WO2021191882A1 - Antimalarial agent, methods and uses thereof - Google Patents

Antimalarial agent, methods and uses thereof Download PDF

Info

Publication number
WO2021191882A1
WO2021191882A1 PCT/IB2021/052585 IB2021052585W WO2021191882A1 WO 2021191882 A1 WO2021191882 A1 WO 2021191882A1 IB 2021052585 W IB2021052585 W IB 2021052585W WO 2021191882 A1 WO2021191882 A1 WO 2021191882A1
Authority
WO
WIPO (PCT)
Prior art keywords
previous
compounds according
mmol
dmso
list consisting
Prior art date
Application number
PCT/IB2021/052585
Other languages
French (fr)
Inventor
Marta Sílvia FREITAS DA COSTA
Maria De Fátima MONGINHO BALTAZAR
Maria Fernanda DE JESUS REGO PAIVA PROENÇA
Maria Isabel MENDES VEIGA
Pedro Eduardo MENDES FERREIRA
Carla Sofia MARTINS CALÇADA
Original Assignee
Universidade Do Minho
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 Universidade Do Minho filed Critical Universidade Do Minho
Publication of WO2021191882A1 publication Critical patent/WO2021191882A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/73Unsubstituted amino or imino radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates

Definitions

  • the present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate thereof, for use in medicine, in particular as antimalarial agent.
  • the present disclosure also relates to methods to obtain the compound of formula I.
  • Malaria is caused by Plasmodium parasites. Five species are capable of infecting humans: P. falciparum, P. vivax, P. ovale, P. malariae and P. knowlesi. P. falciparum is the most virulent form and is responsible for majority of the death toll.
  • Most of the drugs currently used for treating malaria patients include artemisinins, aminoquinolines, arylaminoalcohols and anti-folate drugs.
  • the present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate thereof, for use in medicine, in particular as antimalarial agent.
  • the present disclosure also relates to methods to obtain the compound of formula I.
  • the present disclosure relates to antimalarial agents and to methods of preparing them.
  • the advantage of the present disclosure is that the compound disclosed is highly active in different parasite stages, for both drug sensitive and multidrug resistant malaria strains. This advantage is relevant to counteract development of drug resistance mechanisms.
  • This disclosure further describes the method of synthesizing and isolating the novel class of styrylpyridine and 3-arylacrylate-based antimalarial compounds.
  • the present disclosure relates to novel antimalarial agents, in particular, the present disclosure describes the unique method of synthesising new styrylpyridine and 3-arylacrylate-based compounds by combining at least two different molecules thus giving rise to compounds with an interesting associated biological profile.
  • the solution herein presented relates to the synthesis of new compounds and their antimalarial activity by in vitro screening using survival and growth in vitro assays of P. falciparum cultures using SYBR ® Green DNA staining. The activity of the compounds was explored at different levels.
  • the present disclosure describes new antimalarial styrylpyridine and 3- arylacrylate-based compounds and the toxicity effect of these novel compounds in non- neoplastic cell lines MCF-10A (breast) and HK2 (kidney).
  • styrylpyridines show a multistage action at the erythrocytic P. falciparum cycle.
  • styrylpiridines show similar action on P. falciparum 3D7 sensitive line as well as P. falciparum Dd2 multidrug resistant line. [0016] In an embodiment, styrylpiridines induce oxidative cell death.
  • the Caenorhabditis elegans (C. elegans nematode) model was used as an early-stage drug discovery process for toxicity screening.
  • the compound of the present subject-matter exhibits a unique and promising antimalarialprofile.
  • the present disclosure describes a compound of the formula I, a pharmaceutically acceptable salt, ester, solvate, hydrate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph comprising the following formula: wherein:
  • R 1 , R 2 , R 3 and R 4 are independently selected from each other;
  • R 1 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl derivative, ketone, ester, carboxyl group, heterocyclic ring;
  • R 2 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring;
  • R 3 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring;
  • R 4 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring.
  • the present disclosure describes a compound of the formula I, a pharmaceutically acceptable salt, ester, solvate, hydrate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph comprising the following formula: wherein
  • R 1 , R 2 , R 3 and R 4 are independently selected from each other;
  • R 1 is selected from a list consisting of: H, alkyl, alkoxyl, halogen, hydroxyl, carbonyl
  • R 2 is selected from a list consisting of: H, hydroxyl, carbonyl
  • R 3 is selected from a list consisting of: H, hydroxyl, carbonyl, cyano, carboxyl;
  • R 4 is selected from a list consisting of: amine, amide.
  • R 3 is a H, CN, COOH or COO-.
  • R 3 is selected from a list consisting of: H, cyano.
  • R 4 is selected from a list consisting of: heteroarylamine, amide, preferably heteroarylamine, heteroarylamide.
  • R 4 is selected from the following list: iminiopyridin-1(4H)- yl)vinyl unit, ethyl pyridin-4(1H)-ylidene unit, iminiopyridin-1(4H)-yl unit or 4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl unit.
  • the compound disclosed is for use in medicine or veterinary.
  • the compound disclosed is for use in the prevention, therapy or treatment of P. falciparum infection.
  • the compound disclosed is for use in the prevention, therapy or treatment of parasite infection.
  • the compound disclosed is for the prevention, therapy or treatment of malaria diseases.
  • the compound disclosed is for the prevention, therapy or treatment of P. falciparum infection.
  • a pharmaceutical composition comprising the disclosed compound in a therapeutically effectively amount and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition comprising the disclosed compound wherein the pharmaceutically acceptable excipient is a carrier, adjuvant, excipient or mixtures thereof.
  • the pharmaceutical composition comprising the disclosed compound may further comprise an anti-viral compound, preferably wherein the antiviral compound is an anti-HIV compound or anti-tuberculosis compound.
  • the pharmaceutical composition comprising the disclosed compound may further comprise a second anti-malarial agent, preferably wherein the second anti-malarial agent is selected from the group consisting of: Chloroquine, Fansidar, Amodiaquine, Quinine, Halofantrine, Mefloquine, Artemether/Artesunate and Malarone.
  • a second anti-malarial agent selected from the group consisting of: Chloroquine, Fansidar, Amodiaquine, Quinine, Halofantrine, Mefloquine, Artemether/Artesunate and Malarone.
  • Another aspect of the present disclosure relates to a method for preparing the disclosed compound comprising the following steps:
  • the present disclosure relates to a method for obtaining the compound, that comprises the following steps:
  • Another aspect of the present disclosure relates to a method of preparing the compound of the present disclosure comprising: adding pyridinium salt to a solution of aldehyde in aqueous base and ethanol or water in order to obtain a first mixture; stirring at room temperature to 110 °C for 2 h 55 min to 87.5 hours; cooling to the first mixture down to room temperature; filtering the cooled first mixture to obtain precipitate; washing the precipitate with water to obtain 3-arylacrylate-based compound or derivatives thereof.
  • the method may further comprise: heating a solution of 3-(2-hydroxyphenyl)-2-pyridyl)acrylate in DMSO at 130 °C for a duration from 35 minutes to 3 hours to obtain a reaction mixture; adding 1 mL of water to the reaction mixture; filtering the reaction mixture to obtain precipitate; washing the precipitate with water to obtain pure styrylpyridine derivatives.
  • the word "comprise” and variations of the word are not intended to exclude other technical features, additives, components, or steps. Additional objects, advantages and features of the invention will become apparent to those skilled in the art upon examination of the description or may be learned by practice of the invention.
  • the following examples and drawings are provided by way of illustration, and they are not intended to be limiting of the present invention.
  • the present invention covers all possible combinations of particular and preferred embodiments described herein.
  • an alkyl group is defined as a univalent group derived from alkanes by removal of a hydrogen atom from any carbon atom -CnH 2n+1 .
  • the groups derived by removal of a hydrogen atom from a terminal carbon atom of unbranched alkanes form a subclass of normal alkyl (n-alkyl) groups H (CH 2 ) n .
  • the groups RCH 2 , R 2 CH (R ⁇ H), and R 3 C (R ⁇ H) are primary, secondary and tertiary alkyl groups respectively.
  • An aryl group is derived from arenes (monocyclic and polycyclic aromatic hydrocarbons) by removal of a hydrogen atom from a ring carbon atom.
  • Alkyl includes “lower alkyl” and extends to cover carbon fragments having up to 30 carbon atoms.
  • alkyl groups include octyl, nonyl, norbornyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, eicosyl, 3,7-diethyl-2,2-dimethyl-4 - propylnonyl, 2-(cyclododecyl)ethyl, adamantyl, and the like.
  • Lower alkyl means alkyl groups with 1 to 7 carbon atoms.
  • lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl, pentyl, hexyl, heptyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, 2- methylcyclopropyl, cyclopropylmethyl, and the like.
  • halogen refers to an element selected from the list consisting of: fluorine (F), chlorine (Cl), bromine (Br), iodine (I), astatine (At).
  • heterocyclic ring denotes a ring wherein at least one of the atoms forming the ring backbone is not a carbon. Unless otherwise indicated, a heterocyclic ring can be a saturated, partially unsaturated, or fully unsaturated ring. "Saturated heterocyclic ring” refers to a heterocyclic ring containing only single bonds between ring members. "Partially saturated heterocyclic ring” refers to a non-aromatic heterocyclic ring containing at least one double bond. The term “heteroaromatic ring” denotes a fully unsaturated aromatic ring in which at least one atom forming the ring backbone is not a carbon.
  • heteroaromatic ring typically contains no more than 4 nitrogens, no more than 1 oxygen, and no more than 1 sulfur. Unless otherwise indicated, heteroaromatic rings can be attached through any available carbon or nitrogen by replacement of a hydrogen on said carbon or nitrogen.
  • heteroaromatic bicyclic ring system denotes a ring system consisting of two fused rings in which at least one of the two rings is a heteroaromatic ring as defined above.
