WO2010144101A1 - Next generation quinoloine methanols - Google Patents

Next generation quinoloine methanols Download PDF

Info

Publication number
WO2010144101A1
WO2010144101A1 PCT/US2009/055796 US2009055796W WO2010144101A1 WO 2010144101 A1 WO2010144101 A1 WO 2010144101A1 US 2009055796 W US2009055796 W US 2009055796W WO 2010144101 A1 WO2010144101 A1 WO 2010144101A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl group
mefloquine
therapeutic composition
group
cyclic
Prior art date
Application number
PCT/US2009/055796
Other languages
French (fr)
Inventor
Geoffrey S. Dow
William F. Mccalmont
Erin E. Milner
Original Assignee
The United States Of America, As Represented By The Secretary Of The Army, On Behalf Of U.S.
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 The United States Of America, As Represented By The Secretary Of The Army, On Behalf Of U.S. filed Critical The United States Of America, As Represented By The Secretary Of The Army, On Behalf Of U.S.
Publication of WO2010144101A1 publication Critical patent/WO2010144101A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms 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
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to new quinoline methanol derivatives. They are useful in the treatment or prevention of malaria, microbial, parasitic, protozoan, bacterial, and fungal diseases and conditions.
  • mefloquine Commercially available quinoline methanols, such as mefloquine, have considerable clinical utility as antimalarials as a consequence of their potency against chloroquine-resistant parasites, oral bioavailability, and long plasma half- lives. Mefloquine has also shown effectiveness as an antimicrobial, antiparasitic, antiprotozoan, antibacterial, and antifungal agent. But, mefloquine is not without its adverse side effects.
  • Mefloquine is associated with severe depression, anxiety, paranoia, aggression, nightmares, insomnia, seizures, birth defects, peripheral motor- sensory neuropathy, vestibular (balance) damage, and various central nervous system problems. Effects to the CNS are permanent.
  • Mefloquine is known to cross the blood-brain barrier and accumulate as much as 30-fold in the central nervous system (CNS). It acts as a blood schizonticide, but the exact mechanism(s) of action of mefloquine is unknown. The adverse effects of mefloquine presumably occur because it crosses the blood-brain barrier and accumulates in the CNS where it is known to interact with numerous neurological targets. Therefore, a potential way to eliminate the poor CNS tolerability of mefloquine is to limit its passage across the blood-brain barrier and its accumulation in the brain.
  • mefloquine In addition to the aforementioned adverse effects of melfoquine, resistance by Plasmodium spp., the parasites responsible for malaria, to mefloquine is becoming more prevalent. Further, mefloquine is relatively expensive compared to other antimalarials. As a result, the clinical utility of mefloquine is limited. New compounds are needed.
  • the present invention provides quinoline methanol derivatives that are less able to cross the blood-brain barrier than mefloquine. These derivatives will be more clinically useful than mefloquine because they have the suitable structures and physiochemical properties that maintain or improve their therapeutic activity, but limit their exposure to the CNS.
  • the present invention provides therapeutic compositions that comprise new quinoline methanol compounds derived from modification of the mefloquine skeleton. As compared to mefloquine, these new quinoline methanols are more useful pharmacological agents for the prevention or treatment of malaria, or other microbial, parasitic, protozoan, bacterial and fungal diseases, because they are less able to cross the blood-brain barrier yet retain equal or better therapeutic activity.
  • the quinoline methanol compounds of the present invention differ from mefloquine in that the piperidine ring is replaced by non-piperidine functional groups at the 4 position, and the trifluromethyl group at the 8 position of the quinoline ring may be replaced with various combinations of hydrogen or additional trifluromethyl groups at the 6, or 7, or 8 positions or combinations thereof.
  • ( ) n represents one, two, or more carbon atoms
  • R 1 is a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, or a triazole
  • R 2 is a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, and a triazole
  • R 3 represents at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a trifluromethyl group, a OH group, an oxygen atoms
  • a trifluromethyl group is at position 8, more preferably a trifluromethyl group is at position 8 and position 6 or 7; and even more preferably a trifluromethyl group is present at positions 6, 7, and 8.
  • R 1 nor R 2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH 2 - CHOH-CH 2 -CH 3 , CH 2 -CH 2 -CHOH-CH 3 , CH 2 -CH 2 -CH 2 -CH 2 OH, CH 2 OH, and CH 2 -CH 2 - COOH.
  • R 1 or R 2 may be selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH 2 -CHOH-CH 2 -CH 3 , CH 2 -CH 2 -CHOH-CH 3 , CH 2 -CH 2 - CH 2 -CH 2 OH, CH 2 OH, CH 2 -CH 2 -COOH, other straight chain alkyl groups, cyclic alkyl groups, straight chain alkyl groups containing a nitrogen atom, or cyclic alkyl groups containing a nitrogen atom.
  • LogP is the partition coefficient reflecting the relative solubility in octanol versus water
  • PSA is the polar surface area of a molecule
  • FRB is the number of freely rotatable bonds in a molecule.
  • a quinoline methanol compound of the invention exhibits an in vitro permeability across the blood-brain barrier that is less than 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, or even 50% of the relative rate of mefloquine under similar conditions in vitro.
  • the quinoline methanol compounds exhibits an in vitro permeability across the blood- brain barrier that is less than 80% of the relative rate of mefloquine.
  • a quinoline methanol compound of the invention exhibits an in vivo total or free brain concentration that is less than 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, or even 50% of the relative concentration of mefloquine under similar conditions in vivo.
  • the quinoline methanol compounds exhibits an in vivo total or free brain concentration that is less than 80% of the relative concentration of mefloquine.
  • the R 1 and R 2 groups are joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring.
  • the R 1 and R 2 groups are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
  • the invention provides therapeutic compositions comprising a quinoline methanol compound of Formula I
  • R 1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
  • R 2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
  • R 2 is selected from the group consisting
  • R 1 and R 2 are either joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring; or alternatively, R 1 and R 2 are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
  • both R 1 and R 2 contain a heterocycle or heteroatom.
  • R 1 or R 2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH 2 - CHOH-CH 2 -CH 3 .
  • CH 2 -CH 2 -CHOH-CH 3 CH 2 -CH 2 -CH 2 -CH 2 OH, CH 2 OH, and CH 2 -CH 2 - COOH.
  • the therapeutic compositions of the invention also include a pharmaceutically- acceptable carrier or physiologically-acceptable carrier such as a diluent, adjuvant, or a combination thereof.
  • the present invention provides therapeutic compositions that are less neurotoxic than mefloquine and that are antimalarials at least as efficacious as mefloquine against Plasmodium spp..
  • these therapeutic compositions are also at least as efficacious as mefloquine against other diseases or conditions associated with infection by microbes, parasites, protozoans, bacteria, or fungi.
  • the present invention provides therapeutic compositions that can be used as antimalarial, antimicrobial, antiparasitic, antiprotozoan. antibacterial, or antifungal agents either alone, in combination with one another, or in combination with other therapies or compositions known in the art.
  • the compounds of the present invention alleviate neurotoxicity and improve therapeutic activity, while retaining the desirable properties of a practical and useful pharmacological agent, These principles are broadly applicable to the treatment and prevention of any of the conditions including infectious disease and immune disease against which mefloquine can be applied.
  • the present invention provides mefloquine analog compounds and methods for identifying and making these less neurotoxic mefloquine analogs that also retain the properties of useful drug substances for treatment of a variety of diseases and conditions.
  • Quinoline methanol compounds and therapeutic compositions of the present invention may be included in a pharmaceutical preparation that is administered to a subject by at least one mode selected from the group consisting of oral, topcial, parenteral, subcutaneous, intramuscular, intradermal, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intraccervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal.
  • compositions according to the invention may be administered once or several times, also intermittently, for instance on a daily or weekly basis for several days, weeks, or months in different dosages and by a combination of different routes.
  • the invention also provides methods of making therapeutic compositions and quinoline methanol compounds that are less neurotoxic than mefloquine as well as being efficacious as antimalarials, antimicrobials, antiparasitics, antiprotozoans, antibacterials, or antifungals. Preferably, these compounds may be utilized as preventative measures against or as treatment for malaria and other diseases and infections. [0030]
  • the invention further provides methods of using the described therapeutic compositions to treat, reduce, or prevent diseases or conditions associated with infection by microbes, parasites, preferably Plasmodium spp., protozoans, bacteria, or fungii.
  • the therapeutic compositions may be used alone, in combination with one another, or in combination with other therapies or compositions.
  • the invention provides a method of treating (e.g. curing, suppressing, ameliorating, delaying or preventing the onset of, or preventing recurrence or relapse of) or preventing a microbial, parasitic, protozoan, bacterial, or fungal associated disease or condition in a subject.
  • the method includes: administering to a subject a quinoline methanol compound of the invention in an effective amount sufficient to treat or prevent the disease or condition.
  • the quinoline methanol compound or derivative can be administered to the subject alone or in combination with other therapeutic modalities.
  • the invention provides a method of reducing the incidence of or severity of a clinical sign associated with a microbial, parasitic (preferably malaria, more preferably a Plasmodium falciparum infection), protozoan, bacterial, or fungal associated disease or condition, comprising the step of administering a therapeutic composition of the invention such that the incidence of or the severity of a clinical sign of malaria is reduced by at least 10%, preferably by 15%, 20%, 25%, 30%, 35%, 40% 45%, 50% or more, relative to a subject that has not received the therapeutic composition.
  • a microbial, parasitic preferably malaria, more preferably a Plasmodium falciparum infection
  • protozoan preferably preferably a Plasmodium falciparum infection
  • protozoan bacterial, or fungal associated disease or condition
  • Such clinical signs include fever, chills, sweats, headaches, muscle pains, nausea, vomiting, elevated temperature, perspiration, tiredness, confusion, coma, neurologic focal signs, severe anemia, and respiratory difficulties.
  • kits for administering a therapeutic composition of the invention comprising a set of printed instructions; a dispenser capable of administering a therapeutic composition to a subject; and at least one quinoline methanol compound of the invention that is therapeutically effective against at least one clinical sign associated with a microbial, parasitic (preferably malaria, more preferably a Plasmodium falciparum infection), protozoan, bacterial, or fungal associated disease or condition.
  • Kits of the invention may further comprise a physiologically acceptable vehicle, carrier molecule, adjuvant, or combination thereof.
  • a quinoline methanol compound of the invention and a physiologically acceptable vehicle, carrier molecule, adjuvant, or combination may be supplied in the kit either separately or in a premixed combination.
  • the term "effective amount” refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity or duration of a disease or disorder, or one or more symptoms thereof, prevent the advancement of a disease or disorder, cause the regression of a disease or disorder, prevent the recurrence, development, onset, or progression of one or more symptoms associated with a disease or disorder, or enhance or improve the prophylaxis or treatment of another therapy or therapeutic agent.
  • a "subject” is a mammal, preferably a human, in need of either prophylactic or treatment for a microbial, parasitic, protozoan, bacterial, or fungal associated infection, disease, or condition.
  • Protection against disease and similar phrases, mean a response against a disease or condition generated by administration of one or more therapeutic compositions of the invention, or a combination thereof, that results in fewer deleterious effects than would be expected in a non-immunized subject that has been exposed to disease or infection. That is, the severity of the deleterious effects of the infection are lessened in a vaccinated subject. Infection may be reduced, slowed, or possibly fully prevented, in a vaccinated subject.
  • complete prevention of infection it is specifically stated. If complete prevention is not stated then the term includes partial prevention.
  • “reduction of the incidence and/or severity of clinical signs” or “reduction of clinical symptoms” means, but is not limited to, reducing the number of infected subjects in a group, reducing or eliminating the number of subjects exhibiting clinical signs of infection, or reducing the severity of any clinical signs that are present in one or more subjects, in comparison to wild-type infection.
  • these clinical signs are reduced in one or more subjects receiving the therapeutic composition of the present invention by at least 10% in comparison to subjects not receiving the composition and that become infected. More preferably clinical signs are reduced in subjects receiving a composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
  • the term "increased protection” herein means, but is not limited to, a statistically significant reduction of one or more clinical symptoms which are associated with infection by an infectious agent, preferably a Plasmodium spp,, respectively, in a vaccinated group of subjects vs. a non-vaccinated control group of subjects.
  • the term "statistically significant reduction of clinical symptoms” means, but is not limited to, the frequency in the incidence of at least one clinical symptom in the vaccinated group of subjects is at least 10%, preferably 20%, more preferably 30%, even more preferably 50%, and even more preferably 70% lower than in the non-vaccinated control group after the challenge the infectious agent.
  • compositions used herein may incorporate known injectable, physiologically acceptable sterile solutions.
  • aqueous isotonic solutions e.g. saline or plasma protein solutions
  • the immunogenic and vaccine compositions of the present invention can include pharmaceutical- or veterinary- acceptable carriers, diluents, isotonic agents, stabilizers, or adjuvants.
