor growth (fig. 32C-32D).
We further resected tumors from a375/TxR xenograft study and performed H & E staining to observe the extent of tumor necrosis. As shown in fig. 33A, images of H & E stained tissues showed more necrotic cells in the 5v treated tumors compared to vehicle treated or paclitaxel treated tumors, indicating that 5v can induce a375/TxR tumor necrosis. To further confirm the effect of 5v on tumor cell proliferation, angiogenesis and apoptosis in vivo, we performed IHC staining to detect Ki67, CD31 and cleaved caspase-3 expression in tumors obtained in a375/TxR xenograft model. Ki67 and CD31 expression was significantly reduced in 5v treated tumors compared to tumors treated with vehicle or paclitaxel, indicating that 5v significantly inhibited tumor proliferation and angiogenesis in tumor-bearing mice (fig. 33A-33B). However, paclitaxel-treated tumors had slightly elevated Ki67 expression relative to vehicle group, while CD31 expression was not significantly altered (fig. 33B). In addition, IHC staining results of cleaved caspase-3 showed that 5v (2 mg/kg, 4 mg/kg) significantly increased the proportion of cleaved caspase-3 positive cells in 5v treated tumors in a dose-dependent manner, thus indicating that 5v had a pro-apoptotic effect in vivo (FIGS. 33A-33B). Taken together, these data show that 5v as a microtubule polymerization inhibitor inhibits tumor growth by inhibiting tumor proliferation, targeting tumor angiogenesis, and inducing apoptosis.
5v inhibits spontaneous metastasis of A375/TxR xenografts without significant toxicity to major organs: we newly established a375/TxR xenograft models were shown to have spontaneous metastasis to the lung and liver. Therefore, in this study we also focused on the efficacy of 5v in inhibiting spontaneous metastasis. When dissected, we found that almost all mice grew in the axillary lymph nodes. We therefore collected and imaged all axillary lymph nodes (fig. 34A). Visually, 5v could potentially inhibit tumor metastasis into axillary lymph nodes. In addition, metastasis was extensively detected in the whole lung of vehicle or paclitaxel-treated group, while 5v (2 mg/kg or 4 mg/kg) treated group significantly reduced the number and size of tumor nodules in the lung (fig. 37). In addition, H of mouse lung or liver&E staining indicated that 5v significantly inhibited spontaneous lung or liver metastasis, as indicated by the yellow arrow in the representative image (fig. 37). As shown in fig. 34B-34C and representative anti-mitochondrial stained lung or liver images (fig. 34D), both 2mg/kg 5v and 4mg/kg 5v resulted in significant inhibition of lung or liver metastasis compared to vehicle, indicating that 5v was not only effective against the growth of a375/TxR tumor, but also significantly inhibited tumor metastasis to lymph nodes, lung and liver.
We have shown that 5v has no acute toxicity at doses of 2mg/kg or 4mg/kg (FIG. 32B). To determine if 5v is toxic to major organs, we further studied 5v toxicity by staining major organs (heart, kidney and spleen) with H & E, since both the lungs and liver of mice have a375/TxR tumor mass in the study and it is difficult to rule out the effects of metastasis on organ damage. The results showed that after three weeks of 5v (2 mg/kg or 4 mg/kg) treatment, there was no significant damage to the major organs of the mice, and that the 5v treated organs were similar to the H & E staining results of the vehicle and paclitaxel treated organs (FIG. 35). In summary, 5v exhibits potent anti-tumor efficacy, anti-spontaneous metastatic activity and low toxicity in vitro and in vivo, and is worthy of further investigation.
Discussion of the invention: in our effort to find new CBSI, we identified 5v as a new tubulin polymerization inhibitor. In our previous study (example 5), we reported that 5m and 5t have very strong antiproliferative efficacy both in vitro and in vivo. Thus, we hypothesize that the new analogs of 5m and 5t designed by replacing the methyl moiety of 5m with the ethylamine moiety of 5t can achieve greater cytotoxicity and potency than either 5m or 5 t. Before any in vitro or in vivo experiments were performed, we used X-ray crystallography, tubulin polymerization assays, and immunofluorescence assays to verify if the newly synthesized 5v was CBSI. As expected, 5v can target the colchicine binding site and inhibit tubulin polymerization (fig. 29A). The following MTS and colony formation assays showed, as we hypothesized, that 5v had a higher cytotoxic antiproliferative activity than 5m or 5t against a panel of cancer cells and taxol-resistant sublines, with sub-nanomolar levels of IC 50 The value (FIG. 30), and its efficacy is similar to that of paclitaxel, an anticancer drug used in clinic to treat solid tumors. We then performed scratch assays, annexin V/PI staining and cell cycle analysis experiments to further confirm the effect of 5V as CBSI on other aspects of cell growth inhibition such as cell migration, apoptosis and cell cycle arrest. In fact, 5v induced cell migration inhibition, cell cycle arrest and apoptosis in paclitaxel resistant a375/TxR cells (fig. 31).