  • carbocyclic ring denotes a ring wherein the atoms forming the ring backbone are selected only from carbon. Unless otherwise indicated, a carbocyclic ring can be a saturated, partially unsaturated, or fully unsaturated ring. When a fully unsaturated carbocyclic ring satisfies Huckel's rule, then said ring is also called an "aromatic ring". "Saturated carbocyclic ring” refers to a ring having a backbone consisting of carbon atoms linked to one another by single bonds; unless otherwise specified, the remaining carbon valences are occupied by hydrogen atoms.
  • Figure 1 shows the in vivo toxicity of the compounds using C. elegans model.
  • the present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate or prodrug thereof, for use in medicine, in particular as antimalarial agent.
  • the present solution describes a new styrylpyridine and 3-arylacrylate-based compounds, the method of synthesizing these compounds by combining at least two different molecules thus giving rise to compounds with an interesting associated biological profile.
  • the multi-parasite stage action of styrylpyridine was determined through the analysis of IC 50 for the different stages on P. falciparum in Dd2 parasite line. The results show IC 50 values as low as 3.7 nM (Table 3). Table 3. Multi erythrocytic antimalarial action of MCA-205 compound on P. falciparum Dd2 line.
  • IC 50 values of the five most active compounds were determined in non-neoplastic MCF-10A (breast) and HK2 (kidney) cell lines, as part of the evaluation of the selective cytotoxicity exhibited by the compounds.
  • Selectivity index (SI) was calculated for each tested compound and are presented in Tables 4 and 5 below.
  • Plasmodium falciparum lines were obtained from MR4- Malaria Resources and maintained at ⁇ 4% haematocrit with human red blood cells in RPMI-1640, supplemented with 2mM L-glutamine, 200 ⁇ M hypoxanthine, 0.25 ⁇ g/mL gentamycin, 25mM HEPES, 0.2% NaHCO 3 , and 0.25% Albumax II.
  • Parasite cultures were maintained at 37°C under a humidified controlled atmosphere of 5% O 2 /5% CO 2 /90% N 2 .
  • Parasite growth was monitored through Giemsa-stained blood smears.
  • Parasite synchronization was performed with 5% sorbitol for 15 minutes at 37 °C. To obtain highly synchronous cultures, sorbitol treatment was applied after 6 - 8 hours.
  • synchronized ring-stage parasite was incubated at 37°C with 0.2% starting parasitemia and 1% haematocrit. After 72h, to determine the parasite growth, the parasites were stained with SYBR Green (Thermofisher) in PBS lx for 30 minutes and then fluorescence analysed and growth normalized to the controls with no drugs. The IC 50 values were calculated using nonlinear regression analysis with GraphPad prism 6 software.
  • non-neoplastic breast cell line MCF-10A was obtained from ATCC (American Type Culture Collection) and cultured in Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F12, Gibco) supplemented with 5% heating inactivated FBS (Gibco), 1% antibiotic solution (Penicillin-Streptomycin, Gibco), 1% steroid hormone (Hydrocortisone, Sigma-Aldrich), 0.1% peptide hormone (Insulin, Sigma-Aldrich) and 0.01% protein complex (Cholera Toxin, Gibco).
  • the Kidney cell line HK2 was also obtained from ATCC and cultured in RPMI 1640 medium (Biochrom ® - Merck Millipore), supplemented with 10% heat inactivated FBS (Biochrom ® - Merck Millipore) and 1% antibiotic/antimitotic mixture (Invitrogen ® ). All cells were grown in a humidified incubator at 37 °C and 5% CO 2 . For all assays, DMSO (Dimethyl Sulfoxide, Sigma-Aldrich) controls were used.
  • cell viability assays were performed. MCF-10A cells were plated at 3000 cells per mL and HK2 cells were plated at 2000 cells per mL, in triplicate, using 96-multiwell culture plate. The cells were then allowed to adhere in complete medium, over a period of 18-20 hours. Cells were subsequently treated with 5 different concentrations (60, 45, 30, 10 and 5 ⁇ M) of compounds or control in fresh medium. After 72h of incubation, Sulforhodamine B assay was used according to manufacturer's instructions. The data was log-transformed and the concentration of each compound that decreased the number of viable cells to 50% (IC 50 ) relative to control was calculated using the GraphPad Prism 6 software.
  • selectivity index (SI) value was calculated using the IC 50 values of the five best compounds for MCF10A and HK2 cell lines versus the IC 50 values calculated for P. falciparum Dd2 and 3D7, using the following mathematical formula:
  • SI (IC 50 normal cell line - IC 50 malaria)/ IC 50 malaria
  • C. elegans drug toxicity assay was performed using the five most active compounds.
  • _C. elegans wild-type strain N2 (Bristol strain) was cultured in nematode growth medium (NGM) plates seeded with Escherichia coli OP50 strain, at 20 °C.
  • NNM nematode growth medium
  • the toxicity of compounds in vivo was determined in the wild-type N2 Bristol strain, using the food clearance assay. Briefly, C.
  • elegans were cultured in liquid culture, using 96-well plates, in the presence of inactivated OP50 and with different concentrations of each compound (from 100 ⁇ M to 0.05 ⁇ M). The consumption of food (OD 595 nm) was measured daily for 7 days. In the assay, DMSO was used as drug vehicle at a final drug concentration of 1%. C. elegans treated with 1% and 5% DMSO were used as non-toxic and toxic concentration controls, respectively.
  • the reactions of all the chemical compounds were monitored by thin layer chromatography (TLC) using silica gel 60 plates (Macherey-Nagel, 0.2 mm) with fluorescence indicator.
  • TLC thin layer chromatography
  • silica gel 60 plates Macherey-Nagel, 0.2 mm
  • fluorescence indicator fluorescence indicator
  • a UV chamber CN-6 Vilber Lourmat
  • a hot plate stirrer IKAMAG RCT was used with appropriate magnetic stirring and at different temperatures according to the specific procedure.
  • Solvents were evaporated in a Buchi RE 11 rotary evaporator with vacuum and variable bath temperature.
  • IR spectra were recorded in FT-IR Bomem MB 104 using nujol mulls and NaCI cells. Melting points were determined in a Stuart SMP3 apparatus and were not corrected.
  • pyridinium salt 2 (0.03-0.19 g; 1-1.6 eq) was added to a solution/suspension of aldehyde 1 (0.01-0.19 g) in an aqueous solution of NaHCO 3 (0.01 or 0.1 M; 2-3 mL)/mixture of an aqueous solution of NaHCO 3 (0.01 or 0.1 M; 2-3 mL) and ethanol (0.2-0.3 mL) or in water (2-7 mL).
  • the reaction mixture was stirred at a temperature between 20 °C and 110 °C for a duration from 55 min to 87.5 hours. After cooling to room temperature in an ice bath, the precipitated solid was filtered and washed with water leading to the pure 3-arylacrylate-based compound or MC317 (17- 98%).
  • 3-(2-hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin-1(4H)- yl)acrylate MCA-195 was synthesised.
  • 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a 120 mg; 0.709 mmol; 1.2 eq.
  • 3- methoxysalicylaldehyde 1 90.8 mg; 0.591 mmol
  • NaHCO 3 0.01M 3 mL
  • 3-(5-chloro-2-hydroxyphenyl)-2-(4-iminiopyridin-1(4H)- yl)acrylate MCA-201 was synthesised.
  • 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (163 mg; 0.963 mmol; 1.2 eq.) was added to a white suspension of 5- chlorosalicylaldehyde 1 (128 mg; 0.803 mmol) in ethanol (0.2 mL) and aqueous solution of NaHCO 3 0.01M (3 mL). The suspension was stirred at 110 °C and a homogeneous solution was obtained after 55 min. A light brown solid started to precipitate after 30 minutes.
  • the brown solid was identified as 3-(5-bromo-2-hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin- 1(4H)-yl)acrylate MCB-145 (260 mg; 0.746 mmol).
  • 2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)-3-(2- hydroxyphenyl)acrylate MCA-64 was synthesised.
  • 1-(Cyanomethyl)-4- (ethoxycarbonylamino)pyridinium chloride 2b 143 mg; 0.590 mmol; 1.6 eq.
  • the yellow solution was stirred at 80 °C and after 2 h and 45 min, a yellow solid precipitated from the reaction mixture.
  • the product (0.05 g; 0.14 mmol) was identified as 3-(2-hydroxy-4-methoxylphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-360.
  • 3-(2-hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1- ium-1-yl)acrylate MCB-314 was synthesised.
  • 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.031 g; 0.18 mmol; 1.6 eq.) was added to a solution of salicylaldehyde 1 (0.01 g; 0.12 mmol; 12 m ⁇ ) in an aqueous solution of NaHC0 3 0.01M (1 mL) and heated at 80 °C. After 1 h and 30 min, a beige solid started to precipitate from the reaction mixture.
  • 4-(2-cyano-2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MC- 317 was synthesised.
  • 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.18 g; 1.04 mmol; 1.2 eq.) was added to a solution of 4-hydroxybenzaldehyde 1 (0.11 g; 0.89 mmol) in an aqueous solution of NaHCO 3 0,05M (1 mL) and propylamine (54.6 mg; 0.92 mmol; 76m ⁇ ).
  • ethyl (1-(2-hydroxystyryl)pyridin-4(lH)-ylidene)carbamate MCA- 189 was synthesised.
  • a yellow suspension of 2-(4-((ethoxycarbonyl)amino)pyridin- l-ium-1-yl)-3-(2-hydroxyphenyl)acrylate MCA-64 (96.9 mg; 0.263 mmol) in DMSO (0.2 mL) was stirred at 110 °C and a red solution was obtained 15 minutes later.
  • the reaction was complete after 1 h and 45 min (by TLC).
  • An orange suspension was formed after addition of water (1 mL) and the solid was filtered and washed with water.
  • ethyl 4-chloro-2-(2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MCA-206 was synthesised.
  • a suspension of 3-(5-chloro-2-hydroxyphenyl)-2-(4- iminiopyridin-1(4H)-yl)acrylate MCA-201 (88.9 mg; 0.319 mmol) in DMSO (0.2) was stirred at 130 °C.
  • ethyl (1-(5-bromo-2-hydroxystyryl)pyridin-4(1H)- ylidene)carbamate MCA-116 was synthesised.
  • a suspension of 3-(5-bromo-2- hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-122 (115 mg; 0.283 mg) in DMSO (0.2 mL) was stirred at 130 °C.
  • the starting material was solubilized 10 minutes later and after 1 h and 50 min, TLC confirmed the absence of starting material and the addition of water (1mL) led to an orange solid.
  • 2,4-difluoro-6-(2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MCA-249 was synthesised.
  • a suspension of 3-(3,5-difluoro-2-hydroxyphenyl)-2-(4- iminiopyridin-1(4H)-yl)acrylate MCB-156 (0.03 g; 0.10 mmol) in DMSO (0.2 mL) and water (6.0 m ⁇ ) was stirred at 130 °C. After 1 h and 15 min the reaction was complete (by TLC). Addition of water (2 mL) led to an orange solid that was filtered and washed with water.
  • ethyl (1-(3,5-difluoro-2-hydroxystyryl)pyridin-4(lH)- ylidene)carbamate MCB-376 was synthesised.
  • a yellow suspension of 3-(3,5-difluoro-2- hydroxylphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-192 (0.02 g; 0.06 mmol) in DMSO (0.2 mL) was stirred at 100 °C. After 30 min the reaction temperature was raised to 130 °C and maintained for a further hour.

