CA2926928A1 - Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents - Google Patents

Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents Download PDF

Info

Publication number
CA2926928A1
CA2926928A1 CA2926928A CA2926928A CA2926928A1 CA 2926928 A1 CA2926928 A1 CA 2926928A1 CA 2926928 A CA2926928 A CA 2926928A CA 2926928 A CA2926928 A CA 2926928A CA 2926928 A1 CA2926928 A1 CA 2926928A1
Authority
CA
Canada
Prior art keywords
cancer
compound
chosen
acetyl
propiolyl
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.)
Abandoned
Application number
CA2926928A
Other languages
French (fr)
Inventor
Gervais Berube
Carlos Reyes-Moreno
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.)
3R VALO SEC
Original Assignee
3R VALO SEC
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 3R VALO SEC filed Critical 3R VALO SEC
Priority to CA2926928A priority Critical patent/CA2926928A1/en
Publication of CA2926928A1 publication Critical patent/CA2926928A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/44Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members
    • C07D207/444Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5
    • C07D207/448Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5 with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms, e.g. maleimide
    • C07D207/452Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5 with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms, e.g. maleimide with hydrocarbon radicals, substituted by hetero atoms, directly attached to the ring nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/36Oxygen or sulfur atoms
    • C07D207/402,5-Pyrrolidine-diones
    • C07D207/4162,5-Pyrrolidine-diones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms

Landscapes

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

Abstract

There are provided compounds of formula (I) (see formula I) in which R1, R2, R3, R4 and Q can represent various different possibilities.
These compounds can be useful as anticancer agents as well as anti-inflammatory agents and/or anti-metastatic agents.

Description

= CA 02926928 2016-04-11 AMINOBENZOIC ACID DERIVATIVES FOR USE AS ANTI-INFLAMMATORY
AGENTS, ANTI-METASTATIC AGENTS AND/OR ANTICANCER AGENTS
FIELD OF THE DISCLOSURE
[0001] This disclosure relates to the field of active agents.
More particularly, this disclosure relates to anti-inflammatory agents, anti-metastatic agents and anticancer agents.
BACKGROUND OF THE DISCLOSURE
[0002] There are several methods used to treat cancer.' The most common are:
surgery, chemotherapy, radiation therapy, targeted therapy and immunotherapy."

Other procedures are based on stem cell transplant, photodynamic therapy, and cryogenic therapy. Lasers are nowadays a useful tool during surgery of localized cancers. Many of these methods are quite effective. However, most present important side effects.7'8 Hence, the need to discover alternative therapeutics and treatment modalities. Particularly, compounds and treatment protocols that could simultaneously attack cancer on diverse fronts (initiation, propagation, metastasis etc.) are of great interest.
SUMMARY OF THE DISCLOSURE
[0003] According to one aspect, there are included compounds of formula (1) :

(I) wherein R1 is H, alkyl or halogen;
R2 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
Q is QA or QB;

õN
QA= R6 .
( QB .
= a single bond or a double bond;
R5 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R6 is H, Boc, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;

= CA 02926928 2016-04-11 R7 is a substitued or unsubstituted member chosen from C1-C8 alkyl, C3-C8 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinoly1;
R3 and R4 are independently chosen from H, -SR8 and ¨NR9R10, or R3 and R4 are joined together to form a 5-7 membered ring that optionally comprises an heteroatom chosen from N, S and 0;
R8 is H, C1-C8 alkyl, -(CH2)õNHBoc, or -(CH2)nNH2 wherein n = 1 to 6;
R9 is H or CI-C8 alkyl;
R10 is H, Cl-C8 alkyl, acetyl, propiolyl, butyryl, isobutyryl, or benzoyl;
wherein R2, R5, R6 and R7, when substituted, are substituted with at least one substituent chosen from -0R9, -F, -Cl, -Br, -I, acetyl, propiolyl, butyryl, isobutyryl, benzoyl, -NO2, CI-Cs alkyl, methoxycarbonyl-, or alkyloxycarbonyl-;
or an enantiomer, diastereoisomer, racemic mixture, pharmaceutically acceptable salt, solvate or prodrug thereof.
[0004] According to another aspect, there is included a composition comprising a pharmaceutically acceptable carrier and at least one compound of the present disclosure.
[0005] According to another aspect, there is included a method for treating cancer or at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma, said method comprising administering to a subject in need thereof an effective amount of at least one compound of the present disclosure.
[0006] According to another aspect, there is included a method for reducing the risks of developing cancer or for reducing the risk of developing at least one cancer in a subject, the cancer being, for example, chosen from breast cancer, uterine = CA 02926928 2016-04-11 cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma, said method comprising administering to the subject an effective amount of at least one compound of the present disclosure.
[0007] According to another aspect, there is included a method for inhibiting cancer cell growth, the method comprising administering to a subject in need thereof an effective amount of at least one compound of the present disclosure. For example, the cancer can be chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma.
[0008] According to another aspect, there is included the use of at least one compound of the present disclosure for treating cancer or for treating at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma
[0009] According to another aspect, there is included the use of at least one compound of the present disclosure for reducing the risks of developing cancer or for reducing the risks of developing at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma
[0010] According to another aspect, there is included the use of at least one compound of the present disclosure in the manufacture of a medicament for treating cancer or for treating at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma
[0011] According to another aspect, there is included the use of at least one compound of the present disclosure in the manufacture of a medicament for reducing the risks of developing cancer or for reducing the risks of developing at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma
[0012] According to another aspect, there is included the use of at least one compound of the present disclosure for inhibiting cancer cell growth. For example, the cancer can be chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma.
[0013] According to another aspect, there is included the use of at least one compound of the present disclosure in the manufacture of a medicament for inhibiting = CA 02926928 2016-04-11 cancer cell growth. For example, the cancer can be chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer, bladder cancer, and melanoma.
BRIEF DESCRIPTION OF FIGURES
[0014]
The following drawings represent in a non-limitative manner examples of specific embodiments in which Figure 1 represents images (a) and graphical analysis (b) showing Western blot analysis to determine the expression level of phosphorylated STAT1 and STAT3 in human macrophages (MO) pretreated for 30 min with vehicle (DMSO) or compounds 1 and 1A, and then washed and recovered immediately (t= 0) or after 30 min of activation with either 50 U/mL IFNy or 25 ng/mL IL6. The ratio ofphosphorylated/no phosphorylated proteins was calculated from densitometric analysis of each sample to evaluate the relative activation of pSTAT1 or pSTAT3. * p < 0.05 and ** p <
0.01 denote significant differences between treatments;
Figure 2 represents images (a) and graphical analysis (b) showing flow cytometry analysis to determine the expression level of MHC-II and CD40 surface antigens in resting and IFNy-activated hMitIs untreated and pretreated with compounds 1 (10 i_tM) and 1A (25 M). * p < 0.05 and ** p < 0.01 denote significant differences between treatments;
Figure 3 represents images (a) and graphical analysis (b) showing scratch wound healing assays to determine the motility of hM(13, monolayers cultured for 3 h with vehicle (DMSO) or compounds 1 (10 ?AM) and 1A (25 04), and then activated for h with vehicle (PBS) or 25 ng/mL IL6. The images of the scratch were acquired at t=
O h and t= 48 h by fluorescence microscopy. Five fields were taken randomly for each different treatment. All observations were performed at 5x magnification. Cell motility was expressed as percent (%) of control of motile cells at t= 48 h relative to motile cells at t= 0 h. * p < 0.05 and ** p < 0.01 denote significant differences between treatments;

Figure 4 is a graphical representation of NO production in the macrophage-like J774A.1 cells following a pro-inflammatory stimulation by IFNI, and TNFa after pretreatment with vehicle (DMSO) and the derivatives 1 and 1A. * p < 0.05 and ** p < 0.01 denote significant differences between treatments;
Figure 5 is a graphical representation of relative cell viability on the murine BCa cell line MB49-I following a pro-inflammatory stimulation by IFNy and TNFa after pretreatment with vehicle (DMSO) and anti-inflammatory derivatives 1 and 1A at different concentrations. * p < 0.05 and ** p < 0.01 denote significant differences between treatments;
Figure 6 is a graphical representation of NO production by the murine BCa cell line MB49-I following a pro-inflammatory stimulation by IFN7 and TNFa after pretreatment with vehicle (DMSO) and derivatives 1 and 1A and various precursor intermediates (3, 4, 6), reagents (2, PABA) and maleic anhydride (MA) as well as the left hand (7) and right hand (9) of the main anti-inflammatory compound 1. *p < 0.05 and ** p < 0.01 denote significant differences between treatments. (See publication no.11 for a complete description of the molecules shown in this figure);
Figure 7 refers to episodic intraperitoneal injections of AL-549 in C57B1/6J
mice (n=4) during 3 weeks have no effect on normal development and viability (A), as well as in organs weight (B) and hematocrit (C);
Figure 8 represents images showing histological analysis of liver sections from C57B1/6J mice (n=4) after episodic intraperitoneal injections of AL-549 during weeks. Treatment does not induce features of liver dysfunction such as hepatocellular injury, inflammation, fibrosis and steatosis (haematoxylin and eosin, 100 x and 400 x magnification);
Figure 9 refers to episodic intraperitoneal injections of AL-549 in C57B1/6J
mice (n=6) affects tumor development of MB49-I cells subcutaneously implanted in mice.
*p < 0.01 compared to control;

Figure 10 refers to episodic intraperitoneal injections of AL-549 in C57B1/6J
mice (n=6) reduce the size of MB49-I tumors subcutaneously implanted in C57B1/6J
mice (A) and the number of lung metastases (B). *p < 0.01 compared to control;
Figure 11 represents (A) Western blot analysis: AL-549 efficiently inhibited TNFa/NFKB and IL6/STAT3 signaling pathways in murine UBC MB49-I cells at lower doses (10 mM). (B) Luciferase assay: AL-549 efficiently inhibited NFkB
activation. MB49-I cells were transfected with a NFKB-responsive luciferase construct encoding the firefly luciferase reporter gene under the control of CMV
promoter and tandem repeats of the NFKB transcriptional response element. * p <
0.01 denote significant difference compared to control;
Figure 12 refers to AL-549 efficiently inhibited (A) Ml-induced human UBC T24 cell invasion (matrigel in Boyden chamber) and 1L6-induced motility (scratch assay) in T24 cells (B). * p < 0.05 denote significant difference compared to control (M1 DMSO);
Figure 13 refers to AL-549 efficiently inhibited MDA-MB-241 breast cancer cell invasion (matrigel in 3D cultures). * p < 0.01 denote significant difference compared to day 0 (DO.);
Figure 14 is a graphical representation of relative cell viability on MB49 and cell lines after pretreatment (3h) with either vehicle (DMSO) or AL-549 at different concentrations. * p < 0.05 denote significant difference compared to control (without AL-549);
Figure 15 is a graphical representation of LPS/IFNy-induced NO production in MB49-I cells after pretreatment with vehicle (DMSO) and the molecules AL-549 (30 iaM) and ML-19 (10, 20, 37.5 and 50 p,M). * p < 0.01 denote significant difference compared to positive control (DMSO + LPS /IFNy);