Our further in vivo studies showed that 5v was able to inhibit the growth of a375/TxR xenografts in a dose-dependent manner without causing acute toxicity, and H & E staining of tumors in this study showed that 5v (2 mg/kg or 4 mg/kg) induced tumor necrosis in vivo (fig. 32 and 33A). Furthermore, IHC staining with cell proliferation marker Ki67, prognostic angiogenesis marker CD31 and apoptosis marker cleaved caspase-3 showed that all doses of 5v treatment significantly reduced the percentage of Ki67 positive area and CD31 positive area relative to vehicle or paclitaxel treated tumors and increased the percentage of cleaved caspase-3 positive area in a dose dependent manner in 5v treated tumors (fig. 33). However, unlike its cytotoxicity in vitro, 5v showed weaker in vivo efficacy than 5m (2 mg/kg:62.3% tumor growth inhibition compared to 70.5% of 5 m; 4mg/kg:76.6% tumor growth inhibition compared to 88.2% of 5 m), and efficacy similar to 5t (2 mg/kg:62.3% tumor growth inhibition compared to 64.6% of 5 m; 4mg/kg:76.6% tumor growth inhibition compared to 78.4% of 5 t), unlike what we expected. One reason for reduced efficacy of 5v in vivo may be low metabolic stability.
As shown in table 7, 5v did not show improved metabolic stability compared to 5m with a half-life of 53.6 minutes in human liver microsomes, 8.0 minutes in rat liver microsomes and 14.4 minutes in mouse liver microsomes, but had reduced half-lives in human liver microsomes (24.7 minutes), rat liver microsomes (3.6 minutes; not shown) and mouse liver microsomes (6.33 minutes).
Furthermore, clearance of 5v was higher than 5m in human, rat and mouse liver microsomes, further reducing the metabolic stability of 5 v. Despite the low in vivo metabolic stability, 5v remains a promising CBSI with sub-nanomolar efficacy, which can be used as a payload for antibody-drug conjugates or to load drug nanoparticles through its ethylamine moiety.
At the same time, we also demonstrated that 5v can inhibit spontaneous axillary lymph node, lung and liver metastasis from subcutaneous tumors (fig. 34 and 37). However, as shown in fig. 34A, there was no growth of axillary lymph nodes from one mouse in the vehicle group. When we collected the lungs and liver of this mouse, we found that the number and size of tumor nodules present on the lungs or liver were greater than the number and size of other mice in the vehicle group, suggesting that this particular mouse may be defective in spontaneous metastasis into the axillary lymph nodes, but more prone to migrate to the lungs or liver (fig. 38). Thus, when assessing the area of spontaneous metastasis present in the lungs or liver in the vehicle group, we excluded the mice. Thus, 5v is effective not only in inhibiting the growth of primary tumors, but also in inhibiting metastasis at doses of 2mg/kg or 4 mg/kg. And 5v deserves further investigation into many other solid tumor types, such as prostate, lung and ovarian cancer. Our previous studies have shown that CBSI represented by 17ya, 5m and 5t and 5v can effectively inhibit tumor metastasis to the liver, thus demonstrating the potential role of CBSI in inhibiting liver metastasis, which deserves further investigation. We further assessed toxicity of 5v using H & E staining, demonstrating that 5v was not toxic to major organs at doses of 2mg/kg and 4mg/kg intravenously 2 times/week (FIG. 35). And according to its tolerability study we show that 5v has no acute toxicity when given 10mg/kg intravenously twice weekly (figure 36). Although 5v is not more effective than 5m at the same in vivo dose level, it is safer than 5m, which is toxic when higher doses (5 mg/kg) are administered, further supporting the development of new CBSI based on the discovery of 5 v.
In summary, we synthesized analogues of CBSI 5m and 5t that we previously reported, and named 5v. We then obtained a 5v high resolution X-ray crystal structure and identified it as CBSI that inhibited tubulin polymerization. We also showed that 5v effectively inhibited the growth of various cancer cell lines, overcome paclitaxel resistance, and have the effects of inhibiting cancer cell migration, inducing apoptosis and G2/M cell cycle arrest in vitro. Furthermore, in vivo studies showed that 5v has strong anti-tumor and anti-spontaneous metastasis efficacy in a375/TxR xenograft mouse model without causing toxicity to major organs in the mice. Thus, preclinical evaluation of 5v strongly supports the development of 5v as a next generation tubule inhibitor and deserves further investigation.
Example 11: treatment of head and neck cancer with dihydroquinoxalinones of the invention
A set of dihydroquinoxalinones was tested in two head and neck cancer cell lines a-253 and Detroit 562. A-253 cells are human salivary epidermoid carcinoma cell lines, while Detroit 562 is Head and Neck Squamous Cell Carcinoma (HNSCC). Oral cancer is the most common type of head and neck cancer, and more than 90% of oral cancers are oral or oropharyngeal Squamous Cell Carcinoma (SCC). The major clinical dilemma is the efficiency of chemotherapy targeting Head and Neck Squamous Cell Carcinoma (HNSCC). Failure to successfully complete cancer treatment using standard surgical procedures results in continued research into new compounds with cytostatic activity and fewer side effects due to advanced stages of HNSCC and/or resistance of SCC cells to conventional chemotherapy and/or radiation therapy.
In this experiment, four compounds (SP-I-104, 5m, 12k and 5v HCl) were tested for cytotoxicity in vitro in two head and neck cancer cell lines A-253 and Detroit 562. As can be seen in fig. 39A, three of the four compounds (5 m, 12k and 5v HCl) produced ICs at low nM to high pM levels 50 Cytotoxicity value, thereby demonstrating the ability to effectively inhibit head and neck cancer growth. For example, IC of these three compounds 50 The values range from 0.52nM to 3.2nM. Fig. 39B and 39C show graphical representations of data. From these preliminary screening experiments, 12k was selected as a lead for further study and compared to the structurally unrelated CBSI compound 17 ya. In these experiments, 12k unexpectedly had about 10-fold higher potency (fig. 40A and 40B) than compound 17ya (fig. 40C and 40D) in the a-253 and Detroit 562 cell lines. For example, at therapeutic levels as low as 0.5nM, 12k can almost completely prevent proliferation of head and neck cancer cells, while 17ya at 5nM is required to achieve similar antiproliferative properties. The result diagrams are shown in fig. 40A to 40D. Similarly, as shown in fig. 41A and 41B, respectively, 12k also showed potent ability to prevent colony formation, with almost complete inhibition observed in 1nM in a-253 cells compared to 2nM in Detroit 562 cells.