Landscapes

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

Abstract

The present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate thereof, for use in medicine, in particular as antimalarial agent. The present disclosure also relates to methods to obtain the compound of formula I.

Description

D E S C R I P T I O N
ANTIMALARIAL AGENT, METHODS AND USES THEREOF
TECHNICAL FIELD
[0001] The present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate thereof, for use in medicine, in particular as antimalarial agent.
[0002] Furthermore, the present disclosure also relates to methods to obtain the compound of formula I.
TECHNICAL BACKGROUND
[0003] Malaria affects the whole world, with half of the global population at risk of infection.
[0004] Malaria is caused by Plasmodium parasites. Five species are capable of infecting humans: P. falciparum, P. vivax, P. ovale, P. malariae and P. knowlesi. P. falciparum is the most virulent form and is responsible for majority of the death toll.
[0005] Most of the drugs currently used for treating malaria patients include artemisinins, aminoquinolines, arylaminoalcohols and anti-folate drugs.
[0006] Presently, there is a growing need for antimalarial agents, either for monotherapy or as multi-drug therapies as drug-resistant strains are emerging worldwide, in particular mefloquine and chloroquine resistant strains. Therefore, new agents able to kill the resistant malaria parasites are warranted.
[0007] These facts are disclosed in order to illustrate the technical problem addressed by the present disclosure. GENERAL DESCRIPTION
[0008] The present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate thereof, for use in medicine, in particular as antimalarial agent.
[0009] Furthermore, the present disclosure also relates to methods to obtain the compound of formula I.
[0010] The present disclosure relates to antimalarial agents and to methods of preparing them. The advantage of the present disclosure is that the compound disclosed is highly active in different parasite stages, for both drug sensitive and multidrug resistant malaria strains. This advantage is relevant to counteract development of drug resistance mechanisms. This disclosure further describes the method of synthesizing and isolating the novel class of styrylpyridine and 3-arylacrylate-based antimalarial compounds.
[0011] The present disclosure relates to novel antimalarial agents, in particular, the present disclosure describes the unique method of synthesising new styrylpyridine and 3-arylacrylate-based compounds by combining at least two different molecules thus giving rise to compounds with an interesting associated biological profile.
[0012] Furthermore, the solution herein presented relates to the synthesis of new compounds and their antimalarial activity by in vitro screening using survival and growth in vitro assays of P. falciparum cultures using SYBR® Green DNA staining. The activity of the compounds was explored at different levels.
[0013] The present disclosure describes new antimalarial styrylpyridine and 3- arylacrylate-based compounds and the toxicity effect of these novel compounds in non- neoplastic cell lines MCF-10A (breast) and HK2 (kidney).
[0014] In an embodiment, styrylpyridines show a multistage action at the erythrocytic P. falciparum cycle.
[0015] In an embodiment, styrylpiridines show similar action on P. falciparum 3D7 sensitive line as well as P. falciparum Dd2 multidrug resistant line. [0016] In an embodiment, styrylpiridines induce oxidative cell death.
[0017] In an embodiment, the Caenorhabditis elegans (C. elegans nematode) model was used as an early-stage drug discovery process for toxicity screening.
[0018] The compound of the present subject-matter exhibits a unique and promising antimalarialprofile.
[0019] The present disclosure describes a compound of the formula I, a pharmaceutically acceptable salt, ester, solvate, hydrate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph comprising the following formula:
Figure imgf000004_0001
wherein:
R1, R2, R3 and R4are independently selected from each other;
R1 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl derivative, ketone, ester, carboxyl group, heterocyclic ring;
R2 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring;
R3 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring;
R4 is selected from a H, alkyl, aryl, alkoxyl, acetyl, acyl, halogen, nitro, hydroxyl, amine, amide, carbonyl, ketone, ester, carboxyl group, heterocyclic ring.
[0020] The present disclosure describes a compound of the formula I, a pharmaceutically acceptable salt, ester, solvate, hydrate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph comprising the following formula:
Figure imgf000005_0001
wherein
R1, R2, R3 and R4are independently selected from each other;
R1 is selected from a list consisting of: H, alkyl, alkoxyl, halogen, hydroxyl, carbonyl; R2 is selected from a list consisting of: H, hydroxyl, carbonyl;
R3 is selected from a list consisting of: H, hydroxyl, carbonyl, cyano, carboxyl;
R4 is selected from a list consisting of: amine, amide.
[0021] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R1 is selected from a list consisting of: H, fluoro, bromo, chloro, halo-methoxy.
[0022] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R3 is a H, CN, COOH or COO-. Preferably, wherein R3 is selected from a list consisting of: H, cyano.
[0023] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R2 is a hydroxyl.
[0024] In an embodiment, R4 is selected from a list consisting of: heteroarylamine, amide, preferably heteroarylamine, heteroarylamide.
[0025] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R4 is a pyridine or a pyridinium salt derivative.
[0026] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R4 is a pyridine-moiety. [0027] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein R4 is selected from the following list: iminiopyridin-1(4H)- yl)vinyl unit, ethyl pyridin-4(1H)-ylidene unit, iminiopyridin-1(4H)-yl unit or 4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl unit.
[0028] In an embodiment, the compound or a pharmaceutically acceptable, salt, hydrate, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer, dimer, or polymorph wherein the compound is selected from the following list:
Figure imgf000006_0001
Figure imgf000007_0001
[0029] In an embodiment, the compound disclosed is for use in medicine or veterinary. [0030] In an embodiment, the compound disclosed is for use in the prevention, therapy or treatment of P. falciparum infection.
[0031] In an embodiment, the compound disclosed is for use in the prevention, therapy or treatment of parasite infection.
[0032] In an embodiment, the compound disclosed is for the prevention, therapy or treatment of malaria diseases.
[0033] In an embodiment, the compound disclosed is for the prevention, therapy or treatment of P. falciparum infection.
[0034] In an embodiment, a pharmaceutical composition comprising the disclosed compound in a therapeutically effectively amount and a pharmaceutically acceptable excipient.
[0035] In an embodiment, a pharmaceutical composition comprising the disclosed compound wherein the pharmaceutically acceptable excipient is a carrier, adjuvant, excipient or mixtures thereof.
[0036] In embodiment, the pharmaceutical composition comprising the disclosed compound may further comprise an anti-viral compound, preferably wherein the antiviral compound is an anti-HIV compound or anti-tuberculosis compound.
[0037] In embodiment, the pharmaceutical composition comprising the disclosed compound may further comprise a second anti-malarial agent, preferably wherein the second anti-malarial agent is selected from the group consisting of: Chloroquine, Fansidar, Amodiaquine, Quinine, Halofantrine, Mefloquine, Artemether/Artesunate and Malarone.
[0038] Another aspect of the present disclosure relates to a method for preparing the disclosed compound comprising the following steps:
Adding pyridinium salt (0.03-0.19 g; 1-1.6 eq) to a solution/suspension of aldehyde (0.966-1.02 g) in aqueous base (NaHC03 (0.01-0.1 M; 1-3 mL) and ethanol (0.2-0.3 mL) or water (2-7 mL); Stirring at a temperature between 20 °C and 110 °C for a duration of 2 h 55 min to 87.5 hours;
Cooling to room temperature in an ice bath, filtering the precipitate and washing with water, to obtain pure 3-arylacrylate-based compound or derivatives thereof.
[0039] In an embodiment, for the preparation of the styrylpyridine derivative, the present disclosure relates to a method for obtaining the compound, that comprises the following steps:
Heating a solution of 3-(2-hydroxyphenyl)-2-pyridyl)acrylate (0.02-0.14 g), in DMSO (0.2-0.7 mL) at 130 °C for a duration from 35 minutes to 3 hours;
Adding 1 mL of water to the reaction mixture;
Filtering and washing with water to isolate the pure styrylpyridine derivatives.
[0040] Another aspect of the present disclosure, relates to a method of preparing the compound of the present disclosure comprising: adding pyridinium salt to a solution of aldehyde in aqueous base and ethanol or water in order to obtain a first mixture; stirring at room temperature to 110 °C for 2 h 55 min to 87.5 hours; cooling to the first mixture down to room temperature; filtering the cooled first mixture to obtain precipitate; washing the precipitate with water to obtain 3-arylacrylate-based compound or derivatives thereof.
[0041] In an embodiment, the method may further comprise: heating a solution of 3-(2-hydroxyphenyl)-2-pyridyl)acrylate in DMSO at 130 °C for a duration from 35 minutes to 3 hours to obtain a reaction mixture; adding 1 mL of water to the reaction mixture; filtering the reaction mixture to obtain precipitate; washing the precipitate with water to obtain pure styrylpyridine derivatives. [0042] Throughout the description and claims the word "comprise" and variations of the word, are not intended to exclude other technical features, additives, components, or steps. Additional objects, advantages and features of the invention will become apparent to those skilled in the art upon examination of the description or may be learned by practice of the invention. The following examples and drawings are provided by way of illustration, and they are not intended to be limiting of the present invention. Furthermore, the present invention covers all possible combinations of particular and preferred embodiments described herein.
[0043] Based on the International Union of Pure and Applied Chemistry (lUPAC) definitions, an alkyl group is defined as a univalent group derived from alkanes by removal of a hydrogen atom from any carbon atom -CnH2n+1. The groups derived by removal of a hydrogen atom from a terminal carbon atom of unbranched alkanes form a subclass of normal alkyl (n-alkyl) groups H (CH2)n. The groups RCH2, R2CH (R ≠ H), and R3C (R ≠ H) are primary, secondary and tertiary alkyl groups respectively. An aryl group is derived from arenes (monocyclic and polycyclic aromatic hydrocarbons) by removal of a hydrogen atom from a ring carbon atom.
[0044] "Alkyl" includes "lower alkyl" and extends to cover carbon fragments having up to 30 carbon atoms. Examples of alkyl groups include octyl, nonyl, norbornyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, eicosyl, 3,7-diethyl-2,2-dimethyl-4 - propylnonyl, 2-(cyclododecyl)ethyl, adamantyl, and the like.
[0045] "Lower alkyl" means alkyl groups with 1 to 7 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl, pentyl, hexyl, heptyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, 2- methylcyclopropyl, cyclopropylmethyl, and the like.
[0046] In the present disclosure, halogen refers to an element selected from the list consisting of: fluorine (F), chlorine (Cl), bromine (Br), iodine (I), astatine (At).
[0047] In the present disclosure, the term "heterocyclic ring" denotes a ring wherein at least one of the atoms forming the ring backbone is not a carbon. Unless otherwise indicated, a heterocyclic ring can be a saturated, partially unsaturated, or fully unsaturated ring. "Saturated heterocyclic ring" refers to a heterocyclic ring containing only single bonds between ring members. "Partially saturated heterocyclic ring" refers to a non-aromatic heterocyclic ring containing at least one double bond. The term "heteroaromatic ring" denotes a fully unsaturated aromatic ring in which at least one atom forming the ring backbone is not a carbon. Typically, a heteroaromatic ring contains no more than 4 nitrogens, no more than 1 oxygen, and no more than 1 sulfur. Unless otherwise indicated, heteroaromatic rings can be attached through any available carbon or nitrogen by replacement of a hydrogen on said carbon or nitrogen. The term "heteroaromatic bicyclic ring system" denotes a ring system consisting of two fused rings in which at least one of the two rings is a heteroaromatic ring as defined above.
[0048] The term "carbocyclic ring" denotes a ring wherein the atoms forming the ring backbone are selected only from carbon. Unless otherwise indicated, a carbocyclic ring can be a saturated, partially unsaturated, or fully unsaturated ring. When a fully unsaturated carbocyclic ring satisfies Huckel's rule, then said ring is also called an "aromatic ring". "Saturated carbocyclic ring" refers to a ring having a backbone consisting of carbon atoms linked to one another by single bonds; unless otherwise specified, the remaining carbon valences are occupied by hydrogen atoms.
BRIEF DESCRIPTION OF THE DRAWINGS
[0049] The following figures provide preferred embodiments for the present disclosure and should not be seen as limiting the scope of the disclosure.
[0050] Figure 1: shows the in vivo toxicity of the compounds using C. elegans model.
DETAILED DESCRIPTION
[0051] The present disclosure relates to compound of the formula I; or a pharmaceutically acceptable salt, ester, solvate or prodrug thereof, for use in medicine, in particular as antimalarial agent. [0052] The present solution describes a new styrylpyridine and 3-arylacrylate-based compounds, the method of synthesizing these compounds by combining at least two different molecules thus giving rise to compounds with an interesting associated biological profile.
[0053] In an embodiment, 19 different styrylpyridine and 3-arylacrylate-based compounds were isolated. Table 1 shows the structure of the styrylpyridine and 3- arylacrylate-based scaffolds, as well as the 19 synthesised and isolated styrylpyridine and 3-arylacrylate-based compounds.
Table 1. Structure of the styrylpyridine and 3-arylacrylate-based compounds.
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0002
Figure imgf000014_0001
[0054] In an embodiment, antimalarial activity on P. falciparum was determined using the multidrug resistant parasite line Dd2 and susceptible parasite line 3D7. The results are described in Table 2 below.
Table 2. Antimalarial action of compounds described on Table 1.
Figure imgf000015_0001
[0055] In an embodiment, the multi-parasite stage action of styrylpyridine was determined through the analysis of IC50 for the different stages on P. falciparum in Dd2 parasite line. The results show IC50 values as low as 3.7 nM (Table 3). Table 3. Multi erythrocytic antimalarial action of MCA-205 compound on P. falciparum Dd2 line.
Figure imgf000016_0001
[0056] In an embodiment, IC50 values of the five most active compounds were determined in non-neoplastic MCF-10A (breast) and HK2 (kidney) cell lines, as part of the evaluation of the selective cytotoxicity exhibited by the compounds. Selectivity index (SI) was calculated for each tested compound and are presented in Tables 4 and 5 below.
Table 4. IC50 values (μM) for non-neoplastic cell line MCF-10A and selectivity index (SI) of the compounds for Dd2 and SD7 parasite strains.
Figure imgf000016_0002
Table 5. IC50 values (μM) for non-neoplastic cell line HK2 and selectivity index (SI) of the compounds for P. falciparum Dd2 and SD7 strains.
Figure imgf000016_0003
Figure imgf000017_0001
[0057] For the non-neoplastic cell lines MCF-10A and HK2, the IC50 values were all in the micromolar range and high SI values were obtained, leading to excellent SI values (higherthan 10). These values indicated a promising profile of the compounds regarding toxicity for normal cells.
[0058] In an embodiment, Plasmodium falciparum lines were obtained from MR4- Malaria Resources and maintained at ~ 4% haematocrit with human red blood cells in RPMI-1640, supplemented with 2mM L-glutamine, 200μM hypoxanthine, 0.25 μg/mL gentamycin, 25mM HEPES, 0.2% NaHCO3, and 0.25% Albumax II. Parasite cultures were maintained at 37°C under a humidified controlled atmosphere of 5% O2/5% CO2/90% N2. Parasite growth was monitored through Giemsa-stained blood smears. Parasite synchronization was performed with 5% sorbitol for 15 minutes at 37 °C. To obtain highly synchronous cultures, sorbitol treatment was applied after 6 - 8 hours.
[0059] In an embodiment, in order to determine antimalarial activities, synchronized ring-stage parasite was incubated at 37°C with 0.2% starting parasitemia and 1% haematocrit. After 72h, to determine the parasite growth, the parasites were stained with SYBR Green (Thermofisher) in PBS lx for 30 minutes and then fluorescence analysed and growth normalized to the controls with no drugs. The IC50 values were calculated using nonlinear regression analysis with GraphPad prism 6 software.
[0060] In an embodiment, non-neoplastic breast cell line MCF-10A was obtained from ATCC (American Type Culture Collection) and cultured in Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F12, Gibco) supplemented with 5% heating inactivated FBS (Gibco), 1% antibiotic solution (Penicillin-Streptomycin, Gibco), 1% steroid hormone (Hydrocortisone, Sigma-Aldrich), 0.1% peptide hormone (Insulin, Sigma-Aldrich) and 0.01% protein complex (Cholera Toxin, Gibco). The Kidney cell line HK2 was also obtained from ATCC and cultured in RPMI 1640 medium (Biochrom®- Merck Millipore), supplemented with 10% heat inactivated FBS (Biochrom® - Merck Millipore) and 1% antibiotic/antimitotic mixture (Invitrogen®). All cells were grown in a humidified incubator at 37 °C and 5% CO2. For all assays, DMSO (Dimethyl Sulfoxide, Sigma-Aldrich) controls were used.
[0061] In an embodiment, cell viability assays were performed. MCF-10A cells were plated at 3000 cells per mL and HK2 cells were plated at 2000 cells per mL, in triplicate, using 96-multiwell culture plate. The cells were then allowed to adhere in complete medium, over a period of 18-20 hours. Cells were subsequently treated with 5 different concentrations (60, 45, 30, 10 and 5 μM) of compounds or control in fresh medium. After 72h of incubation, Sulforhodamine B assay was used according to manufacturer's instructions. The data was log-transformed and the concentration of each compound that decreased the number of viable cells to 50% (IC50) relative to control was calculated using the GraphPad Prism 6 software.
[0062] In an embodiment, selectivity index (SI) value was calculated using the IC50 values of the five best compounds for MCF10A and HK2 cell lines versus the IC50 values calculated for P. falciparum Dd2 and 3D7, using the following mathematical formula:
SI = (IC50 normal cell line - IC50 malaria)/ IC50 malaria For SI values >1, cytotoxicity for malaria is higher than for non-neoplastic cell lines.
[0063] In an embodiment, C. elegans drug toxicity assay was performed using the five most active compounds._C. elegans wild-type strain N2 (Bristol strain) was cultured in nematode growth medium (NGM) plates seeded with Escherichia coli OP50 strain, at 20 °C. To obtain populations of eggs, adult animals were submitted to a bleaching technique (treatment with a 20% Alkaline Hypochlorite Solution). The toxicity of compounds in vivo was determined in the wild-type N2 Bristol strain, using the food clearance assay. Briefly, C. elegans were cultured in liquid culture, using 96-well plates, in the presence of inactivated OP50 and with different concentrations of each compound (from 100 μM to 0.05 μM). The consumption of food (OD 595 nm) was measured daily for 7 days. In the assay, DMSO was used as drug vehicle at a final drug concentration of 1%. C. elegans treated with 1% and 5% DMSO were used as non-toxic and toxic concentration controls, respectively.
[0064] In an embodiment, the reactions of all the chemical compounds were monitored by thin layer chromatography (TLC) using silica gel 60 plates (Macherey-Nagel, 0.2 mm) with fluorescence indicator. For revelation, a UV chamber (CN-6 Vilber Lourmat) with a 254 nm lamp was used. A hot plate stirrer IKAMAG RCT was used with appropriate magnetic stirring and at different temperatures according to the specific procedure. Solvents were evaporated in a Buchi RE 11 rotary evaporator with vacuum and variable bath temperature. IR spectra were recorded in FT-IR Bomem MB 104 using nujol mulls and NaCI cells. Melting points were determined in a Stuart SMP3 apparatus and were not corrected. Elemental analysis was performed on a LECO CHNS-932 instrument. NMR spectra were obtained in Bruker Avance III (at 400 MHz for 1H NMR and 100 MHz for 13C NMR), at 25 °C and using deuterated dimethylsulfoxide (DMSO-d6) as solvent. Chemical shifts were recorded in parts per million (ppm) using the residual solvent peak as an internal standard.
Figure imgf000020_0001
[0065] In an embodiment, pyridinium salt 2 (0.03-0.19 g; 1-1.6 eq) was added to a solution/suspension of aldehyde 1 (0.01-0.19 g) in an aqueous solution of NaHCO3 (0.01 or 0.1 M; 2-3 mL)/mixture of an aqueous solution of NaHCO3 (0.01 or 0.1 M; 2-3 mL) and ethanol (0.2-0.3 mL) or in water (2-7 mL). The reaction mixture was stirred at a temperature between 20 °C and 110 °C for a duration from 55 min to 87.5 hours. After cooling to room temperature in an ice bath, the precipitated solid was filtered and washed with water leading to the pure 3-arylacrylate-based compound or MC317 (17- 98%).