  • a pharmaceutical- or veterinary- acceptable carrier includes any and all solvents, dispersion media, coatings, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like.
  • stabilizing agents for use in the present invention include stabilizers for lyophilization or freeze-drying.
  • Disposents can include water, saline, dextrose, ethanol, glycerol, and the like.
  • Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others.
  • Stabilizers include albumin and alkali salts of ethylendiamintetracetic acid, among others.
  • FIG. 1 General structure motifs.
  • FIG. 3 Exemplary active compounds that embody the diamine motif.
  • FIG. 4 Compounds WR318973 and WR177000.
  • FIG. 5 Diamine quinoline methanols with reduced brain penetration as compared to mefloquine.
  • FIG. 6 Diamines with additional lipophilicity.
  • FIG. 7 Putative active metabolites of WR318973.
  • FIG. 8 Generic Diamine Quinoline Methanols of Interest.
  • FIG. 9 Proposed synthetic route to construct N-oxide amine derivatives of WR318973.
  • FIG. 10 Proposed synthetic route to construct N-hydroxyl amine analogs of
  • FIG. 11 Structures of straight chain ethylene diamines.
  • FIG. 12 Synthetic scheme for straight chain ethylene diamines.
  • FIG. 13 Structure of pyrrolidine ethylene diamines.
  • FIG. 14 Synthetic schemes for pyrrolidine ethylene diamines.
  • FIG. 15 Structure of bicyclic ethylene diamines.
  • FIG. 16 Synthetic schemes for bicyclic ethylene diamine 37.
  • FIG. 17 Synthetic schemes for bicyclic ethylene diamine 41.
  • FIG. 18 Synthetic schemes for bicyclic ethylene diamine 50.
  • FIG. 19 Generic bicyclic compounds of interest.
  • FIG. 20 Epoxide opening scheme [EtOH, uw, 30min, 130oC, 25Ow].
  • FIG. 21 Remaining diamine quinoline methanols of interest.
  • FIG. 22 Structures and IC90s of the most potent quinoline methanols. IC90s are against the mefloquine resistant Pf C2A strain.
  • FIG. 23 Reagents and conditions: (a) 1. POBr 3 , 75°C - 15O°C (91%) (b) n-BuLi, N,N-dimethylformamide.THF, -78°C (64%) (c) NaH, Me 3 SO, DMSO, THF (82%) (d) NaH, ROH, THF, O°C to rt (72-89%) (e) NaH, RSH, THF, O°C to rt (81-84%) (f) RMgBr, ether or THF, -78°C to rt (88-93%) (g) amine, 200 proof EtOH, microwaves, 25Ow, 13O°C, 15-30 min (18-97%).
  • FIG. 24 Actual versus calculated LogBB Values for Structurally Diverse
  • the invention provides new quinoline methanol derivatives that are less able to cross the blood-brain barrier as compared to mefloquine and still retain the same, or even improved, therapeutic efficacy as mefloquine. These new compounds are expected to be useful pharmacological agents for the prevention or treatment of malaria, and other microbial, parasitic, protozoan, bacterial and fungal diseases and potentially of Parkinson's disease or diseases associated with prions.
  • the present invention relates to several new quinoline methanol compounds and pharmaceutical compositions containing one or more compounds within these.
  • the invention embodies a series of new antimalarial compounds in which the quinoline methanol scaffold has been optimized in terms of the structure modifications and physiochemical properties required for excellent oral bioavailability, equivalent and/or improved potency as compared to mefloquine and reduced potential for blood brain barrier penetrability/brain uptake than mefloquine. These properties were mediated by the incorporation of 4 position side chains imparting lower lipophilicity and higher polar surface area to the compounds.
  • the general structure of these new compounds is provided in Formula I.
  • ( ) n represents one, two, or more carbon atoms
  • R 1 is a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, or a cyclic alkyl group containing a nitrogen atom
  • R 2 is a hydrogen atom, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, or a cyclic alkyl group containing a nitrogen atom
  • R 3 represents at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a trifluromethyl group, a OH group, an oxygen atom, a hydrogen atom, or a combination thereof.
  • a trifluromethyl group is at position 8, more preferably a trifluromethyl group is at position 8 and position 6 or 7
  • R 1 nor R 2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH 2 - CHOH-CH 2 -CH 3 , CH 2 -CH 2 -CHOH-CH 3 , CH 2 -CH 2 -CH 2 -CH 2 OH, CH 2 OH, and CH 2 -CH 2 - COOH.
  • Rj or R 2 may be selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH 2 -CHOH-CH 2 -CH 3 , CH 2 -CH 2 -CHOH-CH 3 , CH 2 -CH 2 - CH 2 -CH 2 OH, CH 2 OH, CH 2 -CH 2 -COOH, other straight chain alkyl groups, cyclic alkyl groups, straight chain alkyl groups containing a nitrogen atom, or cyclic alkyl groups containing a nitrogen atom.
  • compositions may comprise an effective amount of one or more of these compounds, and a pharmaceutically acceptable carrier.
  • effective amount of these compounds will vary based upon the use for which the composition is intended.
  • compositions within the scope of the present invention may be administered to a patient via any conventional route of administration.
  • Selection of a suitable, pharmaceutically acceptable carrier is well within the skill of the art depending on the method of administration sought to be employed - i.e., oral, intravenous, etc.
  • These compounds and compositions containing them may be used in methods for preventing and/or treating malaria, methods for preventing and/or treating other infectious diseases, or methods for preventing and/or treating certain immunological conditions.
  • compounds and compositions within the scope of the present invention may be used in the intermittent preventive treatment of malaria.
  • Reference to treatment herein includes intermittent preventive treatment.
  • the invention is suitable for use against all species of malaria (i.e., blood stages of all malaria parasites).
  • the invention is suitable for use against diseases caused by bacteria, protozoa, parasites, and fungi, and against some autoimmune diseases (i.e.. rheumatoid arthritis).
  • R 1 , R 3 , R 4 , and R 5 in these formulas represent substituents that render a quinoline methanol less able to cross the blood- brain barrier than mefloquine - see, for example, Tables 1-3 for illustrative substituents.
  • This reduction is because quinoline methanols represented by these formulas exhibit lower LogP, or higher polar surface area (PSA), or have a greater number of freely rotatable bonds (FRBs), or a combination of all three of these properties as compared to mefloquine.
  • R represents a substitution at one or more of the 2, 3, 4, or 5 positions of the imidazole ring or triazole ring.
  • R 2 denotes any substituent at the 6, 7 or 8 positions of the quinoline ring, including but not limited to CF 3 in the 8 position.
  • Series C, and Series D contain one or more carbon atoms positioned between the C-OH and the N atoms of the 4 position side chain as shown in FIG. 1.
  • suitable substituents are those that render quinoline methanols less able to cross the blood-brain barrier relative to mefloquine without imparting a loss of potency.
  • the presence of a second nitrogen atom on the side chain makes such analogs less able to penetrate the blood-brain barrier.
  • diamine analogs such as WR318746, WR318744 and WR319535 exhibit lower brain concentrations 5 min after i.v. dosing than mefloquine (FIG. 5). Brain concentrations were 100, 243, 199 and 550 ng.g respectively for WR319535, WR318744 and WR318744 and mefloquine respectively. As a consequence, they are likely to be better tolerated than mefloquine due to lower exposure of multiple vulnerable CNS targets to the drugs. More generally, quinoline methanols with the requisite diamine motif are more likely to exhibit less blood-brain barrier penetrability than non- diamines and be better tolerated as a consequence.
  • the reduction in lipophilicity associated with the second nitrogen is also associated with improved pharmacokinetic properties.
  • the clearance of WR177000 is 2681 ml/hr/kg versus 912 ml/hr/kg for WR318973. This means that the plasma concentration of WR318973, 24 h after dosing are still measurable at 83 ng/ml, whereas this is not the case for WR177000.
  • the proportion of compound remaining 24 h after an i.v. 5 mg/kg dose is comparable to mefloquine.
  • diamine quinoline methanols as antimalarials is partially dependent on their potency. Potency against malaria parasites is assessed by measuring an IC50 against well- characterized laboratory strains of P. falciparum such as W2 and C2A. Diamine quinoline methanols are unique in that the reduction in lipophilicity generated does not compromise the utility of the compounds by reducing potency to the point of inactivity. Furthermore, the potency of diamines is dependent on restricting access to the second amine, the positioning of the two nitrogen atoms relative to each other and cyclization of the side chain. For example, WR318972 (FIG.
  • WR318746 have IC90s of 30 and 69 ng/ml against Pf W2 versus 490 ng/ml for WR318973 because access to the second amine is restricted. This trend is even more prominent when one considers an analog such as WR308396, with an IC90 of 6 ng/ml, in which a large lipophilic group is added to the terminal portion of the side chain. Also, analogs in which the second amine is three atoms from the first nitrogen are more potent. For example, the IC90 of WR308782 (FIG. 6) is 69 ng/ml compared to 6 ng/ml for WR308396. Finally, cyclization of the side chain improves potency.
  • WR308621 has an IC90 of 46 ng/ml versus 490 ng/ml for WR318973.
  • diamines in which access to the second amine is restricted, in which the terminal nitrogen is cyclized and/or in which the second nitrogen is three atoms from the first, are particularly favored.
  • a quinoline methanol for malaria or other indications is dependent on oral bioavailability.
  • Diamine quinoline antimalarials can be inferred to be bioavailable based on in vivo efficacy studies.
  • WR318973 cured 4 of 5 mice infected with P. berghei after oral administration of a single dose of 320 mg/kg was administered orally (100% of untreated controls are not cured). This result compares favorably to mefloquine, in which 11/18 mice were cured at the same dose.
  • This observation suggests that WR318973 is sufficiently orally bioavailable that enough of the compound reaches the blood to eliminate malaria parasites.
  • WR318973 is effective in vivo despite lower potency relative to other related diamines. It is possible that some of its effects in mice may be mediated by putative diamine active metabolites. These are illustrated in FIG. 7.
  • R groups 1 through 6 represent any functionality that improves the intrinsic properties of the diamine scaffold including but not limited to aliphatic or aromatic or heterocyclic or cyclic substituents. They could be methyl groups that block access to the second amine or the second nitrogen could be incorporated into a cyclic structure in order to improve potency.
  • the substituents might increase the molecular weight of the compound in order to render them PGP substrates and thus less penetrant of the blood-brain barrier.
  • the substituents could result in additional increases in polar surface area and or decreases in lipophilicty in order to further decrease permeability.
  • blocking groups could be added to sterically hinder sites of metabolic attack.
  • R 8 denotes any substituent attached to the 6, or 7, or 8 positions of the quinoline ring, including but not limited to CF 3 at the 8 position.
  • R 7 is intended to be a substituent that renders the compound a tertiary amine, including, but not limited to compounds in which the substituent at R 7 is joined to another R 1-6 substituent to create a cyclic species. Substituents at R 1 , R 2 and R 7 might be O or OH groups in order that the compounds be mono or di hydroxyl amines or N- oxides.
  • Figure 2 outlines the strategy used to synthesize a 4-position library of methanol quinoline compounds.
  • the structure of mefloquine is indicated at (1).
  • the intermediate scaffold 4-(oxiran-2-yl)-2,8-bis(trifluoromethyl)quinoline (3) was synthesized from bis(trifluoromethyl)quinolin-4-ol (2) by the addition of POBr 3 at 75°C to 15O°C for 2 hours with 91 % yield.
  • the resulting 4-bromo-2,8-bis(trifluoromethyl)quinoline was dissolved in tetrahydrofuran, cooled to -78°C and subjected to a n-butyllithium.
  • N, N,-dimethylformamide was subsequently added to afford 2,8-bis(trifluoromethyl)quinoline-4-carbaldehyde.
  • Utilization of Corey's dimethylsulfonium methylide provided racemic epoxide (3).
  • the epoxide (3) can also be purchased commercially from Bioblocks (San Diego, California).
  • the quinoline scaffold (3) was diversified at the 4-position in a single step with commercially available nucleophiles.
  • N-oxide compounds (3) and (4) In order to construct the N-oxide compounds (3) and (4) from WR318973 (1), the benzylic alcohol and the first amine on the side chain must be protected (FIG. 9).
  • Mixed acetal (2) will be constructed from isobutyraldehyde utilizing the standard protocol in dichloromethane, which allows the second amine to be oxidized with a variety of oxidants. As shown in FIG. 9, t-butyl peroxide promoted oxidation is of interest since it was shown to not oxidize heterocyclic nitrogen atoms (see J. D. Fields and P. J. Kropp. Journal of Organic Chemistry, 2000, 65, 5937-5941).
  • the desired tosyl-protected aziridine 13 is constructed from the corresponding amino alcohol (Loethar W. Bieber and Mariea C. F. de Araujo. Molecules, 2002, 7, 902-906 incorporated herein by reference) or commercially available aziridines.
  • the tosyl group protects the secondary amine, activates the aziridine to nucleophilic attack (SN2), and is an ultraviolet tag to monitor the reaction.
  • Tosyl-protected aziridine 13 (l.Oeq) is dissolved in ethanol (200 proof, 0.25M), amine 14 (l.leq) is added, and the reaction mixture is subjected to microwaves (130°C, 250 watts) for 30 minutes.
  • 1.25M HCl/EtOH 1.5eq
  • 2,2,6, 6-Tetramethylpiperidine (3.0eq) is added to increase the pH ⁇ 9, followed by the addition of epoxide 7, and the reaction mixture is further subjected to microwaves (13O°C, 250 watts) for 30 minutes.
  • the ethanol is removed in vacuo and the reaction mixture is purified via normal phase preparative thin layer chromatography in 95:5 dichlomethane: methanol to yield 28-43% yield over three steps. The conditions can be optimized to increase yields.
  • Pyrrolidine ethylene diamines as depicted in FIG. 13 are also particularly favored embodiments of the present invention. These can be synthesized using the following described methods or variations thereof as illustrated in FIG. 14. By utilizing chiral enolates of pseudoephedrine amides 18 and azirides such as 13, substituted pyrrolidin-2-ones 20 can be prepared in >99% enantiomeric excess (see Jose L. Vicario, Dolores Badia, and Luisa Carrillo, J. Org. Chem. 2001, 66, 5801-5807, incorporated herein by reference).
  • a three-step sequence will be employed to transform pyrrolidin-2-one 20 to ⁇ -methylaminonitrile 25, which can be reduced directly to amine 26 or treated with Grignard reagent to afford substituted pyrrolidine 27.
  • substituted pyrrolidine 27 23, 24, or 26
  • utilization of our microwave-assisted epoxide opening and subsequent deprotection will be utilized to afford HCl salt 28.
  • Bicyclic ethylene diamines are also particularly favored embodiments of the present invention (FIG. 15). These can be synthesized as follows or similar methods thereof as outlined in FIGs. 16-20.
  • FIG. 16 illustrates the proposed synthesis of bicyclic amines such as 37. Utilizing similar strategies employed in the synthesis of the pyrrolidines (FIG. 14), carboxyglutamic acid derivatives such as 31 can be subjected to Swartz' reagent (Cp 2 ZrHCl) and cyanotrimethylsilane (TMSCN) to afford the corresponding ⁇ -aminonitrile 32 (Q. Xia and B. Ganem. Tetrahedron Letters, 2002, 43, 1597-1598, incorporated herein by reference).