Furthermore, the anti-mitotic mechanism of action was confirmed by western blot analysis, which showed that apoptosis was induced over time in the tested head and neck cancer cell types. After treatment of these cell lines with 100nM 12k for 72 hours, the level of apoptosis markers is elevated, such as lysis of PARP (c-PARP) and cas 3 (c-cas 3). FIGS. 42A and 42B illustrate these results for cell lines A-253 and Detroit 562, respectively. This data suggests that anticancer activity in head and neck cancer works through induced apoptosis, consistent with the known mechanism of action of colchicine binding site inhibitors of these compounds.
Example 12: synthesis of 5v and 5v HCl
Compound 5v (4- (2- (ethylamino) -6, 7-dihydro-5H-cyclopenta [ d ] pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one) was synthesized by the scheme shown in fig. 43.
Chemical:4-chloro-2- (methylthio) -6, 7-dihydro-5H-cyclopenta [ d ]]Pyrimidine 1 (4) (Aware et al, "cyclyl-pyrimidine based analogues as novel and potent IGF-1R inhibitor," European Journal of Medicinal Chemistry,2015,92,246-256) was obtained from pyrimidine (3) and ethyl 2-oxocyclopentanecarboxylate (1) and (4) was coupled with 4-methoxy-2-nitroaniline (5) in anhydrous IPA in the presence of a catalytic amount of HCl (3 drops to 4 drops) to give 2-methylthio 4- (4-methoxy-2-nitrophenyl) aminopyrimidine (6) in good yield. (Cui et al, "In vivo and mechanistic studies on antitumor lead 7-methoxy-4- (2-methylquinazolin-4-yl) -3, 4-dihydroquinazolin-2 (1H) -one and its modification as a novel class of tubulin-binding tuner-vascular disrupting agents," j. Med. Chem.,2017,60,5586-5598.) the nitro groups are then reduced to amines (7) in catalytic AcOH at 0 ℃ with the aid of zinc powder. The resulting amine was immediately coupled with chloroacetyl chloride to yield 8. Next, compound 8 undergoes intramolecular cyclization in anhydrous THF in the presence of NaH to yield intermediate 9, which is oxidized in methanol and water in the presence of oxone to yield 10. Finally, the sulfone group on the pyrimidine ring of 10 is replaced with ethylamine at 100 ℃ to 110 ℃ in the presence of anhydrous 1, 4-dioxane to give 5v.
General procedure
All nonaqueous reactions were carried out in oven-dried glassware under an inert atmosphere of dry nitrogen. All reagents and solvents were purchased from Aldrich (St. Louis, mo.), alfa-Aesar (Ward H)ill, MA), combi-Blocks (San Diego, CA), ark Pharm (Libertyville, IL), and were used without further purification. Analytical thin layer chromatography was performed on silica gel GHLF 10cm×20cm Analtech TLC Uniplates (Analtech, newark, DE) and visualized by fluorescence quenching under UV light. The compound was purified using silica gel (60 mesh-120 mesh or 100 mesh-200 mesh). Recording on a Varian Inova-500 spectrometer (400 MHz) (Agilent Technologies, santa Clara, calif.) or a Bruker ascase 400 (400 MHz) (Billerica, mass.) spectrometer 1 H NMR 13 C NMR spectrum. Chemical shifts are reported in ppm on delta scale and referenced to the appropriate solvent residual peak (CDCl) 3 For the following 1 H is 7.27ppm and for 13 C is 77.23ppm, and DMSO-d 6 For the following 1 H is 2.50ppm and for 13 C is 39.51 ppm), all coupling constants (J) are given in hertz (Hz). Mass spectra were collected in positive and negative modes on a Bruker amazon SL electrospray/ion trap instrument. High Resolution Mass Spectrometer (HRMS) data were obtained on a Waters Xevo G2-S qTOF (Milford, MA) system equipped with an Acquity I type UPLC system. Porcine brain tubulin (catalog number T-238P) was purchased from Cytoskeleton, inc. All test compounds were found to be 95% pure by 1H NMR and HPLC. The HPLC method for determining purity is as follows: compound purity was analyzed using an Agilent 1100HPLC system (Santa Clara, CA) with a Zorbax SB-C18 column (particle size 3.5 μm,4.6mm x 150 mm) from Agilent. The mobile phase consisted of water (a) with 0.1% formic acid and acetonitrile (B) with 0.1% formic acid. A flow rate of 1mL/min was used. Gradient elution was started from 50% B. It reaches 100% B from 0 to 9 minutes, remains from 9 to 12 minutes, then decreases from 12 to 15 minutes to 50% B and stops. Compound purity was monitored with a DAD detector set at 254 nm.