[0066] In an embodiment, 3-(2-hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin-1(4H)- yl)acrylate MCA-195 was synthesised. 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (120 mg; 0.709 mmol; 1.2 eq.) was added to a yellow suspension of 3- methoxysalicylaldehyde 1 (90.8 mg; 0.591 mmol) in an aqueous solution of NaHCO3 0.01M (3 mL) leading to a yellow suspension. The reaction mixture was heated at 110 °C, leading to a brownish solid suspension after 50 minutes. After 2 h, analysis of the reaction mixture by TLC showed the absence of starting materials and the green solid suspension was filtered and washed with water, analysed and identified as the pure product 3-(2-hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin-1(4H)-yl)acrylate MCA-195 (125 mg; 0.437 mmol). Beige solid; yield 74 %; mp 219-220 °C; 1H NMR (400 MHz, DMSO- d6) δ 13.00 (s; 1H; OH), 8.20 (d; J =7.8Hz; 4H), 7.79 (d; J =7.8Hz), 6.90 (dd; J1 = 3.3Hz; J2 =1.5Hz), 6.89 (s, 1H), 6.87 (d; J = 1.8Hz), 6.72 (t; J = 7.8Hz), 3.74 (s; 3H); 13C NMR 100 MHz, DMSO- d6) δ 164.23, 158.71, 150.45, 147.41, 143.50, 139.32, 128.47, 123.96, 122.62, 118.06, 112.61, 108.34; IR (Nujol mull) v 3289, 3084, 1664, 1598, 1533 cm-1; Anal. Calcd for C15H13N2O4: C, 56.00; H, 4.39; N, 8.71. Found: C, 56.05; H, 4.42; N, 8.75.
[0067] In an embodiment, 3-(5-chloro-2-hydroxyphenyl)-2-(4-iminiopyridin-1(4H)- yl)acrylate MCA-201 was synthesised. 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (163 mg; 0.963 mmol; 1.2 eq.) was added to a white suspension of 5- chlorosalicylaldehyde 1 (128 mg; 0.803 mmol) in ethanol (0.2 mL) and aqueous solution of NaHCO3 0.01M (3 mL). The suspension was stirred at 110 °C and a homogeneous solution was obtained after 55 min. A light brown solid started to precipitate after 30 minutes. After stirring for a further 2 h 40 min, the brown solid was filtered, washed with water and identified as 3-(5-chloro-2-hydroxyphenyl)-2-(4-iminiopyridin-1(4H)- yl)acrylate MCA-201 (141 mg; 0.484 mmol). Brown solid; yield 60 %; mp 229-230 °C; 1H NMR (400 MHz, DMSO-d6) δ 12.65 (s; 1H; OH), 8.24 (brs; 2H), 8.21 (d; J = 7.6Hz; 2H), 7.54 (d; J=2.8Hz; 1H), 7.19 (dd; J1 = 8.8Hz; J2 = 2.8Hz; 1H), 6.84 (s; 1H), 6.80 (s; 1H), 6.81 (d; J = 7.6Hz; 2H); 13C NMR (75 MHz, DMSO-d6) δ 163.67, 158.81, 156.10, 143.04, 140.44, 131.00, 129.76, 125.47, 123.72, 121.00, 120.55, 108.36; IR (Nujol mull) v 3368, 3334, 1656, 1638, 1607, 1589, 1537 cm-1; Anal. Calcd for C14H11N2O3CI : C, 57.83; H, 3.79; N, 9.64. Found: C, 57.59; H, 4.01; N, 9.42.
[0068] In an embodiment, 3-(5-bromo-2-hydroxy-3-methoxyphenyl)-2-(4- iminiopyridin-1(4H)-yl)acrylate MCB-145 was synthesised. 4-Amino-1-
(cyanomethyl)pyridin-1-ium chloride 2a (163 mg; 0.959 mmol; 1.2 eq.) was added to a brown suspension of 5-bromo-3-methoxysalicylaldehyde 1 (190 mg; 0.799mm) in ethanol (0.2 mL) and aqueous solution of NaHCO3 0.01M (2mL). The reaction mixture was stirred at 110 °C and after 35 min the aldehyde was partially solubilized. An orange solid started to precipitate after 3 h and 40 min and was isolated by filtration. The brown solid was identified as 3-(5-bromo-2-hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin- 1(4H)-yl)acrylate MCB-145 (260 mg; 0.746 mmol). Brown solid; yield 90 %; mp 238-239 °C; 1H NMR (400 MHz, DMSO- d6) δ 13.24 (brs; 1H; OH), 8.20 (d; J = 8.00Hz; 2H), 7.18 (d; J = 2.4Hz; 1H), 7.01 (d; J = 2.4Hz; 1H), 6.85 (s; 1H), 6.80 (d; J = 8.00Hz), 3.76 (s; 3H); 13C NMR (75 MHz, DMSO-d6) δ 163.85, 158.81, 151.33, 146.83, 143.04, 140.04, 126.48, 125.37, 123.96, 114.97, 108.81, 108.50, 55.98; IR (Nujol mull) v 3370, 3149, 1656, 1599, 1537, 1465 cm-1; Anal. Calcd for C15H13N2O4 Br: C, 49.33; H, 3.60; N, 7.67. Found: C, 49.48;
H, 3.65; N, 7.73.
[0069] In an embodiment, 3-(5-bromo-2-hydroxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-122 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (92.4 mg; 0.392 mmol;
1.1 eq.) was added to a white suspension of 5-bromosalicylaldehyde 1 (59.6 mg; 0.700 mmol) in ethanol (0.2 mL) and in an aqueous solution of NaHCO30.01M (2 mL). The white suspension was initially stirred at room temperature for 21 h and then heated in a water bath at 40 °C, precipitating solid after 7.5 h. The starting materials were absent after 59 hours, as shown byTLC analysis. The white solid was filtered, washed with water and identified as 3-(5-bromo-2-hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1- ium-1-yl)acrylate MCA-122 (122 mg; 0.301 mmol). White solid; yield 79%; mp 178-179 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.23 (brs; 1H; OH), 11.41 (brs; 1H; NH), 8.80 (d; J = 7.8Hz; 2H), 7.88 (d; J = 7.8Hz; 2H), 7.80 (s; 1H), 7.67 (d; J = 2.4Hz; 1H), 7.35 (dd; J1 = 8.8Hz; J2 = 2.4Hz; 1H), 7.04 (s; 1H), 6.80 (d; J = 8.8Hz; 1H), 4.26 (q; J = 7.2Hz; 2H), 1.29 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 162.84, 156.80, 152.97, 152.79, 145.43, 140.20, 134.41, 133.18, 128.13, 123.78, 121.21, 112.79, 109.43, 62.16, 14.09; IR (Nujol mull) v 3139, 3084, 1742, 1624, 1606, 1533, 1513 cm-1; Anal. Calcd for C17H15N2O5Br: C, 50.12;
H, 3.69; N, 6.88. Found: C, 50.33; H, 3.88; N, 6.88.
[0070] In an embodiment, 3-(5-chloro-2-hydroxy-3-methoxyphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-125 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (101 mg; 0.419 mmol;
1.1 eq.) was added to a beige suspension of 5-chloro-3-methoxysalicylaldehyde 1 (73.2 mg; 3.81 mmol) in an aqueous solution of NaHCO30.01M (2 mL) and ethanol (0.2 mL). The suspension was stirred in a water bath at 40° C for 1 hour, leading to a beige solution. The reaction was followed by TLC and after 16 h, the aldehyde was no longer present. The solid was filtered, washed with water and identified as a mixture of MCA- 125 and the starting material 2b (112 mg; 286 mmol). Water (20 mL) was added to the solid mixture and vigorously stirred at room temperature for 2 h. The solid was filtered, washed with water and identified as 3-(5-chloro-2-hydroxy-3-methoxyphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-125 (120 mg; 0.306 mmol). White solid; yield 80%; mp 201-202 °C; 1H NMR (400 MHz, DMSO-d6) δ 11.32 (brs; 1H, NH), 8.77 (d; J = 7.2Hz; 2H), 7.86 (d; J = 7.2Hz; 2H), 7.07 (d; J = 2.4Hz; 1H), 7.04 (s; 1H), 6,95 (d; J = 2.4Hz; 1H), 4.24 (q; J = 7.6Hz; 2H), 3.77 (s; 3H), 1.29 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 163.08, 153.24, 153.01, 151.20, 146.65, 145.44 (2C), 140.03, 128.79, 122.74, 122.61, 121.49, 112.86, 112.75, 62.15, 56.01, 14.13; IR (Nujol mull) v 3134, 3069, 1753, 1656, 1600, 1532 cm-1; Anal. Calcd for C18H17N2O6CI: C, 55.04; H, 4.36; N, 7.13. Found: C, 55.12; H, 4.39; N, 7.17.
[0071] In an embodiment, 2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)-3-(2- hydroxyphenyl)acrylate MCA-64 was synthesised. 1-(Cyanomethyl)-4- (ethoxycarbonylamino)pyridinium chloride 2b (143 mg; 0.590 mmol; 1.6 eq.) was added to a solution of salicylaldehyde 1 (45.5 mg; 39.2 μL; 0.369 mmol) in water (2.5 mL). The yellow solution was stirred at 80 °C and after 2 h and 45 min, a yellow solid precipitated from the reaction mixture. After 6 h 30 min, the reaction was complete, by TLC analysis. The beige solid was filtered and washed with water and identified as 2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)-3-(2-hydroxyphenyl)acrylate MCA-64 (119 mg; 0.361 mmol). Beige solid; yield 98%; mp 185-186 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.15 (brs; 1H; OH), 11.40 (brs; 1H; NH), 8.78 (d; J = 7.6Hz; 2H), 7.88 (d; J = 7.6Hz; 2H), 7.39 (dd; J1 = 7.6Hz, J2 = 1.2Hz; 1H), 7.23 (dt; J1 = 8.4Hz; J2 = 1.6Hz; 1H), 7.09 (s; 1H), 6.84 (t; J = 8.4Hz; 1H), 6.80 (dd; J1 = 1.2Hz; 1H), 4.26 (q; J = 7.2Hz; 2H), 1.28 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 163.33, 162.17, 157.57, 152.81 (2C), 145.66 (2C), 139.24, 133.03, 131.01, 130.54, 121.67, 119.46, 118.63, 112.78 (2C), 14.12; IR (Nujol mull) v 3124, 1773, 1659, 1635, 1615, 1581, 1534, 1461 cm-1; Anal. Calcd for C17H16N2O5: C, 62.19; H, 4.91; N, 8.53. Found: C, 62.19; H, 4.92; N, 8.53. [0072] In an embodiment, 3-(2,3-dihydroxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-204 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (122 mg; 0.503 mmol; 1 eq.) was added to a solution of 3-hydroxysalicylaldehyde 1 (46.4 mg; 0.336 mmol) in water (7 mL) resulting in a yellow solution. After 20 minutes under reflux, a yellow solid precipitated from the reaction mixture. After 3h and 30 min, the reaction was complete (by TLC) and the solid was filtered and washed with water. The product (61.1 mg; 0.177 mmol) was identified as 3-(2,3-dihydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin- 1-ium-1-yl)acrylate MCA-204. Yellow solid; yield 53%; mp 247-248 °C; 1H NMR (400 MHz, DMSO-d6) δ 14.02 (brs; 1H; OH), 11.13 (brs; 1H; NH), 8.75 (d; J = 7.2Hz; 2H), 7.85 (d; J = 7.2Hz; 2H), 7.13 (s; 1H), 6.75 (dd; J1 = 5.6Hz; J2 = 1.6Hz; 1H), 6.72 (dd; J1 = 6.8Hz; J2 = 1.6Hz; 1H), 6.64 (t; J = 7.6Hz; 1H), 4.25 (q; J = 6.8Hz; 2H), 1.29 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO- d6) δ163.53, 153.21, 153.12,148.49, 146.29, 145.82 (2C), 138.42, 132.52, 123.21, 121.69, 118.48, 115.56, 113. 17 (2C), 62.05, 14.12; IR (Nujol mull) v 3130, 3069, 1738, 1645, 1527 cm-1; Anal. Calcd for C17H16N2O6: C, 59.30; H, 4.68; N, 8.14. Found: C, 59.41; H, 4.70; N, 8.16.
[0073] In an embodiment, 3-(5-chloro-2-hydroxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-103 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (98.8 mg; 0.409 mmol; 1 eq.) was added to a suspension of 5-chlorosalicylaldehyde 1 (58.2 mg; 0.372 mmol) in an aqueous solution of NaHC030.01M ( 3 mL). The suspension was stirred at 110 °C and after 15 min, a yellow solid precipitated from the reaction mixture. After 3 h and 15 minutes the solid was filtered, washed with water and identified by 1H NMR as product MCA-103 slightly contaminated (125 mg; 344 mmol). Water (20 mL) was added to the isolated solid and the suspension was vigorously stirred at room temperature for 2 h. The product was filtered, washed with water and identified as 3-(5-chloro-2- hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-103 (109 mg; 0.300 mmol). Yellow solid; yield 73%; mp 190-191 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.11 (s; 1H; OH), 11.90 (s; 1H; NH), 8.81 (d; J = 7.2Hz; 2H), 7.88 (d; J = 7.2Hz; 2H), 7.56 (d; J=2.4Hz; 1H), 7.30 (dd; Ji = 8.7Hz; J2 = 2.7Hz; 1H), 7.04 (s; 1H), 6.86 (d; J = 8.7Hz; 1H), 4.26 (q; J = 7.2Hz; 2H), 1.29 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 162.90, 156.33, 154.98, 152.81, 145.94 (2C), 140.33, 131.97, 130.40, 128.00, 123.20, 121.95, 120.70, 112.84 (2C), 62.20, 14.13; IR (Nujol mull) v 3128, 3069, 1743, 1703, 1652, 1637, 1606, 1590, 1525 cm-1; Anal. Calcd for C17H15N2O5CI: C, 56.27; H, 4.14; N, 7.72. Found: C, 56.03; H, 4.36; N, 8.00.
[0074] In an embodiment, 3-(2-hydroxy-5-methylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-335 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (0.14 g; 0.59 mmol; 1.6 eq.) was added to a solution of 5-methylsalicylaldehyde 1 (0.05 g; 0.38 mmol) in an aqueous solution of NaHCO3 0.1M (2 mL) and heated at 80 °C. After 15 minutes, a beige solid precipitated from the reaction mixture. After stirring for a further 15 minutes the reaction was complete (by TLC) and the solid was filtered and washed with water. The product (0.09 g; 0.28 mmol) was identified as 3-(2-hydroxy-5-methylphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-335. Yellow solid; yield 75%; mp 180-182 °C; 1H NMR (400 MHz, DMSO- d6) δ 12.93 (s, 1H, OH), 11.34 (s, 1H, NH); 8.77 (d, J = 7,6Hz, 2H), 7.87 (d, J = 7,2Hz, 2H), 7.16 (d, J = 2Hz, 1H), 7.06 (s, 1H), 7.04 (dd, J = 6Hz, J = 2Hz, 1H), 6.74 (d, J = 8Hz, 1H), 2.19 (s, 3H), 4.26 (q, J = 7,2Hz, 2H), 1.29 (t, J = 7,2Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ 163.30, 155.28, 152.86, 145.66, 139.25, 132.94, 131.74, 130.76, 126.98, 121.43, 119.47, 112.76, 62.15, 19.96, 14.12; IR (Nujol mull) v 3131, 1737, 1653, 1636, 1526 cm-1; Anal. Calcd for C18H18N2O5: C, 63.15; H, 5.30; N, 8.18. Found: C, 63.27; H, 5.42; N, 8.10.
[0075] In an embodiment, 3-(2-hydroxy-4-methoxylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-360 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (0.13 g; 0.53 mmol; 1.6 eq.) was added to a solution of 4-methoxylsalicylaldehyde 1 (0.05 g; 0.33 mmol) in an aqueous solution of NaHCO3 0.1M (3 mL) and heated at 90 °C. After 25 minutes, a beige solid precipitated from the reaction mixture. After stirring for a further 30 minutes, the reaction was complete (by TLC) and the solid was filtered and washed with water. The product (0.05 g; 0.14 mmol) was identified as 3-(2-hydroxy-4-methoxylphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-360. Yellow solid; yield 43%; mp 202-204 °C; 1H NMR (400 MHz, DMSO-d6) δ 14.15 (s, 1H, OH), 11.34 (s, 1H, NH), 8.74 (d, J = 7,2Hz, 2H), 7.85 (d, J = 7,6Hz, 2H), 7,25 (d, J = 8,8Hz, 1H), 7.05 (s, 1H), 6.42 (dd, J = 6Hz, J=2,8Hz, 1H), 6.37 (d, J = 2,8Hz, 1H), 4.26 (q, J = 7,2Hz, 2H), 3.73 (s, 3H), 1.29
(t, J = 6,8Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ163.68, 162.28, 160.27, 152.90,
152.72, 146.05, 136.68, 135.33, 132.43,114.34, 112.64, 106.03, 103.76, 62.10, 55.07, 14.11; IR (Nujol mull) v 1732, 1645, 1526 cm-1; Anal. Calcd for C18H18N2O6: C, 60.33; H, 5.06; N, 7.82. Found: C, 60.35; H, 5.10; N, 7.84.
[0076] In an embodiment, 3-(3,5-difluoro-2-hydroxylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-192 was synthesised. 1- (Cyanomethyl)-4-(ethoxycarbonylamino)pyridinium chloride 2b (0.24 g; 0.97 mmol; 1.2 eq.) was added to a solution of 3,5-difluorosalicylaldehyde 1 (0.13 g; 0.81 mmol) in an aqueous solution of NaHCO3 0.1M (3 mL) and ethanol (0.2 mL), heated at 100 °C. After 45 minutes, a beige solid precipitated from the reaction mixture. After stirring for a further 3 h and 15 minutes the reaction was complete (by TLC) and the solid was filtered and washed with water. The product (0.22 g; 0.61 mmol) was identified as 3-(3,5- difluoro-2-hydroxylphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-192. Yellow solid; yield 76%; mp 179-180 °C; 1H NMR (400 MHz, DMSO-d6) δ 14.15 (s, 1H, OH), 11.43 (s, 1H, NH), 8.82 (d, J = 7.2Hz, 2H), 7.88 (d, J = 6.8Hz, 2H), 7.16 (s, 1H), 4.26 (q, J = 7.2Hz, 2H), 1.29 (t, J = 7.2Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ 162.98, 145.58 (2C), 129.51, 113.13, 112.89 (2C), 105.80, 62.14, 14.13; IR (Nujol mull) v 3211, 3127, 3066, 1753, 1640, 1613, 1596, 1533 cm-1; Anal. Calcd for C17H14N2O5F2: C, 56.05; H, 3.87; N, 7.69. Found: C, 56.07; H, 3.87; N, 7.71.
[0077] In an embodiment, 3-(3,5-difluoro-2-hydroxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-156 was synthesised. 4- Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.24 g; 0.97 mmol; 1.2 eq.) was added to a solution of 3,5-difluorosalicylaldehyde 1 (0.13 g; 0.81 mmol) in an aqueous solution of NaHCO30.01M (3 mL) and ethanol (0.2 mL) and heated at 110 °C. After 45 minutes, a beige solid started to precipitate from the reaction mixture. After 3h and 15 min the reaction was complete (by TLC) and the solid was filtered and washed with water. The product (0.22 g; 0.61 mmol) was identified as 3-(3,5-difluoro-2-hydroxyphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-156. Beige solid; yield 76%; mp 210-211 °C; 1H NMR (400 MHz, DMSO-d6) δ 14.14 (s, 1H, OH), 8.23 (d, J = 7.2Hz, 2H), 8.19 (s, 2H)6.95 (s, 1H), 6.80 (d, J = 7.6Hz, 2H); 13C NMR (75 MHz, DMSO-d6) δ 163.23, 158.59, 158.16 (d, J = 43.0Hz), 143.94 (d, J = 4.0Hz), 142.70, 140.12, 125.75, 111.80 (d, J = 21.0Hz), 108.10, 103.88 (d, J = 25.0Hz), Cy and Cy (not observed); IR (Nujol mull) v 3243, 3178, 3087, 1681, 1651, 1638, 1608, 1535, 1514 cm-1; Anal. Calcd for C14H10N2O3F2: C, 57.54; H, 3.45; N, 9.59. Found: C, 57.55; H, 3.49; N, 9.68.
[0078] In an embodiment, 3-(2-hydroxy-5-methoxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-193 was synthesised. 4- Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.16 g; 0.96 mmol; 1.6 eq.) was added to a solution of 5-methoxysalicylaldehyde 1 (0.09 g; 0.60 mmol) in an aqueous solution of NaHCO3 0.01M (3 mL) and ethanol (0.3 mL) and heated at 110 °C. After 1 h, a green solid started to precipitate from the reaction mixture. After stirring for a further 2h, the reaction was complete (by TLC). The solid was filtered, washed with water and identified as 3-(2-hydroxy-5-methoxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1- yl)acrylate MCB-193 (0.13 g; 0.44 mmol). Greenish solid; yield 74%; mp 206-207 °C; 1H NMR (400 MHz, DMSO-d6) δ 12,46 (brs; 1H; OH), 8.21 (d, J = 7.6Hz, 2H), 6.79 (d, J = 7.6Hz, 2H), 6.98 (d, J = 2.8Hz, 1H), 6.81 (dd, J = 1.6Hz, J = 1.2Hz, 1H), 6.75 (d, J = 9.2Hz, 1H), 6.84 (s; 1H), 3.66 (s; 3H); 13C NMR (75 MHz, DMSO-d6) δ 163.95, 158.65, 151.56, 150.95, 143.40, 139.91, 127.58, 122.42, 119.95, 116.19, 115.83, 108.29, 55.35; IR (Nujol mull) v 3291, 3080, 3052, 1672, 1650, 1598, 1502 cm-1; Anal. Calcd for C15H14N2O4: C, 62.93; H, 4.93; N, 9.79. Found: C, 62.93; H, 4.96; N, 9.77.
[0079] In an embodiment, 3-(5-bromo-2-hydroxyphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-147 was synthesised. 4- Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.16 g; 0.96 mmol; 1.2 eq.) was added to a solution of 5-bromosalicylaldehyde 1 (0.16 g; 0.80 mmol) in an aqueous solution of NaHCO30.01M (3 mL) and ethanol (0.2 mL) and heated at 110 °C. After 30 min, a yellow solid started to precipitate from the reaction mixture. After stirring for a further 3.5 h, the reaction was complete (by TLC). The solid was filtered, washed with water and identified as 3-(5-bromo-2-hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium- 1-yl)acrylate MCB-147 (0.24 g; 0.66 mmol). Green solid; yield 82%; mp 220-221 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.29 (s, 1H, OH), 8.22 (d, J = 7.6Hz, 2H), 8.16 (s, 2H), 7.61 (d, J = 2.4Hz, 1H), 7.31 (dd, J = 6.4Hz, J = 2,4Hz, 1H), 6.83 (s, 1H), 6,79 (d, J = 7.6Hz, 2H), 6.76 (s, 1H); 13C NMR (75 MHz, DMSO-d6) δ 163.66, 158.73, 156.60, 143.25 (2C), 140.34, 133.99, 133.26, 132.63, 125.74, 124.41, 121.19, 108.36 (2C); IR (Nujol mull) v 3393, 3322, 3292, 3145, 3087, 3060, 1654, 1637, 1605, 1589, 1536, 1509 cm-1; Anal. Calcd for C14H11N2O3Br: C, 50.17; H, 3.31; N, 8.36. Found: C, 50.20; H, 3.41; N, 8.42.
[0080] In an embodiment, 3-(2-hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1- ium-1-yl)acrylate MCB-314 was synthesised. 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.031 g; 0.18 mmol; 1.6 eq.) was added to a solution of salicylaldehyde 1 (0.01 g; 0.12 mmol; 12 mΐ) in an aqueous solution of NaHC03 0.01M (1 mL) and heated at 80 °C. After 1 h and 30 min, a beige solid started to precipitate from the reaction mixture. After stirring for a further 2.5 h, the reaction was complete (by TLC). The solid was filtered, washed with water and identified as 3-(2-hydroxyphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-314 (0.03 g; 0.11 mmol). White solid; yield 97%; mp 208-210 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.16 (s, 1H, OH), 8.19 (d, J = 7,6Hz, 2H), 8.15 (s, 2H), 6.88 (s, 1H); 13C NMR (75 MHz, DMSO-d6) δ 164.10, 158.67, 157.30, 143.47, 139.34, 132.54, 130.42, 128.30, 122.18, 119.34, 118.51, 108.31; IR (Nujol mull) v 1657, 1580, 1531 cm-1; Anal. Calcd for C14H12N2O3: C, 65.62; H, 4.72; N, 10.93. Found: C, 65.65; H, 4.76; N, 10.94.
[0081] In an embodiment, 3-(2-hydroxy-3-methylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-324 was synthesised. 4- Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.10 g; 0.59 mmol; 1.6 eq.) was added to a solution of 3-methylsalicylaldehyde 1 (0.05 g; 0.37 mmol; 45 mί) in an aqueous solution of NaHC030.01M (2 mL) and heated at 80 °C. After 55 min, a green solid started to precipitate and the reaction was complete by TLC. The solid was filtered, washed with water and identified as 3-(2-hydroxy-3-methylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-324 (0.10 g; 0.36 mmol). Green solid; yield 98%; mp 203-205 °C; 1H NMR (400 MHz, DMSO-d6) δ 13.71 (s, 1H, OH), 8.18 (d, J = 7.6Hz, 2H), 8.13 (s, 2H), 6.94 (s, 1H), 6.78 (d, J = 7.6Hz, 2H), 6.70 (t, J = 7.6Hz, 1H); 13C NMR (75 MHz, DMSO-d6) δ 164.30, 158.68, 155.94, 143.86, 138.60, 131.33, 131.27, 130.75, 128.08, 121.85, 118.10, 108.24, 16.94; IR (Nujol mull) v 3130, 3069, 1738, 1645, 1527 cm-1 Anal. Calcd for C15H14N2O3: C, 66.66; H, 5.22; N, 10.36. Found: C, 66.71; H, 5.34; N, 10.27.
[0082] In an embodiment, 3-(2-hydroxy-5-methylphenyl)-2-(4-
((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-325 was synthesised. 4- Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.10 g; 0.59 mmol; 1.6 eq.) was added to a solution of 5-methylsalicylaldehyde 1 (0.05 g; 0.37 mmol) in an aqueous solution of NaHCO30,01M (2 mL) and heated at 80 °C. After 3h and 50 min, a green solid started to precipitate and after 20 more minutes, the reaction was complete by TLC. The solid was filtered, washed with water and identified as 3-(2-hydroxy-5-methylphenyl)-2-(4- ((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-325 (0.08 g; 0.28 mmol). White solid; yield 76%; mp 204-206 °C; 3H NMR (400 MHz, DMSO-d6) δ 12.88 (s, 1H, OH), 8.19 (d, J = 7.6Hz, 2H), 8.14 (s, 2H), 7.13 (d, J = 2Hz, 1H), 6.99 (dd, J = 6Hz, J = 2Hz, 1H), 6.83 (s, 1H), 6.79 (d, J = 7.6Hz, 2H), 6.71 (d, J = 8Hz, 1H), 2.17 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ164.10, 158.65, 154.98, 143.48 (2C), 139.36, 132.48, 131.12, 128.47, 126.85, 121.89, 119.27, 108.29 (2C), 19.99; IR (Nujol mull) v 3130, 3069, 1738, 1645, 1527 cm- 3; Anal. Calcd for C15H14N2O3: C, 66.66; H, 5.22; N, 10.36. Found: C, 66.67; H, 5.26; N, 10.31.
[0083] In an embodiment, 4-(2-cyano-2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MC- 317 was synthesised. 4-Amino-1-(cyanomethyl)pyridin-1-ium chloride 2a (0.18 g; 1.04 mmol; 1.2 eq.) was added to a solution of 4-hydroxybenzaldehyde 1 (0.11 g; 0.89 mmol) in an aqueous solution of NaHCO30,05M (1 mL) and propylamine (54.6 mg; 0.92 mmol; 76mί). After stirring at room temperature for 4 h, an orange solid started to precipitate and after 30 more minutes, the solid was filtered and washed with water (18.4 mg). The mother-liquor was further stirred at room temperature for 15 h and a second crop of an orange solid was filtered and washed with water (18.0 mg). The two crops were identified as 4-(2-cyano-2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MC-317 (36.4 mg; 0.153 mmol). Orange solid; yield 17%; mp 192-194 °C; 3H NMR (400 MHz, DMSO-d6) δ 8.30-9.50 (brs; 2H), 8.26 (d; J = 7.2Hz; 2H), 7.44 (d; J = 8.8Hz; 2H), 7.30 (s; 1H), 6.83 (d; J = 7.2Hz; 2H), 6.12 (d; J = 9.2Hz; 2H); 13C NMR (75 MHz, DMSO-d6) δ 178.41, 158.77, 144.18, 143.04 (2C), 133.41 (2C), 120.78 (2C), 118.70, 109.73 (2C), 109.12, 95.47; IR (Nujol mull) v 3160, 3099, 2236, 1712, 1641, 1515 cm-1; Anal. Calcd for C14H11N3O: C, 70.87; H, 4.67; N, 17.71. Found: C, 70.91; H, 4.65; N, 17.68.
[0084] In an embodiment, a solution of 3-(2-hydroxyphenyl)-2-pyridyl)acrylate (0.02- 0.14 g), in DMSO (0.2-0.7 mL) was stirred at 100-130 °C for 1 h and 10 min to 2 h. A solid precipitated from the reaction mixture by the addition of water and the product was filtered and washed with water leading to the pure styrylpyridine derivative (60-89%).
[0085] In an embodiment, ethyl (1-(2-hydroxystyryl)pyridin-4(lH)-ylidene)carbamate MCA- 189 was synthesised. A yellow suspension of 2-(4-((ethoxycarbonyl)amino)pyridin- l-ium-1-yl)-3-(2-hydroxyphenyl)acrylate MCA-64 (96.9 mg; 0.263 mmol) in DMSO (0.2 mL) was stirred at 110 °C and a red solution was obtained 15 minutes later. The reaction was complete after 1 h and 45 min (by TLC). An orange suspension was formed after addition of water (1 mL) and the solid was filtered and washed with water. The product was identified as ethyl (1-(2-hydroxystyryl)pyridin-4(lH)-ylidene)carbamate MCA-189 (64.0 mg; 0.224 mmol). Orange solid; yield 89%; mp 176-177 °C; 1H NMR (400 MHz, DMSO-d6) δ 8.20 (d; J = 7.8Hz; 2H),7.70 (d; J = 14.4Hz), 7.15 (d; J = 1.5Hz; 1H), 7.10 (d; J = 9.9Hz; 1H), 7.06 (d; J = 7.8Hz; 2H),6.88 (dd; J1 = 7.8Hz; J2 = 0.9Hz; 1H), 6.82 (dt; Ji = 7.2Hz; J2 = 0.9Hz), 3.98 (q, J = 6.9Hz; 2H), 3.83 (s; 3H), 1.16 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 164.22, 162.47, 155.38, 137.16 (2C), 129.46, 129. 25, 127.86, 120.82, 119.28, 115.89, 115.77, 115.56 (2C), 54.53, 14.68; IR (Nujol mull) v l738, 1665, 1614, 1510 cm-1; Anal. Calcd for C16H16N2O3: C, 67.59; H, 5.67; N, 9.85. Found: C, 67.60; H, 5.66; N, 9.95.
[0086] In an embodiment, ethyl 4-chloro-2-(2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MCA-206 was synthesised. A suspension of 3-(5-chloro-2-hydroxyphenyl)-2-(4- iminiopyridin-1(4H)-yl)acrylate MCA-201 (88.9 mg; 0.319 mmol) in DMSO (0.2) was stirred at 130 °C. After 35 min the starting material was partially solubilized and after a further hour, the brown solid was isolated by addition of water (1.5 mL) and identified as 4-chloro-2-(2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MCA-206 (59.1 mg; 0.256 mmol). Brown solid; yield 80%; mp 197-198 °C; 1H NMR (400 MHz, DMSO-d6) δ 38.19 (d; J = 7.8Hz; 2H), 7.79 (d; J= 14.7Hz; 1H), 7.60 (d; J = 2.4Hz; 1H), 7.24 (dd; J1 = 8.4Hz; J2 = 2.4Hz; 1H), 7.07 (d; J = 7.8Hz; 2H), 7.02 (s; 1H), 6.80 (d; J = 8.7Hz; 1H), 4.00 (q; J = 6.9Hz; 2H), 1.17 (t; J = 7.2Hz); 13C NMR (75 MHz, DMSO-d6) δ 163.70, 162.13, 155.25, 137.11 (2C), 131.34, 130.30, 129.51, 123.43, 118.06, 115.40 (2C), 114.08, 109,89, 59.62, 14.59; IR (Nujol mull) v3150, 3069, 1656, 1605, 1589, 1537 cm-1; Anal. Calcd for C13H11N2OCI: C, 63.04; H, 4.88; N, 11.31. Found: C, 63.09; H, 4.93; N, 11.45.
[0087] In an embodiment, ethyl (1-(5-bromo-2-hydroxystyryl)pyridin-4(1H)- ylidene)carbamate MCA-116 was synthesised. A suspension of 3-(5-bromo-2- hydroxyphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCA-122 (115 mg; 0.283 mg) in DMSO (0.2 mL) was stirred at 130 °C. The starting material was solubilized 10 minutes later and after 1 h and 50 min, TLC confirmed the absence of starting material and the addition of water (1mL) led to an orange solid. The solid was filtered, washed with water and identified as ethyl (1-(5-bromo-2-hydroxystyryl)pyridin- 4(lH)-ylidene)carbamate MCA-116 (82.8 mg; 0.228 mmol). Orange solid; yield 81%; mp 212-213 °C; 1H NMR (400 MHz, DMSO-d6) δ 8.18 (d; J = 7.6Hz; 2H), 7.72 (d; J = 14.4Hz; 1H), 7.15 (d; J = 2.4Hz; 1H), 7.07 (d; J = 14.4Hz; 1H), 7.04 (s; 2H), 6.95 (d; J = 2Hz; 1H), 4.00 (q; J = 7.2Hz; 2H), 3.83 (s; 3H), 1.17 (t; J = 6,8Hz); 13C NMR (75 MHz, DMSO-d6) δ 163.72, 162.18, 148.78, 143.43, 137.04 (2C), 130.38, 122.67, 122.33, 117.64, 115.38 (2C), 113.27, 111.28, 59.60, 54.24, 14.57; IR (Nujol mull) vl742, 1683, 1623, 1514 cm-1; Anal. Calcd for C16H15N2O3Br: C, 52.91; H, 4.16; N, 7.71. Found: C, 53.00; H, 4.21; N, 7.79.
[0088] In an embodiment, 4-bromo-2-(2-(4-iminiopyridin-1(4H)-yl)vinyl)-6- methoxyphenolate MCA-205 was synthesised. A yellow suspension of 3-(5-bromo-2- hydroxy-3-methoxyphenyl)-2-(4-iminiopyridin-1(4H)-yl)acrylate MCB-145 (137 mg; 0.393 mmol) in DMSO (0.7 mL) was stirred at 130 °C, leading to a red solution 20 minutes later. After 50 min the reaction was complete (by TLC). Addition of water (1 mL) led to an orange solid that was filtered and washed with water. The product was identified as 4-bromo-2-(2-(4-iminiopyridin-1(4H)-yl)vinyl)-6-methoxyphenolate MCA-205 (112 mg; 0.368 mmol). Orange solid; yield 60%; mp 200-201 °C; 1H NMR (400 MHz, DMSO-d6) δ 8.35 (d; J = 6.6Hz; 2H), 8.18 (d; J =14.1Hz; 1H), 7.11 (d; J = 13.8Hz; 1H) 6.93 (d; J = 2.4Hz; 1H), 6.87 (d; J = 6.6Hz; 2H), 6.63 (d; J = 2.4Hz; 1H), 3.53 (s; 3H); 13C NMR (75 MHz, DMSO-d6) δ 158.00, 156.51, 152.03, 149.44, 139.30 (2C), 126.06, 122.82, 121.15, 113.60, 109.65 (2C), 100.32, 55.62; IR (Nujol mull) v3269, 3053, 1645, 1533 cm-1; Anal. Calcd for C14H11N2O2Br: C, 51.56; H, 3.40; N, 8.59. Found: C, 51.59; H, 3.40; N, 8.64.
[0089] In an embodiment, 2,4-difluoro-6-(2-(4-iminiopyridin-1(4H)-yl)vinyl)phenolate MCA-249 was synthesised. A suspension of 3-(3,5-difluoro-2-hydroxyphenyl)-2-(4- iminiopyridin-1(4H)-yl)acrylate MCB-156 (0.03 g; 0.10 mmol) in DMSO (0.2 mL) and water (6.0 mί) was stirred at 130 °C. After 1 h and 15 min the reaction was complete (by TLC). Addition of water (2 mL) led to an orange solid that was filtered and washed with water. The product was identified as 2,4-difluoro-6-(2-(4-iminiopyridin-1(4H)- yl)vinyl)phenolate MCA-249 (0.02 g; 0.07 mmol). Orange solid; yield 75%; mp 210-211 °C; 1H NMR (400 MHz, DMSO-d6) δ 8.81 (brs, 2H), 8.35 (brs, 2H), 8.25 (d; J = 14.0Hz; 1H), 7.17 (d; J = 14.0Hz; 1H), 6.72-6.94 (m; 2H), 6.86 (brs; 2H); 13C NMR (75 MHz, DMSO-d6) d 158.65, 153.79 (dd, J = 224.0Hz, J = 22.0Hz), 152.63 (d, J = 13.0Hz), 148.21 (dd, J = 212.0Hz, J = 13.0Hz), 139.3, 127.06, 121.30, 109.65, 108.30 (dd, J = 18.0Hz, J = 2.0Hz), 103.10 (dd, J = 24.0Hz, J = 3.0Hz; IR (Nujol mull) v 1653, 1647, 1540 cm-1; Anal. Calcd for C13H10N2OF2: C, 62.90; H, 4.06; N, 11.29. Found: C, 62.92; H, 4.04; N, 11.31.
[0090] In an embodiment, ethyl (1-(3,5-difluoro-2-hydroxystyryl)pyridin-4(lH)- ylidene)carbamate MCB-376 was synthesised. A yellow suspension of 3-(3,5-difluoro-2- hydroxylphenyl)-2-(4-((ethoxycarbonyl)amino)pyridin-1-ium-1-yl)acrylate MCB-192 (0.02 g; 0.06 mmol) in DMSO (0.2 mL) was stirred at 100 °C. After 30 min the reaction temperature was raised to 130 °C and maintained for a further hour. An orange suspension was formed after addition of water (1 mL) and the solid was filtered and washed with water, leading to ethyl (1-(3,5-difluoro-2-hydroxystyryl)pyridin-4(lH)- ylidene)carbamate MCB-376 (11.0 mg; 0.033 mmol). Orange solid; yield 53%; mp 170- 171 °C; 1H NMR (400 MHz, DMSO-d6) δ 8.25 (d; J = 7.6Hz; 2H), 7.85 (d, J = 14.4Hz, 1H), 7.09-7.18 (m, 5H), 4.03 (q, J = 7.2Hz, 2H), 1.19 (t, J = 7.2Hz, 3H); 13C NMR (75 MHz, DMSO- de) d 162.57, 161.23, 150.48, 137.50, 130.91, 124.26 (d, J = 11.0Hz), 113.82, 108.06 (d, J = 26.0Hz), 103.51 (d, J = 27.0Hz), 59.94, 14.53; IR (Nujol mull) v 1733, 1682 cm-1; Anal. Calcd for C16H14N2O3F2: C, 60.00; H, 4.41; N, 8.75. Found: C, 60.10; H, 4.42; N, 8.71. [0091] All references recited in this document are incorporated herein in their entirety by reference, as if each and every reference had been incorporated by reference individually.
[0092] Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and/or the understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise. It is also to be understood that unless otherwise indicated or otherwise evident from the context and/or the understanding of one of ordinary skill in the art, values expressed as ranges can assume any subrange within the given range, wherein the endpoints of the subrange are expressed to the same degree of accuracy as the tenth of the unit of the lower limit of the range.
[0093] In addition, it is to be understood that any particular embodiment of the present invention may be explicitly excluded from any one or more of the claims. Where ranges are given, any value within the range may explicitly be excluded from any one or more of the claims. Any embodiment, element, feature, application, or aspect of the compositions and/or methods of the invention, can be excluded from any one or more claims.
[0094] Antimalarial and other in vitro assays were funded by Fundação para a Ciencia e Tecnologia (FCT) trough projects PTDC/SAU-PAR/28066/2017, UIDB/50026/2020, Ul DP/50026/2020 and grants SFRH/BPD/76614/2011 and PD/BD/127826/2016; by the projects NORTE-01-0145-FEDER-000013 and NORTE-01-0145-FEDER-000023, supported by Norte Portugal Regional Operational Programme (NORTE2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF). Also trough the Merieux Research Grant.
[0095] Synthesis and spectroscopic characterization of the compounds were funded by The NMR spectrometer Bruker Avance III 400, part of the National NMR Network (RNRMN) and purchased within the framework of the National Program for Scientific Reequipment, contract REDE/1517/RMN/2005 with funds from POCI2010 (FEDER) and FCT; FEDERCOMPETE through Centro de Quimica (F-COMP-01-0124-FEDER-022716, PEst-C/QUI/U 10686/2011, U I D/QU 1/00686/2013 and UID/QU 1/0686/2016); FCT trough BPD grant (SFRH/BPD/79609/2011).
[0096] The present disclosure should not be seen in any way restricted to the embodiments described and a person with ordinary skill in the art will foresee many possibilities to modifications thereof.
[0097] The above described embodiments are combinable. The following claims further set out particular embodiments of the disclosure.