  • Swartz' reagent Cp 2 ZrHCl
  • TMSCN cyanotrimethylsilane
  • FIG.19 Some specific bicyclic compounds of interest are presented in FIG.19.
  • compounds such as Compound 50 can be made as outlined in FIG 18.
  • Methyl-4- hydroxybenzoate 40 (FIG. 17)was reduced to the corresponding aliphatic cyclohexane and oxidized to ketone 43.
  • a Bucherer-Bergs reaction was utilized to establish the amine and carboxylic acid group in a cis configuration with spirohydantoinic compound 44. Subsequent saponification, formation of the acid chloride, and cyclization led to lactam 46.
  • diamines of interest may also be synthesized by epoxide opening as depicted in FIG. 20.
  • the diamine can be purchased commercially or constructed in house and utilized in microwave-assisted epoxide opening.
  • Table 1 Summary of physiochemical properties and biological data for novel quinoline methanols (NGQM - Next Generation Quinoline Methanols). The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
  • Table 2 Summary of physiochemical property and biological data for novel quinoline methanols. The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
  • Table 3 In vitro Plasmodium falciparum (PF) screening and in vivo efficacy and toxicity data for selected compounds of interest. The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
  • Table 7 Properties of different functional groups present in active and inactive amines.
  • Table 8 Plasmodium falciparum IC90 (ng/ml) values resulting from probing hydroxyl utility.
  • Table 15 In vivo efficacy of selected quinoline methanols in the P. berghei mouse model and permeability across MDRl-transfected MDCK cell monolayers.
  • LogP is the partition coefficient reflecting the relative solubility of a drug in octanol versus water. The higher the value, the lower the water solubility. Generally a reduction in the LogP is associated with reduced permeability across the blood brain barrier. LogP can be predicted from the structure of a compound using standard physiochemical prediction software (e.g. ACD).
  • PSA is the polar surface area of a molecule and is a reflection of the polarity of the molecule. Generally, higher PSA is associated with reduced permeability across the blood brain barrier. PSA can be predicted from the structure of a compound using standard physiochemical prediction software (e.g. ACD).
  • FRBs is the number of freely rotatable bonds a compound has. A greater number of freely rotatable bonds generally correlates with lower blood-brain barrier permeability. FRBs can be determined from the structure of a compound using standard physiochemical prediction software (e.g. ACD).
  • PF IC90 (ng/ml) is the 90% inhibitory concentration in ng/ml of a molecule against P. falciparum in an in vitro cell based growth inhibition assay.
  • W2, D6, C235 and C2A strains of drug resistance P. falciparum were used. The lower the value, the more active the molecule.
  • Macrophage IC50 ( ⁇ M) is the 50% inhibitory concentration in micromolar units of a molecule in an in vitro cytotoxicity assay against a rodent macrophage cell line. The lower the value, the more toxic the molecule.
  • Lipinski is the number of violations of Lipinski's rule of 5. This index relates the likelihood that a compound will be orally bioavailable based on its physiochemical properties. The fewer Lipinski violations, the greater the likelihood of a compound being orally bioavailable.
  • P. berghei-mice is the animal model that is used to evaluate the potential utility of new antimalarial compounds in a discovery setting.
  • P. berghei, or Plasmodium berghei is a rodent malaria parasite that induces a lethal infection in mice.
  • quinoline methanols all commercially available compounds clinically effective against human malaria parasites are also active in this model.
  • quinoline methanols brain and plasma concentrations were determined in vivo after intravenous dosing.
  • Groups of 2-4, seven-eight week old male FVB mice were administered a 5 mg/kg dose of quinoline methanol base.
  • the drug was given as a 50 microliter bolus dose in a drug vehicle consisting of 5% w/v glucose, and/or 0-5% dimethylsulfoxide, and/or 0-1% solutol in 20 mM citrate or acetate buffer at pH 3-4 with the specific buffer characteristics being determined for specific compounds based on solubility.
  • a drug vehicle consisting of 5% w/v glucose, and/or 0-5% dimethylsulfoxide, and/or 0-1% solutol in 20 mM citrate or acetate buffer at pH 3-4 with the specific buffer characteristics being determined for specific compounds based on solubility.
  • 60 min, 6h and 24h plasma and brain samples were obtained from the mice and frozen at -8O°C until they were
  • C235 and C2A strains are resistant to multiple drugs.
  • the IC50 and IC90 values represent the concentrations in ng/ml at which the incorporation of tritiated hypoxanthine is inhibited by 50% or 90% respectively. Since hypoxanthine incorporation is essential for parasite growth, inhibition of its action by drug uptake is a measure of the drug's toxicity to the parasite.
  • the LC50 of some of the analogs was determined in vitro against a RAW macrophage cell line.
  • the LC50 is the 50% lethal concentration, and represents the concentration at which colorimetric conversion of a substrate by cell mitochondria is inhibited by 50%. Since mitochondrial activity is indicative of cell viability, the decline substrate conversion is interpreted as the lethal effect of a drug on cells. Thus the lower an LC50, the more potent its toxic effects.
  • Selectivity index relates toxicity to mammalian cells with antimalarial activity by calculation of the ratio of the macrophage LC50 to the IC50 against a parasite line such as W2. The greater the selectivity index the greater the selectivity of the antimalarial effect.
  • MDRI-MDCK monolayers were grown to confluence on collagen-coated, microporous, polycarbonate membranes in 12-well Costar Transwell@ plates.
  • the permeability assay buffer was Hanks Balanced Salt Solution containing 10 mM HEPES and 15 mM glucose at a pH of 7.4.
  • a known p-glycoprotein inhibitor cyclosporin A (CSA) was also added to the assay buffer at 10 mM.
  • the dosing solution concentrations of the test compound were 2.5 and 5.0 mM in the assay buffer. All cell monolayers were first pre-incubated for 30 minutes with assay buffer to saturate any P glycoprotein sites with test compound. After 30 minutes, the buffer was removed, replaced with fresh buffer, and time was recorded as 0.
  • Vr is the volume of the receiver compartment in cm3.
  • Vd is the volume of the donor compartment in cm3.
  • A is the area of the cell monolayer (1.13 cm2 for 12- well Transwell®).
  • CO is the measured concentration of the donor chamber at time 0 in mM.
  • CN is the nominal concentration of the dosing solution in mM.
  • Cr final is the culnulative receiver concentration in mM at the end of the incubation period.
  • Cd ina is the concentration of the donor in mM at the end of the incubation period.
  • Permeability across MDRl -transformed MDCK cell monolayers was determined by Absorption Systems (Exton, Pennsylvania) determined as previously described (Wang et al., 2005, incorporated herein by reference) with appropriate modifications to maximize the permeability of mefloquine. These were the incorporation of 1% BSA into recipient wells, co- incubation with the PgP inhibitor cyclosporine A at 10 ⁇ M and extension of the normal incubation time to 120 min after an initial 30 min incubation. This screen is a standard in vitro assay for blood-brain barrier permeability. All physiochemical properties were calculated using ACD (Version 10, ACD Labs, Toronto, Canada) except LogD (pH 7.4) which was determined using Pipeline Pilot (Version 6.1, Accelrys, San Diego California).
  • WR308245, WR308255, WR308257 and WR308266 were selected because they were approximately equivalent or superior to mefloquine in terms of potency, but contained less steric bulk in their side chains.
  • WR308387, WR.308388, WR308413 and WR.308446 were selected as they were substantially more potent than mefloquine in vitro.
  • WR308396 exhibited slightly lower permeability than mefloquine and half the permeability of WR308387 (Table 5).
  • the inhibition of the A2A and Al receptors by four of the analogs at 200 nM was evaluated (Table 5). In most cases the level of inhibition observed was lower or comparable to that observed with mefloquine. The exception was WR308245 against the Al receptor.
  • Analogs were categorized arbitrarily as having particular structural motifs.
  • the proportion of active (IC90 ⁇ 500 ng/ml or 1000 nM) and inactive compounds containing these functional groups was determined and differences between the groups were tested for significance using Fisher's Exact test (Table 7).
  • the inactive group contained a greater proportion of compounds in which the 4 position amino side chain contained additional heteroatoms, analogs in which the hydroxyl group or amine functionality were replaced, and compounds in which the first nitrogen atom in the side chain was conjugated.
  • the active groups of compounds contained a higher proportion of secondary amines and compounds in which the amino side chain contained no additional heteroatoms.
  • the inactive and active groups contained similar proportions of tertiary and benzyl amines.
  • the goal is to resolve the CNS tolerability issues of mefloquine by reducing partitioning into the central nervous system.
  • this strategy may be counter productive if it results in inadvertent increases in potency against suspected targets of mefloquine in the CNS.
  • Prior studies suggest that mefloquine may exhibit a direct neurotoxic effect in vivo and has potent activity against the A2A receptor (Weiss et at., 2003, Caridha et al., 2008, Gillespie et al., 2008). The most interesting analogs were evaluated against these targets. Most of the analogs tested exhibited lower LC50s against neurons or greater inhibition of the A2A receptor than mefloquine.
  • mefloquine and the compounds evaluated in the in vivo studies showed the same rank order in terms of potency in vitro against the four drug resistant strains of P, falciparum. This may indicate cross-susceptibility to mefloquine.
  • compounds, where they have equivalent potency to mefloquine might not be fully effective if used clinically as single agents for malaria treatment in areas where background resistance to mefloquine if used as monotherapy is prevalent (e.g. the Thai borders), Mefloquine is normally used in combination with artesunate in such regions and remains clinically useful when deployed in this manner (Price et al, 2004).
  • next generation quinoline methanol with a combination (perhaps non-artemisinin) agent in the same manner if needed. For this reason, lack of cross-susceptibility to mefloquine is a desirable rather than a required property of a next generation quinoline methanol.
  • Bis(trifluoromethyl) quinolin-4-ol 8 was melted along with phosphorous oxybromide to provide 4-bromo-2,8-bis(trifluoromethyl) quinoline 9.
  • the resulting white solid was dissolved in tetrahydrofuran, cooled to -78°C and subjected to n- butyllithium. N,N-dimethylformamide was subsequently added to afford 2,8- bis(trifluoromethyl)quinoline-4-carbaldehyde 10.
  • Utilization of Corey's dimethylsulfonium methylide provided racemic epoxide 11, which is also commercially available from Bioblocks (San Diego, Ca).
  • Imidazole and benzo[d]imidazole derivatives were collectively categorized as heterocyclic amino quinoline methanols (HAQMs).
  • HAQMs heterocyclic amino quinoline methanols
  • IC90 values for the four drug resistant P. falciparum strains are essentially the same order of magnitude
  • a promising cross- susceptibility IC90 profile emerges (Table 11).
  • WR308437 and WR308623 in particular illustrated this trend.
  • benzo[d]imidazole derivates a similar trend was observed for WR308682, WR308763, and WR308764.
  • WR308682 has increased potency and a superior selectivity index as compared to mefloquine.
  • the HAQMs exhibited half-lives in the in vitro metabolic stability assays of 4-22 minutes in comparison to > 60 minutes for mefloquine. They are therefore much less metabolically stable than mefloquine. If this translates into shortlived plasma drug concentrations in vivo, it would not bode well for their utility for prophylaxis.
  • alkyl amino quinoline methanols AAQMs
  • mefloquine mefloquine
  • quinine the lipophilic nature of mefloquine (MQ) and quinine are known to correlate with delivery of drug to the parasite.
  • MQ is also known for high-affinity binding to erythrocytes and other cells, which may provide a reservoir of drug and contribute to the long half-life. In general, these compounds were more potent than MQ and displayed a one- log increase in selectivity. Alkyl substitution is paramount.
  • the primary amine WR308314 is nearly devoid of activity, while the addition of methyl, ethyl, propyl, and butyl groups substantially increase efficacy (Table 12).
  • branched alkyl substituents such as i-Pr and i-Bu prove quite active, while t-butyl displays moderate activity.
  • Chain length also appears to affect activity since n-Bu (WR177000) and n-hex (WR308442) have different levels of potency, presumably resulting from the addition of two methylene units.
  • the increase in potency coupled with the reduced cost of goods for these analogs as compared to MQ add to their potential utility.
  • N-methyl (WR308245), i-Pr (WR308257), and t-butyl (WRl 83545) derivatives (Table 12) all displayed favorable metabolic stability profiles presumably due to inhibition of N- dealkylation.
  • WR17700 was chosen as a scaffold, and the initial strategy was to prepare a series of branched alkyl amino quinoline methanols (Table 13). Methyl substitution resulted in half-lives of greater than 60 minutes with human liver microsomes. As for mouse microsomes, the trend in half- life followed Rl > R2 > R3. Overall, most AAQMs demonstrated an increase in potency and a selectivity index superior to MQ, while branching proved to be an efficient strategy for improving metabolic stability.
  • NQM next generation quinoline methanol
  • Alkylaminoquinoline methanols with additional heteroatoms such as WR308412, WR308622, WR308378 and WR308396 exhibited equivalent potency to mefloquine but their activity across different parasite strains also tracked with those of mefloquine.
  • WR308412, WR308622, WR308378 and WR308396 exhibited equivalent potency to mefloquine but their activity across different parasite strains also tracked with those of mefloquine.
  • some evidence of efficacy after oral dosing is desired, as well as, the potential for reduced permeability across the blood brain barrier. The latter trait is essential if the adverse neurological effects of mefloquine are to be avoided in a new series of analogs.
  • the actual log of the ratios of brain to blood concentrations was determined in mice as follows. The compounds were administered intravenously to groups of 2-4 FVB mice at a dose of 5 milligrams per kilogram of bodyweight. At 5 minutes, 60 minutes, 4 h and 24 h, brain and plasma concentrations were measured. The maximum brain and plasma concentrations were calculated, The ratio of maximum brain concentration to plasma concentration was determined. These values were converted to Log units. These values are referred to as the actual LogBB values. They were plotted and are presented in FIG. 24. Linear regression was performed, yielding an r2 value of 0.44 and a significantly non-zero slope of the regression line (P ⁇ 0.0001). This result means that a correlation exists between the calculated and actual values for LogBB, underscoring the utility of the in silico calculation method.

Abstract

The present invention relates to new quinoline methanol derivatives and therapeutic compostions comprising one or more quinoline methanol derivatives. These compositons are useful in the reduction, treatment, or prevention of malaria, microbial, parasitic, protozoan, bacterial, and fungal diseases and conditions. Advantageously, compositions of the invention are less able to cross the blood-brain barrier than mefloquine and as a result produce fewer adverse side effects to the central nervous system as compared to mefloquine.

Description

NEXT GENERATION QUINOLOINE METHANOLS
RELATED APPLICATIONS
[0001] This application relates to and claims priority to U.S. Provisional Patent Application No. 61/268,654, which was filed June 9, 2009, and U.S. Provisional Patent Application No. 61/093,560, which was filed September 2, 2008. All of which are incorporated herein by reference in their entirety. All applications are commonly owned.
BACKGROUND OF THE INVENTION
A. Field of the Invention
[0002] The present invention relates to new quinoline methanol derivatives. They are useful in the treatment or prevention of malaria, microbial, parasitic, protozoan, bacterial, and fungal diseases and conditions.
B. Description of the Related Art
[0003] Commercially available quinoline methanols, such as mefloquine, have considerable clinical utility as antimalarials as a consequence of their potency against chloroquine-resistant parasites, oral bioavailability, and long plasma half- lives. Mefloquine has also shown effectiveness as an antimicrobial, antiparasitic, antiprotozoan, antibacterial, and antifungal agent. But, mefloquine is not without its adverse side effects.
[0004] Among subjects who received mefloquine for treatment, the most frequently observed adverse experiences included: dizziness, myalgia, nausea, fever, headache, vomiting, chills, diarrhea, skin rash, abdominal pain, fatigue, loss of appetite, and tinnitus. Those side effects occurring in less than 1% included bradycardia, hair loss, emotional problems, pruritus, asthenia, transient emotional disturbances and telogen effluvium (loss of resting hair). Seizures have also been reported.
[0005] Among subjects who received mefloquine for prophylaxis of malaria, the most frequently observed adverse experience was vomiting (3%). Dizziness, syncope, extrasystoles and other complaints affecting less than 1% were also reported. But, because these experiences are reported voluntarily from a population of uncertain size, it is likely that their frequency is under reported.
[0006] Mefloquine is associated with severe depression, anxiety, paranoia, aggression, nightmares, insomnia, seizures, birth defects, peripheral motor- sensory neuropathy, vestibular (balance) damage, and various central nervous system problems. Effects to the CNS are permanent.
[0007] The most frequently observed laboratory alterations possibly attributable to administration of mefloquine were decreased hematocrit, transient elevation of transaminases, leukopenia and thrombocytopenia. These alterations were observed in patients with acute malaria who received treatment doses of the drug and were attributed to the disease itself. During prophylactic administration of mefloquine to indigenous populations in malaria-endemic areas, the following occasional alterations in laboratory values were observed: transient elevation of transaminases, leukocytosis or thrombocytopenia.
[0008] Mefloquine is known to cross the blood-brain barrier and accumulate as much as 30-fold in the central nervous system (CNS). It acts as a blood schizonticide, but the exact mechanism(s) of action of mefloquine is unknown. The adverse effects of mefloquine presumably occur because it crosses the blood-brain barrier and accumulates in the CNS where it is known to interact with numerous neurological targets. Therefore, a potential way to eliminate the poor CNS tolerability of mefloquine is to limit its passage across the blood-brain barrier and its accumulation in the brain.
[0009] In addition to the aforementioned adverse effects of melfoquine, resistance by Plasmodium spp., the parasites responsible for malaria, to mefloquine is becoming more prevalent. Further, mefloquine is relatively expensive compared to other antimalarials. As a result, the clinical utility of mefloquine is limited. New compounds are needed.
[0010] The present invention provides quinoline methanol derivatives that are less able to cross the blood-brain barrier than mefloquine. These derivatives will be more clinically useful than mefloquine because they have the suitable structures and physiochemical properties that maintain or improve their therapeutic activity, but limit their exposure to the CNS. SUMMARY OF THE INVENTION
[0011] The present invention provides therapeutic compositions that comprise new quinoline methanol compounds derived from modification of the mefloquine skeleton. As compared to mefloquine, these new quinoline methanols are more useful pharmacological agents for the prevention or treatment of malaria, or other microbial, parasitic, protozoan, bacterial and fungal diseases, because they are less able to cross the blood-brain barrier yet retain equal or better therapeutic activity.
[0012] The quinoline methanol compounds of the present invention differ from mefloquine in that the piperidine ring is replaced by non-piperidine functional groups at the 4 position, and the trifluromethyl group at the 8 position of the quinoline ring may be replaced with various combinations of hydrogen or additional trifluromethyl groups at the 6, or 7, or 8 positions or combinations thereof.
[0013] The quinoline methanol compounds of the invention are represented by Formula I.
Figure imgf000004_0001
In Formula I, ( )n represents one, two, or more carbon atoms; R1 is a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, or a triazole; R2 is a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, and a triazole; and R3 represents at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a trifluromethyl group, a OH group, an oxygen atom, a hydrogen atom, or a combination thereof. Preferably, a trifluromethyl group is at position 8, more preferably a trifluromethyl group is at position 8 and position 6 or 7; and even more preferably a trifluromethyl group is present at positions 6, 7, and 8. [0014] Notably, when ( )n represents one carbon atom then neither R1 nor R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2- CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2- COOH. Whereas, in quinoline methanol compounds where ( )n represents two or more carbon atoms either R1 or R2 may be selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2- CH2-CH2OH, CH2OH, CH2-CH2-COOH, other straight chain alkyl groups, cyclic alkyl groups, straight chain alkyl groups containing a nitrogen atom, or cyclic alkyl groups containing a nitrogen atom.
[0015] Exemplary quinoline methanol compounds of the invention are presented in Tables 1-3 herein. It will be understood in the art that the invention is not limited to the exemplary compounds herein but includes other compounds that are represented by Formula I.
[0016] The therapeutic compositions of the invention comprise a pharmaceutically- acceptable carrier, adjuvant, or combination thereof, and at least one quinoline methanol compound as represented by Formula I and having a calculated log ratio of brain:blood concentration (cLogBB) that is less than the cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18. LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule.
[0017] A quinoline methanol compound of the invention exhibits an in vitro permeability across the blood-brain barrier that is less than 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, or even 50% of the relative rate of mefloquine under similar conditions in vitro. Preferably, the quinoline methanol compounds exhibits an in vitro permeability across the blood- brain barrier that is less than 80% of the relative rate of mefloquine. [0018] In another aspect, a quinoline methanol compound of the invention exhibits an in vivo total or free brain concentration that is less than 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, or even 50% of the relative concentration of mefloquine under similar conditions in vivo. Preferably, the quinoline methanol compounds exhibits an in vivo total or free brain concentration that is less than 80% of the relative concentration of mefloquine.
[0019] In certain quinoline methanol compounds of the invention, the R1 and R2 groups are joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring.
[0020] In other quinoline methanol compounds of the invention, the R1 and R2 groups are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
[0021] In another aspect, the invention provides therapeutic compositions comprising a quinoline methanol compound of Formula I
Figure imgf000006_0001
where ( )n represents one, two, or more carbon atoms; R1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms; R2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms; R3 is at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a CF3 group, a OH group, an oxygen atom, and a hydrogen atom. Further, R1 and R2 are either joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring; or alternatively, R1 and R2 are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
[0022] In certain compounds of the invention, both R1 and R2 contain a heterocycle or heteroatom.
[0023] For these therapeutic compounds a calculated log ratio of brain:blood concentration (cLogBB) of the quinoline methanol compound is less than a cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18, wherein LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule, such that the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 90% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 90% of the relative concentration of mefloquine in vivo.
[0024] Notably, when ( )n represents one carbon atom neither R1 or R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2- CHOH-CH2-CH3. CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2- COOH. The therapeutic compositions of the invention also include a pharmaceutically- acceptable carrier or physiologically-acceptable carrier such as a diluent, adjuvant, or a combination thereof.
[0025] The present invention provides therapeutic compositions that are less neurotoxic than mefloquine and that are antimalarials at least as efficacious as mefloquine against Plasmodium spp.. Preferably, these therapeutic compositions are also at least as efficacious as mefloquine against other diseases or conditions associated with infection by microbes, parasites, protozoans, bacteria, or fungi.
[0026] The present invention provides therapeutic compositions that can be used as antimalarial, antimicrobial, antiparasitic, antiprotozoan. antibacterial, or antifungal agents either alone, in combination with one another, or in combination with other therapies or compositions known in the art.
[0027] The compounds of the present invention alleviate neurotoxicity and improve therapeutic activity, while retaining the desirable properties of a practical and useful pharmacological agent, These principles are broadly applicable to the treatment and prevention of any of the conditions including infectious disease and immune disease against which mefloquine can be applied. Thus, the present invention provides mefloquine analog compounds and methods for identifying and making these less neurotoxic mefloquine analogs that also retain the properties of useful drug substances for treatment of a variety of diseases and conditions.
[0028] Quinoline methanol compounds and therapeutic compositions of the present invention may be included in a pharmaceutical preparation that is administered to a subject by at least one mode selected from the group consisting of oral, topcial, parenteral, subcutaneous, intramuscular, intradermal, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intraccervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal. Preferred routes of administration include oral, topcial, parenteral, subcutaneous, or intramuscular. Depending on the desired duration and effectiveness of the therapy, the compositions according to the invention may be administered once or several times, also intermittently, for instance on a daily or weekly basis for several days, weeks, or months in different dosages and by a combination of different routes.
[0029] The invention also provides methods of making therapeutic compositions and quinoline methanol compounds that are less neurotoxic than mefloquine as well as being efficacious as antimalarials, antimicrobials, antiparasitics, antiprotozoans, antibacterials, or antifungals. Preferably, these compounds may be utilized as preventative measures against or as treatment for malaria and other diseases and infections. [0030] The invention further provides methods of using the described therapeutic compositions to treat, reduce, or prevent diseases or conditions associated with infection by microbes, parasites, preferably Plasmodium spp., protozoans, bacteria, or fungii. The therapeutic compositions may be used alone, in combination with one another, or in combination with other therapies or compositions.
[0031] In another aspect, the invention provides a method of treating (e.g. curing, suppressing, ameliorating, delaying or preventing the onset of, or preventing recurrence or relapse of) or preventing a microbial, parasitic, protozoan, bacterial, or fungal associated disease or condition in a subject. The method includes: administering to a subject a quinoline methanol compound of the invention in an effective amount sufficient to treat or prevent the disease or condition. The quinoline methanol compound or derivative can be administered to the subject alone or in combination with other therapeutic modalities.
[0032] The invention provides a method of reducing the incidence of or severity of a clinical sign associated with a microbial, parasitic (preferably malaria, more preferably a Plasmodium falciparum infection), protozoan, bacterial, or fungal associated disease or condition, comprising the step of administering a therapeutic composition of the invention such that the incidence of or the severity of a clinical sign of malaria is reduced by at least 10%, preferably by 15%, 20%, 25%, 30%, 35%, 40% 45%, 50% or more, relative to a subject that has not received the therapeutic composition. Such clinical signs include fever, chills, sweats, headaches, muscle pains, nausea, vomiting, elevated temperature, perspiration, tiredness, confusion, coma, neurologic focal signs, severe anemia, and respiratory difficulties.
[0033] The invention also provides kits for administering a therapeutic composition of the invention comprising a set of printed instructions; a dispenser capable of administering a therapeutic composition to a subject; and at least one quinoline methanol compound of the invention that is therapeutically effective against at least one clinical sign associated with a microbial, parasitic (preferably malaria, more preferably a Plasmodium falciparum infection), protozoan, bacterial, or fungal associated disease or condition. Kits of the invention may further comprise a physiologically acceptable vehicle, carrier molecule, adjuvant, or combination thereof. A quinoline methanol compound of the invention and a physiologically acceptable vehicle, carrier molecule, adjuvant, or combination may be supplied in the kit either separately or in a premixed combination.
[0034] As used herein, the term "effective amount" refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity or duration of a disease or disorder, or one or more symptoms thereof, prevent the advancement of a disease or disorder, cause the regression of a disease or disorder, prevent the recurrence, development, onset, or progression of one or more symptoms associated with a disease or disorder, or enhance or improve the prophylaxis or treatment of another therapy or therapeutic agent.
[0035] Herein, a "subject" is a mammal, preferably a human, in need of either prophylactic or treatment for a microbial, parasitic, protozoan, bacterial, or fungal associated infection, disease, or condition.
[0036] "Protection against disease" and similar phrases, mean a response against a disease or condition generated by administration of one or more therapeutic compositions of the invention, or a combination thereof, that results in fewer deleterious effects than would be expected in a non-immunized subject that has been exposed to disease or infection. That is, the severity of the deleterious effects of the infection are lessened in a vaccinated subject. Infection may be reduced, slowed, or possibly fully prevented, in a vaccinated subject. Herein, where complete prevention of infection is meant, it is specifically stated. If complete prevention is not stated then the term includes partial prevention.
[0037] Herein, "reduction of the incidence and/or severity of clinical signs" or "reduction of clinical symptoms" means, but is not limited to, reducing the number of infected subjects in a group, reducing or eliminating the number of subjects exhibiting clinical signs of infection, or reducing the severity of any clinical signs that are present in one or more subjects, in comparison to wild-type infection. For example, it should refer to any reduction of pathogen load, pathogen shedding, reduction in pathogen transmission, or reduction of any clinical sign symptomatic of malaria. Preferably these clinical signs are reduced in one or more subjects receiving the therapeutic composition of the present invention by at least 10% in comparison to subjects not receiving the composition and that become infected. More preferably clinical signs are reduced in subjects receiving a composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
[0038] The term "increased protection" herein means, but is not limited to, a statistically significant reduction of one or more clinical symptoms which are associated with infection by an infectious agent, preferably a Plasmodium spp,, respectively, in a vaccinated group of subjects vs. a non-vaccinated control group of subjects. The term "statistically significant reduction of clinical symptoms" means, but is not limited to, the frequency in the incidence of at least one clinical symptom in the vaccinated group of subjects is at least 10%, preferably 20%, more preferably 30%, even more preferably 50%, and even more preferably 70% lower than in the non-vaccinated control group after the challenge the infectious agent.
[0039] Those of skill in the art will understand that the compositions used herein may incorporate known injectable, physiologically acceptable sterile solutions. For preparing a ready-to-use solution for parenteral injection or infusion, aqueous isotonic solutions, e.g. saline or plasma protein solutions, are readily available. In addition, the immunogenic and vaccine compositions of the present invention can include pharmaceutical- or veterinary- acceptable carriers, diluents, isotonic agents, stabilizers, or adjuvants.
[0040] As used herein, "a pharmaceutical- or veterinary- acceptable carrier" includes any and all solvents, dispersion media, coatings, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like. In some preferred embodiments, and especially those that include lyophilized immunogenic compositions, stabilizing agents for use in the present invention include stabilizers for lyophilization or freeze-drying.
[0041] "Diluents" can include water, saline, dextrose, ethanol, glycerol, and the like. Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others. Stabilizers include albumin and alkali salts of ethylendiamintetracetic acid, among others.
[0042] Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRPTION OF THE DRAWINGS
[0043] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
[0044] FIG. 1. General structure motifs.
[0045] FIG. 2. Synthesis of 4-position library.
[0046] FIG. 3. Exemplary active compounds that embody the diamine motif.
[0047] FIG. 4. Compounds WR318973 and WR177000.
[0048] FIG. 5. Diamine quinoline methanols with reduced brain penetration as compared to mefloquine.
[0049] FIG. 6. Diamines with additional lipophilicity.
[0050] FIG. 7. Putative active metabolites of WR318973.
[0051] FIG. 8. Generic Diamine Quinoline Methanols of Interest.
[0052] FIG. 9. Proposed synthetic route to construct N-oxide amine derivatives of WR318973.
[0053] FIG. 10. Proposed synthetic route to construct N-hydroxyl amine analogs of
WR318973.
[0054] FIG. 11. Structures of straight chain ethylene diamines.
[0055] FIG. 12. Synthetic scheme for straight chain ethylene diamines.
[0056] FIG. 13. Structure of pyrrolidine ethylene diamines.
[0057] FIG. 14. Synthetic schemes for pyrrolidine ethylene diamines. [0058] FIG. 15. Structure of bicyclic ethylene diamines.
[0059] FIG. 16. Synthetic schemes for bicyclic ethylene diamine 37.
[0060] FIG. 17. Synthetic schemes for bicyclic ethylene diamine 41.
[0061] FIG. 18. Synthetic schemes for bicyclic ethylene diamine 50.
[0062] FIG. 19. Generic bicyclic compounds of interest.
[0063] FIG. 20. Epoxide opening scheme [EtOH, uw, 30min, 130oC, 25Ow].
[0064] FIG. 21. Remaining diamine quinoline methanols of interest.
[0065] FIG. 22. Structures and IC90s of the most potent quinoline methanols. IC90s are against the mefloquine resistant Pf C2A strain.
[0066] FIG. 23. Reagents and conditions: (a) 1. POBr3, 75°C - 15O°C (91%) (b) n-BuLi, N,N-dimethylformamide.THF, -78°C (64%) (c) NaH, Me3SO, DMSO, THF (82%) (d) NaH, ROH, THF, O°C to rt (72-89%) (e) NaH, RSH, THF, O°C to rt (81-84%) (f) RMgBr, ether or THF, -78°C to rt (88-93%) (g) amine, 200 proof EtOH, microwaves, 25Ow, 13O°C, 15-30 min (18-97%).
[0067] FIG. 24. Actual versus calculated LogBB Values for Structurally Diverse
Quinoline Methanols,
DETAILED DESCRIPTION
[0068] The invention provides new quinoline methanol derivatives that are less able to cross the blood-brain barrier as compared to mefloquine and still retain the same, or even improved, therapeutic efficacy as mefloquine. These new compounds are expected to be useful pharmacological agents for the prevention or treatment of malaria, and other microbial, parasitic, protozoan, bacterial and fungal diseases and potentially of Parkinson's disease or diseases associated with prions.
[0069] The present invention relates to several new quinoline methanol compounds and pharmaceutical compositions containing one or more compounds within these. The invention embodies a series of new antimalarial compounds in which the quinoline methanol scaffold has been optimized in terms of the structure modifications and physiochemical properties required for excellent oral bioavailability, equivalent and/or improved potency as compared to mefloquine and reduced potential for blood brain barrier penetrability/brain uptake than mefloquine. These properties were mediated by the incorporation of 4 position side chains imparting lower lipophilicity and higher polar surface area to the compounds. The general structure of these new compounds is provided in Formula I.
Figure imgf000014_0001
Formula I
In Formula I, ( )n represents one, two, or more carbon atoms; R1 is a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, or a cyclic alkyl group containing a nitrogen atom; R2 is a hydrogen atom, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, or a cyclic alkyl group containing a nitrogen atom; and R3 represents at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a trifluromethyl group, a OH group, an oxygen atom, a hydrogen atom, or a combination thereof. Preferably, a trifluromethyl group is at position 8, more preferably a trifluromethyl group is at position 8 and position 6 or 7; and even more preferably a trifluromethyl group is present at positions 6, 7, and 8.
[0070] Notably, when ( )n represents one carbon atom then neither R1 nor R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2- CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2- COOH. Whereas, in quinoline methanol compounds where ( )n represents two or more carbon atoms either Rj or R2 may be selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2- CH2-CH2OH, CH2OH, CH2-CH2-COOH, other straight chain alkyl groups, cyclic alkyl groups, straight chain alkyl groups containing a nitrogen atom, or cyclic alkyl groups containing a nitrogen atom. [0071] These new compounds have physiochemical properties, such as Log P (water- octanol partition coefficient) values, polar surface area (PSA) and/or freely rotatable bonds (FRBs), which limit the ability of these compounds to penetrate the blood brain barrier and enter the central nervous system (CNS). These new compounds have equivalent and/or improved activity over known drugs from this general class of compounds - i.e., mefloquine.
[0072] Conventional quinoline methanols, such as mefloquine, penetrate the CNS and cause adverse CNS effects. The new compounds within the scope of the present invention have been engineered to avoid or limit penetration of the CNS, and thereby prevent or reduce CNS adverse events. They, therefore, offer a potential advantage over existing drugs.
[0073] The novelty of these compounds is found in their biological and chemical properties. There are no drugs presently available from this general class of compounds (quinoline methanols) with the same pharmaceutical activity which also exhibit propensity to not cross the blood-brain barrier. In addition, the chemical structures of these compounds are different from those compounds within the class that are available commercially or that might be reported elsewhere.
[0074] The compounds within the scope of the invention may be employed in pharmaceutical compositions, wherein said compositions may comprise an effective amount of one or more of these compounds, and a pharmaceutically acceptable carrier. The effective amount of these compounds will vary based upon the use for which the composition is intended.
[0075] The compounds and compositions within the scope of the present invention may be administered to a patient via any conventional route of administration. Selection of a suitable, pharmaceutically acceptable carrier is well within the skill of the art depending on the method of administration sought to be employed - i.e., oral, intravenous, etc.
[0076] These compounds and compositions containing them may be used in methods for preventing and/or treating malaria, methods for preventing and/or treating other infectious diseases, or methods for preventing and/or treating certain immunological conditions. In addition, compounds and compositions within the scope of the present invention may be used in the intermittent preventive treatment of malaria. Reference to treatment herein includes intermittent preventive treatment. [0077] The invention is suitable for use against all species of malaria (i.e., blood stages of all malaria parasites). In addition, the invention is suitable for use against diseases caused by bacteria, protozoa, parasites, and fungi, and against some autoimmune diseases (i.e.. rheumatoid arthritis).
[0078] Compounds within the scope of the present invention are generally represented by
Formula I. Subsets of compounds of Formula I are further described by the formulas set forth a as Series A, Series B, Series C, and Series D in FIG. 1. In general, R1, R3, R4, and R5 in these formulas represent substituents that render a quinoline methanol less able to cross the blood- brain barrier than mefloquine - see, for example, Tables 1-3 for illustrative substituents. This reduction is because quinoline methanols represented by these formulas exhibit lower LogP, or higher polar surface area (PSA), or have a greater number of freely rotatable bonds (FRBs), or a combination of all three of these properties as compared to mefloquine. These changes in physiochemical properties are known to correlate with lower blood-brain barrier permeability. (In Tables 1 and 2 the combined effects of different PSA, LogP and FRBs for each analog is expressed as the parameter cLogBB or the calculated log ratio of brain:blood concentration. Use of cLogBB is one method available to assess the sought after properties herein. The individual values of LogP, PSA and FRBs can be derived from the chemical structures using standard physiochemical prediction software (e.g. ACD). Furthermore, this reduced penetration of the blood-brain barrier is achieved without loss of functionality critical to maintaining intrinsic activity which is essential for their utility as potential drugs. R and R (see FIG. 1, Series B) may include cyclic and alkyl tertiary amines. They may also join one to the other to form a substituted or unsubstituted heterocyclic ring. R (see FIG. 1, Series C and D) represents a substitution at one or more of the 2, 3, 4, or 5 positions of the imidazole ring or triazole ring.
[0079] R2 denotes any substituent at the 6, 7 or 8 positions of the quinoline ring, including but not limited to CF3 in the 8 position.
[0080] The notation ( )n indicates that the structures represented by Series A, Series B,
Series C, and Series D contain one or more carbon atoms positioned between the C-OH and the N atoms of the 4 position side chain as shown in FIG. 1. Analogues wherein n=l or 2 were found to be of particular interest; however, the invention is not so limited. Any suitable value for "n" wherein the compounds illustrate the desirable properties described herein is within the scope of the present invention.
[0081] Compounds within the scope of the present invention that are of particular interest are set forth as general structures in FIG.l, and specific exemplary compounds are described in Table 1, Table 2 and Table 3. Herein, compounds of particular interest are referred to as diamine quinoline methanols (see FIG. 1). These compounds are illustrative of the types of compounds within the scope of the invention. The present invention, however, is not intended to be limited to the compounds identified in these tables. One having ordinary skill in the art, and equipped with the description set forth herein, would know or be able to identify what substituents would be suitable for attaining the sought after properties of interest herein. As set forth above, for example, "with reference to R1, R3, R4, and R5 suitable substituents are those that render quinoline methanols less able to cross the blood-brain barrier relative to mefloquine without imparting a loss of potency.
[0082] The analogs and compounds described herein were made in a single step nucleophilic substitution using heat or microwave radiation. They were made from a commercially available epoxide, 2-[2,8-bis(trifluromethyl)-4-quinolyl]oxirane (obtained from BioBlocks, San Diego, California) and commercially available nucleophiles (mostly amines obtained from Maybridge, Cornwall, United Kingdom). See FIG. 2.
[0083] Compounds within the scope of the invention may be made using any suitable method. One skilled in the art would be able to identify and determine other methods of making the compounds within the scope of the present invention. The method of making the compounds herein is not intended to be so limited.
A. Pharmacological Properties of Diamine Quinoline Methanol Compounds
[0084] A series of in vitro and in vivo permeability/brain uptake and susceptibility/efficacy and in vivo PK studies were executed. These studies, outlined below and detailed in the Examples herein, reduce to practice, the utility of diamine quinoline methanols, since they are less penetrant than non-diamine comparators and maintain useful potency and efficacy and plasma levels relative to mefloquine. [0085] Specific diamines such as WR318973 cross the blood-brain barrier to a lesser extent than similar compounds that do not contain a second nitrogen atom (FIG. 4). For example, 5 min after an intravenous (i.v.) dose of 5 mg/kg administered to FVB mice, brain concentrations of WR318973 were 129 ng/g compared to 8811 ng/g for WR177000.
[0086] More generically, the presence of a second nitrogen atom on the side chain makes such analogs less able to penetrate the blood-brain barrier. For example, diamine analogs such as WR318746, WR318744 and WR319535 exhibit lower brain concentrations 5 min after i.v. dosing than mefloquine (FIG. 5). Brain concentrations were 100, 243, 199 and 550 ng.g respectively for WR319535, WR318744 and WR318744 and mefloquine respectively. As a consequence, they are likely to be better tolerated than mefloquine due to lower exposure of multiple vulnerable CNS targets to the drugs. More generally, quinoline methanols with the requisite diamine motif are more likely to exhibit less blood-brain barrier penetrability than non- diamines and be better tolerated as a consequence.
[0087] The reduction in lipophilicity associated with the second nitrogen is also associated with improved pharmacokinetic properties. For example, the clearance of WR177000 is 2681 ml/hr/kg versus 912 ml/hr/kg for WR318973. This means that the plasma concentration of WR318973, 24 h after dosing are still measurable at 83 ng/ml, whereas this is not the case for WR177000. In some diamines the proportion of compound remaining 24 h after an i.v. 5 mg/kg dose is comparable to mefloquine. For example 26% of WR308621 in plasma remained at 24 h versus 26-31% for the mefloquine isomers, meaning that, generically, diamine quinoline methanols may be potentially applicable for treatment indications such as intermittent preventive treatment of malaria or malaria prophylaxis where maintenance of adequate plasma concentrations over substantial periods of time is essential.
[0088] The utility of diamine quinoline methanols as antimalarials is partially dependent on their potency. Potency against malaria parasites is assessed by measuring an IC50 against well- characterized laboratory strains of P. falciparum such as W2 and C2A. Diamine quinoline methanols are unique in that the reduction in lipophilicity generated does not compromise the utility of the compounds by reducing potency to the point of inactivity. Furthermore, the potency of diamines is dependent on restricting access to the second amine, the positioning of the two nitrogen atoms relative to each other and cyclization of the side chain. For example, WR318972 (FIG. 6) and WR318746 have IC90s of 30 and 69 ng/ml against Pf W2 versus 490 ng/ml for WR318973 because access to the second amine is restricted. This trend is even more prominent when one considers an analog such as WR308396, with an IC90 of 6 ng/ml, in which a large lipophilic group is added to the terminal portion of the side chain. Also, analogs in which the second amine is three atoms from the first nitrogen are more potent. For example, the IC90 of WR308782 (FIG. 6) is 69 ng/ml compared to 6 ng/ml for WR308396. Finally, cyclization of the side chain improves potency. As an example, WR308621 has an IC90 of 46 ng/ml versus 490 ng/ml for WR318973. Thus diamines in which access to the second amine is restricted, in which the terminal nitrogen is cyclized and/or in which the second nitrogen is three atoms from the first, are particularly favored.
[0089] The utility of a quinoline methanol for malaria or other indications is dependent on oral bioavailability. Diamine quinoline antimalarials can be inferred to be bioavailable based on in vivo efficacy studies. WR318973 cured 4 of 5 mice infected with P. berghei after oral administration of a single dose of 320 mg/kg was administered orally (100% of untreated controls are not cured). This result compares favorably to mefloquine, in which 11/18 mice were cured at the same dose. This observation suggests that WR318973 is sufficiently orally bioavailable that enough of the compound reaches the blood to eliminate malaria parasites. Interestingly, WR318973 is effective in vivo despite lower potency relative to other related diamines. It is possible that some of its effects in mice may be mediated by putative diamine active metabolites. These are illustrated in FIG. 7.
[0090] Based on the biological data, diamines of the generic structure outlined below as
Series C, D and E (FIG. 8) are particularly favored embodiments of the present invention. Generally R groups 1 through 6 represent any functionality that improves the intrinsic properties of the diamine scaffold including but not limited to aliphatic or aromatic or heterocyclic or cyclic substituents. They could be methyl groups that block access to the second amine or the second nitrogen could be incorporated into a cyclic structure in order to improve potency. The substituents might increase the molecular weight of the compound in order to render them PGP substrates and thus less penetrant of the blood-brain barrier. The substituents could result in additional increases in polar surface area and or decreases in lipophilicty in order to further decrease permeability. Alternatively blocking groups could be added to sterically hinder sites of metabolic attack. The symbol ( )n indicates that there are 1 or more carbon atoms in the side chain either between the hydroxyl and first nitrogen and/or between the first and second nitrogen atoms. Compounds with two carbon atoms between the nitrogen atoms are particularly favored. R8 denotes any substituent attached to the 6, or 7, or 8 positions of the quinoline ring, including but not limited to CF3 at the 8 position. R7 is intended to be a substituent that renders the compound a tertiary amine, including, but not limited to compounds in which the substituent at R7 is joined to another R1-6 substituent to create a cyclic species. Substituents at R1, R2 and R7 might be O or OH groups in order that the compounds be mono or di hydroxyl amines or N- oxides.
B. Construction of Quinoline Methanol Compounds
[0091] Figure 2 outlines the strategy used to synthesize a 4-position library of methanol quinoline compounds. The structure of mefloquine is indicated at (1). The intermediate scaffold 4-(oxiran-2-yl)-2,8-bis(trifluoromethyl)quinoline (3) was synthesized from bis(trifluoromethyl)quinolin-4-ol (2) by the addition of POBr3 at 75°C to 15O°C for 2 hours with 91 % yield. The resulting 4-bromo-2,8-bis(trifluoromethyl)quinoline was dissolved in tetrahydrofuran, cooled to -78°C and subjected to a n-butyllithium. N, N,-dimethylformamide was subsequently added to afford 2,8-bis(trifluoromethyl)quinoline-4-carbaldehyde. Utilization of Corey's dimethylsulfonium methylide provided racemic epoxide (3). The epoxide (3) can also be purchased commercially from Bioblocks (San Diego, California). The quinoline scaffold (3) was diversified at the 4-position in a single step with commercially available nucleophiles.
[0092] Well over two hundred quinoline methanols were synthesized in 20 mg quantities and submitted for in vitro screening. Compounds exhibiting IC90s < 250 nM (approximately equivalent to the upper limit of mefloquine) against Pf C2A and an LC50 against mammalian cells no worse than mefloquine were considered for in vivo studies. The structures of these compounds were examined and ten interesting compounds were selected for further in vivo, ADME and CNS screens. Based on these data, a single compound was selected as a lead around which to synthesize new series of compounds. [0093] Compounds particularly favored can be constructed using the methodologies outlined below. In order to construct the N-oxide compounds (3) and (4) from WR318973 (1), the benzylic alcohol and the first amine on the side chain must be protected (FIG. 9). Mixed acetal (2) will be constructed from isobutyraldehyde utilizing the standard protocol in dichloromethane, which allows the second amine to be oxidized with a variety of oxidants. As shown in FIG. 9, t-butyl peroxide promoted oxidation is of interest since it was shown to not oxidize heterocyclic nitrogen atoms (see J. D. Fields and P. J. Kropp. Journal of Organic Chemistry, 2000, 65, 5937-5941). Another possibility will entail the use of 2- sulfonyloxaziridines (Davis' reagents) for the transformation to N-oxides (see W. W. Zajac. T. R. Walters, and M. G. Darcy, Journal of Organic Chemistry, 1988, 53. 5856-5860). If these routes are not successful, there are a variety of procedures to form N-oxide (3) or its tautomer (4). In order to de-protect the mixed acetal, trimethylsilyl iodide in acetonitrile can be utilized. To form dioxide (6), the benzylic alcohol can be protected as the silyl ether. Similar oxidation conditions can be tested followed by tetrabutylammonium fluoride or some other mild deprotection in order to remove the triethyl silane. To form mono-oxide tautomers (9) and (10), a similar sequence can be employed starting with epoxide (7). Utilizing a microwave-assisted nucleophilic epoxide opening, which has been optimized, followed by selective protection of the benzylic alcohol.
[0094] The synthetic routes utilized to construct the putative metabolites are illustrated in FIG. 10. In order to form the N-hydroxyl amine, a procedure reported by Fields and Kropp (J.
D. Fields and P. J. Kropp, Journal of Organic Chemistry, 2000. 65, 5937-5941, incorporated herein by reference) can be used. Despite excess oxidant, only the N-hydroxyl amine was isolated when utilizing OXONE on silica. Trimethylsilyl iodide and acetonitrile (G. A. Olah and
S. C. Narang, Tetrahedron, 1982, 38, 2225, incorporated herein by reference), can be utilized as a mild deprotection strategy for the mixed acetal and t-butyl carbamate, respectfully.
[0095] Particularly favored are so called straight chain ethylene diamines depicted in FIG. 11. These can be made by a three-component- 1 -pot synthesis as outlined in FIG. 12. First, the desired tosyl-protected aziridine 13, with various R substitution patterns, is constructed from the corresponding amino alcohol (Loethar W. Bieber and Mariea C. F. de Araujo. Molecules, 2002, 7, 902-906 incorporated herein by reference) or commercially available aziridines. The tosyl group protects the secondary amine, activates the aziridine to nucleophilic attack (SN2), and is an ultraviolet tag to monitor the reaction. Tosyl-protected aziridine 13 (l.Oeq) is dissolved in ethanol (200 proof, 0.25M), amine 14 (l.leq) is added, and the reaction mixture is subjected to microwaves (130°C, 250 watts) for 30 minutes. In order to remove the tosyl group, 1.25M HCl/EtOH (1.5eq) is added and the reaction mixture is further subjected to microwaves (13O°C, 250 watts) for 30 minutes. 2,2,6, 6-Tetramethylpiperidine (3.0eq) is added to increase the pH ~ 9, followed by the addition of epoxide 7, and the reaction mixture is further subjected to microwaves (13O°C, 250 watts) for 30 minutes. The ethanol is removed in vacuo and the reaction mixture is purified via normal phase preparative thin layer chromatography in 95:5 dichlomethane: methanol to yield 28-43% yield over three steps. The conditions can be optimized to increase yields.
[0096] Pyrrolidine ethylene diamines as depicted in FIG. 13 are also particularly favored embodiments of the present invention. These can be synthesized using the following described methods or variations thereof as illustrated in FIG. 14. By utilizing chiral enolates of pseudoephedrine amides 18 and azirides such as 13, substituted pyrrolidin-2-ones 20 can be prepared in >99% enantiomeric excess (see Jose L. Vicario, Dolores Badia, and Luisa Carrillo, J. Org. Chem. 2001, 66, 5801-5807, incorporated herein by reference). Treating pyrrolidin-2-one 20 with Swartz' reagent (Cp2ZrHCl) and cyanotrimethylsilane (TMSCN) affords the corresponding α-aminonitrile 21 (see Q. Xia and B. Ganem. Tetrahedron Letters, 2002, 43, 1597-1598, incorporated herein by reference). Subsequent protection of the amine will afford 22, followed by either direct reduction to 23 or treatment with Grignard reagent to yield gem- dimethyl substituted pyrrolidine 24. In order to install a methyl group alpha to the amine, a three-step sequence will be employed to transform pyrrolidin-2-one 20 to α-methylaminonitrile 25, which can be reduced directly to amine 26 or treated with Grignard reagent to afford substituted pyrrolidine 27. With substituted pyrrolidine 27 (23, 24, or 26) in hand, utilization of our microwave-assisted epoxide opening and subsequent deprotection will be utilized to afford HCl salt 28.
[0097] Bicyclic ethylene diamines are also particularly favored embodiments of the present invention (FIG. 15). These can be synthesized as follows or similar methods thereof as outlined in FIGs. 16-20. FIG. 16 illustrates the proposed synthesis of bicyclic amines such as 37. Utilizing similar strategies employed in the synthesis of the pyrrolidines (FIG. 14), carboxyglutamic acid derivatives such as 31 can be subjected to Swartz' reagent (Cp2ZrHCl) and cyanotrimethylsilane (TMSCN) to afford the corresponding α-aminonitrile 32 (Q. Xia and B. Ganem. Tetrahedron Letters, 2002, 43, 1597-1598, incorporated herein by reference). Subsequent protecting and ring closing metathesis utilized Grubb's catalyst (see Handbook of Metathesis; Grubbs, R. H., Ed.; Wiley- VCH: Weinheim, Germany, 2003, incorporated herein by reference) should afford 34. Reduction to the primary amine should yield 35, while treatment with Grignard and reduction of the olefin should yield 36. The bicyclic diamine can be utilized in microwave-assisted epoxide opening to yield 37.
[0098] The synthesis of compound 41 will proceed in a similar manner as outlined in Fig.
12, where aziridine 13 and commercially available amine 38 will be subjected to microwaves to yield 39 as shown in FIG. 17. Deprotection under acidic conditions and microwaves should yield 40. After adjustment of the pH with 2,2,6.6-tetramethylpiperidine and subsequent microwave-assisted epoxide opening should yield 41.
[0099] Some specific bicyclic compounds of interest are presented in FIG.19. As an example, compounds such as Compound 50 can be made as outlined in FIG 18. Methyl-4- hydroxybenzoate 40 (FIG. 17)was reduced to the corresponding aliphatic cyclohexane and oxidized to ketone 43. A Bucherer-Bergs reaction was utilized to establish the amine and carboxylic acid group in a cis configuration with spirohydantoinic compound 44. Subsequent saponification, formation of the acid chloride, and cyclization led to lactam 46. Protection as the benzylamine provided 47, which was reduced utilizing diphenylsilane and rhodium catalysts provided bicyclic compound 48 (D. Casabona and C. Cativiela. Tetrahedron, 2006, 62, 10000- 10004, incorporated herein by reference). From this point, the amide should be formed upon treatment with ammonia followed by reduction to primary amine 49, which will be subjected to microwave-assisted epoxide opening and subsequent reduction to yield compound 50.
[00100] Other diamines of interest, depicted in FIG. 21, may also be synthesized by epoxide opening as depicted in FIG. 20. The diamine can be purchased commercially or constructed in house and utilized in microwave-assisted epoxide opening. C. In vivo Screening of Compounds
[00101] Compounds were tested at two different doses in groups of five mice: 160 mg per kilogram of bodyweight per day for three days, and as a single dose of 320 mg/kg. All dosing was oral. Details of tests are provided in the Examples.
[00102] The activity of a compound in this model is reflected as number cures relative to the number of mice experimentally infected (#cures/#tested). Control mice (those administered no drug) died or were humanely euthanized. The control data are not shown. The term #toxic/#infected represents how many of five mice in each treatment group experienced signs of acute toxicity. The results of the tests conducted are set forth in Table 3.
D. Table Legends
[00103] Table 1: Summary of physiochemical properties and biological data for novel quinoline methanols (NGQM - Next Generation Quinoline Methanols). The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
[00104] Table 2: Summary of physiochemical property and biological data for novel quinoline methanols. The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
[00105] Table 3: In vitro Plasmodium falciparum (PF) screening and in vivo efficacy and toxicity data for selected compounds of interest. The properties for which data are provided are defined, with a short description of methodology used to derive them outlined in the "Definitions" herein.
[00106] Table 4. Physiological properties of the 4-position library,
[00107] Table 5. Pharmacological data for compounds selected for further screening.
[00108] Table 6. Physiochemical properties of potent, active and inactive compounds.
[00109] Table 7. Properties of different functional groups present in active and inactive amines. [00110] Table 8. Plasmodium falciparum IC90 (ng/ml) values resulting from probing hydroxyl utility.
[00111] Table 9. Plasmodium falciparum IC90 (ng/ml) values resulting from probing amine utility.
[00112] Table 10. Plasmodium falciparum IC90 (ng/ml) values for phenyl, benzyl, and phenethylamino QMs.
[00113] Table 11. Plasmodium falciparum IC90 (ng/ml) values and selectivity of heterocyclic amino quinoline methanols (HAQMs).
[00114] Table 12. Plasmodium falciparum IC90 (ng/ml) values for alkyl amino quinoline methanols (AAQMs).
[00115] Table 13. Plasmodium falciparum IC90 (ng/ml) values for additional branched alkyl amino quinoline methanols (AAQMs).
[00116] Table 14. Plasmodium falciparum IC90 (ng/ml) values for alkyl amino quinoline methanols containing additional heteroatoms (AAQMHs).
[00117] Table 15. In vivo efficacy of selected quinoline methanols in the P. berghei mouse model and permeability across MDRl-transfected MDCK cell monolayers.
E. Definitions
[00118] "LogP" is the partition coefficient reflecting the relative solubility of a drug in octanol versus water. The higher the value, the lower the water solubility. Generally a reduction in the LogP is associated with reduced permeability across the blood brain barrier. LogP can be predicted from the structure of a compound using standard physiochemical prediction software (e.g. ACD).
[00119] "PSA" is the polar surface area of a molecule and is a reflection of the polarity of the molecule. Generally, higher PSA is associated with reduced permeability across the blood brain barrier. PSA can be predicted from the structure of a compound using standard physiochemical prediction software (e.g. ACD). [00120] "FRBs" is the number of freely rotatable bonds a compound has. A greater number of freely rotatable bonds generally correlates with lower blood-brain barrier permeability. FRBs can be determined from the structure of a compound using standard physiochemical prediction software (e.g. ACD).
[00121] "cLogBB" is the calculated log ratio of the brain:plasma concentration of a drug calculated using the following formula: cLogBB = (0.205 * LogP) - (0.0094*PSA) - (0.055 * FRBs) + 0.18. This is a composite index that combines the effects of PSA, LogP, and FRBs.
[00122] "PF IC90 (ng/ml)" is the 90% inhibitory concentration in ng/ml of a molecule against P. falciparum in an in vitro cell based growth inhibition assay. Four different strains of drug resistance P. falciparum (W2, D6, C235 and C2A) were used. The lower the value, the more active the molecule.
[00123] "Macrophage IC50 (μM)" is the 50% inhibitory concentration in micromolar units of a molecule in an in vitro cytotoxicity assay against a rodent macrophage cell line. The lower the value, the more toxic the molecule.
[00124] "Lip. Viol." is the number of violations of Lipinski's rule of 5. This index relates the likelihood that a compound will be orally bioavailable based on its physiochemical properties. The fewer Lipinski violations, the greater the likelihood of a compound being orally bioavailable.
[00125] "P. berghei-mice" is the animal model that is used to evaluate the potential utility of new antimalarial compounds in a discovery setting. P. berghei, or Plasmodium berghei, is a rodent malaria parasite that induces a lethal infection in mice. For quinoline methanols, all commercially available compounds clinically effective against human malaria parasites are also active in this model.
[00126] Herein, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including", as well as other forms such as "includes" and "included" is not limiting.
[00127] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs at the time of filing. The meaning and scope of terms should be clear; however, in the event of any latent ambiguity, definitions provided herein take precedent over any dictionary or extrinsic definition. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. All patents and publications referred to herein are incorporated by reference herein.
EXAMPLES
[00128] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventors to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
A. Materials and Methods 1. In Vivo PK Studies
[00129] For selected quinoline methanols brain and plasma concentrations were determined in vivo after intravenous dosing. Groups of 2-4, seven-eight week old male FVB mice were administered a 5 mg/kg dose of quinoline methanol base. The drug was given as a 50 microliter bolus dose in a drug vehicle consisting of 5% w/v glucose, and/or 0-5% dimethylsulfoxide, and/or 0-1% solutol in 20 mM citrate or acetate buffer at pH 3-4 with the specific buffer characteristics being determined for specific compounds based on solubility. At 5 min, 60 min, 6h and 24h plasma and brain samples were obtained from the mice and frozen at -8O°C until they were analyzed. On the day of analysis, whole brains were homogenized in 3 parts of aqueous solution containing 0.1 M EDTA and 0.4 g/L KF (inhibitors of hydrolytic enzymes to reduce the potential for ex vivo tissue-mediated degradation of the test compounds). Calibration standards were prepared by spiking blank brain homogenate and blank plasma with the test compound. Both samples and standards were processed by adding acetonitrile (to precipitate proteins) and centrifuging to separate the supernate for analysis. An aliquot of supernatant from each brain homogenate and plasma sample was analysed to determine quinoline methanol concentrations by liquid chromatography/mass spectrometry. Drug concentration-time data was analyzed using WinNonLin to generate PK parameters using a non-compartmental model.
2. In Vitro Susceptibility Studies
[00130] The 50% and 90% inhibitory concentrations (IC50 and IC90) of the analogs were determined against four drug resistant strains of Plasmodium falciparum in vitro. These strains were W2, D6, C235 and C2A. W2 is chloroquine-resistant, D6 is mefloquine resistant, and the
C235 and C2A strains are resistant to multiple drugs. The IC50 and IC90 values represent the concentrations in ng/ml at which the incorporation of tritiated hypoxanthine is inhibited by 50% or 90% respectively. Since hypoxanthine incorporation is essential for parasite growth, inhibition of its action by drug uptake is a measure of the drug's toxicity to the parasite.
Therefore the lower the IC50 or IC90 of a drug, the greater is its potency.
3. In Vitro Toxicity Assays
[00131] The LC50 of some of the analogs was determined in vitro against a RAW macrophage cell line. The LC50 is the 50% lethal concentration, and represents the concentration at which colorimetric conversion of a substrate by cell mitochondria is inhibited by 50%. Since mitochondrial activity is indicative of cell viability, the decline substrate conversion is interpreted as the lethal effect of a drug on cells. Thus the lower an LC50, the more potent its toxic effects. Selectivity index relates toxicity to mammalian cells with antimalarial activity by calculation of the ratio of the macrophage LC50 to the IC50 against a parasite line such as W2. The greater the selectivity index the greater the selectivity of the antimalarial effect.
4. MDCK Permeability Assay:
[00132] MDRI-MDCK monolayers were grown to confluence on collagen-coated, microporous, polycarbonate membranes in 12-well Costar Transwell@ plates. The permeability assay buffer was Hanks Balanced Salt Solution containing 10 mM HEPES and 15 mM glucose at a pH of 7.4. A known p-glycoprotein inhibitor cyclosporin A (CSA) was also added to the assay buffer at 10 mM. The dosing solution concentrations of the test compound were 2.5 and 5.0 mM in the assay buffer. All cell monolayers were first pre-incubated for 30 minutes with assay buffer to saturate any P glycoprotein sites with test compound. After 30 minutes, the buffer was removed, replaced with fresh buffer, and time was recorded as 0. Cell monolayers were dosed on the apical side (A-to-B) or basolateral side (B-to-A) and incubated at 37°C with 5% CO2 in a humidified incubator. After 2 hours, aliquots were taken from the receiver chambers. Samples were taken from the donor chamber at 0 and 2 hours. Each determination was performed in duplicate. The lucifer yellow flux was also measured for each monolayer to ensure no damage was inflicted to the cell monolayers during the flux period. All samples were assayed by LCIMSIMS using electrospray ionization. The apparent permeability, Papp, and percent recovery were calculated as follows:
Papp = (dCϊ Idt) x W(A x CO) (I)
Percent Recovery = 100 x ((Vr x Crfinal) + (Vd x Cdfina1)) / (Vd x CN) (2) where, JCr Idt is the slope of the cumulative concentration in the receiver compartment versus time in μM s-1.
Vr is the volume of the receiver compartment in cm3.
Vd is the volume of the donor compartment in cm3.
A is the area of the cell monolayer (1.13 cm2 for 12- well Transwell®).
CO is the measured concentration of the donor chamber at time 0 in mM.
CN is the nominal concentration of the dosing solution in mM.
Crfinal is the culnulative receiver concentration in mM at the end of the incubation period.
Cd ina is the concentration of the donor in mM at the end of the incubation period.
EXAMPLE l: Physiochemical Properties and Biological Data
Methods
[00133] Over two hundred next generation quinoline methanols were synthesized. All analogs were modified at the 4 position. The synthesis was designed to provide rapid access to a broad range of chemotypes at the 4-position in a single step from the 4-(oxiran-2-yl)-2,8- bis(trifluoromethyl)quinoline scaffold using the general reaction scheme outlined in FIG. 2. A small subset of compounds was synthesized by alternative means (data not shown). Compounds were designed to be rule of 5 (R05) compliant (Lipinski, 2000, J. Pharmacol. Toxicol. Methods 44: 235-49, incorporated herein by reference) and to encompass the widest feasible range of LogD, LogP, freely rotatable bonds (FRBs), polar surface area (PSA), hydrogen bond donors (HBDs) and hydrogen bond acceptors (HBAs, see Table 4). The in vitro IC90 values of the compounds against four different strains of P. falciparum. W2, D6, C235 and C2A, and LC50 values in a cytotoxicity assay were determined as described herein (Dow et al., 2008a).
[00134] Interesting compounds were evaluated in several secondary assays including metabolic stability assessments using in vitro liver microsomes, drug-drug interaction assays, in vivo efficacy in the P. berghei mouse model as single and three day dose regimens and against neurons in vitro as previously described by Dow et al. ,2006a and Dow et al., 2008a, incorporated herein by reference. Inhibition of the A2A and Al receptors was determined at 200 nM in duplicate by Caliper Biosciences (Hanover, Maryland). The A2A and Al receptors are suspected to be two of many targets of mefloquine in vivo (Gillespie et al., 2008, Weiss et al., 2003). Permeability across MDRl -transformed MDCK cell monolayers was determined by Absorption Systems (Exton, Pennsylvania) determined as previously described (Wang et al., 2005, incorporated herein by reference) with appropriate modifications to maximize the permeability of mefloquine. These were the incorporation of 1% BSA into recipient wells, co- incubation with the PgP inhibitor cyclosporine A at 10 μM and extension of the normal incubation time to 120 min after an initial 30 min incubation. This screen is a standard in vitro assay for blood-brain barrier permeability. All physiochemical properties were calculated using ACD (Version 10, ACD Labs, Toronto, Canada) except LogD (pH 7.4) which was determined using Pipeline Pilot (Version 6.1, Accelrys, San Diego California).
Results
Synthesis and primary screening.
[00135] One hundred ninety eight quinoline methanols were synthesized and subjected to the screening paradigm laid out in FIG.2. The ranges of physiochemical properties represented by the library are outlined in Table 4. The primary screening data and physiochemical properties of all compounds are presented in Tables 1 and 2. Of an original 198 compounds, 24 (12%) exhibited IC90s less than 250 nM and cytotoxicity similar or better than mefloquine. The structures of these compounds and their IC90s are presented in FIG. 22. Ten of these compounds were selected for screening in various secondary assays based on their chemical structures. WR308278 and WR308396 were selected as they were the only compounds containing heteroatoms in the side chain that were more potent than mefloquine. WR308245, WR308255, WR308257 and WR308266 were selected because they were approximately equivalent or superior to mefloquine in terms of potency, but contained less steric bulk in their side chains. WR308387, WR.308388, WR308413 and WR.308446 were selected as they were substantially more potent than mefloquine in vitro.
Secondary screening.
[00136] The rank order of mefloquine and selected quinoline methanols in terms of potency against the four strains of Pf were similar in most cases (Table 5). Most of the compounds screened were more metabolically labile than mefloquine (Table 5). The exceptions were those in which the side chain was presumably less susceptible to N-dealkylation (WR308245 and WR308257) and the diamine WR308396. In the in vivo experiments, only WR308245 and WR308257 exhibited similar cure rates to mefloquine after administration of a single dose of 320 mg/kg. There was a significant correlation between the half- life in mouse microsomes to the therapeutic outcomes in mice after administration of a single dose (r = 0.51, P = O 014) of 320 mg/kg but not with the three day regimen. The failure of WR308396 in vivo despite its metabolic stability is an outlier relative to this general trend. All the compounds except WR308245 and WR308255 exhibited more potent inhibitory effects on cP450 2D6 than the other isoforms. All the compounds were less neurotoxic than mefloquine WR308245, WR308257, WR308278 and WR308387 were more permeable across MDCK-MDRI cell monolayers than mefloquine (Table 5). WR308396 exhibited slightly lower permeability than mefloquine and half the permeability of WR308387 (Table 5). The inhibition of the A2A and Al receptors by four of the analogs at 200 nM was evaluated (Table 5). In most cases the level of inhibition observed was lower or comparable to that observed with mefloquine. The exception was WR308245 against the Al receptor.
Relationship between activity and physiochemical properties.
[00137] The physiochemical properties amongst inactive compounds, active compounds with IC90s < 500 ng/ml or 1000 nM and active compounds with IC90s < 250 nM were compared. The threshold for inactivity was set at an IC90 > 500 (or the approximate corresponding concentration of 1000 nM) since this was the highest concentration tested in the assay. There were no significant differences amongst these groups in terms of LogD, HBDs and FRBs (Table 6). Lo gP was significantly higher. PSA significantly lower, and the number of HBAs significantly lower in active compounds (Table 6). The most active compounds exhibited lower molecular weights (MW) than less potent and inactive compounds.
Structural characteristics of active and inactive compounds.
[00138] Analogs were categorized arbitrarily as having particular structural motifs. The proportion of active (IC90 < 500 ng/ml or 1000 nM) and inactive compounds containing these functional groups was determined and differences between the groups were tested for significance using Fisher's Exact test (Table 7). The inactive group contained a greater proportion of compounds in which the 4 position amino side chain contained additional heteroatoms, analogs in which the hydroxyl group or amine functionality were replaced, and compounds in which the first nitrogen atom in the side chain was conjugated. The active groups of compounds contained a higher proportion of secondary amines and compounds in which the amino side chain contained no additional heteroatoms. The inactive and active groups contained similar proportions of tertiary and benzyl amines.
Discussion
[00139] From a structural standpoint several attributes appear to be required for activity in quinoline methanols. The 4-position side chain hydroxyl and amine functionality is essential. Generally, conjugated amines were ineffective so these should be avoided. Lower molecular weight secondary amines with no additional heteroatoms in the side chain were the most potent (e.g. WR177000. WR308446 and WR308387). However, there are two barriers to the utility of these compounds as drugs. First, they exhibit higher relative permeability than mefloquine across MDCK-MDRl cell monolayers. Second, they are metabolically labile and this is reflected in poorer efficacy outcomes in vivo, confirming earlier observations (Dow et al., 2006b). Both of these limitations are likely related to their higher lipophilicity relative to mefloquine. Both could potentially be resolved by the inclusion of appropriate substituents in the side chain to lower lipophilicity. [00140] Permeability across the blood-brain barrier can be reduced by decreasing lipophilicity (lower LogP and LogD), increasing PSA, and increasing HBDs and HBAs (Kearns and Di, 2008). A priori it was a reasonable expectation that these characteristics might also be associated with higher IC90s (and therefore lower potency) as a consequence of lower permeability across parasite membranes. The data suggest this relationship may be more complicated. Significant differences between inactive and active compounds were observed for LogP, HBAs and PSA. This was not the case for HBDs and LogD. It can be rationalized that the first set of data has increasing lipophilicity resulting in increased potency, presumably due to increased permeability. LogD incorporates ionization state at physiological pH whereas LogP does not. Almost all the analogs, active and inactive, contain a basic side chain and thus are likely to be protonated at physiological pH. The HBD trends are more counter-intuitive given that increased HBDs should lower lipophilicity and therefore potency. This may be an artifact imposed by the constraints of R05 compliance, as library contains only seven compounds with greater than three HBDs. Mefloquine has two HBDs. The sample sizes may be insufficient to show a statistical association.
[00141] From the perspective of synthesizing a next generation of quinoline methanols, these observations indicate that incorporation of HBDs (rather than HBAs) into the side chain may be the most viable way to optimize potency and blood-brain barrier permeability. To this end, WR308396 is probably a more useful starting scaffold than WR308278 because it was less, rather than more, permeable relative to an appropriate comparator (WR.308387). However, there may be a limit to how many H-bond donors can be added before decreasing lipophilicity results in decreased potency. It is well known that decreasing lipophilicity increases metabolic stability and decreases clearance in vivo (Kerns and Di, 2008). Therefore these same structural modifications may improve efficacy in vivo. However it is also possible that such changes will have a counteracting effect in terms of reducing volumes of distribution (and therefore half-life).
[00142] The goal is to resolve the CNS tolerability issues of mefloquine by reducing partitioning into the central nervous system. However, this strategy may be counter productive if it results in inadvertent increases in potency against suspected targets of mefloquine in the CNS. Prior studies suggest that mefloquine may exhibit a direct neurotoxic effect in vivo and has potent activity against the A2A receptor (Weiss et at., 2003, Caridha et al., 2008, Gillespie et al., 2008). The most interesting analogs were evaluated against these targets. Most of the analogs tested exhibited lower LC50s against neurons or greater inhibition of the A2A receptor than mefloquine.
[00143] Generally mefloquine and the compounds evaluated in the in vivo studies showed the same rank order in terms of potency in vitro against the four drug resistant strains of P, falciparum. This may indicate cross-susceptibility to mefloquine. One can speculate that, all other factors being equal, such compounds, where they have equivalent potency to mefloquine, might not be fully effective if used clinically as single agents for malaria treatment in areas where background resistance to mefloquine if used as monotherapy is prevalent (e.g. the Thai borders), Mefloquine is normally used in combination with artesunate in such regions and remains clinically useful when deployed in this manner (Price et al, 2004). The intent would be to use a next generation quinoline methanol with a combination (perhaps non-artemisinin) agent in the same manner if needed. For this reason, lack of cross-susceptibility to mefloquine is a desirable rather than a required property of a next generation quinoline methanol.
EXAMPLE 2: Structure Activity Relationships
[00144] In order to efficiently prepare analogs with the 2,8-trifluoromethyl quinoline core, the vast majority of derivatives were constructed according to the procedure outlined in FIG. 23. Large quantities of the 2,8-bis-trifluoromethyl- 1H-quinolin-4-one intermediate 8 were prepared following the protocol of the Lutze group (see Ohnmacht et al. 1971. Antimalarials. 7. Bis(trifluoromethyl)-α-(2-piperidyl)-4-quinolinemethanols. J Med Chem VoI 14, No 10: pp 926- 928] incorporated herein by reference). Bis(trifluoromethyl) quinolin-4-ol 8 was melted along with phosphorous oxybromide to provide 4-bromo-2,8-bis(trifluoromethyl) quinoline 9. The resulting white solid was dissolved in tetrahydrofuran, cooled to -78°C and subjected to n- butyllithium. N,N-dimethylformamide was subsequently added to afford 2,8- bis(trifluoromethyl)quinoline-4-carbaldehyde 10. Utilization of Corey's dimethylsulfonium methylide provided racemic epoxide 11, which is also commercially available from Bioblocks (San Diego, Ca).
[00145] Quinoline scaffold 11 was diversified at the 4-position through a regio selective SN2 nucleophilic ring opening mechanism. To prepare ethers P2a and thioethers 12b, the requisite alcohol or thiol was added to a suspension of sodium hydride in THF and cooled to O°C prior to adding epoxide 11. After screening a variety of carbon nucleophiles, it was determined that Grignard reagents were clearly superior for constructing 12c. Epoxide 11 was dissolved in ether or THF, depending on the solvent associated with the commercially available Grignard reagent, and cooled to -78°C prior to adding the Grignard reagent and subsequently allowed to warm to O°C. Generally, the nitrogen-based analogs were prepared via a microwave-assisted epoxide opening. Epoxide 11 was dissolved in ethanol; the requisite amine was added and the solution subjected to microwaves at 13O°C.
Results and Discussion
[00146] In an effort to systematically explore the 4-position, the utility of the amino alcohol was tested. It appears the benzylic alcohol is essential for activity since the addition of a methyl blocking group results in loss of activity (compare mefloquine WR142490 to WR308038, Table 8). Substitution of hydroxyl with either a thiol or amino group results in a loss of activity relative to mefloquine (compare nzefloquine WR142490 to WR308393 and WR308392, Table 8).
[00147] Once the alcohol was deemed essential, the utility of the amine was explored. As shown in Table 9, while n-butyl amine (WR177000) was quite efficacious, alternate heteroatoms such as oxygen (WR308633), sulfur (WR308632), and carbon (WR308653) were devoid of activity. Based upon these results, nitrogen-based moieties were explored at the 4-position while generating the remainder of the empirical library. In regards to nitrogen functionalization, the data suggest that the nature of the 4-position amine and the degree of electron density around the nitrogen are most strongly associated with activity. The remainder of the discussion explores the relationship between 4-position amine substituents and antimalarial activity.
[00148] Antimalarial and selectivity data for a subset of compounds with benzene side chains are presented in Table 10. In order to maintain consistency with the numbering system of polysubstituted benzenes, nitrogen fbnctionalization is denoted as position 1 on the ring and the R-groups are numbered accordingly beginning with R2. Not surprisingly, the data suggest resonance and inductive effects are influencing efficacy. For example, phenyl amine (WR308251) has a much lower activity then benzyl amine (WR308252) and phenethylamino QM (WR308253). This suggests the reduction of electron density around the amine, due to the adjacent benzene ring, diminishes potency. In addition all of phenyl amines tested were nearly devoid of activity and the addition of electron withdrawing groups fbrther decreased activity.
[00149] Utilizing unsubstituted WR308252 as the reference point, a modest trend was observed for benzyl amines. A 4-methyl (WR308375) substitution increased activity, suggesting additional liphophilicity or steric bulk is beneficial. Electron donating and withdrawing groups had little effect on the structure activity relationship. For example, 3,5-fluoro (WR308414) and
3-piperidine (WR308518) analogs were more active than analogs possessing methoxy
(WR308395 and WR308506), chloro (WR308400 and WR308371), and thiophene ( W 3 08590) substituents.
[00150] It is well known that activation mechanisms of amines may lead to toxicity. Arylamines in particular are known to form N-hydroxyl amines, which produce nitrenium intermediates that subsequently react with nucleophiles such as DNA to cause toxicity. The in vitro assay systems used to generate the data presented in Table 10 would not likely detect such toxicity. At a more generic level, however, it is encouraging that there is no apparent relationship between intrinsic antimalarial activity and selectivity. All of the compounds except WR308251 and WR308253 exhibited improved selectivity indexes relative to mefloquine.
[00151] Imidazole and benzo[d]imidazole derivatives were collectively categorized as heterocyclic amino quinoline methanols (HAQMs). When the IC90 values for the four drug resistant P. falciparum strains are essentially the same order of magnitude, a promising cross- susceptibility IC90 profile emerges (Table 11). WR308437 and WR308623 in particular illustrated this trend. In regards to benzo[d]imidazole derivates, a similar trend was observed for WR308682, WR308763, and WR308764. In particular, WR308682 has increased potency and a superior selectivity index as compared to mefloquine. The HAQMs exhibited half-lives in the in vitro metabolic stability assays of 4-22 minutes in comparison to > 60 minutes for mefloquine. They are therefore much less metabolically stable than mefloquine. If this translates into shortlived plasma drug concentrations in vivo, it would not bode well for their utility for prophylaxis.
[00152] In regards to alkyl amino quinoline methanols (AAQMs), an intricate relationship between steric bulk and liphophilicity of the alkyl groups have emerged and merits further study to determine the discrete contributions to efficacy. Although the mechanism of action for mefloquine is not fully understood, the lipophilic nature of mefloquine (MQ) and quinine are known to correlate with delivery of drug to the parasite. MQ is also known for high-affinity binding to erythrocytes and other cells, which may provide a reservoir of drug and contribute to the long half-life. In general, these compounds were more potent than MQ and displayed a one- log increase in selectivity. Alkyl substitution is paramount. For example, the primary amine WR308314 is nearly devoid of activity, while the addition of methyl, ethyl, propyl, and butyl groups substantially increase efficacy (Table 12). Interestingly, branched alkyl substituents such as i-Pr and i-Bu prove quite active, while t-butyl displays moderate activity. Chain length also appears to affect activity since n-Bu (WR177000) and n-hex (WR308442) have different levels of potency, presumably resulting from the addition of two methylene units. In general, the increase in potency coupled with the reduced cost of goods for these analogs as compared to MQ add to their potential utility.
[00153] N-methyl (WR308245), i-Pr (WR308257), and t-butyl (WRl 83545) derivatives (Table 12) all displayed favorable metabolic stability profiles presumably due to inhibition of N- dealkylation. In an effort to probe structural modifications to improve stability, it was useful to probe the site of metabolism. Therefore, WR17700 was chosen as a scaffold, and the initial strategy was to prepare a series of branched alkyl amino quinoline methanols (Table 13). Methyl substitution resulted in half-lives of greater than 60 minutes with human liver microsomes. As for mouse microsomes, the trend in half- life followed Rl > R2 > R3. Overall, most AAQMs demonstrated an increase in potency and a selectivity index superior to MQ, while branching proved to be an efficient strategy for improving metabolic stability.
[00154] A variety of alkyl amino quinoline methanols containing additional heteroatoms (AAQMHs) were also constructed (Table 14). In doing so, it became apparent efficacy is reduced by the addition of an alcohol, acid, fluorine, or amide within the side-chain. In particular, when the hydroxyl of WR308258 is transformed into the methyl ether (WR308412) potency increases by nearly an order of magnitude. Ether WR308622 and thioether WR308278 were more potent and demonstrated a superior selectivity index compared to mefloquine. The presence of a benzyl amine (WR308396) was also advantageous. Interestingly, the presence of a primary amine within WR308426 and WR308384 resulted in decreased potency. Of this series, the ethers, thioethers, and secondary amines have proved most promising. A series was also constructed based upon varying the polar surface area of the efficacious dibutyl amine (WR176990; Table 12). Once the corresponding acid, ester, alcohol and amide were prepared (WR308147, WR308320, WR308321, WR308318; Table 14, respectively), it was apparent that these modifications did not increase potency.
[00155] This next generation quinoline methanol (NGQM) library was constructed based upon a 4-position scaffold, the goal of achieving greater or equivalent potency, improved selectivity and a lack of cross-susceptibility to mefloquine. One or more of the chemotypes investigated here displayed one or all of these characteristics and could potentially be considered early lead compounds, Analogs with phenyl-containing side chains such as WR308375 and WR308253 have equivalent or greater potency and selectivity relative to mefloquine with different cross-susceptibility profiles. The same is also the case for imidazole-containing analogs such as WR308407, WR308681, and WR308682. Alkylaminoquinoline methanols with additional heteroatoms such as WR308412, WR308622, WR308378 and WR308396 exhibited equivalent potency to mefloquine but their activity across different parasite strains also tracked with those of mefloquine. However, to be considered a potential lead compound some evidence of efficacy after oral dosing is desired, as well as, the potential for reduced permeability across the blood brain barrier. The latter trait is essential if the adverse neurological effects of mefloquine are to be avoided in a new series of analogs.
[00156] Representative compounds from the mentioned list were evaluated for their efficacy in vivo against P. berghei in mice. The bidirectional permeability of the same compounds was measured across MDRl -transformed MDCK cell monolayers in the presence of the PgP inhibitor cyclosporin A. This screen is normally used as a surrogate of potential permeability across the blood-brain barrier. Here data for compounds (WR308437, WR308412 and WR308622) not previously described is reported. In an earlier study it was shown that the thioether WR308278, the long chain alkyl compound WR177000 and the short chain alkyl compound WR308245 were active in vivo after oral dosing but exhibited greater permeability relative to mefloquine across MDCK cell monolayers (Table 15). The latter property suggests greater propensity for accumulation into the central nervous system. This was also the case for WR308427, WR308412 and WR308622 in the present study. Taking all the data into consideration, only WR308396 exhibited efficacy in vivo after oral dosing, lower or equivalent permeability across MDCK cell monolayers relative to mefloquine, and reduced permeability across MDCK cell monolayers relative to lipophilic alkyl compounds such as WR177000 lacking the diamine motif. These observations suggest a hypothesis for further investigation. Diamine quinoline methanols will retain the potency of mefloquine but exhibit significantly reduced permeability across the blood:brain barrier.
Conclusion
[00157] Structure activity relationships were investigated amongst a library of 200 A- position analogs of mefloquine. The 4-position alcohol and at least one amine met the minimum requirement for equivalent potency to mefloquine. Decreased electron density around the first side chain amine greatly diminished potency. Moderately enhanced potency, increased selectivity and altered cross-susceptibility patterns were achieved with imidazole and benzene containing analogs. Ten-fold greater than potency and selectivity than mefloquine without altered cross-susceptibility patterns were achievable in analogs with alkyl side chains. Introduction of heteroatoms into these latter analogs generally reduced potency, although select analogs exhibited equivalent potency to mefloquine.
[00158] All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. AU such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the following claims. EXAMPLE 3: Actual and Calculated Log Ratios of a Drug
[00159] A selection of 29 structurally diverse quinoline methanols was synthesized using the methods described. The log of the ratio of brain to blood concentrations was calculated (the cLogBB value) based on the equation cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18. These were compared to the actual log of the ratio of brain to blood concentration.
[00160] The actual log of the ratios of brain to blood concentrations was determined in mice as follows. The compounds were administered intravenously to groups of 2-4 FVB mice at a dose of 5 milligrams per kilogram of bodyweight. At 5 minutes, 60 minutes, 4 h and 24 h, brain and plasma concentrations were measured. The maximum brain and plasma concentrations were calculated, The ratio of maximum brain concentration to plasma concentration was determined. These values were converted to Log units. These values are referred to as the actual LogBB values. They were plotted and are presented in FIG. 24. Linear regression was performed, yielding an r2 value of 0.44 and a significantly non-zero slope of the regression line (P < 0.0001). This result means that a correlation exists between the calculated and actual values for LogBB, underscoring the utility of the in silico calculation method.
REFERENCES
[00161] The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
Caridha, D., et al., (2008) Mefloquine-induced disruption of calcium homeostasis in mammalian cells is similar to that induced by ionomycin. Antimicrob Agents Chemother. 52: 684-93. Clam, D. E. (2003) in silico prediction of blood-brain barrier permeation. Drug Discovery
Today, 8: 927-933.
Doan, K. M. M., et al., (2002) Passive Permeability and P-Glycoprotein-Mediated Emux Differentiate Central Nervous System (CNS) and Non-CNS Marketed Drugs. J. of
Pharmacology and Experimental Therapeutics, 303: 1029-1037.
Dow, G., et al., (2006a) Mefloquine induces dose related neurological effects in a rat model. Antimicrob Agents Chemother, 50; 1045-53. Dow, G. S., et al, (2008a) Antimalarial activity of phenylthiazolyl-bearing hydroxamate-based histone deacetylase inhibitors. Antimicrob Agents Chemother, 52: 3467-77. Dow, G. S., et al., (2006b) Utility of alkylaminoquinolinyl methanols as new antimalarial drugs.
Antimicrob Agents Chemother, 50: 4132-43. Dow, G. S., et al.,. (2008b) Clinical development of new prophylactic antimalarial drugs after the
5th Amendment to the Declaration of Helsinki. Theraputics and Clinical Risk
Management, 4. Glllespie, R. J., et al.. (2008) Antagonists of the human adenosine A2A receptor. Part 1 : Discovery and synthesis of thieno[3,2-dlpyrimidine-4-methanone derivatives. Bioorg Med Chem Lett, 18, 2916-9. Kerns, E. H. and Di, L. (2008) Drug-like properties: concepts, structure design and methods,
Burlington, MA, Elsevier Inc. Lanteri, C. (2009) Antimalarial activity of alkylaminoquinoline methanols. Antimicrobial
Agents and Chemotherapy, In press.
Lipinski, C. A. (2000) Drug-like properties and the causes of poor solubility and poor permeability. J Pharmacol Toxicol Methods, 44, 235-49.
Pardridge, W. M. (1995) Transport of small molecules through blood-brain barrier: biology and methodology. Advanced Drug Delivery Reviews, 15: 536. Pardridge, W. M. (1998) CNS drug design based on principles of blood-brain barrier transport. J
Neurochem. 70: 1781-92. Price, R N, et al., (2004) Mefloquine resistance in Plasmodium falciparum and increased pfmdrl gene copy number. Lancet, 364: 438-47. Rendi- Wagner, P., et al., (2002) Unexpected frequency, duration and spectrum of adverse events after therapeutic dose of mefloquine in healthy adults. Acta Trop. 81: 167-73. Schellenberg, D., et al.. (2006) The IPTi Consortium: research for policy and action. Trends Parasitol, 22, 296-300.
Shanks, G. D., et al., (2007) Drug-free holidays: pre-travel versus during travel malaria chemoprophylaxis. Am J Trop Med Hyg, 77, 1-2. Wang, Q., et al., 2005) Evaluation of the MDR-MDCK cell line as a permeability screen for the blood-brain barrier. Int JPharm, 288,349-59. Weiss, S. M., et al, 2003) Discovery of nonxanthine adenosine A2A receptor antagonists for the treatment of Parkinson's disease. Neurology, 61, S101-6.
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Ill
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001

Claims

CLAIMSWhat is claimed is:
1. A therapeutic composition comprising a quinoline methanol compound of Formula I
Figure imgf000143_0001
Formula I wherein ( )n represents one, two, or more carbon atoms;
R1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
R2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
R3 is at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a CF3 group, a OH group, an oxygen atom, and a hydrogen atom; wherein a calculated log ratio of brain:blood concentration (cLogBB) of the quinoline methanol compound is less than a cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18, wherein LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule, such that the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 90% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 90% of the relative concentration of mefloquine in vivo; and wherein when ( )n represents one carbon atom neither R1 or R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2-COOH; and a pharmaceutically- acceptable carrier.
2. The therapeutic composition of claim 1 wherein ( )n is one carbon atom.
3. The therapeutic composition of claim 1 wherein ( )n is two carbon atoms.
4. The therapeutic composition of claim 1 wherein R1 and R2 are joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring.
5. The therapeutic composition of claim 1 wherein R1 and R2 are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
6. The therapeutic composition of claim 1 wherein R1 and R2 contain a heterocycle.
7. The therapeutic composition of claim 1 wherein R3 is a substitution at the 8 position of the quinoline ring and is a CF3 group.
8. The therapeutic composition of claim 1 wherein R1 is a hydrogen atom and R2 is a side chain containing a nitrogen atom.
9. The therapeutic composition of claim 1 wherein R1 is a hydrogen atom and R2 is a side chain with a single nitrogen atom as the only heteroatom.
10. The therapeutic composition of claim 1 wherein R1 is a hydrogen atom and R2 is a side chain with a single nitrogen atom as the only heteroatom and wherein there are two carbon atoms between a first nitrogen and a second nitrogen on the R2 side chain.
11. The therapeutic composition of claim 1 wherein R1 is any side chain that does not contain a heteroatom, and R2 is any side chain containing a nitrogen atom.
12. The therapeutic composition of claim 1 wherein R1 is any side chain that does not contain a heteroatom, and R2 is any side chain with a single nitrogen atom as the only heteroatom.
13. The therapeutic composition of claim 1 wherein R1 is any side chain that does not contain a heteroatom, and R2 is any side chain with a single nitrogen atom as the only heteroatom and wherein there are two carbon atoms between a first nitrogen and a second nitrogen on the R2 side chain.
14. The therapeutic composition of claim 1 wherein the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 85% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 85% of the relative rate of mefloquine in vivo.
15. The therapeutic composition of claim 1 wherein the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 80% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 80% of the relative rate of mefloquine in vivo.
16. A method of treating or preventing a disease or condition associated with infection comprising administering to a subject an effective amount of at least one therapeutic compound of claim 1.
17. The method of claim 15 wherein the infection is caused by a pathogen selected from the group consisting of a microbe, parasite, protozoan, bacterium, fungus, or combination thereof.
18. The method of claim 16 wherein the parasite is a Plasmodium spp..
19. The method of claim 15 wherein the therapeutic compound is administered to a subject by at least one mode selected from the group consisting of oral, topcial, parenteral, subcutaneous, intramuscular, intradermal, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intraccervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal.
20. The method of claim 15 wherein the subject is a human.
21. The method of claim 15 wherein the subject is an animal.
22. The method of claim 15 wherein two or more therapeutic compounds are administered.
23. The method of claim 15 wherein the therapeutic compound is administered in combination with another therapeutic modality.
24. A method of reducing the incidence of or severity of a clinical sign associated with a disease or condition comprising administering a therapeutic composition of the invention to a subject such that incidence or severity of a clinical sign of the disease or condition is reduced by at least 10% relative to a subject that has not received the therapeutic composition.
25. The method of claim 23, wherein the disease or condition is microbial, parasitic, protozoan, bacterial, or fungal in nature.
26. The method of claim 23, wherein the the incidence or the severity of a clinical sign is reduced by at least 20%.
27. The method of claim 23, wherein the the incidence or the severity of a clinical sign is reduced by at least 30%.
28. The method of claim 23, wherein the clinical sign is selected from the group consisting of fever, chills, sweats, headaches, muscle pains, nausea, vomiting, elevated temperature, perspiration, tiredness, confusion, coma, neurologic focal signs, severe anemia, and respiratory difficulties.
29. A kit comprising a set of printed instructions; a dispenser capable of administering a therapeutic composition to a subject; and at least one quinoline methanol compound of
Formula I
Figure imgf000147_0001
Formula I wherein ( )n represents one, two, or more carbon atoms;
R1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, and a triazole;
R2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, an imidazole, and a triazole;
R3 is at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a CF3 group, a OH group, an oxygen atom, and a hydrogen atom; wherein a calculated log ratio of brain:blood concentration (cLogBB) of the quinoline methanol compound is less than a cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18, wherein LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule, such that the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 90% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 90% of the relative concentration of mefloquine in vivo; and wherein when ( )n represents one carbon atom neither R1 or R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2-COOH.
30. The kit of claim 28 further comprising a physiologically acceptable vehicle, carrier molecule, adjuvant, or combination thereof.
31. The kit of claim 28 wherein ( )n is one carbon atom.
32. The kit of claim 28 wherein ( )n is two carbon atoms.
33. The kit of claim 28 wherein R1 and R2 are joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring.
34. The kit of claim 28 wherein R1 and R2 are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine.
35. The kit of claim 28 wherein R3 is a substitution at the 8 position of the quinoline ring and is a CF3 group.
36. The kit of claim 28 wherein the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 85% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 85% of the relative rate of mefloquine in vivo.
37. The kit of claim 28 wherein the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 80% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 80% of the relative rate of mefloquine in vivo.
38. A therapeutic composition comprising a quinoline methanol compound of Formula I
Figure imgf000149_0001
Formula I wherein ( )n represents one, two, or more carbon atoms;
R1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
R2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
R3 is at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a CF3 group, a OH group, an oxygen atom, and a hydrogen atom; wherein R1 and R2 are joined directly or through linking atoms to form a substituted imidazole ring, unsubstituted imidazole ring, substituted triazole ring, or unsubstituted triazole ring; wherein a calculated log ratio of brain:blood concentration (cLogBB) of the quinoline methanol compound is less than a cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18, wherein LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule, such that the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 90% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 90% of the relative concentration of mefloquine in vivo; and wherein when ( )n represents one carbon atom neither R1 or R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2-COOH; and a pharmaceutically- acceptable carrier.
39. A therapeutic composition comprising a quinoline methanol compound of Formula I
Figure imgf000150_0001
Formula I wherein ( )n represents one, two, or more carbon atoms;
R1 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain alkyl group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms;
R2 is selected from the group consisting of a hydrogen atom, a side chain containing heterocycle, a straight chain group, a cyclic alkyl group, a straight chain alkyl group containing a nitrogen atom, a cyclic alkyl group containing a nitrogen atom, a cyclic amine, a cyclic chain alkyl group containing one or more heteroatoms, and a straight chain alkyl group containing one or more heteroatoms; R3 is at least one substitution at the 6 or 7 or 8 position of the quinoline ring, wherein the substitution is selected from the group consisting of a CF3 group, a OH group, an oxygen atom, and a hydrogen atom; wherein R1 and R2 are joined directly or through linking atoms to form a substituted or unsubstituted cyclic amine; wherein a calculated log ratio of brain:blood concentration (cLogBB) of the quinoline methanol compound is less than a cLogBB of mefloquine as determined by cLogBB = (0.205*LogP) - (0.0094*PSA) - (0.055*FRBs) + 0.18, wherein LogP is the partition coefficient reflecting the relative solubility in octanol versus water; PSA is the polar surface area of a molecule; and FRB is the number of freely rotatable bonds in a molecule, such that the therapeutic composition exhibits in vitro permeability across a blood-brain barrier at less than 90% of the relative rate of mefloquine in vitro, or exhibits in vivo total or free brain concentrations at less than 90% of the relative concentration of mefloquine in vivo; and wherein when ( )n represents one carbon atom neither R1 or R2 are selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, hydroxy, cyclopropyl, CH2-CHOH-CH2-CH3, CH2-CH2-CHOH-CH3, CH2-CH2-CH2-CH2OH, CH2OH, and CH2-CH2-COOH; and a pharmaceutically- acceptable carrier.
PCT/US2009/055796 2009-06-09 2009-09-02 Next generation quinoloine methanols WO2010144101A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US26865409P 2009-06-09 2009-06-09
US61/268,654 2009-06-09
US9356009P 2009-09-02 2009-09-02
US61/093,560 2009-09-02

Publications (1)

Publication Number Publication Date
WO2010144101A1 true WO2010144101A1 (en) 2010-12-16

Family

ID=43309139

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/055796 WO2010144101A1 (en) 2009-06-09 2009-09-02 Next generation quinoloine methanols

Country Status (1)

Country Link
WO (1) WO2010144101A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012068560A2 (en) * 2010-11-18 2012-05-24 Jenrin Discovery 4-quinolinemethanols as anti-malarial agents
EP2487157A1 (en) * 2011-02-11 2012-08-15 Université de Picardie Jules Verne Enantioselective synthesis method of 4-aminoalcoholquinoline derivatives and the use
WO2019086614A1 (en) 2017-11-03 2019-05-09 Universite Amiens Picardie Jules Verne Novel aminopyridinemethanol compounds and their use

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6583156B1 (en) * 1998-09-04 2003-06-24 Vernalis Research Limited 4-Quinolinemethanol derivatives as purine receptor antagonists (1)
US20050148645A1 (en) * 2003-11-21 2005-07-07 Lin Ai J. 2-Guanidinylimidazolidinedione compounds and methods of making and using thereof
US7342026B2 (en) * 2001-01-17 2008-03-11 Institut De Recherche Pour Le Developpement Substituted quinolines for the treatment of protozoa and retrovirus co-infections
US20080188462A1 (en) * 2005-02-17 2008-08-07 Peyton David H Quinoline Derivatives And Uses Thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6583156B1 (en) * 1998-09-04 2003-06-24 Vernalis Research Limited 4-Quinolinemethanol derivatives as purine receptor antagonists (1)
US7342026B2 (en) * 2001-01-17 2008-03-11 Institut De Recherche Pour Le Developpement Substituted quinolines for the treatment of protozoa and retrovirus co-infections
US20050148645A1 (en) * 2003-11-21 2005-07-07 Lin Ai J. 2-Guanidinylimidazolidinedione compounds and methods of making and using thereof
US20080188462A1 (en) * 2005-02-17 2008-08-07 Peyton David H Quinoline Derivatives And Uses Thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DOW ET AL.: "Utility of Alkylaminoquinolinyl Methanols as New Antimalarial Drugs", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 50, no. 12, December 2006 (2006-12-01), pages 4132 - 4143, XP008095290, DOI: doi:10.1128/AAC.00631-06 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012068560A2 (en) * 2010-11-18 2012-05-24 Jenrin Discovery 4-quinolinemethanols as anti-malarial agents
US20120178717A1 (en) * 2010-11-18 2012-07-12 Jenrin Discovery 4-quinolinemethanols as anti-malarial agents
WO2012068560A3 (en) * 2010-11-18 2013-08-15 Jenrin Discovery 4-quinolinemethanols as anti-malarial agents
US8716265B2 (en) * 2010-11-18 2014-05-06 Jenrin Discovery, Inc. 4-quinolinemethanols as anti-malarial agents
EP2487157A1 (en) * 2011-02-11 2012-08-15 Université de Picardie Jules Verne Enantioselective synthesis method of 4-aminoalcoholquinoline derivatives and the use
WO2012107532A1 (en) * 2011-02-11 2012-08-16 Université de Picardie Jules Verne 4-aminoalcoholquinoline derivatives, enantioselective synthesis methods and the use thereof
WO2019086614A1 (en) 2017-11-03 2019-05-09 Universite Amiens Picardie Jules Verne Novel aminopyridinemethanol compounds and their use

Similar Documents

Publication Publication Date Title
Valdés Acridine and acridinones: old and new structures with antimalarial activity
Maryanoff et al. Pyrroloisoquinoline antidepressants. 2. In-depth exploration of structure-activity relationships
JP3038155B2 (en) Tinnitus treatment with neuroprotective agents
KR20060005334A (en) Aryl-substituted diazabicycloalkanes as nicotinic acetylcholine agonists
Surase et al. Identification and synthesis of novel inhibitors of mycobacterium ATP synthase
WO2017103615A1 (en) Combination product
NO20051901L (en) New antimycobacterial compositions and pyrrole derivatives such as antimycobacterial compositions
AU2023241329A1 (en) Psychotropic agents and uses thereof
US20080085915A1 (en) Compounds and methods for the treatment of gastrointestinal and central nervous system disorders
Tian et al. Astemizole analogues with reduced hERG inhibition as potent antimalarial compounds
WO2010144101A1 (en) Next generation quinoloine methanols
WO2021060453A1 (en) Crosslinked optically active secondary amine derivative
Taft et al. Discovery and preclinical pharmacology of INE963, a potent and fast-acting blood-stage antimalarial with a high barrier to resistance and potential for single-dose cures in uncomplicated malaria
ZA200201762B (en) Thienoisoxazolyl- and thienylpyrrazolyl-phenoxy substituted propyl derivatives useful as D4 antagonists.
ZA200306108B (en) Beta-carboline derivatives and its pharmaceutical use against depression and anxiety.
WO2010144434A1 (en) Derivatives of mefloquine and associated methods for making and using
SG181992A1 (en) Sulfone compounds as 5-ht6 receptor ligands
WO2010144102A1 (en) Pentafluorosulfanyl analogs of mefloquine
Dassonville-Klimpt et al. Mefloquine derivatives: synthesis, mechanisms of action, antimicrobial activities
Ma et al. Synthesis and Biological Evaluation of 1, 3, 3, 4‐Tetrasubstituted Pyrrolidine CCR5 Receptor Antagonists. Discovery of a Potent and Orally Bioavailable Anti‐HIV Agent
EP2913324A1 (en) Fluorine substituted cyclic amine compounds and preparation methods, pharmaceutical compositions, and uses thereof
Lin et al. Design, synthesis, and characterization of novel CXCR4 antagonists featuring cyclic amines
US20140315919A1 (en) Highly selective sigma receptor ligands
Varoli et al. Diphenidol-related diamines as novel muscarinic M4 receptor antagonists
WO2020089478A1 (en) New tetrahydropyrimidodiazepin and tetrahydropyridodiazepin compounds for treating pain and pain related conditions

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: 09845942

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: 09845942

Country of ref document: EP

Kind code of ref document: A1