Chemical synthesis:
4- (2- (ethylamino) -6, 7-dihydro-5H-cyclopenta [ d ]]Synthesis of pyrimidin-4-yl) -7-methoxy-3, 4-dihydroquinoxalin-2 (1H) -one (5 v-HCl). A mixture of 10 (100 mg,0.26 mmol), ethylamine (36 mg,0.80 mmol) in 1, 4-dioxane was heated in a sealed tube to 110℃for 6 hours. The mixture was poured into ice water, and the solid was collected by filtration, usingWashed with water (5X 10 mL) and dried. The crude product was purified by column chromatography to give pure salt-free 5v (80 mg) as an off-white solid. Ether HCl (0.5M HCl in diethyl ether) 0.52mL (1.1 mol) was added to a secondary amine 5v in CH 2 Cl 2 In the solution of (2) and in N 2 Stirring was carried out at room temperature for 5 hours under an atmosphere. Then, the solvent was removed and the resulting hydrochloride salt of the amine (5 v-HCl) (50 mg,56% yield) was dried under high vacuum; 1 H NMR(400MHz,DMSO-d 6 )δ10.84(s,1H),8.04(bs,2H),7.08(d,J=8.1Hz,1H),6.65-6.63(m,2H),3.88(bs,1H),3.76-3.72(m,4H),3.44-3.41(m,2H),2.83(bs,1H),2.08(bs,1H),1.88(bs,2H),1.18(t,J=6Hz,2H); 13 C NMR(100MHz,DMSO-d 6 )δ167.56,158.48,133.92,125.18,119.29,107.38,102.11,55.90,49.70,36.50,31.18,22.44,14.75;HRMS[C 18 H 22 N 5 O 2 + ]calculated 340.1773, measured 340.1777; HPLC purity 97.0%; decomposing at 210 ℃ to 211 ℃.
Example 13: treatment of metastatic properties that have been transferred to the brain (BrnMets) with brain penetrating agent compounds of the invention Breast Cancer (MBC)
Challenges in MBC treatment and limitations of FDA approved tubulin inhibitors: despite significant advances in breast cancer treatment, effective treatment of Metastatic Breast Cancer (MBC) remains challenging. The major metastatic sites in MBC generally include bone (41%), lung (22%), liver (8%) and brain (7%). While the precise distribution among these primary metastatic sites depends on the specific breast cancer molecular subtype, the most common site is bone and the most refractory site is brain. In general, about 70% of women with ER negative MBC (TNBC or HER2 positive) will develop brain metastases (BrnMets) within 5 years of diagnosis, with metastases at any other site, whereas ER positive MBC is a much larger patient population, the burden of which can ultimately lead to BrnMets. Patients with bone destruction lesions (osteolytic) are particularly susceptible to fractures and chronic pain. In addition to liver metastases, where surgical resection is the standard treatment, systemic treatment is the primary treatment option for patients with stage IV, with limited efficacy. Thus, there is a significant unmet medical need in developing new systemic/targeted therapies for the effective treatment of MBC metastases, particularly brain metastases (BrnMets) and bone metastases.
Tubulin inhibitors such as paclitaxel (paclitaxel/Taxol) and docetaxel (docetaxel/Taxotere) are classical systemic drugs and are widely used in the treatment of patients suffering from metastatic disease. The three most characteristic binding sites in tubulin are the taxane site, the vinca site and the colchicine site. Currently, all FDA approved tubulin inhibitors for cancer treatment bind to taxane sites (e.g., paclitaxel) or vinca sites (e.g., vinblastine). However, their clinical use is associated with several limitations. First, they are often good substrates for drug efflux pumps, including P-glycoprotein (P-gp), breast Cancer Resistance Protein (BCRP), or multi-drug resistance protein (MRP). They are also susceptible to resistance mediated by β3-tubulin overexpression. Thus, multi-drug resistance (MDR) is often developed after long-term drug administration. Although newer taxanes, such as cabazitaxel (cabazitaxel), are not susceptible to P-gp mediated drug efflux, they are still susceptible to beta 3-tubulin mediated resistance. Second, approved tubulin inhibitors have dose-limiting hematopoietic and cumulative neurotoxicity, including peripheral neuropathy, leading to treatment window narrowing. Third, these approved drugs have poor water solubility, which requires the use of surfactants (e.g., cremophor EL) in their formulations. Surfactants may cause additional side effects and require pre-drug treatment with corticosteroids/antihistamines. The emerging data indicate that corticosteroids activate tumor-promoting stress response pathways and enrich cancer stem cell-like activity in Triple Negative Breast Cancers (TNBC). Efforts have been made to develop new generation taxane drugs, including oral formulations of paclitaxel (Oraxol) and chemically modified paclitaxel (tesetaxel), but both were not approved by the FDA early in 2021, possibly due to the inherent limitations of taxane stents. Since tubulin inhibitors are first-line drugs for stage IV breast cancer, there is a strong clinical need to develop new tubulin inhibitors for more effective treatment. The compounds of the present invention, which are Colchicine Binding Site Inhibitors (CBSI), effectively act as anti-tubulin agents, which lack the therapeutic limitations of taxanes and vinca alkaloids and variants thereof.