Claims

C L A I M S
1. A compound of the formula (I) or a pharmaceutically acceptable salt, solvate, N- oxide, stereoisomer, diastereoisomer, enantiomer, atropisomer or polymorph
Figure imgf000035_0001
wherein
R1, R2, R3 and R4are independently selected from each other;
R1 is selected from a list consisting of: H, alkyl, alkoxyl, halogen, hydroxyl, carbonyl;
R2 is selected from a list consisting of: H, hydroxyl, carbonyl;
R3 is selected from a list consisting of: H, hydroxyl, carbonyl, cyano, carboxyl;
R4 is selected from a list consisting of: amine, amide.
2. The compounds according to any of the previous claims wherein R1 is selected from a list consisting of: H, hydroxyl, carbonyl, fluoro, bromo, chloro, halo-methoxy, alkyl.
3. The compounds according to any of the previous claims wherein R1 is selected from a list consisting of: H, fluoro, bromo, chloro, halo-methoxy.
4. The compounds according to any of the previous claims wherein R2 is a hydroxyl.
5. The compounds according to any of the previous claims wherein R3 is selected from a list consisting of: H, cyano.
6. The compounds according to any of the previous claims R4 is selected from a list consisting of: heteroarylamine, amide, preferably heteroarylamine, heteroarylamide.
7. The compounds according to any of the previous claims wherein R4 is a pyridine- moiety.
8. The compounds according to the previous claim wherein the compound is selected from a list consisting of:
Figure imgf000036_0001
Figure imgf000037_0001
9. The compounds according to any of the previous claims wherein the salt is a hydrochloride salt.
10. The compounds according to any of the previous claims for use in medicine or veterinary.
11. The compounds according to any of the previous claims for use in the prevention, therapy, or treatment of infection.
12. The compounds according to any of the previous claims for use in the prevention, therapy, or treatment of parasite infection, preferably P. falciparum infection.
13. The compounds according to any of any one of the previous claims for use in the prevention, therapy or treatment of malaria diseases.
14. Pharmaceutical composition comprising a compound as defined in any one of claims 1-13 in a therapeutically effectively amount and a pharmaceutically acceptable excipient.
15. Pharmaceutical composition according to the previous claim wherein the pharmaceutically acceptable excipient is a carrier, adjuvant, excipient or mixtures thereof.
16. Pharmaceutical composition according to any of the previous claims 13-15 comprising a further anti-viral agent, preferably an anti-HIV agent or anti- tuberculosis agent.
17. Pharmaceutical composition according to any of the previous claims 13-16 comprising a second anti-malarial agent, preferably wherein the second anti- malarial agent is selected from the group consisting of: Chloroquine, Fansidar, Amodiaquine, Quinine, Halofantrine, Mefloquine, Artemether/Artesunate and Malarone.
PCT/IB2021/052585 2020-03-27 2021-03-29 Antimalarial agent, methods and uses thereof WO2021191882A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
PT116202 2020-03-27
PT11620220 2020-03-27
EP20166887 2020-03-30
EP20166887.8 2020-03-30
EP2020059155 2020-03-31
EPPCT/EP2020/059155 2020-03-31

Publications (1)

Publication Number Publication Date
WO2021191882A1 true WO2021191882A1 (en) 2021-09-30

Family

ID=75674880

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/052585 WO2021191882A1 (en) 2020-03-27 2021-03-29 Antimalarial agent, methods and uses thereof

Country Status (1)

Country Link
WO (1) WO2021191882A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2012978A1 (en) * 1968-07-15 1970-03-27 May & Baker Ltd Combined anthelmintics based on quaternary ammonium - salts
GB1221061A (en) * 1967-01-18 1971-02-03 May & Baker Ltd Quaternary ammonium derivatives, their use as anthelmintics and methods for their preparation
WO2004000783A1 (en) * 2002-06-24 2003-12-31 Merck & Co., Inc. Aminoalkylphenols, methods of using and making the same
WO2013063243A1 (en) * 2011-10-25 2013-05-02 New York University Small molecule malarial aldolase-trap enhancers and glideosome inhibitors
CN103992310A (en) * 2013-05-14 2014-08-20 中国医学科学院医药生物技术研究所 Group of substituted benzoheterocycle amine derivatives and preparation method and related application thereof as IMPDH (inosine monophosphate dehydrogenase) inhibitor
CN107318839A (en) * 2016-04-28 2017-11-07 闫合 A kind of bactericide and its application

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1221061A (en) * 1967-01-18 1971-02-03 May & Baker Ltd Quaternary ammonium derivatives, their use as anthelmintics and methods for their preparation
FR2012978A1 (en) * 1968-07-15 1970-03-27 May & Baker Ltd Combined anthelmintics based on quaternary ammonium - salts
WO2004000783A1 (en) * 2002-06-24 2003-12-31 Merck & Co., Inc. Aminoalkylphenols, methods of using and making the same
WO2013063243A1 (en) * 2011-10-25 2013-05-02 New York University Small molecule malarial aldolase-trap enhancers and glideosome inhibitors
CN103992310A (en) * 2013-05-14 2014-08-20 中国医学科学院医药生物技术研究所 Group of substituted benzoheterocycle amine derivatives and preparation method and related application thereof as IMPDH (inosine monophosphate dehydrogenase) inhibitor
CN107318839A (en) * 2016-04-28 2017-11-07 闫合 A kind of bactericide and its application

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
ARAI S ET AL: "SYNTHESIS AND REACTION OF METHYLBENZO[a]QUINOLIZINIUM SALTS [1]", JOURNAL OF HETEROCYCLIC CHEMISTRY, WILEY-BLACKWELL PUBLISHING, INC, US, vol. 27, no. 4, May 1990 (1990-05-01), pages 1073 - 1078, XP000652730, ISSN: 0022-152X, DOI: 10.1002/JHET.5570270448 *
CHENG PI ET AL: "Synthesis and biological evaluation ofN-acetyl-[beta]-aryl-1,2-didehydroethylamines as new HIV-1 RT inhibitors in vitro", BIORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 17, no. 16, 15 August 2007 (2007-08-15), pages 4476 - 4480, XP029094789, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2007.06.008 *
DUNCAN W G ET AL: "2-(omega-Aminoalkyl)-4-t-butyl-6-phenylphenols as Antimalarial Agents", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 12, no. 2, July 1969 (1969-07-01), pages 711/712, XP002094647, ISSN: 0022-2623, DOI: 10.1021/JM00304A044 *
GOULAOUIC-DUBOIS C ET AL: "Protection of amines by the pyridine-2-sulfonyl group and its cleavage under mild conditions (SmI2 or electrolysis)", JOURNAL OF ORGANIC CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, vol. 60, September 1995 (1995-09-01), pages 5969 - 5972, XP002364116, ISSN: 0022-3263, DOI: 10.1021/JO00123A041 *
MADDALI L. N. RAO ET AL: "Rh-Catalyzed Deformylative Coupling of Salicylaldehydes with Acrylates and Acrylamides", JOURNAL OF ORGANIC CHEMISTRY, vol. 84, no. 9, 9 April 2019 (2019-04-09), US, pages 5677 - 5683, XP055714953, ISSN: 0022-3263, DOI: 10.1021/acs.joc.9b00602 *
MCFARLAND JAMES W. ET AL: "Novel anthelmintic agents. III. 1-(2-Arylvinyl)pyridinium salts", JOURNAL OF MEDICINAL CHEMISTRY, vol. 12, no. 6, November 1969 (1969-11-01), US, pages 1079 - 1086, XP055804474, ISSN: 0022-2623, Retrieved from the Internet <URL:https://pubs.acs.org/doi/pdf/10.1021/jm00306a024> DOI: 10.1021/jm00306a024 *
O' CALLAGHAN ET AL: "Isomerisation of benzopyran-2-imines in [2H6]dimethyl sulfoxide", J. CHEM. SOC. PERKIN TRANS, February 1998 (1998-02-01), pages 425 - 429, XP055714981, Retrieved from the Internet <URL:https://pubs.rsc.org/en/content/articlepdf/1998/p2/a703747c> [retrieved on 20201109] *
PAUL BHASKAR ET AL: "Tandem Transformation of Nitro Compounds into N -Methylated Amines: Greener Strategy for the Utilization of Methanol as a Methylating Agent", CHEMSUSCHEM, vol. 10, no. 11, 18 May 2017 (2017-05-18), DE, pages 2370 - 2374, XP055804435, ISSN: 1864-5631, Retrieved from the Internet <URL:https://api.wiley.com/onlinelibrary/tdm/v1/articles/10.1002%2Fcssc.201700503> DOI: 10.1002/cssc.201700503 *
ROKHYATOU SECK ET AL: "Synthesis and Antimalarial Activity of Cinnamic Acid Derivatives", JOURNAL OF CHEMICAL AND PHARMACEUTICAL RESEARCH, October 2018 (2018-10-01), pages 1 - 8, XP055714959, Retrieved from the Internet <URL:http://www.jocpr.com/articles/synthesis-and-antimalarial-activity-of-cinnamic-acid-derivatives.pdf> [retrieved on 20200715] *
SANYASI SITHA ET AL: "Enhanced Nonlinear Optical Response in Zwitterionic Molecules: A Computational Study on the Role of Orbital Interactions through [sigma] Bonds 1", JOURNAL OF PHYSICAL CHEMISTRY. A, MOLECULES, SPECTROSCOPY,KINETICS, ENVIRONMENT AND GENERAL THEORY, vol. 105, no. 38, September 2001 (2001-09-01), US, pages 8727 - 8733, XP055699743, ISSN: 1089-5639, DOI: 10.1021/jp011134h *
SARIKA PATHAK SHARMA ET AL: "In vitro antibacterial and antimalarial activity of dehydrophenylalanine-containing undecapeptides alone and in combination with drugs", INTERNATIONAL JOURNAL OF ANTIMICROBIAL AGENTS, ELSEVIER, AMSTERDAM, NL, vol. 39, no. 2, 6 October 2011 (2011-10-06), pages 146 - 152, XP028434348, ISSN: 0924-8579, [retrieved on 20111104], DOI: 10.1016/J.IJANTIMICAG.2011.10.008 *
VOLKOV ALEXEY ET AL: "Catalytic Reductive Dehydration of Tertiary Amides to Enamines under Hydrosilylation Conditions", ORGANIC LETTERS ., vol. 16, no. 3, 7 February 2014 (2014-02-07), US, pages 680 - 683, XP055804471, ISSN: 1523-7060, Retrieved from the Internet <URL:https://pubs.acs.org/doi/pdf/10.1021/ol403302g> DOI: 10.1021/ol403302g *

Similar Documents

Publication Publication Date Title
RU2270194C2 (en) BENZOPHENONES AS INHIBITORS OF IL-β AND TNF-α, PHARMACEUTICAL COMPOSITION AND TREATMENT METHOD
AU696109B2 (en) Aminoquinoline derivatives
WO2008141843A1 (en) Novel sulphoximine-substituted quinazoline and quinazoline derivatives as kinase inhibitors
AU2016324013B2 (en) Substituted 1H-indole-2-carboxamide compounds as indoleamine-2,3-dioxygenase inhibitors
JP6951406B2 (en) Flabagulin derivative
EP2949647A1 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing same
FI100530B (en) Process for the preparation of substituted diaminophthalimides and their analogues
US20110105514A1 (en) 5,6,7,8-tetrahydro-imidazo[1,5-a]pyrazine compounds
JP2018531230A6 (en) Pharmaceutical compounds
JP2013543504A (en) 1,2-Dihydro-4-hydroxy-2-oxo-quinoline-3-carboxyanilide as an AHR activator
WO2017198196A1 (en) Quinoline derivative having anti-tumor activity
CN1251102A (en) Tetrahydropyrido compounds
US7189714B2 (en) N-type calcium channel antagonists for the treatment of pain
JP2009542585A (en) Quinoline derivatives, methods for their preparation, uses thereof, and pharmaceuticals comprising them
JP6606806B2 (en) Deuterated quinazolinone compound and drug composition containing the compound
CN107739368B (en) N-substituted-5- ((4-substituted pyrimidine-2-yl) amino) indole derivatives, and preparation method and application thereof
CN1830964A (en) 4-Substituting anilino-3-nitroquinoline compounds, prepn. method and use thereof
WO2021191882A1 (en) Antimalarial agent, methods and uses thereof
JP2009511548A (en) Novel pyrimidine-2,4-diamine derivatives and their use as modulators of calcium-dependent potassium channels of small conductance
CN114409636B (en) Quinazolinone compound or pharmaceutically acceptable salt thereof, and preparation method and application thereof
CN107652218B (en) Dihydropyrrolidone derivative and application thereof in preparation of antitumor drugs
CN107635991B (en) Furoquinolinediones as inhibitors of TDP2
CA3230542A1 (en) Novel ras inhibitors
Muscia et al. Evaluation of antiparasitic, antituberculosis and antiangiogenic activities of 3-aminoquinolin-2-one derivatives
JP6943886B2 (en) Condensed pyrimidinopiperidin derivative, and its production method and application

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21721599

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21721599

Country of ref document: EP

Kind code of ref document: A1