= CA 02926928 2016-04-11 Figure 16 is a graphical representation of LPS/IFNy-induced NO production in MB49-I cells after pretreatment with vehicle (DMSO) and the molecules AL-549 (37.5 M) and ML-28 (25, 37.5 and 50 *
p < 0.01 denote significant difference compared to positive control (DMSO + LPS /IFNy). ** p < 0.05 denote significant difference compared to AL-549 at 37.5 IAM (+ LPS /IFNy);
Figure 17 refers to episodic intraperitoneal injections of ML-28 in C57BI/6J
mice (n=8) affects tumor development of MB49-I cells subcutaneously implanted in mice.
*p < 0.05 and **p < 0.01 compared to control;
Figure 18 refers to Non-muscle invasive UBC MB49 cells produced low levels of iNOS (A) and NO (B) relative to highly invasive MB49-I cells in response toTNFcc but not IL6. *p < 0.01 compared to control;
Figure 19 Loss of basal and TNFa-induced iNOS expression (A) and NO production (B) by shRNA affects MB49-I tumor development (C) subcutaneously implanted in C57BI/6J mice. *p < 0.01 compared to control; and Figure 20 refers to episodic intraperitoneal injections of AL-549 in C57B1/6J
mice (n=6) reduce the size of non-muscle invasive MB49 tumors subcutaneously implanted in C57B1/6J mice. *p < 0.001 compared to Ctl (ANOVA, Mann Whitney Test).
[0015] In figures 1 to 6: Compounds 1 and 1A correspond to 4 (AL-549) and (AL-361) of the present application.
DETAILED DESCRIPTION OF THE DISCLOSURE
[0016]
The present disclosure concerns the discovery of small aminobenzoic acid derivatives showing anti-inflammatory, anti-metastatic and anticancer properties in vitro and in vivo. It describes the synthetic methodology to make these derivatives from readily available ortho-, meta- and para-benzoic acid and their biological = CA 02926928 2016-04-11 applications for the treatment of a several types of cancers. In addition, this disclosure relates to different pharmaceutical compositions comprising these compounds.
The compounds and the pharmaceutical composition of this disclosure have been shown to possess anticancerous activity on various types of cancers. Furthermore, this disclosure provides novel treatment modalities against cancer. The unique biological properties of these compounds may be advantageously used to provide compounds with anticancer activity against cancers including but not limited to breast, prostate, ovarian and bladder cancers.
[0017] The term a "therapeutically effective amount", "effective amount" or a "sufficient amount" of a compound of the present disclosure is a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, including clinical results, and, as such, an "effective amount" or synonym thereto depends upon the context in which it is being applied.
For example, in the context of treating cancer, for example, it is an amount of the compound sufficient to achieve such treatment of the cancer as compared to the response obtained without administration of the compound. The amount of a given compound of the present disclosure that will correspond to an effective amount will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. Also, as used herein, a "therapeutically effective amount" , "effective amount or a "sufficient amount of a compound of the present disclosure is an amount which inhibits, suppresses or reduces a cancer (e.g., as determined by clinical symptoms or the amount of cancerous cells) in a subject as compared to a control.
[0018] The term "subject" as used herein includes all members of the animal kingdom including human. According to one embodiment, the subject is a human.
[0019] The term "alkyl" as used herein means straight and/or branched chain, saturated alkyl groups containing from one to n carbon atoms and includes (depending on the identity of n) methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like, wherein n is the maximum number of carbon atoms in the group.
[0020] The expression an alkyl component of a naturally occurring amino acid" as used herein refers to the portion of a naturally occurring amino acid that is comprised between the carbon of the carbonyl group of the amino acid and the nitrogen atom of the amino acid.
[0021] The expression "compound(s) of the present disclosure" as used in the present document refers to compounds of formulae I, IA, IB, IC, ID and IE, presented in the present disclosure, isomers thereof, such as stereoisomers (for example, enantiomers, diastereoisomers, including racemic mixtures) or tautomers, or to pharmaceutically acceptable salts, solvates, hydrates and/or prodrugs of these compounds, isomers of these latter compounds, or racemic mixtures of these latter compounds. The expression "compound(s) of the present disclosure" also refers to mixtures of the various compounds or variants mentioned in the present paragraph.
[0022] The term "halogen" as used herein comprises fluoro, chloro, bromo and iodo.
[0023] It is to be clear that the present disclosure includes isomers, racemic mixtures, pharmaceutically acceptable salts, solvates, hydrates and prodrugs of compounds described therein and mixtures comprising two or more of such compounds.
[0024] The compounds of the disclosure may have at least one asymmetric centre. Where the compounds according to the present document possess more than one asymmetric centre, they may exist as diastereomers. It is to be understood that all such isomers and mixtures thereof in any proportion are encompassed within the scope of the present disclosure. It is to be understood that while the stereochemistry of the compounds of the present disclosure may be as provided for in any given compound listed herein, such compounds of the disclosure may also contain certain amounts (for example less than 30%, less than 20%, less than 10%, or less than 5%) of compounds of the present disclosure having alternate stereochemistry.
[0025] The term "suitable", as in for example, "suitable counter anion" or "suitable reaction conditions" means that the selection of the particular group or conditions would depend on the specific synthetic manipulation to be performed and the identity of the molecule but the selection would be well within the skill of a person trained in the art. All process steps described herein are to be conducted under conditions suitable to provide the product shown. A person skilled in the art would understand that all reaction conditions, including, for example, reaction solvent, reaction time, reaction temperature, reaction pressure, reactant ratio and whether or not the reaction should be performed under an anhydrous or inert atmosphere, can be varied to optimize the yield of the desired product and it is within their skill to do so.
[0026] The expression "pharmaceutically acceptable means compatible with the treatment of subjects such as animals or humans.
[0027] The expression "pharmaceutically acceptable salt means an acid addition salt or basic addition salt which is suitable for or compatible with the treatment of subjects such as animals or humans.
[0028] The expression "pharmaceutically acceptable acid addition salt as used herein means any non-toxic organic or inorganic salt of any compound of the present disclosure, or any of its intermediates. Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids that form suitable salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as para-toluene sulfonic and methanesulfonic acids. Either the mono or di-acid salts can be formed, and such salts may exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of the compounds of the present disclosure are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. The selection of the appropriate salt will be known to one skilled in the art. Other non-pharmaceutically acceptable salts, e.g. oxalates, may be used, for example, in the isolation of the compounds of the present disclosure, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt. In embodiments of the present disclosure, the pharmaceutically acceptable acid addition salt is the hydrochloride salt.
[0029] The term "pharmaceutically acceptable basic addition salt" as used herein means any non-toxic organic or inorganic base addition salt of any acid compound of the disclosure, or any of its intermediates. Acidic compounds of the disclosure that may form a basic addition salt include, for example, where R
is CO2H.
Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxide. Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethylamine and picoline or ammonia. The selection of the appropriate salt will be known to a person skilled in the art. Other non-pharmaceutically acceptable basic addition salts, may be used, for example, in the isolation of the compounds of the disclosure, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
[0030] The formation of a desired compound salt is achieved using standard techniques. For example, the neutral compound is treated with an acid or base in a suitable solvent and the formed salt is isolated by filtration, extraction or any other suitable method.
[0031] The term "solvate" as used herein means a compound of the present disclosure, wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered.
Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate". The formation of solvates of the compounds of the present disclosure will vary depending on the compound and the solvate. In general, solvates are formed by dissolving the compound in the appropriate solvent and isolating the solvate by cooling or using an antisolvent. The solvate is typically dried or azeotroped under ambient conditions.
[0032] Compounds of the present disclosure include prodrugs. In general, such prodrugs will be functional derivatives of these compounds which are readily convertible in vivo into the compound from which it is notionally derived.
Prodrugs of the compounds of the present disclosure may be conventional esters formed with available hydroxy, or amino group. For example, an available OH or nitrogen in a compound of the present disclosure may be acylated using an activated acid in the presence of a base, and optionally, in inert solvent (e.g. an acid chloride in pyridine).
Some common esters which have been utilized as prodrugs are phenyl esters, aliphatic = CA 02926928 2016-04-11 (C8-C24) esters, acyloxymethyl esters, carbamates and amino acid esters. In certain instances, the prodrugs of the compounds of the present disclosure are those in which one or more of the hydroxy groups in the compounds is masked as groups which can be converted to hydroxy groups in vivo. Conventional procedures for the selection and preparation of suitable prodrugs are described, for example, in "Design of Prodrugs" ed. H. Bundgaard, Elsevier, 1985.
[0033] Compounds of the present disclosure include radiolabeled forms, for example, compounds labeled by incorporation within the structure 2H, 3H, 14C, 15N, or a radioactive halogen such as 1251. A radiolabeled compound of the compounds of the present disclosure may be prepared using standard methods known in the art.
[0034] As used herein, and as well understood in the art, "treatment" or "treating" is an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" or "treating" can also mean prolonging survival as compared to expected survival if not receiving treatment.
[0035] "Palliating" a disease or disorder, means that the extent and/or undesirable clinical manifestations of a disorder or a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder.
[0036] In understanding the scope of the present disclosure, the term "comprising" and its derivatives, as used herein, are intended to be open ended terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but do not exclude the presence of other unstated features, elements, components, groups, integers and/or steps. The foregoing also applies to words having similar meanings such as the terms, "including", "having" and their derivatives. Finally, terms of degree such as "substantially", "about" and "approximately" as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of = CA 02926928 2016-04-11 degree should be construed as including a deviation of at least 5% of the modified term if this deviation would not negate the meaning of the word it modifies.
[0037] In an embodiment of the present disclosure, there are included compounds of formula (IA) :

R3 \R2 (IA) wherein R2, R3, R4 and Q are as previously defined.
[0038] In another embodiment of the present disclosure, there are included compounds of formula (IB) :