The studies presented herein have focused on structural optimization to produce potent and highly brain-permeable tubulin inhibitors that can also overcome taxane resistance for MBC brain metastasis (BrnMets) treatment. This ability will not only prolong survival and improve QOL in MBC patients, but also in patients of other cancer types who currently use tubulin inhibitors. We screened for new analogs in vitro using a panel of TNBC and her2+ conventional cell lines (including taxane-sensitive and taxane-resistant cell lines) and early passage cells derived from taxane-sensitive and taxane-resistant PDX models, as well as normal cells. We also determined brain permeability, maximum tolerability (not shown) and optimum Pharmacokinetics (PK) of the most active compounds in this series to select the best compounds for downstream in vivo efficacy studies. We selected 5m from the compounds of the other examples as the overall best compound for in vivo evaluation. In vivo evaluation involved the use of taxane-sensitive, well-characterized preclinical models that developed BrnMets (MDA-MB-231 BrM sub-line) after intracardiac injection to score delays in metastasis progression and increases in Overall Survival (OS) in response to treatment. The reduction of BrnMets burden and progression is highly likely to significantly improve PFS, OS and QOL in MBC patients. In addition, patients diagnosed with other types of metastatic solid tumors, where tubulin inhibitors are the current standard of care (SOC), may also benefit from the present invention.
The BrnMets material presented here is divided into three types of experiments: 1) brain penetration of 5m was determined as measured by brain to plasma concentration ratio (B/P), 2) delay in transfer progression was measured with 5m compared to vehicle (all animals were euthanized at the same time point), and 3) comparison of OS of 5m compared to vehicle (euthanized animals when dying due to transfer).
The increased B/P ratio of the compounds of the invention.In the blood brain barrier penetration study, male NSG mice were injected by tail vein at 4mg/kg5m (four mice) or 12k (three mice)Intravenous administration. After one hour, the mice were anesthetized, blood was collected by cardiac puncture, and brain samples were collected after perfusion with saline to remove all blood in the brain. The plasma of the blood sample is treated by centrifugation. Brain samples were homogenized using homogenization buffer (1:2 methanol: PBS) at a ratio of 1:9 (1 gram brain tissue to 9mL buffer). Following established protocols, tissue samples were kept at-80 ℃ until LC-MS/MS analysis [ PMID:22760659]. The concentration (nM) ratio of 5m or 12k to plasma (P) in brain (B) was defined as the B/P profile, measured as 4.56 for 5m and 0.45 for 12k (FIG. 44). As a comparison, compound 17ya (evaluated in nude mice) had much lower brain penetration (B/P ratio of 17ya was only 0.054 at 1 hour, only 0.089 at 4 hours, [ PMID: 22760659) ])。
Most breast cancer patients die from tumor metastasis, and therefore, durable treatment of Metastatic Breast Cancer (MBC) must have good efficacy in treating metastasis. Of the four major sites of MBC metastasis (bone, lung, liver and brain), brain metastases (BrnMets) are the most challenging sites for treatment because of the need for drug availability across the blood: brain barrier (BBB). Compound 17ya has limited BBB permeability and therefore it is unlikely to be optimal for the treatment of BrnMets. Interestingly, azixa (an effective CBSI) is rapidly and widely distributed into the brain, exhibiting 14-fold higher brain exposure relative to plasma and an elimination half-life similar to plasma [ PMID:19296653]. Although Azixa cannot be used as a cancer therapeutic due to its toxic metabolite, its radiolabeled version with carbon-11 is currently in clinical trials as a PET imaging agent (e.g., NCT 04575727) for neurodegenerative disease applications because of its very high brain penetration capacity. We hypothesize that proper hybridization of 17ya (metabolically stable, good safety profile, but low brain penetration) and Azixa (potent, excellent brain penetration, but metabolically unstable and highly toxic to the heart and liver) will "dial out" their respective limitations to produce metabolically stable, low toxic and brain permeable CBSI. Through extensive pharmaceutical chemistry we have found a scaffold for dihydroquinoxalinones, as exemplified throughout the examples, with 5m as the lead drug currently used for the treatment of BrnMets (fig. 45). 5m maintains the ability to overcome taxane resistance (see fig. 10A-10D, 13A-13D, 19A, 19B, 19E, etc.), and has a brain to plasma (B/P) distribution ratio of 4.56 (NSG mice, 4mg/kg, IV bolus, n=4, see the previous paragraph for details). Furthermore, FIG. 19C shows that 5m is able to overcome resistance to 17ya in the TNBC cell line MDA-MB-231/VxR.
These results indicate that, unlike standard therapeutic chemotherapy, 5m and other compounds of the invention are able to reach the site of action in the brain, which is a prerequisite for effective treatment of MBC BrnMets. Furthermore, due to their lack of sensitivity to taxane/vinca resistance mechanisms and 17ya resistance mechanisms, the compounds of the present invention will be able to treat BrnMets patients in severely pre-treated patients, even if these patients have failed in taxane, vinca and/or 17ya therapies.
Effect of the Compounds of the invention on growth of breast cancer transferred to the brain in mice
After confirming brain penetration by 5m, we tested the ability to prevent MBC transfer to the brain and treat BrnMets progression to preferentially induce BrnMets using the widely used MDA-MB-231BrM (BrM) cell line after Intracardiac (IC) injection (PMID: 19421193). The most effective method of enriching breast cancer BrnMets in mice is by Intracardiac (IC) injection of a single cell suspension of tumor cells into hyperimmune compromised NSG mice. As shown throughout the examples, 5m and its analogs have a broad range of anticancer activity, including in many taxane resistance models. For example, the TNBC model that is commonly used is the MDA-MB-231 cell line (see FIG. 30A) and its taxane-resistant subline MDA-MB-231/TxR (see FIG. 30B).
Consistent with clinical observations that about 95% of MBC patients diagnosed with BrnMets have metastases at least one other site, typically at multiple extracranial sites (such as bone, liver, and lung), the BrM model rapidly produces BrnMets, but will also develop other metastases over time. Mortality is driven primarily by BrnMets in the BrM model. Importantly, the BrM model expresses a luciferase reporter gene to enable the use of PMID: the method in 31645441 performs longitudinal bioimaging and tracks the transfer pattern over time. All bioimaging data are shown as total flux (photons/s) calculated using the Living Image software. At harvest, all organs were collected and bioimaged ex vivo to confirm the location of the metastasis signal observed in intact mice and to quantify the organ-specific metastasis burden.
It was exciting that in the pilot study 5m reduced the 231-BrM BrnMet load (showing bioimaging of total luminous flux; all mice were harvested 28 days after treatment) (see fig. 46A).
Procedure: in experiment 1, the MDA-MB-231-BrM2 (BrM 2) brain metastasis enrichment sub-line was obtained from Memorial Sloan Kettering Cancer Care (MSKCC). A total of 200,000 cellular ICs were injected into NSG female mice from 8 weeks to 9 weeks old. Treatment was started 48 hours later and IV administration was 2 x/week. The initial dose was 1.5mg/kg, but mice lost weight and had diarrhea, so the dose was reduced to 1.0mg/kg. All mice were harvested simultaneously on day 24 of treatment.
Figure 47 shows that 5m treatment allowed mice to maintain body weight throughout day 24, while vehicle-treated mice steadily decreased in body weight from day 15 to day 24. Weight loss is common when mice are ill due to metastatic load. In addition, weight loss is associated with signs and symptoms of brain metastases, including sleepiness, difficulty walking, head tilting, etc.
In fig. 48, brains were excised after ex vivo bioimaging in vehicle-treated (top) and 5 m-treated (bottom) mice shown harvested on day 24. Quantitative analysis showed that the 5m treatment reduced the total photon flux from an average of 6.4×107p/s to 2.2×107p/s compared to vehicle treatment, and this difference was statistically significant (p=0.044). In fig. 49A and 49B, the in vivo head imaging (right panel of each panel treatment) and the subsequent ex vivo head imaging (left panel of each panel treatment) of whole mice for the same animals are shown for vehicle and 5m treated animals. The reduction in photon flux in the ex vivo brain can be further appreciated using the same capture time (1 minute); photon flux observed in 5m treated ex vivo brains (see rightmost image; 1.51X107 p/s) was reduced compared to vehicle treated brains (second plot from left; 6.01X107 p/s). Animals were imaged using Perkin Elmer XMRS instrument.
Figures 50A-50C also show that BrM cells will transfer to bone, lung and spleen. Extracranial metastases were observed in both treatment groups (fig. 50A, 50B and 50C, respectively), but treatment with 5m reduced or delayed metastatic growth of MBC, not only to the brain (as described above), but also statistically reduced metastases to bone, lung and spleen, as determined by reduced total photon flux measured ex vivo in these organs in the same experiment.
The compounds of the invention are useful in mice (euthanized animals only when moribund) with breast cancer metastasis to the brain Effect of overall survival
In an independent study to measure survival, brM2 cells (100,000) ICs were injected into 8-to 9-week old mice and treatment was initiated after 24 hours. Treatment was started 24 hours later at a dose of 1.0mg/kg, given twice weekly via the IV route. The reduced number of injected cells is selected in an attempt to increase the duration of the treatment. In this study, mice were harvested only when they met the euthanasia criteria to generate Kaplan-Meier survival curves. Not only did 5m significantly reduce brain signals in vivo over time, similar to the data from experiment #1 shown in fig. 46A-46B above, but Overall Survival (OS) was significantly improved at an impressive risk ratio (hr=5.13) (p=0.018) as shown in fig. 51.
In this survival study (experiment # 2), mice treated with 5m survived longer, as indicated by the% change in body weight over time in figure 52. In addition, the 5m cohort increased body weight on day 21 of dosing, while vehicle-treated mice began to lose body weight on day 11. The% change in body weight in both cohorts appeared to change in the latter phase of the experiment, as the average reflects animals that remain alive only on those days, and as extreme weight loss (15-20%) was the primary euthanasia criterion. Arrows indicate days of administration of 5m or vehicle.
Consistent with previous studies in which mice were harvested on the same day, mice in the survival study suffered from brain tumors, measured as an increase in total photon flux in the brain over time. Figure 53 shows that 5m treatment delayed the progression of metastasis in the brain as observed by the average total photon flux decreasing at each time point greater than 14 days (the signal tracked in the figure was from brain alone). Despite the high variability of the data, the effect of the 5m treatment was statistically significant at day 28 (p-value 0.0141).
In fig. 54, in vivo imaging (using the same biological imaging capture time) of a single representative mouse (n=1, therefore error bar free) from each cohort was tracked over time. Again, by day 14, vehicle-treated mice began to show differences in total luminous flux compared to the 5 m-treated mice, again indicating that 5m delayed the transfer progression, and that on day 28 the transfer was increased 6.7-fold, although each mouse showed similar BrnMets luminous flux onset values (7.5 x 10 5 Compared with 6.3X10 5 ). In addition, representative vehicle-treated mice died at day 28, while 5 m-treated mice survived until day 35. Animals were imaged using Perkin Elmer XMRS instrument.