N-Q

(IB) wherein R2, R3, R4 and Q are as previously defined.
[0039] In a further embodiment of the present disclosure, there are included compounds of formula (IC) :

= CA 02926928 2016-04-11 N¨NT5 (1C) wherein R2, R5 and R6 are as previously defined.
[0040] In still a further embodiment of the present disclosure, there are included compounds of formula (ID) :

(R4 N¨N

(ID) wherein R2, R3, R4 and R7 are as previously defined.
[0041] In still a further embodiment of the present disclosure, there are included compounds of formula (IE) :

= CA 02926928 2016-04-11 H
N
111 / ________________________________________________________ 7;---------<
( (1E) wherein R2 and R7 are as previously defined.
[0042] In still a further embodiment of the present disclosure, R2 is H, unsubstituted member chosen from acetyl and propiolyl;
Q is QA;
R5 is H, unsubstituted member chosen from acetyl and propiolyl; and R6 is Boc, H, or an unsubstituted member chosen from acetyl and propiolyl.
[0043] In still a further embodiment of the present disclosure, R2 is H or unsubstituted member chosen from acetyl and propiolyl; and R7 is an unsubstituted member chosen from C1-C8 alkyl, C3-C6 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl.
[0044] For example, in a further embodiment of the present disclosure, the compound of formula I is as previously defined with the proviso that the compound is different from o o o N . 0 NHNH4 ( NHNH2*HC1 0 and N
NHNH2*CF3CO2H

.
[0045] However, accodring to an embodiment the above three excluded compounds are not to be excluded from the scope of the various uses and methods as previously described.

' CA 02926928 2016-04-11
[0046] According to another embodiment, the above three excluded compounds are to be excluded from the scope of the various uses and methods as previously described.
[0047] In still a further embodiment of the present disclosure, there are included compounds of the following formulas:

O 9 o CI
NHNH--`< / 0 (1\I * 0 N * p O CI _____________ 0 __ 0 4a 4b ( = 0 )0LR --/, I N =0 NHN NHNBocIr-R ----O o ).
o R (i31 8 (R = CH3, ML-28) 10 (R = CH3, ML-33B) o ili\I .11 o Boc * *
---- N¨N. NHNHBoc O R-i >0 o OR 12 (ML-19) 11 (R = CH3, ML-31E) H /c1\1 leo Boc-NS HN-NH
0 'Boc 13 (ML-21)
[0048] For example, the compound can be chosen from = CA 02926928 2016-04-11 =

1-11\1 X.0 H3 NHN N N CH3 NHNBoc N¨NBoc 0 0 /t 0 H3C-41, HN-NH N¨NH
0 and 0 H3C--
[0049] Schemes 1 to 4 represent examples of synthetic routes used for the preparation of the compounds of the present disclosure. The reaction conditions of each step are presented directly in the schemes.
[0050] Using para-amino benzoic acid (1) as the starting material derivative 4 was made in a three-step reaction sequence (Scheme 1).911 Para-amino benzoic acid (1 or PABA) was first reacted with maleic anhydride (MA) in dry acetone to give the diacid (2) with 90% yield. Cyclisation to form the maleimide was accomplished with acetic anhydride and sodium acetate to give compound 3 with 89% yield after hydrolysis of the mixed anhydride intermediate with water. Finally, activation of acid 3 with /so-butyl chloroformate in the presence of pyridine was done followed by treatment with tert-butyl carbazate gave the desired anti-inflammatory derivative 4 with 54% yield. Deprotection of 4 with hydrochloric acid in ether gave the hydrochloride salt 5 with 46% yield after recrystallization. Example 1 shows the preparation of compounds 4 and 5. This reaction sequence can be used to produce other derivatives starting from unsubstituted or substituted ortho-, meta- and para-benzoic acid starting materials. Hence, this approach lead to the synthesis of N'-[3-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro-benzoyll-hydrazine carboxylic acid tert-butyl ester (4a) and N'44-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-2-chloro-benzoy1]-hydrazine carboxylic acid tert-butyl ester (4b) shown in examples 2 and 3, respectively.

H2N= 0 OH
a (c1\1=

1 2 (90%) 3 (88%) N O lN
NHNH¨µ( ( NHNH2*HC1 4 (AL-549, 54%) 5 (AL-361, 46%) Reagents and conditions: a) Maleic anhydride (MA), dry acetone, methanol, 22 C, 1 h; b) 1) Ac20, AcONa, 50 C, 2 h; 2) H20, 70 C, 2 h; c) 1) iso-butyl chloroformate, Et3N, CH2C12, 0 C, 1 h and 22 C, 1 h; 2) tert-butyl carbazate, CH2C12, 22 C, 12 h; d) HC1, dioxane, 22 C, 5 h.
Scheme 1
[0051] Further transformations of derivative 4 can lead to novel compounds with anti-inflammatory, anti-metastatic and anticancer activities (Scheme 2).
However, catalytic hydrogenation of 4 led to compound 12 which lost its anti-inflammatory activity (see example 4). Otherwise, compound 4 is transformed into its trifluoroacetate salt 6 upon treatment with trifluoroacetic acid in dichloromethane.
The crude material 6 can be reacted either with acetyl chloride or acetic anhydride (or any relevant anhydride or acid chloride) to give derivative 8. Of note, derivatives 7 and 9 were likely produced but not isolated in example 5. Alternatively, compound 4 can be acylated with a relevant anhydride or acid chloride to yield compound 10 and 11. Interestingly, using acetyl chloride, compound 4 was transformed efficienly into the diacetylated derivative 11 (R = CH3, ML-3113) and, with acetic anhydride the main product of the reaction was the monoacetylated derivative 10 (R = CH3, ML-33B). Derivative 10 can be deprotected to give 7 and derivative 11 can lead to derivative 9. It is possible to produce efficienly compound 7 and 9 via a two-step sequence from 4.

' CA 02926928 2016-04-11 o O o - o R
-----O O b ---k - HN-NH
i N * --0- N il ¨4'7 l N .
---\ NHNHBoc NHNH2*CF3CO2H --1 o O - o - o 4 6 (ML-15) 7 O o +
b 1 N li IN 0 0 (( lik )1.-R
NHNHBoc O. NHNr.R
0 . 'c / 0,..\N
12 (ML-19) % o o 0 \
O o CDSCN 8 (R =
CH3, ML-28) N . O + -1---AN * o =Boc c.
O +
NHNBoc 0 0 R¨ 0 N II
R 0 0 R (from 11 to yield 9) NNH R
O
.' 10 (R = CH3, ML-33B) 11 (R = CH3, ML-31E) 9 0 R
Reagents and conditions: a) I-FA, CH2C12, 22 C, 0.5 h; b) Relevant anhydride or acid chloride, Et0Ac, 22 C, 30 min to 2 h; c) H2, Pd/C, CH3OH, 22 C, 3 h.
Scheme 2
[0052] Scheme 3 presents the methodology leading to 13 the alkylhydrazones or arylhydrazones derivatives following the procedure described by Taha et a/.12 Accordingly, compound 5 can be treated with a relevant aldehyde (alkyl aldehydes (linear or branched), benzaldehyde or substituted benzaldehydes or other arylaldehydes) under acidic conditions at reflux in butanol (or other solvent) to give the desired derivatives of general structure 13.

N a 1\1 le FINH 7 ¨1.-- HN¨N--=\

R = alkyl or aromatic (unsubstituted or substituted) Reagents and conditions: a) RCHO, H+, Butanol, reflux, 1 to 5 h.
Scheme 3
[0053] On Scheme 4, derivative 4 (or any other maleimides described herein) can be reacted with an appropriate diene (butadiene (unsubstituted or substituted), cyclopentadiene, cyclohexadiene cycloheptadiene, furane, thiophene, pyrrole, N-= CA 02926928 2016-04-11 alkylpyrrole) to give the desired cycloadducts (Diels-Alder products) such as 14, 15 and 16. This reaction can be performed by heating the pure reagents (diene and dienophile) either neat or in solution, with or without pressure as it is described in example 10.
H
= N
H o HN¨Q
a NH-Q ¨0.- or or N
NH-Q