In survival studies, other sites of extracranial metastases were not significantly inhibited by 5m, as expected, because the euthanasia criteria were based on morbidity/survival and the cause of death was metastasis. In fig. 55, which includes a table, the survival of each queue was tracked over time and plotted on a kaplan-meyer survival curve. The table shows the day of euthanasia for each of the six mice in each cohort. All vehicle-treated mice died at 30 days, while all 5 m-treated mice survived for more than 30 days. The median survival of 5m treated mice was 36.5 days, while vehicle treated mice were 25 days. Finally, the kaplan-Meier curve shows a statistically significant increase in survival up to the 5m incidence treatment cohort, with a risk ratio of 5.13, and a p-value of 0.018.
In conclusion, 5m and the compounds of the present invention show brain penetrating agents and maintain very strong CBSI activity in vivo. In addition, the ability to delay progression of breast cancer metastasis has been demonstrated to include inhibition of metastasis to the brain, bone, spleen, and lung. In a separate study, these compounds were shown to increase OS due to their ability to inhibit metastasis.
While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those skilled in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims (32)

1. A compound having the structure of formula I:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl) s,-NHPh、-NH(C 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
2. A compound having the structure of formula IA:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl group),
-N(C 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocycle having at least one N, O or S atom, wherein said cycloalkyl ring or heterocycle can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocycle can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
3. A compound having the structure of formula II:
wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halide, and,C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, whichWherein the substituents of the cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof, and a pharmaceutically acceptable excipient.
4. A compound of formula I represented by any one of the following compounds 5j-5r, 5t-5v or 12a-12m and 12o-12 q:
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
5. A compound having the structure of formula III:
wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
6. A compound of formula III represented by any one of the following compounds:
Or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
7. A method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula I wherein the structure of formula I is
Wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein the cycloalkyl ring or heterocyclic ring can optionally haveHaving at least one unsaturation, wherein the cycloalkyl ring or heterocycle may be optionally substituted, wherein the substituents of the cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
8. The method of claim 7, wherein the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer.
9. The method of claim 8, wherein the cancer has metastasized to the brain.
10. The method of any one of claims 7 to 9, wherein the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
11. The method of claim 10, wherein the breast cancer is any one of triple negative breast cancer, HER2 positive breast cancer, or ER positive breast cancer that has metastasized to the brain (BrnMets).
12. A method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula I wherein the structure of formula IA is
Wherein the method comprises the steps of
R 1 Is a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, C 1 -C 4 Haloalkoxy, ph, O (C) 5 -C 10 Aryl), OPh, (C) 1 -C 3 Alkyl) phenyl, -O (C) 1 -C 3 Alkyl) phenyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 2 is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl group),
-N(C 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halogenChemical compound, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Taken together form a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocycle having at least one N, O or S atom, wherein said cycloalkyl ring or heterocycle can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocycle can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocycle include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
13. The method of claim 12, wherein the cancer is at least one of: a drug resistant tumor; metastatic cancer;
or drug resistant cancer.
14. The method of claim 13, wherein the cancer has metastasized to the brain.
15. The method of any one of claims 12 to 14, wherein the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
16. The method of claim 15, wherein the breast cancer is any one of triple negative breast cancer, HER2 positive breast cancer, or ER positive breast cancer that has metastasized to the brain (BrnMets).
17. A method of treating cancer in a subject in need thereof by administering to the subject a therapeutically effective amount of: a compound of the structure of formula II, wherein the structure of formula II is
Wherein the method comprises the steps of
R 2 Is at least one of the following: hydrogen, halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 3 is hydrogen, halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 、-NH(C 1 -C 4 Heteroalkyl), -NHPh, -NH (C) 3 -C 10 Aryl) -NH (C) 3 -C 10 Heteroaryl), -NH (C) 3 -C 10 Cycloalkyl), -NH (C) 3 -C 10 Heterocyclyl), hydroxy, cyano, NCS, C 3 -C 6 Heterocyclyl or C 2 -C 5 An ether, wherein the heterocyclyl has at least one of O, N or S, and wherein the heterocyclyl can be optionally substituted, wherein the substituents of the heterocyclyl include halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
R 4 and R is 5 Is at least one of the following: halides, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano, C 2 -C 5 Ethers, or
Wherein R when taken together 4 And R is 5 Forming a 5-or 6-membered cycloalkyl ring or a 5-or 6-membered heterocyclic ring having at least one N, O or S atom, wherein said cycloalkyl ring or heterocyclic ring can optionally have at least one unsaturation, wherein said cycloalkyl ring or heterocyclic ring can optionally be substituted, wherein the substituents of said cycloalkyl ring or heterocyclic ring comprise a halide, C 1 -C 4 Alkyl, C 1 -C 4 Alkoxy, C 1 -C 4 Haloalkyl, -NH 2 、-NH(C 1 -C 4 Alkyl), -N (C) 1 -C 4 Alkyl group 2 Hydroxy, cyano or C 2 -C 5 An ether;
provided that if R 4 And R is 5 Taken together to form a phenyl ring, said phenyl ring being substituted, or if R 4 And R is 5 Taken together to form a pyridine ring, R 3 Not chlorine; and is also provided with
n is 1 to 3;
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof, and a pharmaceutically acceptable excipient.
18. The method of claim 17, wherein the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer.
19. The method of claim 18, wherein the cancer has metastasized to the brain.
20. The method of any one of claims 17 to 19, wherein the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
21. The method of claim 20, wherein the breast cancer is any one of triple negative breast cancer, HER2 positive breast cancer, or ER positive breast cancer that has metastasized to the brain (BrnMets).