4 H X = 0, S, NH or NR

NH-Q

n=1,2or3 Relevant diene = butadiene (unsubstituted or substituted), cyclopentadiene, cyclohexadiene, furane, thiophene, pyrrole, N-alkyl pyrrole.
Reagents and conditions: a) Relevant diene, toluene, 4, 3 h.
Scheme 4
[0054] As it can be appreciated by the skilled artisan, the above synthetic schemes are not intended to be a comprehensive list of all means by which the compounds described and claimed in this application may be synthesized.
Further methods can also potentially be used to prepare the compounds of the present disclosure.
[0055] The compounds of the present disclosure may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
[0056] The compounds of the present disclosure may contain one or more asymmetric carbon atoms and thus may occur as racemates and racemic mixtures, single enantiomer, diastereomeric mixtures and individual diastereoisomers.
All such isomeric forms of these compounds are expressly included in the present disclosure.
Each stereogenic carbon may be of the R or S configuration.
[0057] In the present disclosure, the following abbreviations are used:
Abbreviation Meaning Ac20 Acetic anhydride AcONa Sodium acetate CH2C12 Dichloromethane Boc t-Butyloxycarbonyl Et20 Diethyl ether Et0Ac Ethyl acetate Et3N Triethyl amine Hour HC1 Chlorhydric acid i.p. Intraperitoneal Meta MA Maleic anhydride Me0H Methanol min Minute mmol Millimole MTT 3-(4,5-Dimethylthiazol-2-y1)-2,5-diphenyltetrazolium bromide NMR Nuclear magnetic resonance o Ortho Para Phe Phenyl s.c. Subcutaneously TLC Thin layer chromatography TFA Trifluoroacetic acid UBC Urothelial bladder cancer EXAMPLES
[0058] This section also describes the synthesis of several compounds that are presented in this document. These examples are not to be construed as limiting the scope of the present disclosure in any way.
Materials and Methods - Chemistry
[0059] Anhydrous reactions were performed under an inert atmosphere, the set-up assembled and cooled under dry nitrogen. Unless otherwise noted, starting material, reactant and solvents were obtained commercially and were used as such or purified and dried by standard means.I3 Organic solutions were dried over magnesium sulfate, evaporated on a rotatory evaporator and under reduced pressure. All reactions were monitored by UV fluorescence, or staining with iodine. Commercial TLC
plates were Sigma T 6145 (polyester silica gel 60 A, 0.25mm). Flash column chromatography was performed according to the method of Still and co-workers on Merck grade 60 silica gel, 230-400 mesh." All solvents used in chromatography had been distilled prior to use.
[0060] The infrared spectra were taken on a Nicolet Impact 420 FT-IR. Mass spectral assays were obtained using a MS model 6210, Agilent technology instrument.
The high resolution mass spectra (HRMS) were obtained by TOF (time of flight) using ESI (electrospray ionization) using the positive mode (ESI+).
(Plateforme analytique pour molecules organiques de l'Universite du Quebec à Montreal).
[0061] Nuclear magnetic resonance (NMR) spectra were recorded on a Varian 200 MHz NMR apparatus. Samples were dissolved in deuterochloroform (CDC13), deuteroacetone (acetone-d6) or deuterodimethylsulfoxide (DMSO-d6) for data acquisition using tetramethylsilane or chloroform as internal standard (TMS, 8 0.0 ppm for 1H-NMR and CDC13 8 77.0 ppm for 13C-NMR). Chemical shifts (8) are expressed in parts per million (ppm), the coupling constants (J) are expressed in hertz (Hz). Multiplicities are described by the following abbreviations: s for singlet, d for doublet, dd for doublet of doublets, t for triplet, dt for doublet of triplets, q for quartet, dq for doublet of quartets, m for multiplet, #m for several multiplets, br for a broad signal.
[0062] The following compounds were prepared from a relevant aminobenzoic acid derivative using the procedures summarized in schemes 1, 2, 3 or 4.
Example 1. Preparation of N'44-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoyli-hydrazine carboxylic acid tert-butyl ester (4 or AL-549), its hydrochloric acid salt (5 or AL-361) and its trifluoroacetic acid salt (6 or ML-15) Step A. Synthesis of 4-(3-carboxy-acryloylamino)-benzoic acid (2)
[0063] 4-Aminobenzoic acid (1, 5.34 g, 38.93 mmol) was dissolved in dry acetone (12 mL) to which was added methanol (l mL). Maleic anhydride (1.05 eq.) dissolved in dry acetone was added to the first solution. The reaction mixture was stirred for a period of 2 h allowing sufficient time for the complete precipitation of the diacid 2. The precipitate was filtered and washed twice with acetone (2 x 2 mL) and dried in a desiccator overnight. The crude diacid 2 (9.16 g, 90%) was sufficiently pure to be use without further purification at the next step. IR (v, cm-1): 3500-(CO2H), 1686 cm-1 (C=0); I H NMR (DMSO-d6, 6 ppm): 12.79 (br s, 2 H, 2 x CO21-1), 10.58 (s, 1H, NH), 7.89 and 7.71 (2 x d, J=8.6 Hz, 4H, aromatic), 6.48 and 6.30 (2 x d, J=12.2 Hz, 2H, maleimide); I3C NMR (DMSO-d6, 6 ppm): 167.4, 167.3, 164.1, 143.2, 132.1, 130.9 (2), 130.6, 126.0, 119.2 (2); ESI+ HRMS: (M+H)+
calculated for CiiHi0N05= 236.0553; found = 236.0558.
Step B. Synthesis of 4-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoic acid (3)
[0064] The diacid 2 (2.01 g, 8.54 mmol) was treated with acetic anhydride (4.0 mL, 36.28 mmol) and anhydrous sodium acetate (350 mg, 4.27 mmol) and the mixture heated at 50 C for 2 h. Afterwards, the solution was evaporated to dryness and stirred with water at 70 C for a period of 2 h. The resulting precipitate was filtered and dried in a desiccator overnight to yield 1.65 g (89%) of maleimide 4. The spectral data of this derivative correspond to those reported in the literature.91 IR (v, cm-1): 3475-2600 (CO2H), 1715 (C=0), 1704 (C=0); 1H NMR (acetone-d6, 6 ppm):
8.14 and 7.57 (2 x d, J=8.6 Hz, 4H, aromatic), 7.08 (s, 2H, maleimide); 13C
NMR
(acetone-d6, 6 ppm): 169.3 (2), 166.2, 136.2, 134.7 (2), 130.1 (2), 129.3, 125.8 (2);
ESI+ HRMS: (M+H)+ calculated for CI IH8N04 = 218.0448; found = 218.0447.
Step C. Synthesis of N'44-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoyli-hydrazine carboxylic acid tert-butyl ester (4 or AL-549)
[0065] Derivative 4 was synthesized using a modified procedure reported by Willner et al.'s as it is also described by Lau et al. and Hamelin-Morrissette et al.9-11 A cooled suspension (0 C) of molecule 3 (211 mg, 0.97 mmol) in methylene chloride (4.5 mL) was treated with triethylamine (190 pt, 1.36 mmol) and isobutyl chloroformate (175 IAL, 1.34 mmol). The mixture was stirrred for 1 h at 0 C
and at room temperature (22 C) for about 1 h. Afterwards, tert-butyl carbazate (128 mg, 0.97 mmol) dissolved in methylene chloride (0.8 mL) was added dropwise to the mixture and stirred for an additional 12 h at 22 C. The reaction mixture was diluted with ethyl acetate (55 mL) and methylene chloride (20 mL) and washed twice with saturated NaHCO3 (2 x 50 mL), twice with 0.1 N HCI (2 x 50 mL), twice with saturated NaC1 (2 x 50 mL), and finally with H20 (50 mL). The organic phase was dried (MgSO4) and evaporated to give crude derivative 4. The product was purified by flash chromatography, using a mixture of hexanes / acetone (3/2), to yield 173 mg (54%) of 4. The spectral data of this derivative correspond to those reported in the literature.1 IR (v, cm-I): 3360-3240 (NH), 3087 (C=C), 2988 (CH, aliphatic), (C=0), 1706 (C=0); IH NMR (acetone-d6, 6 ppm): 9.05 (s, 1H, NH), 8.02 and 7.53 (2 x d, J=8.6 Hz, 4H, aromatic), 7.07 (s, 2H, maleimide), 2.84 (br s, 1H, NH), 1.45 (s, 9H, 3 x CH3); 13C NMR (acetone-d6, 6 ppm): 169.3 (2), 166.0, 155.7, 135.1, 134.6 (2), 131.6, 127.9 (2), 125.9 (2), 79.6, 27.5 (3); ESI+ HRMS: (M+Na)+
calculated for = CA 02926928 2016-04-11 C16Hi7N3Na05 = 354.1060; found = 354.1072; (M -2-methylpropene +H)+ calculated for C12H11 N305 = 276.0620; found = 276.0627.
Step D. Synthesis of 4-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoic acid hydrazide hydrochloride (or 4-maleimidbenzoic acid hydrazide hydrochloride) (5 or AL-361)
[0066] The hydrolysis of 4 was performed using a similar procedure reported by Heindel et al. for the cleavage of maleimidoacetic acid (tert-butyloxycarbonyl) hydrazide with hydrochloric acid to form maleimidoacetic acid hydrazide hydrochloride.16 To a solution of 4 (2.41 g, 7.27 mmol) dissolved in dry dioxane (30 mL) was added a solution of hydrochloric acid (60 mL, 1.0 M in diethyl ether, mmol). The mixture was stirred at room temperature for a period of 5 hours.
Afterwards, 150 mL of hexanes were added to complete the precipitation of the hydrochloride salt 5. The crude precipitated was filtered, washed with hexanes and recrystallized twice with a mixture of methanol / isopropyl alcohol / hexanes (8 / 3 /
10) to yield 1.7 g (46%) of the desired material. IR (v, cm-I): 3200-2500 (CO2H), 3269 (NH), 1702 (C=0), 1693 (C=0); 1H NMR (DMSO-d6, 6 ppm): 8.06 and 7.52 (2 x d, J=8.8 Hz, 4H, aromatic), 7.21 (s, 2H, maleimide); 13C NMR (DMSO-d6, 6 ppm):
170.0 (2), 165.6, 135.9, 135.4 (2), 129.6, 129.0 (2), 126.8 (2); ES1+ HRMS:
(M+H)+
calculated for CiiHi0N303 = 232.0717; found = 232.0717 and ESI+ HRMS: (M+H)+
calculated for Ci1HiiCIN303 = 268.0483; found = 268.0483.
Step E. Synthesis of 4-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoic acid hydrazide trifluoroacetic acid salt (6 or ML-15)
[0067] A solution of 4 (106 mg, 0.32 mmol) dissolved in trifluoroacetic acid (0.5 mL) was stirred at 0 C for a period of 30 minutes. Afterwards, the excess trifluoroacetic acid was removed under vacum at 22 C to give compound 6 quantitatively. IR (v, cm-I): 3500-2500 (CO2H), 3277 (NH), 1710 (C=0); IH NMR
(DMSO-d6, 6 ppm): 11.62 (br s, 1H, NHNH3+CF3CO2-), 8.63 (br s, NH3), 8.01 and = CA 02926928 2016-04-11 7.56 (2 x d, J=8.5 Hz, 4H, aromatic), 7.24 (s, 2H, maleimide); I3C NMR (DMSO-d6, 6 ppm): 169.6 (2), 165.4, 135.5, 135.0 (2), 129.4, 128.4 (2), 126.5 (2).
Example 2. Preparation of Nr-13-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro-benzoy1Fhydrazine carboxylic acid tert-butyl ester (4a) Following the procedure of Example 1, steps A ¨ C described above using 3-amino-4-chloro benzoic acid as the starting material instead of 4-amino benzoic acid derivative 4a was prepared efficiently.
Step A. Synthesis of 3-(cis-3-carboxy-acryloylamino)-4-chloro benzoic acid (2a)
[0068] Spectral data for 2a: IR (v, cm-1): 3500-2500 (CO2H), 3310 (NH), 1696 (C=0); I H NMR (DMSO-d6, 6 ppm): 13.08 (s, 2H, 2 x CO2H), 10.13 (s, 1H, NH), 8.37 (s, 1H, aromatic) 7.72 and 7.61 (2 x d, 2H, J = 10.0 Hz, aromatic), 6.59 and 6.34 (2 x d, 2H, J = 12.0 Hz, maleimide); I3C MNR (DMSO-d6, 6 ppm): 167.5, 166.7, 164.1, 135.0, 131.6, 131.2 (2), 130.4 (2), 127.2 (2).
Step B. Synthesis of 3-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro benzoic acid (3a)
[0069] Spectral data for 3a: IR (v, cm-I): 3300-2500 (CO2H), 3490 (amine), 1674 (C=0). 1H NMR (acetone-d6, 6 ppm): 8.1 (m, 2H, aromatic), 7.76 (d, 1H, J
= 8.0 Hz, aromatic), 7.14 (s, 2H, maleimide) I3C NMR (acetone-d6, 6 ppm): 168.7 (2), 165.0, 143.0, 137.84, 134.9 (2), 132.3, 131.6, 130.6, 130.2.
Step C. Synthesis of N'-13-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro-benzoylPhydrazine carboxylic acid tert-butyl ester (4a) = CA 02926928 2016-04-11
[0070] Spectral data for 4a: IR (v, cm-1): 3494 (amine), 3090 (C=C), 2974 (aliphatic), 1717 (C=0). H NMR (acetone-d6, 6 ppm): 9.71 (s, 1H, NH), 8.05 (dd, 1H, J = 8.2 Hz and J = 1.8 Hz, aromatic) 7.95 (d, 1H, J = 1.8 Hz, aromatic), 7.75 (d, 1H, J = 8.6 Hz, aromatic), 7.11 (s, 2H, maleimide), 2.91 (s, 1H, NH), 1.43 (s, 9H, 3 x CH3); I3C NMR (acetone-d6, 6 ppm): 169.5 (2), 166.0, 156.5, 135.0 (2), 134.0 (2), 130.0, 130.7, 129.0 (2), 80.0, 28.1(3).
Example 3. Preparation of N'44-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-2-chloro-benzoylphydrazine carboxylic acid tert-butyl ester (4b) Following the procedure of Example I, steps A ¨ C described above using 4-amino-2-chloro benzoic acid as the starting material instead of 4-amino benzoic acid derivative 4b was prepared efficiently.
Step A. Synthesis of 4-(cis-3-carboxy-acryloylamino)-2-chloro benzoic acid (2b)
[0071] Spectral data for 2b : IR (v, cm-I): 3500-2500 (CO2H), 3262 (NH), 1689 (C=0); I H NMR (DMSO-d6, 6 ppm): 13.8 (s, 2H, 2 x CO2H), 10.7 (s, 1H, NH), 7.85 (m, 2H, aromatic) 7.55 (dd, 1H, J = 10.0 Hz and J = 2.0 Hz, aromatic), 6.45 and 6.31 (2 x d, 2H, J = 12.0 Hz, maleimide); 13C MNR (DMSO-d6, 6 ppm): 168.0, 166.0, 164.0, 143.0, 136.5, 135.0, 132.0, 131.5, 126.0, 121.5, 119.2.
Step B. Synthesis of 3-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro benzoic acid (3b)
[0072] Spectral data for 3b : IR (v, cm-I): 3300-2500 (CO2H), 3470 (amine), 1723 (C=0). I H NMR (acetone-d6, 6 ppm): 7.90 (d, 1H, J = 8.6 Hz, aromatic), 7,67 = CA 02926928 2016-04-11 (d, 1H, J = 2.2 Hz, aromatic), 7.50 (dd, 1H, J = 8.3 Hz and J = 2.0 Hz, aromatic) 7.11 (s, 2H, maleimide); 13C NMR (acetone-d6, 6 ppm): 168.0 (2), 165.0, 138.0, 136.0 (2), 135.0, 132.0, 128.0, 124.0, 118Ø
Step C.
Synthesis of N'-13-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-4-chloro-benzoy1]-hydrazine carboxylic acid tert-butyl ester (4b)
[0073]
Spectral data for 4b : IR (v, cm-1): 3473 (amine), 3090 (C=C), 2984 (aliphatic), 1708 (C=0). I H NMR (acetone-d6, 6 ppm): 9.03 (s, 1H, NH), 7.65(d, 1H, J = 8.0 Hz, aromatic) 7.59 (d, 1H, J = 1.6 Hz, aromatic), 7.45 (dd, 1H, J =
8.2 and J --1.8 Hz aromatic), 7.08 (s, 2H, maleimide), 2.96 (s, 1H, NH), 1.47 (s. 9H, 3 x CH3);
I3C NMR (acetone-d6, 6 ppm): 169.0 (2), 166.0, 156.0, 135.0 (2), 134.6, 133.0, 131.0, 127.9(2), 125.9, 80.0, 27.5 (3).
Example 4. Preparation of N'-14-(2,5-dioxo-pyrrolidin-1-y1)-benzoy1]-hydrazine carboxylic acid tert-butyl ester (12 or ML-19)
[0074]
Maleimide 4 (103 mg, 0.31 mmol) was dissolved in methanol (1 mL) to which was added 5% Pd/C (14 mg). Some hydrogen gas was bubbled during 30 seconds into the mixture. The suspension was stirred vigorously under a hydrogen atmosphere for a period of 3 hours. Of note, a longer period of time is required on a larger scale. Afterwards, the suspension was filtered on silica with a mixture of hexanes / acetone (3/2) as the eluent, to yield 87 mg (84%) of 12. IR (v, cm-1): 3400-3100 (NH), 2981 (CH), 1703 (C=0); 1H NMR (acetone-d6, 6 ppm) : 9.54 (br s, 1H, NH), 8.00 and 7.45 (2 x d, J=8.6 Hz, 4FI, aromatic), 1.45 (s, 9H, 3 x CH3);

(acetone-d6, 6 ppm) 176.2 (2), 165.9, 155.7, 136.1, 132.4, 127.8 (2), 126.7 (2), 79.6, 28.2, 27.5; ESI+ HRMS: (M+H)+ calculated for Ci6H20N305 = 334.1397; found =
334.1391 and ESI+ HRMS: (M+H -C4H9)+ calculated for Cl2F112N305 = 278.0771;
found = 278.0769.

= CA 02926928 2016-04-11 Example 5. Preparation of 4-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoic acid N'-diacetyl-hydrazide (8, R = CH3 or ML-28)
[0075] To a solution of crude 6 (444 mg, 1.28 mmol) dissolved in dichloromethane (10 mL) was added triethylamine (1.07 mL, 780 mg, 7.7 mmol) and acetic anhydride (0.61 mL, 659 mg, 6.4 mmol). The mixtured was stirred at 22 C for about 30 minutes. Afterwards, the organic phase was diluted with ethyl acetate (75 mL) directly into an extraction funnel. The organic phase was washed successively with a 5% sodium bicarbonate aqueous solution (50 mL), with a 10% sodium chloride aoous solution (50 mL) and finnally with water (50 mL). The organic phase was dried with anhydrous magnesium sulfate, filtered and evaporated to the crude material (111 mg). The product was purified by flash column chromatography using a mixture of hexanes / acetone (7/3) to give 75 mg (18%) of the desired material 8, R =
CH3). Of note, different anhydrides or alkaloyl chlorides can be used to produce analogs of this specific derivative. IR (v, cm-1): 3200 (NH), 1702 (C=0), 1662 (C=0); 1H NMR
(acetone-d6, 6 ppm): 10.15 (br s, 1H, NH), 8.09 and 7.61 (2 x d, J = 8.6 Hz, 4H, aromatic), 7.10 (s, 2H, maleimide), 2.41 (s, 6H, 2 x CH3); 13C NMR (acetone-d6, 6 ppm): 171.1 (2), 169.3 (2), 166.0, 135.8, 134.7 (2), 130.8, 128.2 (2), 126.1 (2), 24.2 (2). ESI+ HRMS: (M+H)+ calculated for C15H14N305 = 316.0928; found = 316.0945 and ES1+ HRMS: (M+H -Ac)+ calculated for C13H12N304 =275.08252; found =
275.0856.
Example 6. Preparation of N-acetyl-/V-[4-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoyll-hydrazinecarboxylic acid tert-butyl ester (10, R = CH3 or ML-33B)
[0076] To a solution of 4 (100 mg, 0.30 mmol) dissolved in dichloromethane (3 mL) was added triethylamine (252 tL, 183 mg, 1.81 mmol) and acetic anhydride (71 1AL, 76.7 mg, 0.75mmol). The mixtured was stirred at 22 C for about 30 minutes.
Afterwards, the organic phase was diluted with ethyl acetate (25 mL) directly into an extraction funnel. The organic phase was washed successively with a 5% sodium = CA 02926928 2016-04-11 bicarbonate aqueous solution (10 mL), with a 10% sodium chloride aoous solution (10 mL) and finnally with water (20 mL). The organic phase was dried with anhydrous magnesium sulfate, filtered and evaporated to the crude material (123 mg). The product was purified by flash column chromatography using a mixture of hexanes /
acetone (4/1) to give 22 mg (20%) of the desired material 10, R = CH3). It is noteworthy, that a longer reaction time (2 hours) lead to higher yield of the desired material. However, some diacetylated product (11) is also present as a side product.
IR (v, cm-I): 3286 (NH), 1753 (C=0), 1711 (C=0), 1652 (C=0); 1H NMR (CDC13, 6 ppm): 8.28 (br s, 1H, NH), 7.90 and 7.50 (2 x d, J = 8.6 Hz, 4H, aromatic), 6.89 (s, 2H, maleimide), 2.61 (s, 31-1, CH3), 1.51 (s, 9H, 3 x CH3); I3C NMR (CDC13, 6 ppm):
170.5, 168.9 (2), 165.2, 151.0, 135.0, 134.4 (2), 130.9, 128.3 (2), 125.7 (2), 84.8, 27.8 (3), 25.5; ESI+ HRMS: (M+Na)+ calculated for Ci8H19N3Na06 = 396.1166; found =
396.1165 and ESI+ HRMS: (M+H -tert-Boc)+ calculated for Ci3Hi2N304 = 274.0822;

found = 274.0824.
Example 7. Preparation of N,N'-diacetyl-N'-14-(2,5-dioxo-2,5-dihydro-pyrrol-1-y1)-benzoyll-hydrazinecarboxylic acid tert-butyl ester (11, R =
CH3 or ML-31B)
[0077] To a solution of 4 (100 mg, 0.30 mmol) dissolved in dichloromethane (3 mL) was added triethylamine (252 ttL, 183 mg, 1.81 mmol) and acetyl chloride (54 iAL, 60 mg, 0.75 mmol). The mixtured was stirred at 22 C for about 2 hours.
Afterwards, the organic phase was diluted with ethyl acetate (25 mL) directly into an extraction funnel. The organic phase was washed successively with a 5% sodium bicarbonate aqueous solution (10 mL), with a 10% sodium chloride aoous solution (10 mL) and finally with water (20 mL). The organic phase was dried with anhydrous magnesium sulfate, filtered and evaporated to the crude material (118 mg). The product was purified by flash column chromatography using a mixture of hexanes /
acetone (4/1) to give 100 mg (80%) of the desired material 11, R = CH3). IR
(v, cm-I):
3193 (NH), 1701 (C=0), 1660 (C=0); I H NMR (CDC13, 6 ppm): 7.68 and 7.50 (2 x d, J = 8.6 Hz, 4H, aromatic), 6.89 (s, 2H, maleimide), 2.49 and 2.48 (2 x s, 6H, 2 x CH3), 1.50 (s, 9H, 3 x CH3); 13C NMR (CDC13, 6 ppm): 170.2, 169.9 (2), 168.8 (2), 150.2, = CA 02926928 2016-04-11 134.6, 134.4 (2), 132.9, 128.2 (2), 125.0 (2), 85.9, 27.8 (3), 25.4, 24.6;
ESI+ HRMS:
(M+Na)+ calculated for C201-1211\13Na07 = 438.1272; found = 438.1281 and ESI+
HRMS: (M+H -Ac and -tert-Boc)+ calculated for Ci3H12N304 = 274.0822; found =
274.0833.
Example 8. Preparation of N'-{4-13-(2-tert-butoxycarbonylamino-ethylsulfany1)-2,5-dioxo-pyrrolidin-1-yli-benzoy1}-hydrazinecarboxylic acid ter(-butyl ester (13 or ML-21)
[0078] To a solution of 4 (115 mg, 0.34 mmol) dissolved in methanol (3 mL) was added 2-(Boc-amino) ethanethiol (70 pt, 73 mg, 0.41 mmol). The mixture was stirred at 22 'C for 2 hours and at 50 C for 1 hour. The organic phase was diluted with ethyl acetate (30 mL) directly into an extraction funnel and washed successively with a 5% sodium bicarbonate aqueous solution (2 x 10 mL) and with water (2 x 20 mL).
The organic phase was dried with anhydrous magnesium sulfate, filtered and evaporated to the crude material (191 mg). The product was purified by flash column chromatography using a mixture of hexanes / acetone (3/2) to give 107 mg (61%) of the desired material 15. IR (v, cm-1): 3300 (NH), 1707 (C=0), 1680 (C=0); 1H
NMR
(CDCI3, 6 ppm): 8.80 (br s, 1H, NH), 7.86 and 7.34 (2 x d, J = 8.2 Hz, 4H, aromatic), 6.90 (br s, 1H, NH), 5.07 (br s, 1H, NH), 3.98 (1H, m, -CHS-) 2.6-3.6 (several m, 6H, 3 x -CH2-), 1.50 and 1.45 (2 x s, 18H, 2 x 3 x CH3); 13C NMR (CDC13, 6 ppm):
175.4, 173.2, 165.8, 155.9 (2), 134.8, 131.7, 128.2 (2), 126.4 (2), 82.1,
79.7, 39.4, 38.9, 36.1, 32.7, 28.4 (3), 28.2 (3); ESI+ HRMS: (M+Na)+ calculated for C23H32N4Na07S = 531.1884; found = 531.1881.
Example 9. General procedure for the preparation of derivatives of general structure 13 (see scheme 3) [0079]
Following the procedure described by Taha et al.12 compound 5 can be treated with a relevant aldehyde (alkyl aldehydes (linear or branched), benzaldehyde = CA 02926928 2016-04-11 or substituted benzaldehydes) under acidic conditions at reflux in butanol (or other solvent) to give the desired alkylhydrazones or benzoylhydrazones derivatives.
Example 10. General procedure for the preparation of derivatives 14, 15 or 16 (see scheme 4)
[0080] Derivative 4 (or any other maleimides described herein) can be reacted with an appropriate diene (butadiene (unsubstituted or substituted), cyclopentadiene, cyclohexadiene cycloheptadiene, furane, thiophene, pyrrole, N-alkylpyrrole) to give the desired cycloadducts (DieIs-Alder products) such as for example 14, 15 and 16.
This reaction can be performed by heating the pure reagents (diene and dienophile) either neat or in solution, with or without pressure.
Materials and Methods - Biology In vitro studies (for derivatives 4 (A1-549), 5 (AL-361), 12 (ML-19), 8 (ML-28), etc.)
[0081] Cell culture ¨ general: Biological assays were performed using the human monocytic cell line THP1, the murine macrophage-like cell line J774A.1, and the murine BCa cell line MB49-I. The cells were maintained in RPMI medium supplemented with 10% heat-inactivated fetal bovine serum (FBS) and containing mM sodium pyruvate, 10 mM 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid (HEPES) and 50 lAg/mL gentamycin (referred as 10% FBS RPMI-1640). The cells were maintained at 37 C in a moisture-saturated atmosphere containing 5% CO2.
THP1 cells and J774A.1 cells are the most widely used cell lines to investigate the function and differentiation of monocytes and macrophages (MO) in response to various inflammatory mediators.I7'18 Undifferentiated THP1 cells resemble primary monocytes/M0 isolated from healthy donors or donors with inflammatory diseases, such as diabetes mellitus and atherosclerosis.19 After treatment with phorbol esters, THP1 cells differentiate into MOike cells which mimic native monocyte-derived Ms in several respects.2 As we previously described, green fluorescent protein-expressing (GFP)-THP1 cells were cultured for 18 h in 50 nM phorbol 12-myristate 13-acetate to induce monocyte-to-M43. differentiation.21-23 The J774A.1 cell line was kindly provided by Dr Tatiana Scorza (Universite du Quebec à Montreal, Canada).
This cell line is a macrophage-like cell model which produce large amount of NO in response to IFNy, TNFa, bacterial infection and bacterial products, such as lipopolysaccharide (LPS),I7 The cell line MB49-I is a highly invasive and tumorigenic BCa cell model that was developed by successive in vivo passages of MB49 primary tumors.24
[0082] Cell signaling studies: THP1-derived hM4>s (750x103 cells/mL) were pretreated for 30 min with vehicle (DMSO) or compounds 1 and 1A (both at 10 )1M
and 50 WVI), and then washed and recovered immediately (t= 0 min) or after 30 min of activation with 50 U/mL IFNy and 25 ng/mL IL6. Cell lysates were prepared and analyzed by immunoblotting as described.21-23'25 Briefly, protein samples were resolved by SDS-PAGE under reducing conditions and transferred onto a PVDF
membrane. Blots were first probed with rabbit polyclonal antibodies against phospho-STAT1 (pSTAT1) and pSTAT3 (both at 1:2000) overnight at 4 C. Blots were then incubated with HRP-conjugated goat anti-rabbit IgG Ab (1:3000) for 1 h at room temperature. The same blots were stripped and then probed with anti-STAT1 and anti-STAT3 Abs (both at 1:1000). In both cases, probed molecules were visualized using an enhancement chemiluminescence detection kit (Thermo Fisher Scientific).
[0083] Surface antigen expression analysis: To study membrane receptor expression, THP1-derived hni)s (750x104 cells/mL) were pretreated for 3 h with DMSO or compounds 1 (10 ilM) and 1A (25 [tM), and then left untreated (control) or treated for 48 h with 50 U/mL IFNy. The expression level of MHC-II and CD40 was evaluated by flow cytometry as described.21-23
[0084] Motility assays: The in vitro scratch wound healing assay was performed to study the effects of compounds 1 (10 1AM) and 1A (25 1AM) in 1L6-induced hM,11, cell migration, as described.26 Briefly, THP1-derived hM(I)s (750x103 cells/mL) were seeded into 24-well tissue culture plate to reach ¨70-80%
confluence as a monolayer. The cell monolayers were scraped in a straight line in one direction to create a "scratch" with a p200 pipet tip. To obtain the same field during the image acquisition, another straight line was scratched perpendicular to the first would line to create a cross in each well. Cell debris were removed and the edges of the scratch were smoothed by washing the cells once with 1 mL of Hank's buffer. Cell monolayers were pretreated for 3 h with vehicle (DMSO) or compounds 1 (10 1AM) and 1A (25 M), and then left untreated (control) or treated for 48 h with 25 ng/mL
IL6. Using the cross as reference points the plate was placed under an inverted fluorescence microscope, and the images of the scratch were acquired at t= 0 h and t=
48 h. The number of motile cells was determined using Java-based image processing program ImageJ (National Institutes of Health) and relative cell motility was expressed as percent (%) of control of motile cells at t= 48 h relative to motile cells within the initial wound (at t= 0 h).
[0085] Evaluation of NO production by the Griess reagent method: The MB49-I cells and the J774A.1 cells (25x103 cells/well) were grown and pretreated, as indicated, with various anti-inflammatory derivatives, precursors and mono-functional derivatives for a period of 3 h. Afterwards the cells were washed twice with 10% FBS
RPMI-1640 and then activated to produce NO for a period of 24 h with cytokines INFy and TNFa. NO production was measured using the Griess reagent method as previously described.27 This method involves the detection of nitrite ions (NO2-) formed by the spontaneous oxidation of NO under physiological conditions.
According the manufacture procedure (Life Technologies; # G-7921), equal volumes of sulfanilic acid and N-(1-naphthyl)ethylenediamine are mixed together to form the Griess reagent. In the presence of NO2-, sulfanilic acid is converted to a diazonium salt, which in turn is coupled to N-(1-naphthyl)ethylenediamine to produce a pink coloration that is measured with a spectrophotometer (Biotek, synergy HT) at 548 nm.
[0086] Evaluation of cell proliferation by the MU assay: To evaluate the anti-proliferative activity, cell viability/proliferation MTT assays were performed as previously described.21-23=25'28 Briefly, MB49-I cells (5x103 cells/well) were plated in 96-well plates in 100 viL 10% FBS RPMI-1640 and cultured for 24 h at 37 C and 5%
CO2. Cells were pretreated for a period of 3 h with vehicle (DMSO) or derivatives 1 and 1A at 0, 10, 25, 37.5, and 50 1AM, and then incubated for 24 h in the absence or the presence of INFy and TNFa. At the end of the culture period, 10 tL of 5 mg/mL
methylthiazolyldiphenyl-tetrazolium bromide (MTT) solution was added to each well.
After a 3-h incubation period with MTT reagent, 100 1., of MTT solubilization buffer (10% SDS in 10 mM HC1) was added and plates were placed overnight in the cell incubator before absorbance measure. The optical density was read at 580 nm using the Microplate Reader Manager (from Bio-Rad Laboratories).
[0087] Statistical analyses: For all biological assays, data were presented as mean SD from three independent experiments. Data were analyzed by one-way ANOVA followed by Bonferonni post-test using Prism software, version 3.03 (GraphPad, San Diego, CA). p values of < 0.05 were considered to indicate statistical significance.
In vivo studies (for derivatives 4 (A1-549), 8 (ML-28))
[0088] Urothelial bladder cancer (UBC) is the fifth most common malignancy of all cancers in North America. Although most of detectable tumors are initially non-muscle-invasive and are generally curable by means of chirurgical resection, 27-30%
of them exhibit a lethal phenotype characterized by high histological grade and muscle invasion. This cancer was selected to test derivatives 4 (A1-549), 8 (ML-28).
[0089] Male C57BL/6J mice (6-8 weeks old), each weighing 15-18 g, were used for the experiments (supplied by Charles River). The mice were housed with free access to food and water on a 12:12 h light:dark cycle with the room temperature maintained at 21 C. MB49 cells, MB49-I cells, and iNOS-deficient MB49-I cells (5 x 104 in 100 vtL PBS) were injected subcutaneously (s.c.) into the right flank of the mice. Growth rates of the s.c. tumors were monitored. The size of tumors was determined every 3 days for 24-28 days using a digital caliper and by measuring luciferin luminescence at days 15 and 25 using the IVIS imaging system. A
blinded = CA 02926928 2016-04-11 observer measured tumor length and width. The volume of the tumor was calculated from the formula: Length x width2 x 0.52, where length and width were tumor diameters measured with calipers in mutually perpendicular directions. At a tumor size of approximately 10 mm3 the mice were divided into different groups. A
control group received PBS as treatment. As indicated, other groups were treated at different doses (90, 150 or 300 uM) with an intraperitoneal (i.p.) injection of AL-549 or ML-28 every 3-4 days for 18-20 days.24'27 The results from the in vitro and in vivo experiments are presented in figures 1-19.
REFERENCES
1. LaFond, R. In Cancer: The outlaw cell, 3Ird Ed., ACS publication, Washington, DC, 384 pages (2012) 2. Alderton GK. The tumor microenvironment drives metastasis, Nat. Rev.
Cancer 16, 199 (2016) 3. Steeg PS. Targeting metastasis, Nat. Rev. Cancer 16, 201-218 (2016) 4. Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, Richter C, Zips D, Bortfeld T. Radiation oncology in the era of precision medicine, Nat. Rev. Cancer 16, 234-249 (2016) 5. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment, Nat. Rev. Cancer 12, 237-251 (2012) 6. Scott AM, Wolchok JD, Old Li. Antibody therapy of cancer. Nat. Rev.
Cancer 12, 278-287 (2012) 7. Phillips AP, Mentha JW. US Patent 3,046,301 (October 29, 1959); Deglin JH, Vallerand AH. In Guide des medicaments, ERPI, pp. 424-427 (1995) 8. Remers, WA. Antineoplastic agents, In Wilson and Gisvold's Textbook of organic Medicinal and pharmaceutical chemistry, 9th Ed., Delgado JN, Remers, WA. J.B. Eds, Lippincott, New York, Chapter 8, 321-322 (1989) 9. Lau A, Berube G, Ford CHJ. Conjugation of doxorubicin to monoclonal anti-carcinoembryonic antigen antibody via novel thiol-directed cross-linking reagents, Bioorg. Med. Chem., 3, 1299-1304 (1995) 10. Lau A, Berube G, Ford CHJ, Gallant M. Novel doxorubicin-monoclonal anti-carcinoembryonic antigen antibody immunoconjugate activity in vitro, Bioorg.
Med. Chem., 3, 1305-1312 (1995) 11. Hamelin-Morrissette J, Cloutier S, Girouard J, Belgorosky D, Eijan A-M, Legault J, Reyes-Moreno C, and Berube G. Identification of an anti-inflammatory derivative with anti-cancer potential: The impact of each of its structural components on inflammatory responses in macrophages and bladder cancer cells, Eur. J. Med. Chem., 96, 259-269 (2015) 12. Taha M, Ismail NH Lalani S, Fatmi MQ, Atia-tul-Wahab, Siddiqui S, Khan KM, Imran S, Choudhary MI. Synthesis of novel inhibitors of a-glucosidase based on the benzothiazole skeleton containing benzohydrazide moiety and their molecular docking studies, Eur. J. Med. Chem., 92, 387-400 (2015) 13. Perrin DD, Armarego CF. In Purification of Laboratory Chemicals, 3rd Ed., Pergamon Press, Oxford, New York (1988) 14. Still WC, Kahn M, Mitra A. Rapid chromatographic technique for preparative separations with moderate resolution, i Org. Chem., 43, 2923-2925 (1978) 15. Willner D, Trail PA, Hofstead SJ, King HD, Lasch SJ, Braslawsky GR, Greenfield RS, Kaneko T, Firestone RA. (6-Maleimidocaproyl)hydrazone of doxorubicin. A new derivative for the preparation of immunoconjugates of doxorubicin, Bioconjug. Chem., 4, 521-527 (1993) 16. Heindel ND, Zhao HR, Egolf RA, Chang CH, Schray KJ, Emrich JG, McLaughlin JP, Woo DV, A novel heterobifunctional linker for formyl to thiol coupling, Bioconjug. Chem., 2, 427-430 (1991) 17. Lemaire S, Mingeot-Leclercq MP, Tulkens PM, Van Bambeke F. Study of macrophage functions in murine J774 cells and human activated THP-1 cells exposed to oritavancin, a lipoglycopeptide with high cellular accumulation, Antimicrobial agents and chemotherapy, 58 2059-2066 (2014) 18. Auwerx J, The human leukemia cell line, THP-1: A multifacetted model for the study of monocyte-macrophage differentiation, Experientia, 47, 22-31 (1991) 19. Qin Z, The use of THP-1 cells as a model for mimicking the function and regulation of monocytes and macrophages in the vasculature, Atherosclerosis, 221,2-11 (2012) 20. Daigneault M, Preston JA, Marriott HM, Whyte MK, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages, PloS one, 5, 0008668 (2010) = CA 02926928 2016-04-11 21. Dufresne M, Dumas G, Asselin E, Carrier C, Pouliot M, Reyes-Moreno C, Pro-inflammatory type-1 and anti-inflammatory type-2 macrophages differentially modulate cell survival and invasion of human bladder carcinoma T24 cells, Mol. Immunol., 48 1556-1567 (2011) 22. Dumas G, Dufresne M, Asselin E, Girouard J, Carrier C, Reyes-Moreno C.
CD40 pathway activation reveals dual function for macrophages in human endometrial cancer cell survival and invasion, Cancer Immunol. Immunother., 62, 273-283,(2013) 23. Dallagi A, Girouard 1, Hamelin-Morrissette J, Dadzie R, Laurent L, Vaillancourt C, Lafond J, Carrier C, Reyes-Moreno C. The activating effect of IFN-gamma on monocytes/macrophages is regulated by the LIF-trophoblast-IL-10 axis via Statl inhibition and Stat3 activation, Cell. Mol. Immunol., 12, 326-341 (2015).
24. Fabris VT, Lodillinsky C, Pampena MB, Belgorosky D, Lanari C, Eijan AM.
Cytogenetic characterization of the murine bladder cancer model MB49 and the derived invasive line MB49-I, Cancer Genet., 205, 168-176 (2012).
25. Leduc, Bourassa V, Asselin E, Leclerc P, Lafond, Reyes-Moreno C. Leukemia Inhibitory Factor Regulates Differentiation of Trophoblast-Like BeWo Cells Through the Activation of JAK/STAT and MAPK3/1 MAP Kinase-Signaling Pathways, Biol. Reprod., 86, 54, 1-10 (2012) 26. Menon MB, Ronkina N, Schwermann J, Kotlyarov A, Gaestel M.
Fluorescence-based quantitative scratch wound healing assay demonstrating the role of MAPKAPK-2/3 in fibroblast migration, Cell. Motil. Cytoskeleton, 66, 1041-1047(2009) 27. Belgorosky D, Langle Y, Prack Mc Cormick B, Colombo L, Sandes E, Eijan A.M. Inhibition of nitric oxide is a good therapeutic target for bladder tumors that express iNOS, Nitric Oxide, 36, 11-18 (2014) 28. Carmichael J, Degraff WG, Gazdar AF, Minna JD, Mitchell JD. Evaluation of a tetrazolium-based semiautmated colorimetric assay : Assessment of radiosensitivity. Cancer Res., 47, 943-946 (1987)
[0090]
The present disclosure has been described with regard to specific examples. The description was intended to help the understanding of the present disclosure, rather than to limit its scope. It will be apparent to one skilled in the art that various modifications may be made to the present disclosure without departing from the scope of the present disclosure as described herein, and such modifications are intended to be covered by the present document.

Claims (30)

CLAIMS:
1. A compound of formula (I):
wherein R1 is H, alkyl or halogen;
R2 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
Q is QA or QB;
a single bond or a double bond;
R5 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;

R6 is H, Boc, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R7 is a substitued or unsubstituted member chosen from C1-C8 alkyl, C3-C8 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl;
R3 and R4 are independently chosen from H, -SR8 and ¨NR9R10, or R3 and R4 are joined together to form a 5-7 membered ring that optionally comprises an heteroatom chosen from N, S and O;
R8 is H, C1-C8 alkyl, -(CH2)nNHBoc, or -(CH2)nNH2 wherein n = 1 to 6;
R9 is H or C1-C8 alkyl;
R10 is H, C1-C8 alkyl, acetyl, propiolyl, butyryl, isobutyryl, or benzoyl;
wherein R2, R5, R6 and R7, when substituted, are substituted with at least one substituent chosen from -OR9, -F, -Cl, -Br, -I, acetyl, propiolyl, butyryl, isobutyryl, benzoyl, -NO2, C1-C8 alkyl, methoxycarbonyl-, or alkyloxycarbonyl-;
or an enantiomer, diastereoisomer, racemic mixture, pharmaceutically acceptable salt, solvate or prodrug thereof, with the proviso that the compound is different from
2. The compound of claim 1, wherein said compound is a compound of formula (IA):
wherein R2, R3, R4 and Q are as defined in claim 1.
3. The compound of claim 2, wherein R2 is H, unsubstituted member chosen from acetyl and propiolyl;
Q is QA;
R5 is H, unsubstituted member chosen from acetyl and propiolyl; and R6 is Boc, H, or an unsubstituted member chosen from acetyl and propiolyl.
4. The compound of claim 1, wherein said compound is a compound of formula (IB):

wherein R2, R3, R4 and Q are as defined in claim 1
5. The compound of claim 4, wherein R2 is H, unsubstituted member chosen from acetyl and propiolyl;
Q is QA;
R5 is H, unsubstituted member chosen from acetyl and propiolyl; and R6 is Boc, H, or an unsubstituted member chosen from acetyl and propiolyl.
6. The compound of claim 1, wherein said compound is a compound of formula (IC):
wherein R2, R5 and R6 are as defined in claim 1.
7. The compound of claim 6, wherein R2 is H, unsubstituted member chosen from acetyl and propiolyl;
Q is QA;
R5 is H, unsubstituted member chosen from acetyl and propiolyl; and R6 is Boc, H, or an unsubstituted member chosen from acetyl and propiolyl.
8. The compound of claim 1, wherein said compound is a compound of formula (ID):
wherein R2, R3, R4 and R7 are as defined in claim 1.
9. The compound of claim 8, wherein R2 is H or unsubstituted member chosen from acetyl and propiolyl; and R7 is an unsubstituted member chosen from C1-C8 alkyl, C3-C6 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl.
10. The compound of claim I, wherein said compound is a compound of formula (1E):

wherein R2 and R7 are as defined in claim 1.
11. The compound of claim 10, wherein R2 is H or an unsubstituted member chosen from acetyl and propiolyl; and R7 is an unsubstituted member chosen from C1-C8 alkyl, C3-C6 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl.
12. The compound of claim 1, wherein said compound is chosen from :

13. The compound of claim 1, wherein said compound is chosen from :
<MG>
14. The compound of any one of claims 1 to 13, for use in medicine.
15. The compound of any one of claims 1 to 13, for use in reducing risks of developing at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer and bladder cancer.
16. The compound of any one of claims 1 to 13, for use in the treatment of at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer prostate cancer and bladder cancer.
17. The compound of any one of claims 1 to 13, for use as an anticancer agent.
18. The compound of any one of claims 1 to 13, for use as an anti-inflammatory agent.
19. The compound of any one of claims 1 to 13, for use as an anti-metastatic agent.
20. Use of at least one compound as defined in any one of claims 1 to 13, for reducing the risks of developing at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer and bladder cancer.
21. Use of at least one compound as defined in any one of claims 1 to 13 in the manufacture of a medicament for treating at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer prostate cancer and bladder cancer.
22. Use of at least one compound as defined in any one of claims 1 to 13 in the manufacture of a medicament for reducing the risks of developing of at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer prostate cancer and bladder cancer.
23. Use of at least one compound as defined in any one of claims 1 to 13 for use as an anticancer agent.
24. Use of at least one compound as defined in any one of claims 1 to 13 for use as an anti-inflammatory agent.
25. Use of at least one compound as defined in any one of claims I to 13 for use as an anti-metastatic agent.
26. A method for treating cancer or at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer and bladder cancer, said method comprising administering to a subject in need thereof an effective amount of at least one compound as defined in any one of claims 1 to 13.
27. A method for reducing the risks of developing cancer or for reducing the risk of developing at least one cancer in a subject, the cancer being, for example, chosen from melanoma, breast cancer, uterine cancer, ovarian cancer, prostate cancer and bladder cancer, said method comprising administering to the subject an effective amount of at least one compound as defined in any one of claims 1 to 13.
28. A method for inhibiting cancer cell growth, the method comprising administering to a subject in need thereof an effective amount of at least one compound as defined in any one of claims 1 to 13.
29. A compound of formula (I):
wherein R1 is H, alkyl or halogen;
R2 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
Q is Q A or Q B;
a single bond or a double bond;
R5 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R6 is H, Boc, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R7 is a substitued or unsubstituted member chosen from C1-C8 alkyl, C3-C8 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl;
R3 and R4 are independently chosen from H, -SR8 and ¨NR9R10, or R3 and R4 are joined together to form a 5-7 membered ring that optionally comprises an heteroatom chosen from N, S and O;
R8 is H, C1-C8 alkyl , -(CH2)n NHBoc, or -(CH2)n NH2 wherein n = 1 to 6;
R9 is H or C1-C8 alkyl;
R10 is H, C1-C8 alkyl, acetyl, propiolyl, butyryl, isobutyryl, or benzoyl;

wherein R2, R5, R6 and R7, when substituted, are substituted with at least one substituent chosen from -OR9, -F, -Cl, -Br, -I, acetyl, propiolyl, butyryl, isobutyryl, benzoyl, -NO2, C1-C8 alkyl, methoxycarbonyl-, or alkyloxycarbonyl-;
or an enantiomer, diastereoisomer, racemic mixture, pharmaceutically acceptable salt, solvate or prodrug thereof, for use in the treatment of at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer prostate cancer and bladder cancer.
30. A method for treating cancer or at least one cancer chosen from breast cancer, uterine cancer, ovarian cancer, prostate cancer and bladder cancer, said method comprising administering to a subject in need thereof an effective amount of at least one compound of formula (I):
wherein R1 is H, alkyl or halogen;
R2 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
Q is Q A or Q B;
a single bond or a double bond;
R5 is H, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R6 is H, Boc, or a substitued or unsubstituted member chosen from acetyl, propiolyl, butyryl, isobutyryl and benzoyl;
R7 is a substitued or unsubstituted member chosen from C1-C8 alkyl, C3-C8 cycloalkyl, phenyl, furanyl, thiophenyl, pyridinyl, naphthyl, quinolyl and isoquinolyl;
R3 and R4 are independently chosen from H, -SR8 and ¨NR9R10, or R3 and R4 are joined together to form a 5-7 membered ring that optionally comprises an heteroatom chosen from N, S and O;
R8 is H, C1-C8 alkyl , -(CH2)n NHBoc, or -(CH2)n NH2 wherein n = 1 to 6;
R9 is H or C1-C8 alkyl;

R10 is H, C1 -C8 alkyl, acetyl, propiolyl, butyryl, isobutyryl, or benzoyl;
wherein R2, R5, R6 and R7, when substituted, are substituted with at least one substituent chosen from -OR9, -F, -Cl, -Br, -I, acetyl, propiolyl, butyryl, isobutyryl, benzoyl, -NO2, C1-C8 alkyl, methoxycarbonyl-, or alkyloxycarbonyl-;
or an enantiomer, diastereoisomer, racemic mixture, pharmaceutically acceptable salt, solvate or prodrug thereof.
CA2926928A 2016-04-11 2016-04-11 Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents Abandoned CA2926928A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2926928A CA2926928A1 (en) 2016-04-11 2016-04-11 Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA2926928A CA2926928A1 (en) 2016-04-11 2016-04-11 Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents

Publications (1)

Publication Number Publication Date
CA2926928A1 true CA2926928A1 (en) 2017-10-11

Family

ID=60042915

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2926928A Abandoned CA2926928A1 (en) 2016-04-11 2016-04-11 Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents

Country Status (1)

Country Link
CA (1) CA2926928A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109081799A (en) * 2018-08-27 2018-12-25 连云港锐巴化工有限公司 A kind of rubber special type vulcanizing agent 4- carboxyl phenyl maleimide preparation method
WO2020077459A1 (en) * 2018-10-18 2020-04-23 3R Valo, S.E.C. Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
CN114432301A (en) * 2020-11-03 2022-05-06 北京大学 Covalent inhibitor derivative and application thereof in treating virus infection

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109081799A (en) * 2018-08-27 2018-12-25 连云港锐巴化工有限公司 A kind of rubber special type vulcanizing agent 4- carboxyl phenyl maleimide preparation method
WO2020077459A1 (en) * 2018-10-18 2020-04-23 3R Valo, S.E.C. Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
CN114432301A (en) * 2020-11-03 2022-05-06 北京大学 Covalent inhibitor derivative and application thereof in treating virus infection
CN114432301B (en) * 2020-11-03 2024-05-31 北京大学 Covalent inhibitor derivative and application thereof in treatment of viral infection

Similar Documents

Publication Publication Date Title
US20210002222A1 (en) Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
ES2640003T3 (en) Therapeutic products derived from diglycidyl ether and methods for use
CN105899515B (en) Derivatives of uncialamycin, method of synthesis and use thereof as antitumor agents
US11407762B2 (en) Macrocyclic compounds and uses thereof
KR102343865B1 (en) Compound inhibiting YAP-TEAD interaction and pharmaceutical composition for treating or preventing cancer comprising the same as an active ingredient
US20190328743A1 (en) Ezh2 inhibitors for treating cancer
JP2021523942A (en) Urolicin A and its derivatives for use in therapy
JP2019527701A (en) FGFR4 inhibitor and method for producing and using the same
CA2926928A1 (en) Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
WO2022135546A1 (en) Spirocyclic compound as kras-g12c inhibitor
WO2023125928A1 (en) Menin inhibitor and use thereof
Hamelin-Morrissette et al. Identification of an anti-inflammatory derivative with anti-cancer potential: The impact of each of its structural components on inflammatory responses in macrophages and bladder cancer cells
CN112574174B (en) Protein degradation targeting chimera compound and preparation method and medical application thereof
RU2555999C2 (en) Imidazolidine-2,4-dione derivatives, possessing antiproliferative activity
Kang et al. Discovery of a novel water-soluble, rapid-release triptolide prodrug with improved drug-like properties and high efficacy in human acute myeloid leukemia
CA3021417A1 (en) Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
JP5700324B2 (en) Small molecule anticancer agent that inhibits p53-mdmx interaction
CN113861195B (en) Multi-condensed-ring EGFR inhibitor as well as preparation method and application thereof
JP2019038846A (en) Use of maleimide derivatives for preventing and treating leukemia
WO2014106763A1 (en) Pyridopyrazines as anticancer agents
WO2024140754A1 (en) Naphthylamide compound, and preparation method therefor and use thereof
RU2527526C2 (en) Indolyl-substituted pyrazino-quinolines and using them for treating cancer
JP6082488B1 (en) Preparation of ester of PAK1 blocker acidified by carboxyl group and application to treatment of cancer and other PAK1-dependent diseases
ES2909707T3 (en) Mofezolac and two derivatives for use in the treatment of neuroinflammation
CN118271341A (en) Menin inhibitors and uses thereof

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20210330

EEER Examination request

Effective date: 20210330

EEER Examination request

Effective date: 20210330

FZDE Discontinued

Effective date: 20231011