22. A method of treating cancer in a subject in need thereof by administering a therapeutically effective amount of at least one of the following compounds:
/>
23. the method of claim 22, wherein the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer.
24. The method of claim 23, wherein the cancer has metastasized to the brain.
25. The method of any one of claims 22 to 24, wherein the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
26. The method of claim 25, wherein the breast cancer is any one of triple negative breast cancer, HER2 positive breast cancer, or ER positive breast cancer that has metastasized to the brain (BrnMets).
27. A compound having the structure:
or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
28. A method of treating cancer in a subject in need thereof by administering a therapeutically effective amount of: a compound of the structure:
Or a stereoisomer, pharmaceutically acceptable salt, hydrate, N-oxide, or combination thereof.
29. The method of claim 28, wherein the cancer is at least one of: a drug resistant tumor; metastatic cancer; or drug resistant cancer.
30. The method of claim 29, wherein the cancer has metastasized to the brain.
31. The method of any one of claims 28 to 30, wherein the cancer is at least one of: prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, colon cancer, leukemia, lymphoma, head and neck cancer, pancreatic cancer, esophageal cancer, renal cancer, or CNS cancer.
32. The method of claim 31, wherein the breast cancer is any one of triple negative breast cancer, HER2 positive breast cancer, or ER positive breast cancer that has metastasized to the brain (BrnMets).
CN202280038735.6A 2021-04-20 2022-04-20 Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors Pending CN117440954A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/177,183 2021-04-20
US202263317931P 2022-03-08 2022-03-08
US63/317,931 2022-03-08
PCT/US2022/025637 WO2022226118A1 (en) 2021-04-20 2022-04-20 Metabolically stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors

Publications (1)

Publication Number Publication Date
CN117440954A true CN117440954A (en) 2024-01-23

Family

ID=87932327

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280038735.6A Pending CN117440954A (en) 2021-04-20 2022-04-20 Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors

Country Status (2)

Country Link
US (1) US20230286995A1 (en)
CN (1) CN117440954A (en)

Also Published As

Publication number Publication date
US20230286995A1 (en) 2023-09-14

Similar Documents

Publication Publication Date Title
EP3601281B1 (en) CRYSTALLINE FORMS OF 4-(1-(1,1-DI(PYRIDIN-2-YL)ETHYL)-6-(3,5-DIMETHYLISOXAZOL-4-YL)-1H-&amp;#xA;PYRROLO[3,2-B]PYRIDIN-3-YL)BENZOIC ACID THAT INHIBITS BROMODOMAIN
EP3184519B1 (en) Acid-addition salt of trk-inhibiting compound
TWI433677B (en) Heterocyclic compounds and uses thereof
JP6412920B2 (en) 2-Aminopyrido [4,3-D] pyrimidin-5-one derivatives and their use as Wee-1 inhibitors
US20160340357A1 (en) Solid forms of a compound for modulating kinases
KR102605546B1 (en) Pyrimidopyrimidinones useful as Wee-1 kinase inhibitors
AU2014240003B2 (en) Coumarin derivatives and methods of use in treating hyperproliferative diseases
JP6307088B2 (en) Quinazolinone and isoquinolinone derivatives
US20160145238A1 (en) Heteroaryl substituted pyrazoles
TW201625620A (en) Heterocyclic hydroxamic acids as protein deacetylase inhibitors and dual protein deacetylase-protein kinase inhibitors and methods of use thereof
JP2016522231A (en) Diaminoheteroaryl substituted pyrazoles
CN104837841A (en) Substituted indazol-pyrrolopyrimidines useful in the treatment of hyperproliferative diseases
US9464093B2 (en) Substituted imidazo[4&#39;,5&#39;:4,5]cyclopenta[1,2-e]pyrrolo[1,2-a]pyrazines and oxazolo[4&#39;,5&#39;:4,5]cyclopenta[1,2-e]pyrrolo[1,2-a]pyrazines for treating brain cancer
EP3077392B1 (en) Crystalline forms of n-(4-((3-(2-amino-4-pyrimidinyl) - 2-pyridinyl)oxy)phenyl)-4-(4-methyl - 2-thienyl)-1-phthalazinamine pharmaceutically acceptable salts and uses thereof
Wang et al. Based on 2-(difluoromethyl)-1-[4, 6-di (4-morpholinyl)-1, 3, 5-triazin-2-yl]-1H-benzimidazole (ZSTK474), design, synthesis and biological evaluation of novel PI3Kα selective inhibitors
CN117440954A (en) Metabolic stable pyrimidinyl dihydroquinoxalinones as tubulin polymerization inhibitors
WO2023288002A1 (en) Phthalazinone-based parp-1 inhibitors
WO2023030335A1 (en) Compound as tyk2/jak1 pseudokinase domain inhibitor, and synthesis and use methods
JP2023552368A (en) Y-box binding protein 1 inhibitor
KR20240012386A (en) Metabolically stable pyrimidinyl dihydroquinoxalinone as a tubulin polymerization inhibitor.
US11945814B2 (en) Salt form
KR102053933B1 (en) Compound as TRAP1 inhibitor and pharmaceutical composition for anticancer comprising the same
JP6382964B2 (en) Phthalazine derivatives
US11801243B2 (en) Bromodomain inhibitors for androgen receptor-driven cancers
SK500742020A3 (en) Compound cemtirestat disulfide, prodrug of aldoketoreductase inhibitor, method of its preparation, pharmaceutical composition containing it and its use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination