US20160151380A1 - Therapies for diseases caused by arthropod-borne parasites - Google Patents

Therapies for diseases caused by arthropod-borne parasites Download PDF

Info

Publication number
US20160151380A1
US20160151380A1 US14/904,686 US201414904686A US2016151380A1 US 20160151380 A1 US20160151380 A1 US 20160151380A1 US 201414904686 A US201414904686 A US 201414904686A US 2016151380 A1 US2016151380 A1 US 2016151380A1
Authority
US
United States
Prior art keywords
protein kinase
mammalian
subject
inhibitor
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/904,686
Inventor
Shirley LUCKHART
Cecilia GIULIVI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Priority to US14/904,686 priority Critical patent/US20160151380A1/en
Publication of US20160151380A1 publication Critical patent/US20160151380A1/en
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LUCKHART, Shirley, GIULIVI, Cecilia
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41621,2-Diazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41681,3-Diazoles having a nitrogen attached in position 2, e.g. clonidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56905Protozoa
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/44Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from protozoa
    • G01N2333/445Plasmodium
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2440/00Post-translational modifications [PTMs] in chemical analysis of biological material
    • G01N2440/14Post-translational modifications [PTMs] in chemical analysis of biological material phosphorylation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/20Screening for compounds of potential therapeutic value cell-free 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 disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite.
  • the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in arthropods feeding on recipients of the regimens.
  • the present disclosure provides methods for screening candidate anti-parasitic agents.
  • Chagas disease also known as American trypanosomiasis, is a parasitic disease caused by Trypanosoma cruzi.
  • T. cruzi is a protozoan parasite that is mainly transmitted by blood-feeding insects known as kissing bugs (triatomines) through their feces and contaminated objects, including food, and drink.
  • kissing bugs triatomines
  • Clinically Chagas disease is classified into acute and chronic phases.
  • the acute phase begins when T. cruzi infiltrates the body and starts multiplying within different organs and tissues.
  • the symptoms during the acute phase can be mild and nonspecific, including fever and swelling at the site of infection (e.g., swelling of the eyelid, also referred to as Romana's sign).
  • acute infection can cause severe inflammation of the heart or brain (myocarditis or meningoencephalitis) which can be deadly (Bern et al., supra, 2011).
  • the spectrum of symptoms depends on factors such as the parasite burden and host immunocompetence.
  • the acute phase lasts 4 to 8 weeks, after which initial symptoms cease, and the infection enters a lifelong chronic phase. In the chronic phase, intracellular T.
  • Chagas disease develops into a life-threatening illness characterized by abnormal heart rhythms leading to an increased risk of sudden death, heart failure, and/or gastrointestinal disturbances.
  • the CDC has named Chagas disease as one of five (along with Neurocysticercosis, Toxocariasis, Toxoplasmosis, and Trichomoniasis) neglected parasite infections in the United States as public health priorities based on the severity of the illnesses, the number of people infected, and the ability to prevent and treat the disease.
  • Chagas disease there remains a need for therapy regimens to treat or prevent other diseases caused by arthropod-borne parasites, which are safe and do not encourage development of drug-resistant parasites.
  • new anti-Chagas disease drugs with an improved safety profile are urgently needed.
  • the present disclosure provides methods for treating or preventing a parasitic disease caused by an arthropod-borne parasite by administering a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to the arthropod-borne parasite (e.g., a mammalian subject in need thereof) under conditions effective for treating or preventing the parasitic disease.
  • the arthropod-borne parasite is a protozon parasite.
  • the present disclosure provides methods for treating or preventing a disease caused by a protozoan parasite comprising: administering a first protein kinase inhibitor and a second protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing the disease caused by the protozoan parasite.
  • the present disclosure also provides methods for treating or preventing a disease caused by a protozoan parasite comprising: administering a protein kinase inhibitor and an additional antiprotozoan drug to a mammalian subject in need thereof under conditions effective for treating or preventing the disease caused by the protozoan parasite.
  • the administering reduces activity of at least one mammalian protein kinase.
  • the protein kinase inhibitor comprises a first and a second protein kinase inhibitor.
  • the protozoan parasite is an arthropod-borne parasite selected from the group consisting of Plasmodium sp., Leishmania sp., Babesia sp., and Trypanosoma sp.
  • the parasitic disease is not malaria and the parasite is not a species of Plasmodium .
  • the protein kinase inhibitor reduces activity of an arthropod kinase.
  • the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family.
  • PLC protein kinase C
  • JNK c-Jun N-terminal kinase
  • MAPK mitogen activated protein kinase
  • the subject is infected with a protozoan parasite.
  • the subject is infected with a drug-resistant protozoan parasite (e.g., resistant to currently used anti-parasitic drugs).
  • the subject is acutely infected with a protozoan parasite, while in other embodiments the subject is chronically infected with a protozoan parasite.
  • the subject is experiencing a symptom of the disease prior to the administering step.
  • treating the disease comprises alleviating a symptom of the disease experienced by the subject.
  • the subject is an uninfected individual planning to visit a disease endemic area after (6 hours-72 hours, 1, to or 3 days) the administering step. That is the individual is not infected with but likely to be exposed to a protozoan parasite.
  • the administering commences and concludes before the uninfected individual visits a parasitic disease endemic area.
  • the administering commences before the uninfected individual visits a parasitic disease endemic area, and continues during the duration of their visit. That is in some embodiments, the administering concludes sometime after the uninfected individual leaves the parasitic disease endemic area (e.g., within thirty days of after last potential exposure). In some embodiments, preventing the disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days (between 0 and 30 days). In some embodiments, drug-resistant protozoan parasites (e.g., resistant to currently used anti-parasitic drugs) are known to be present in the disease endemic area.
  • drug-resistant protozoan parasites e.g., resistant to currently used anti-parasitic drugs
  • the present disclosure provides methods for treating or preventing Chagas disease comprising: administering a mammalian protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing Chagas disease.
  • the mammalian protein kinase inhibitor comprises a first kinase inhibitor and a second kinase inhibitor, and the administering reduces activity of a first mammalian protein kinase and a second mammalian protein kinase.
  • the mammalian protein kinase comprises a member of a protein kinase C (PKC) family, a member of the protein kinase B (PKB) family, and/or a member of the phosphoinositide 3-kinase (PI3K) family.
  • the mammalian protein kinase inhibitor is capable of reducing activity of a Triatominae insect kinase.
  • the methods further comprise administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject.
  • the subject is infected with Trypanosoma cruzi .
  • the subject is acutely infected with T.
  • treating Chagas disease comprises alleviating a symptom of Chagas disease experienced by the subject.
  • the methods further comprise administering an effective amount of an additional antiparasitic agent to the mammalian subject.
  • the additional antiparasitic agent comprises one or both of benznidazole and nifurtimox.
  • the subject is an uninfected individual planning to visit a Chagas disease endemic area (e.g., Mexico, Central America or South America) after the administering step. That is the individual is not infected with but likely to be exposed to T. cruzi .
  • the administering commences and concludes before the uninfected individual visits a Chagas disease endemic area. In other embodiments, the administering commences before the uninfected individual visits a Chagas disease endemic area, and continues during the duration of their visit. That is in some embodiments, the administering concludes sometime after the uninfected individual leaves the Chagas disease endemic area (e.g., within thirty days of after last potential exposure). In some embodiments, preventing Chagas disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days.
  • the present disclosure provides method for treating or preventing Chagas disease comprising: administering a protein kinase inhibitor to a subject infected with Trypanosoma cruzi under conditions effective for treating or preventing Chagas disease.
  • the present disclosure provides methods for treating or preventing a parasitic disease comprising: administering a protein kinase inhibitor to a subject infected with an arthropod-borne parasite under conditions effective for treating or preventing a parasitic disease.
  • the present disclosure provides methods of reducing transmission of Trypanosoma cruzi by a Triatominae vector, comprising administering a protein kinase inhibitor to a mammalian subject infected with Trypanosoma cruzi . under conditions effective for reducing transmission of the Trypanosoma cruzi ingested by the Triatominae vector in the subject's blood.
  • the present disclosure also provides methods of reducing transmission of Trypanosoma cruzi by a Triatominae vector, comprising providing a bloodmeal comprising erythrocytes infected with Trypanosoma cruzi to a Triatominae vector in the presence of an effective amount of a protein kinase inhibitor for reducing transmission of the Trypanosoma cruzi ingested by the Triatominae vector in the bloodmeal.
  • the methods further comprise administering an additional anti-parasitic drug to the mammalian subject.
  • the bloodmeal is provided to the Triatominae vector in the further presence of an additional antiparasitic drug.
  • the protein kinase inhibitor comprises a first and a second protein kinase inhibitor. In some embodiments, the protein kinase inhibitor reduces activity of at least one mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some preferred embodiments, the protein kinase inhibitor reduces activity of a Triatominae kinase.
  • PLC protein kinase C
  • JNK c-Jun N-terminal kinase
  • MAPK mitogen activated protein kinase
  • the protein kinase inhibitor reduces activity of a Triatominae kinase.
  • the Triatominae kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms, and p38 MAPK isoforms.
  • the Triatominae kinase comprises aPKC-zeta.
  • the subject is infected with Trypanosoma cruzi .
  • the additional anti-parasitic drug comprises a one or both of benznidazole and nifurtimox.
  • the present disclosure also provides methods of identifying an antiparasitic compound, comprising: measuring activity of a mammalian or insect protein kinase in the presence and absence of a test compound; and identifying the test compound as an antiparasitic compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound.
  • the methods further comprise growing insect cells in vitro in the presence and absence of the test compound and identifying the test compound as insect-cell safe when viability or doubling time of the insect cells is not significantly reduced in the presence as compared to the absence of the test compound.
  • the mammalian protein kinase is a human protein kinase.
  • the human protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family.
  • PLC protein kinase C
  • JNK c-Jun N-terminal kinase
  • MAK mitogen activated protein kinase
  • the methods are suitable for identifying an anti-Chagas disease compound.
  • the insect protein kinase is a Triatominae protein kinase, and the insect cells are Triatominae cells.
  • the Triatominae kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms and p38 MAPK isoforms.
  • the Triatominae kinase comprises aPKC-zeta.
  • the Triatominae cells are Rhodnius sp. cells (e.g., Rhodnius prolixus cells).
  • the present disclosure also provides methods of identifying an anti-Chagas disease compound, comprising: comparing development of parasites in Triatominae vector after consumption of a bloodmeal comprising Trypanosoma cruzi in the presence and absence of a test compound; and identifying the test compound as an anti-Chagas disease compound when the development of parasites is reduced in the Triatominae vector in the presence as compared to the absence of the test compound.
  • the present disclosure provides pharmaceutical compositions comprising a first protein kinase inhibitor, a second protein kinase inhibitor, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitors are present in amounts effective to treat or prevent Chagas disease in a mammalian subject.
  • the present disclosure provides pharmaceutical compositions comprising a protein kinase inhibitor, an additional antiparasitic agent, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitor and the additional antiparasitic agent are present in amounts effective to treat or prevent Chagas disease in a mammalian subject.
  • compositions comprising: a protein kinase inhibitor attached to a mitochondrial-targeting moiety, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the protein kinase inhibitor is present in an amount effective to treat or prevent Chagas disease in a mammalian subject.
  • the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a protein kinase B (PKB) family, and a phosphoinositide 3-kinase (PI3K) family.
  • PLC protein kinase C
  • PBB protein kinase B
  • PI3K phosphoinositide 3-kinase
  • the mammalian protein kinase inhibitor is capable of reducing activity of an arthropod kinase.
  • the methods further comprise administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject.
  • the subject is infected with an arthropod-borne parasite which is not a Plasmodium sp. parasite.
  • the subject is acutely infected with the arthropod-borne parasite, while in other embodiments, the subject is chronically infected with the arthropod-borne parasite.
  • treating the parasitic disease comprises alleviating a symptom of the parasitic disease experienced by the subject.
  • the methods further comprise administering an effective amount of an additional antiparasitic agent to the mammalian subject.
  • the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step commences before the visit.
  • the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step concludes during or after the visit.
  • preventing the parasitic disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days.
  • the protein kinase inhibitor comprises a first and a second protein kinase inhibitor. In some embodiments, the protein kinase inhibitor reduces activity of at least one mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some preferred embodiments, the protein kinase inhibitor reduces activity of an arthropod kinase.
  • PLC protein kinase C
  • JNK c-Jun N-terminal kinase
  • MAPK mitogen activated protein kinase
  • the protein kinase inhibitor reduces activity of an arthropod kinase.
  • the arthropod kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms, and p38 MAPK isoforms.
  • the arthropod kinase comprises aPKC-zeta.
  • the subject is infected with an arthropod-borne parasite.
  • the arthropod-borne parasite is not a species of Plasmodium and the arthropod is not a species of Anopheles.
  • the present disclosure provides, methods of identifying an antiparasitic compound, comprising: measuring activity of a mammalian or insect protein kinase in the presence and absence of a test compound; and identifying the test compound as an antiparasitic compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound, and wherein the insect protein kinase is not a Anopheles sp. kinase.
  • TEP1, APL1, LRIM1 complement-like immune factors
  • LRRD7 leucine-rich repeat gene
  • DEF defensin
  • NOS nitric oxide synthase
  • the present disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor, optionally with one or both of a further protein kinase inhibitor and an antiparasitic agent to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite.
  • a protein kinase inhibitor is administered to the subject, while in other aspects a protein kinase inhibitor and one or both of a further protein kinase inhibitor and an antiparasitic agent are administered to the subject.
  • the present disclosure provides methods for treating or preventing Chagas disease by administration of a protein kinase inhibitor and optionally one or both of a further protein kinase inhibitor and an anti-Chagas disease agent to a mammalian subject infected with or at risk of exposure to Trypanosoma cruzi .
  • the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in Triatominae (e.g., kissing bugs) feeding on recipients of the regimens (e.g., treated mammalian subjects). Additionally, the present disclosure provides methods for screening candidate anti-Chagas disease agents.
  • the disclosure herein pertaining to malaria, Plasmodium sp. and mosquitoes is provided as an exemplary disclosure of embodiments that are applicable in general to diseases caused by arthropod-borne parasites.
  • the disclosure herein pertaining to malaria, Plasmodium sp. and mosquitoes is also generally applicable to Chagas disease (American trypanosomiasis), Trypanosoma cruzi and Triatominae.
  • PKC protein kinase C isoenzymes are an important family of serine/threonine protein kinases that contribute to many diverse cellular and tissue functions, as well as human disease pathologies including cancer development and progression (Bosco et al., Mini Rev Med Chem, 11:185-199, 2011; and Rosse et al., Nat Rev Mol Cell Biol, 11:103-112, 2010).
  • cPKC classical PKC; cPKC ⁇ , cPKC ⁇ and cPKC ⁇ ), nPKC (novel PKC; nPKC ⁇ , nPKC ⁇ , nPKC ⁇ and nPKC ⁇ ) and aPKC (atypical PKC; a PKC ⁇ and aPKC ⁇ ) isoforms.
  • All PKCs are comprised of a C-terminal serine/threonine protein kinase domain belonging to the AGC kinase (protein kinase A/protein kinase G/PKC family kinase) superfamily, and an N-terminal regulatory domain (Parker and Murray-Rust, J Cell Science, 117:131-132, 2004).
  • AGC kinase protein kinase A/protein kinase G/PKC family kinase
  • N-terminal regulatory domain Parker and Murray-Rust, J Cell Science, 117:131-132, 2004.
  • the kinase domain has a C-terminal extension unique to AGC kinases that contains essential ‘priming’ phosphorylation sites required for catalytic activation (Pearce et al., Nat Rev Mol Cell Biol, 11:9-22, 2010), whereas in aPKCs, the most C-terminal of these is replaced by a phospho-mimetic Glu residue.
  • the regulatory domain within all PKC isoforms has an inhibitory region (pseudo-substrate motif) and distinctive arrangements of C1, C2 and/or PB1 (Phox/Bem1) domains (the last being exclusive to the aPKCs) that together are crucial for conferring isoform-specific functions.
  • aPKC isoforms diverge from other PKC family members as their regulatory domains are unresponsive to diacylglycerol, phosphatidylserine and Ca 2+ . Instead they are regulated by protein interactions, for example with polarity proteins Par-6, Par-3 and the Rho family GTPase cdc42 (Suzuki and Ohno, J Cell Sci, 119:979-987, 2003).
  • aPKC has a role in establishing cell polarity and is required for normal cell proliferation, mitotic spindle orientation and migration.
  • aPKC has been considerable interest in aPKCs as drug targets due to their roles in cancer development and progression (Fields and Regala, Pharmacol Res, 55:487-497, 2007).
  • PKC ⁇ is required for epidermal growth factor-induced migration of human breast and lung cancer cells and for colony-stimulating factor 1 chemotaxis of macrophages, among others.
  • PKC ⁇ promotes nicotine-induced migration and invasion of lung cancer cells via phosphorylation of calpains and is involved in other types of cancer.
  • aPKC has been shown to interact with bona fide oncogenes, such as Ras and Notch in the fruit fly Drosophila melanogaster to promote an epithelial cell overgrowth that resembles cancer.
  • aPKC isoenzymes can be inhibited by small-molecule chemicals either through their ATP-binding or allosteric pockets (Knight and Shokat, Chem Biol, 12:621-637, 2005). Only a few selective chemical biology tools inhibit aPKC catalytic activity. These few catalytic inhibitors include the non-selective Gö6983 and Gö6976 compounds and a PKC ⁇ pseudo-substrate peptide containing a membrane-targeting myristoylation site (Roffey et al., Curr Opin Cell Biol, 21:268-279, 2009).
  • PKC ⁇ plays an important role in the activation of TNF-alpha and NF-kB pathways, the latter of which are critical to the balance between host protection and pathology in malaria (Randall and Engwerda, Exp Parasitol, 126:326-331, 2010).
  • mammalian PKC activity has recently been shown to be critical for malaria parasite egress from infected red blood cells (Millholland et al., Cell Host Microbe, 13:15-28, 2013).
  • a PKC inhibitor e.g., a molecule that is directed to the PKC PIF pocket
  • a p38 MAPK inhibitor is used in combination with a p38 MAPK inhibitor to inhibit networked p38 MAPK and PKC ⁇ to enhance control of infection in the human host and transmission via the exposed mosquito.
  • kinase inhibitors such as the JNK inhibitor SP600125 has been shown to reduce malaria disease severity, but without an effect on parasitemia (Anand et al., Parasitol Res, 112:1959-1966, 2013).
  • JNK inhibitor SP600125 has been shown to reduce malaria disease severity, but without an effect on parasitemia (Anand et al., Parasitol Res, 112:1959-1966, 2013).
  • a JNK inhibitor in combination with a PKC inhibitor is used to reduce disease severity and parasitemia.
  • Combinations of small molecule inhibitors (SMIs) of conserved kinases are provided as “next-generation” anti-malarials to control both disease severity and transmission, as well as to reduce risk of development of parasite drug resistance.
  • SIs small molecule inhibitors
  • SMIs of conserved signaling kinases are delivered as orally available therapeutics to malaria patients in endemic countries.
  • the following list of SMI therapeutic attributes is applicable to both pediatric and adult patients: (1) single dose (or few dose) efficacy with inhibition of parasite growth and resolution of disease following treatment, (2) few to no contraindications and side effects, (3) long-term stability under conditions in endemic countries where refrigeration during storage and distribution are limited at best, (4) parasite transmission blocking activity in mosquitoes that feed on treated patients to counter potential “escape” of parasites from treated patients, (5) well-tolerated by the most sensitive populations, children and pregnant women as well as by G6PDH-deficient patients.
  • the actual amount per day for an adult mammal weighing 70 kg is typically between 0.07 and 7,000 mg, or more typically between 0.7 and 700 mg, where this amount can be administered as a single dose per day or as a series of part-doses (such as, for example, two, three, four, five or six) per day, so that the total daily dose is the same.
  • the SMI is selected to provide a formulation that exhibits a high therapeutic index.
  • the therapeutic index is the dose ratio between toxic and therapeutic effects, which can be expressed as the ratio between LD 50 and ED 50 .
  • the LD 50 is the dose lethal to 50% of the population and the ED 50 is the dose therapeutically effective in 50% of the population.
  • the LD 50 and ED 50 are determined by standard pharmaceutical procedures in animal cell cultures or experimental animals.
  • a SMI such as a protein kinase inhibitor
  • a pharmaceutically acceptable salt can be formed with inorganic acids (such as hydrochloric acid, hydroboric acid, nitric acid, sulfuric acid, and phosphoric acid), organic acids (e.g., formic acid, acetic acid, fumaric acid, oxalic acid, tartaric acid, lactic acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, and p-toluenesulfonic acid) or acidic amino acids (such as aspartic acid and glutamic acid).
  • inorganic acids such as hydrochloric acid, hydroboric acid, nitric acid, sulfuric acid, and phosphoric acid
  • organic acids e.g., formic acid, acetic acid, fumaric acid, oxalic acid, tartaric acid, lactic acid, maleic acid, citric acid, succinic acid
  • excipients include but are not limited to binding agents such as hydroxypropylmethylcellulose or povidone (polyvinylpyrollidone), lubricants such as magnesium stearate, and disintegrants such as sodium starch glycollate, croscarmellose sodium, or crospovidone (cross-linked polyvinylpyrollidone).
  • binding agents such as hydroxypropylmethylcellulose or povidone (polyvinylpyrollidone)
  • lubricants such as magnesium stearate
  • disintegrants such as sodium starch glycollate, croscarmellose sodium, or crospovidone (cross-linked polyvinylpyrollidone).
  • kinase inhibitor formulations are comprised of at least two SMIs having high bioavailability (>20%) are administered to a population of parasite-infected patients.
  • particularly preferred SMIs have an EC 50 ⁇ 10 nM in vitro and a single-digit mg/kg activity ED 90 in a human erythrocyte-engrafted SCID mouse model.
  • the kinase inhibitor formulations are either single-drug formulations comprising an SMI as the only active ingredient, or as a multi-drug formulation comprising a first SMI and a second SMI.
  • the patient population is comprised of both children (over six months of age) and adults with confirmed and uncomplicated Plasmodium infection who are able to swallow oral medication.
  • the patient pool does not exclude G6PD-deficient patients.
  • PKs protein kinases
  • PKs protein kinases
  • ERK ERK
  • JNK ERK
  • p38 MAPK protein kinases
  • inhibition of mosquito p38 MAPK significantly reduces malaria parasite development. Without being bound by theory, the inhibition of parasite development is thought to be in response to p38 MAPK-regulated changes in mosquito midgut mitochondrial bioenergetics.
  • SMIs so as to redirect their localization to the mitochondrial compartment.
  • the modified SMIs may result in less drug resistance, catabolism, and off-target effects relative to unmodified SMIs that localize to the cytosolic space (Mourtada et al., PLoS One, 8:e60253, 2013).
  • Suitable modifications to direct delivery of a SMI to a mitochondria include attachment of the SMI to a mitochondrial-targeting moiety, including but are not limited to lipophilic cations, mitochondrial-penetrating peptides, mitochondrial-targeting dyes, liposomes, and nanoparticles (see, e.g., Rin Jean et al., ACS Chem Biol, 9:323-333, 2014).
  • Exemplary mitochondrial-penetrating peptides include but are not limited to SS-31 (Zhao et al., J Biol Chem, 279: 34682-34690, 2004), MPP (Horton et al., Chem Biol 15: 375-382, 2008), and those including 8-15 hydroxyl-containing (Serine, Threonine), positively charged amino acids (Arginine, Lysine) or hydrophobic natural and synthetic amino acids (phenylalanine, cyclohexylalanine)(Horton et al., Chem Biol, 15:375-82, 2008; and Yousif et al., Chembiochem, 10:1939-50, 2009).
  • Exemplary mitochondrial-targeting dyes include but are not limited to rhodamine and the dyes listed in Table V.
  • Another strategy for modification of SMIs for use in the methods and compositions of the present disclosure involve the use of bifunctional inhibitors. These inhibitors tend to be more effective than those solely directed towards either the ATP binding site (Hu et al., Drug Discovery Today, 1:438-447, 1996) or the substrate-binding site (Lee et al., J Am Chem Soc, 126:3394-3395, 2004) because they are directed to both the ATP and substrate binding sites (Ricouart et al., J Med Chem, 34:73-78, 1991).
  • bifunctional protein kinase inhibitors are targeted to mitochondria.
  • the present disclosure further provides methods for treating or preventing malaria comprising administering at least one kinase inhibitor and an additional antimalarial (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing malaria.
  • an additional antimalarial e.g., a compound that is not a kinase inhibitor
  • the kinase inhibitor and the antimalarial are present in a single formulation, while in other embodiments the kinase inhibitor and the antimalarial are present in separate formulations.
  • the additional antimalarial comprises one or more compounds of the antimalarial classes selected from the group consisting of amino alcohols, aminoquinolines, antibiotics, antifolates, endoperoxides, sulfonamides, and others (see, e.g., Delves et al., PLoS Medicine, 9(2) e1001169, 2012).
  • the additional antimalarial compound(s) may be aminoquinolines (including, but not limited to, amodiaquine, naphthoquine, AQ-13, tert-butyl isoquine, hydroxycholoquine, pyronaridine, diethylprimaquine, bulaquine, primaquine, tafenoquine, piperaquine, NPC-1161B, and chloroquine), antibiotics (including, but not limited to, azithromycin, trimethoprim, trimethoprim-sulphamethoxazole, tetracycline, mirincamycin, doxycycline, thiostrepton, fosmidomycin, and clindamycin), endoperoxides (including, but not limited to, artemeter, artesunate, OZ439, OZ277, artemisinin, artemisone, and dihydroartemisinin), antifolates (such as pyrimethamine, proguan
  • the combination therapy includes a fixed-dose artemisinin combination therapy.
  • the artemisinin combination therapy is comprised of one or more kinase inhibitor as well as one or more of artemether, artesunate, dihydroartemisinin, artemisone, and artemisinin.
  • the additional antimalarial counteracts the malarial parasite at the same life stage(s) as the kinase inhibitor. In other embodiments, the additional antimalarial counteracts the malarial parasite at a different life stage from the kinase inhibitor.
  • the present disclosure also provides methods for treating or preventing other parasitic diseases comprising administering at least one kinase inhibitor and an additional antiparasitic agent (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing the parasitic disease.
  • an additional antiparasitic agent e.g., a compound that is not a kinase inhibitor
  • the kinase inhibitor and the antiparasitic agent are present in a single formulation, while in other embodiments the kinase inhibitor and the antiparasitic agent are present in separate formulations.
  • the parasitic disease is not malaria.
  • present disclosure provides methods for treating or preventing Chagas disease comprising administering at least one kinase inhibitor and an additional antiparasitic agent (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing the parasitic disease.
  • an additional antiparasitic agent e.g., a compound that is not a kinase inhibitor
  • the kinase inhibitor and the antiparasitic agent are present in a single formulation, while in other embodiments the kinase inhibitor and the antiparasitic agent are present in separate formulations.
  • the additional antiparasitic agent comprises an azole- or nitro-derivative, such as benznidazole, or nifurtimox.
  • the use of SMIs directed to mammalian kinase pathways is applied to diseases caused by other intracellular parasitic protozoa.
  • a large number of protozoal pathogens spend a portion of their life cycle in a mammalian host, (e.g., human).
  • Diseases caused by infection with other intracellular parasitic protozoa are also treated or prevented by mammalian kinase inhibitors.
  • the present disclosure provides kinase inhibitors and methods for use thereof in mammalian subjects infected with a protozoan parasite selected from the group consisting of Giardia lamblia, Cryptosporidium parvum, Cyclospora cayetanensis, Entamoeba histolytica, Trichomonas tenax, Trichomonas hominis, Trichomonas vaginalis, Trypanosoma brucei gambiense, Trypanosoma brucei rhodesiense, Trypanosoma cruzi, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Toxoplasma gondii, Theileria lawrenci , and Theileria parva .
  • a protozoan parasite selected from the group consisting of Giardia lamblia, Cryptosporidium parvum, Cyclospora cayetanens
  • the protozoan parasite is vectored by an arthropod (e.g., sand fly-transmitted Leishmania , tick-borne Babesia microti , kissing bug-transmitted Trypanosoma cruzi , tsetse-transmitted Trypanosoma brucei ), and the SMI is directed to mammalian and arthropod kinase pathways.
  • the disease is Chagas disease, caused by infection with Trypanosoma cruzi .
  • the present disclosure provides methods for use of kinase inhibitors in hosts infected with a protozoan parasite.
  • an excipient includes one or more excipients.
  • small molecule inhibitor and “SMI” refer to an organic compound (e.g., JNK inhibitor SP600125) of low molecular weight (e.g., less than 900 Daltons, preferably less than 500 Daltons, unless bound to a mitochondrial-targeting moiety in which case the size could be >2 kDa but less than 5 kDa) that inhibits a biological process.
  • Biopolymers such as nucleic acids, proteins, and polysaccharides of greater than 10 kDa are not small molecules, although the nucleotide, amino acid, and saccharide constituents are small molecules.
  • mitochondrial-targeting moiety refers to a functional group of a compound that directs the compound to the mitochondria or to a specific compartment therein.
  • mitochondrial-targeting moieties include but are not limited to lipophilic cations, mitochondrial-penetrating peptides, mitochondrial-targeting dyes, liposomes and nanoparticles.
  • Administration a protein kinase inhibitor in combination with an additional protein kinase inhibitor or an additional anti-parasite drug includes simultaneous (concurrent) and consecutive administration in any order.
  • an “effective amount of” or “under conditions effective for” refers to administration of a protein kinase inhibitor in an amount sufficient to carry out a specifically stated purpose.
  • An “effective amount” may be determined empirically and in a routine manner, in relation to the stated purpose.
  • terapéuticaally effective amount refers to an amount of an agent (e.g., protein kinase inhibitor) effective to “treat” a disease or disorder in a subject (e.g., a mammal such as a human).
  • an agent e.g., protein kinase inhibitor
  • the therapeutically effective amount of the agent reduces a sign or symptom of Chagas disease.
  • treating or “treatment” of a disease refer to executing a protocol, which may include administering one or more drugs to an individual (human or otherwise), in an effort to alleviate signs or symptoms of the disease.
  • treating does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols that have a measurable effect on the individual.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • reduce and “reduction” as used in reference to biological function (e.g., kinase activity, parasite development, etc.) refer to a measurable decrease in the function by preferably at least 50%, more preferably at least 75% and most preferably at least 90%. Depending upon the function, the reduction may be from 10-fold to 1,000,000-fold, or from 10, 100 or 1000-fold to 10,000, 100,000 or 1,000,000-fold.
  • ACPR abbreviations: ACPR (adequate clinical and parasitological response); AMAS (amastigote); HTS (high throughput screen); JNK (c-Jun N-terminal kinase); MAPK (mitogen-activated protein kinase); PI3K (phosphoinositide 3-kinase); PKB (protein kinase B); PKC (protein kinase C); SMI (small molecule inhibitor); TRYP (trypomastigote).
  • Anopheles stephensi ASE, MSQ43
  • Anopheles gambiae 4a3B, SUA
  • These cells are not derived from specific tissues, but rather were selected because they grew continuously under culture conditions after isolation. In general, these cells are phagocyte-like and immune-responsive, but cellular physiology is more muscle cell-like (Giulivi et al., Biochem J, 415:309-16, 2008).
  • Small molecule inhibitors are analyzed using these cells first to establish concentration ranges that inhibit target protein kinases without toxic effects on mosquito cells.
  • SMIs are diluted over a log-range to assay effects on mosquito cell doubling time (about 22 h).
  • Assays are performed in 96 well plates using CellTiter 96® Aqueous Non-Radioactive Cell Proliferation Assay (Promega).
  • HTSs high-throughput screens
  • Anopheles sp. recombinant kinases e.g., PKCs, JNK, and p38 MAPK
  • IMAP immobilized metal ion-affinity fluorescence polarization is used to screen 10,000 kinase-focused collection compounds (Cancer Research Technology) at 30 ⁇ M.
  • An exemplary HTS uses 10 pM of the kinase, 100 nM 5-FAM (fluorescein-amidite)-labeled peptide, and 30 ⁇ M ATP in a buffer consisting of 20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM DTT and 0.25 mM EGTA as described (Kjaer et al., Biochem J, 451:329-342, 2013).
  • SMIs with low IC 50 ⁇ 2 ⁇ M
  • Millipore KinaseProfilerTM service Millipore KinaseProfilerTM service
  • KinomescanTM Discoverx
  • Synthetic peptide targets (MARCKS peptide MO2-58 for cPKC, PKC-delta, PKC-epsilon; sc-3108 peptide for PKC-zeta; sc-3106 for PKC-mu or PKD) are used to distinguish effects of SMIs on PKC activity in Anopheles cells.
  • Synthetic peptide targets (ERKtide, p38 substrate P03-58 or MK-2, and JNK substrate c-Jun) are used to distinguish effects of SMIs on MAPK activity in Anopheles cells. Briefly, cells are treated with SMIs, lysed and prepared for dot blot analysis of kinase activity analyses through detection of phosphorylation of appropriate substrates.
  • GENBANK Accession Nos. of representative Anopheles gambiae kinase sequences are provided in Table 1-1.
  • SMIs Plasmodium growth and mammalian infection assays.
  • SMIs are routinely screened for growth effects on P. falciparum parasites maintained in human erythrocyte culture in vitro. Because parasites are ingested by mosquitoes in the formed of infected erythrocytes, testing SMIs against parasite-infected erythrocytes in vitro provides insight as to whether an SMI of interest affects only the mosquito host or affects both the mosquito host and malaria parasite biology. While SMI effects on the mosquito can all be beneficial in terms of transmission reduction, beneficial effects of SMIs for the malaria patient would necessarily depend on the demonstration of an effect of the SMI on parasite growth in erythrocytes.
  • P. falciparum growth assay Aliquots of ring stage P. falciparum NF54 culture are synchronized using sorbitol 48 h prior to the assay and then plated in 96-well flat-bottom plates in complete RPMI 1640 with HEPES, hypoxanthine, and 10% heat-inactivated human serum. Parasites are treated with an SMI of interest or with an equivalent volume of dimethyl sulfoxide (DMSO) diluent for 50 h in a candle jar in a 37° C. incubator. The assays are terminated by replacing the culture medium with RPMI 1640 with 10% formaldehyde in PBS.
  • DMSO dimethyl sulfoxide
  • Erythrocytes are stained with 10 ⁇ g of propidium iodide/ml in PBS for 1-2 h at room temperature. Infected RBCs are counted with a flow cytometer. The levels of parasite growth in response to treatment are normalized to DMSO controls, which are set to 100%. (see e.g., Pakpour et al., Infect Immun, 80:2141-9, 2012; and Drexler et al., J Exp Biol, 216:208-17, 2013).
  • yoelii 17XNL parasite densities in mouse blood are analyzed using a fluorescence plate reader (96 well plate). Although GFP emission of parasites through live tissue and skin is low, parasite loads can be quantified as described in sampled blood from live mice and in the mouse body following infection with P. y. yoelii 17XNL labeled with GFP-luciferase (obtained from Dr. S. Kappe and described in Miller et al., PLoS ONE, 8(4): e60820, 2013). The bioluminescent signal in live mice infected with P. y.
  • High throughput studies are initiated in the mouse model with IP injection of known SMI doses.
  • SMI formulations are administered daily by IP injection for 1-4 days to Plasmodium -infected mice.
  • SMI formulations are administered daily by IP injection for 3-4 days to mice prior to Plasmodium infection.
  • the effects of SMIs on parasitemia and disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established.
  • the same types of studies of are performed with orally delivered SMIs or control compounds to establish efficacy against infection and disease, as well as to determine pharmacokinetics in blood by high-performance chromatography and/or mass spectrometry of parent compounds.
  • This example provides a description of a clinical study among malaria-infected individuals.
  • the clinical and parasitological response (CPR) in test subjects receiving treatment with candidate kinase inhibitor(s) (test formulation) is compared to the CPR of control subjects receiving treatment with a control formulation (placebo or comparator).
  • Primary Objective to compare the proportions of test and control subjects that are free of parasites in the blood at Day 28 post-treatment.
  • Secondary Objective(s) to compare the proportions of test and control subjects free of parasites in the blood at Day 7 post-treatment; to compare the time of clearance of fever in test and control subjects; and to compare the time to clearance of parasites in the blood in test and control subjects.
  • This study is a subject- and observer-blinded, randomized, controlled study of malaria-infected, but otherwise healthy, adult and children subjects (ages 6 months to 55 years) to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • G6PD Deficient Subjects This study is a subject- and observer-blinded, randomized, controlled study of G6PD-deficient, malaria-infected adult and children subjects (ages 6 months to 55 years) to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • Normal and G6PD-deficient subjects are divided into groups and receive either a once-daily or twice-daily dose, given on study Day 0. Subjects receiving either the once-daily or twice-daily dosing are further divided into groups receiving dosing on Day 0, Days 0-1, Days 0-2, Days 0-3, Days 0-4, Days 0-5, Days 0-6, or Days 0-7. All subjects are asked to return at Days 1, 2, 3, 7, 14, 21, 28 and 48 for clinical evaluation and parasitemia analysis.
  • Inclusion Criteria infection with Plasmodium (including P. falciparum, P. vivax, P. ovale, P. malariae , and P. knowlesi ), ability to swallow oral medication, willingness to comply with the study protocol and ability to adhere to strict dosing requirements.
  • Exclusion Criteria clinically debilitating illness; signs or symptoms of severe or complicated malaria; and treatment with antimalarials within the previous 7 days.
  • ACPR clinical and parasitological response
  • Late clinical failure includes patients not exhibiting ETF who have danger signs or severe malaria in the presence of parasitemia on any day between Day 4 and Day 28, and presence of parasitemia on any day between Day 4 and Day 28 with a temperature of greater than or equal to 37.5° C.
  • Late parasitological failure includes patients not exhibiting ETF or LCF who have parasitemia on any day between Day 7 and Day 28 in the absence of fever (less than 37.5° C.).
  • Preferred test formulations have a clinical efficacy (ACPR) on Day 7 of at least 90%, more preferably at least 95% and most preferably at least 99%. In some aspects, preferred test formulations have clinical efficacy (ACPR) of at least 50% on Day 28, more preferably at least 75% and most preferably at least 95%. Additional efficacy targets are described in Burrows et al., Malaria J, 12:187, 2013.
  • Primary Objective to compare the proportions of test and control subjects free of parasites in the blood at Day 28 post-treatment.
  • G6PD Deficient Subjects This study is a subject- and observer-blinded, randomized, controlled study of G6PD-deficient, but otherwise healthy, adult and children subjects (ages 6 months to 55 years) at risk of contracting malaria by traveling to an endemic area to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age prior to randomization.
  • Subjects are divided into groups and receive either a once-daily or twice-daily dose, given on study Day 0 (1 or 2 days before entering the endemic area). All subjects are asked to obtain a parasitemia analysis and clinical evaluation at Day 28 post-treatment.
  • Inclusion Criteria at risk of contracting malaria by traveling to a malaria endemic area, ability to swallow oral medication, willingness to comply with the study protocol and ability to adhere to strict dosing requirements, age over six months.
  • Exclusion Criteria clinically debilitating illness; signs or symptoms of severe or complicated malaria; and treatment with antimalarials within the previous 7 days.
  • prophylaxis is defined as absence of parasitemia at Day 28 post-treatment.
  • Treatment failure is defined as the presence of parasitemia at or before Day 28.
  • Preferred test formulations have a clinical efficacy (ACPR) of at least 50% on Day 28, more preferably at least 75% and most preferably at least 95%. Additional efficacy targets are described in Burrows et al., Malaria J, 12:187, 2013.
  • SMIs are screened for growth effects on T. cruzi parasites maintained in Vero cells in vitro.
  • Vero cells (1 ⁇ 10 5 /well) seeded on round coverslips into 24-well plates are infected with bloodstream T. cruzi trypomastigotes (TRYP).
  • TRYP bloodstream T. cruzi trypomastigotes
  • the infected cultures are washed to remove free parasites and incubated for another 72 hours with a test SMI (1 to 10 micromolar), a placebo, or control comparator.
  • Cell cultures are set up in triplicate and maintained at 37° C. in 5% CO 2 . Uninfected treated cultures exposed to vehicle (1% DMSO) are used as controls.
  • Bloodstream TRYP are subjected to treatment by incubation of 1.5 ⁇ 10 5 TRYP with 1-10 micromolar of a test SMI, a placebo, or control comparator.
  • Parasites are diluted in RPMI 1640 medium supplemented with 10% FCS, seeded in 96-well microplates, and incubated at 37° C. in a 5% CO 2 atmosphere. After 24 hours, the remaining live parasites are counted in a Neubauer chamber as previously described (Fernandez et al., Experimental Parasitology, 124:172-180, 2010). Trypanocydal effect is determined by counting remaining TRYP at each concentration of drug with respect to a negative control group. Each assay is performed in triplicate.
  • At least two T. cruzi strains are used for the infection assay: one lethal at the acute phase and the other non-lethal at the acute phase. Use of two strains in this matter facilitates evaluation of parasitemia reduction and cure in the acute and chronic phases of infection.
  • SMI formulations are administered daily by IP injection for 5 days to Trypanosoma -infected mice, starting at parasitemia levels above 10 4 TRYP/ml of blood.
  • Control formulations a placebo or comparator, are tested in parallel to the SMI formulations.
  • the effects of SMIs on parasitemia e.g., twice weekly
  • disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established. The same types of studies are performed with orally delivered SMIs.
  • Lethal strain 100 blood TRYP
  • injection intradermally into the hind footpad or alternatively by intraperitoneal inoculation.
  • Non-lethal strain injected intradermally into the hind footpad or alternatively by intraperitoneal inoculation.
  • a fluorescently-labeled non-lethal K98 strain may be used.
  • Primary Objective to compare the proportions of test and control subjects that are free of parasites in the blood at 12 months post-treatment.

Abstract

The present disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite. In some aspects, the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in arthropods feeding on recipients of the regimens. Additionally, the present disclosure provides methods for screening candidate anti-parasitic agents.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 61/923,405, filed Jan. 3, 2014, and U.S. Provisional Application No. 61/845,920, filed Jul. 12, 2013, which are incorporated herein by reference in their entirety for all purposes.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This invention was made with Government support under Grant Nos. AI073745 and AI080799, awarded by National Institutes of Health. The Government has certain rights in the invention.
  • FIELD
  • The present disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite. In some aspects, the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in arthropods feeding on recipients of the regimens. Additionally, the present disclosure provides methods for screening candidate anti-parasitic agents.
  • BACKGROUND
  • Chagas disease, also known as American trypanosomiasis, is a parasitic disease caused by Trypanosoma cruzi. T. cruzi is a protozoan parasite that is mainly transmitted by blood-feeding insects known as kissing bugs (triatomines) through their feces and contaminated objects, including food, and drink. The area where kissing bug species have been reported spans from the southern part of South America up to the Great Lakes in the northern part of the United States (Bern et al., N Engl J Med, 364:2527-2534, 2011).
  • Clinically Chagas disease is classified into acute and chronic phases. The acute phase begins when T. cruzi infiltrates the body and starts multiplying within different organs and tissues. The symptoms during the acute phase can be mild and nonspecific, including fever and swelling at the site of infection (e.g., swelling of the eyelid, also referred to as Romana's sign). However, in some instances, acute infection can cause severe inflammation of the heart or brain (myocarditis or meningoencephalitis) which can be deadly (Bern et al., supra, 2011). The spectrum of symptoms depends on factors such as the parasite burden and host immunocompetence. The acute phase lasts 4 to 8 weeks, after which initial symptoms cease, and the infection enters a lifelong chronic phase. In the chronic phase, intracellular T. cruzi amastigotes remain in infected tissues, especially in cardiac and skeletal muscle, but the parasitemia levels become undetectable by microscopy. Clinical symptoms of chronic Chagas disease can develop decades later in a significant number of chronically infected individuals. In this way, people unaware of their infection can unwittingly transmit the parasite via blood, tissue, and organ donations, as well as from mother to child. In about 30% of chronically-infected individuals Chagas disease develops into a life-threatening illness characterized by abnormal heart rhythms leading to an increased risk of sudden death, heart failure, and/or gastrointestinal disturbances.
  • According to the Centers for Disease Control (CDC), an estimated eight million people living in Mexico, Central and South America are infected. New figures on the prevalence of the disease in endemic countries combined with data on immigration suggests that about 300,000 immigrants in the United States are likely infected with T. cruzi. Thus, Chagas disease is a serious illness affecting a significant portion of the population.
  • Even though T. cruzi was first discovered a century ago, Chagas disease remains largely ignored. There are only two drugs used to treat acute-phase disease, benznidazole (since 1960s) and nifurtimox (since 1970s), both of which are impractical because of low efficacy and frequent side effects. Additionally, neither benznidazole nor nifurtimox is approved for use by the United States Food and Drug Administration (Bern et al., supra, 2011). Treatment of the chronic form of the disease is still limited to the management of symptoms (e.g., antiarrhythmics, steroids, pacemakers, etc.). Even heart transplantation cannot ensure a cure, because the parasites hidden within other organs will eventually reinvade the transplanted heart. Despite the well-recognized limitations of current therapeutic approaches and the general acceptance that specific treatment should be made available to infected individuals, new Chagas disease treatments have not reached the clinic. Moreover, T. cruzi like Plasmodium sp. (arthropod-borne parasite that causes malaria) changes rapidly, thus quickly acquiring resistance to current treatments. For this further reason and because there are no effective Chagas disease treatments that target T. cruzi when it resides within a host cell (see, e.g., Urbina, Acta Trop, 115:55-68, 2010), there is a need for an expanded drug arsenal for targeting T. cruzi at different stages in its life cycle.
  • The CDC has named Chagas disease as one of five (along with Neurocysticercosis, Toxocariasis, Toxoplasmosis, and Trichomoniasis) neglected parasite infections in the United States as public health priorities based on the severity of the illnesses, the number of people infected, and the ability to prevent and treat the disease. Thus, there remains a need for therapy regimens to treat or prevent other diseases caused by arthropod-borne parasites, which are safe and do not encourage development of drug-resistant parasites. In particular, new anti-Chagas disease drugs with an improved safety profile are urgently needed.
  • BRIEF SUMMARY
  • The present disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite. In some aspects, the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in arthropods feeding on recipients of the regimens. Additionally, the present disclosure provides methods for screening candidate anti-parasitic agents.
  • In one aspect, the present disclosure provides methods for treating or preventing a parasitic disease caused by an arthropod-borne parasite by administering a protein kinase inhibitor to a mammalian subject infected with or at risk of exposure to the arthropod-borne parasite (e.g., a mammalian subject in need thereof) under conditions effective for treating or preventing the parasitic disease. In some embodiments, the arthropod-borne parasite is a protozon parasite. In some embodiments, the present disclosure provides methods for treating or preventing a disease caused by a protozoan parasite comprising: administering a first protein kinase inhibitor and a second protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing the disease caused by the protozoan parasite. The present disclosure also provides methods for treating or preventing a disease caused by a protozoan parasite comprising: administering a protein kinase inhibitor and an additional antiprotozoan drug to a mammalian subject in need thereof under conditions effective for treating or preventing the disease caused by the protozoan parasite. In some embodiments, the administering reduces activity of at least one mammalian protein kinase. In some embodiments, the protein kinase inhibitor comprises a first and a second protein kinase inhibitor. In some preferred embodiments, the protozoan parasite is an arthropod-borne parasite selected from the group consisting of Plasmodium sp., Leishmania sp., Babesia sp., and Trypanosoma sp. In some embodiments, the parasitic disease is not malaria and the parasite is not a species of Plasmodium. In some embodiments, the protein kinase inhibitor reduces activity of an arthropod kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some embodiments, the subject is infected with a protozoan parasite. In some embodiments, the subject is infected with a drug-resistant protozoan parasite (e.g., resistant to currently used anti-parasitic drugs). In some embodiments, the subject is acutely infected with a protozoan parasite, while in other embodiments the subject is chronically infected with a protozoan parasite. In some embodiments, the subject is experiencing a symptom of the disease prior to the administering step. In some embodiments, treating the disease comprises alleviating a symptom of the disease experienced by the subject. In some embodiments, the subject is an uninfected individual planning to visit a disease endemic area after (6 hours-72 hours, 1, to or 3 days) the administering step. That is the individual is not infected with but likely to be exposed to a protozoan parasite. In some embodiments, the administering commences and concludes before the uninfected individual visits a parasitic disease endemic area. In other embodiments, the administering commences before the uninfected individual visits a parasitic disease endemic area, and continues during the duration of their visit. That is in some embodiments, the administering concludes sometime after the uninfected individual leaves the parasitic disease endemic area (e.g., within thirty days of after last potential exposure). In some embodiments, preventing the disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days (between 0 and 30 days). In some embodiments, drug-resistant protozoan parasites (e.g., resistant to currently used anti-parasitic drugs) are known to be present in the disease endemic area.
  • Specifically, the present disclosure provides methods for treating or preventing Chagas disease comprising: administering a mammalian protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing Chagas disease. In some embodiments, the mammalian protein kinase inhibitor comprises a first kinase inhibitor and a second kinase inhibitor, and the administering reduces activity of a first mammalian protein kinase and a second mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of a protein kinase C (PKC) family, a member of the protein kinase B (PKB) family, and/or a member of the phosphoinositide 3-kinase (PI3K) family. In some embodiments, the mammalian protein kinase inhibitor is capable of reducing activity of a Triatominae insect kinase. In some embodiments, the methods further comprise administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject. In some embodiments, the subject is infected with Trypanosoma cruzi. In some embodiments, the subject is acutely infected with T. cruzi, while in other embodiments the subject is chronically infected with T. cruzi. In some preferred embodiments, treating Chagas disease comprises alleviating a symptom of Chagas disease experienced by the subject. In some embodiments, the methods further comprise administering an effective amount of an additional antiparasitic agent to the mammalian subject. In some embodiments, the additional antiparasitic agent comprises one or both of benznidazole and nifurtimox. In some embodiments, the subject is an uninfected individual planning to visit a Chagas disease endemic area (e.g., Mexico, Central America or South America) after the administering step. That is the individual is not infected with but likely to be exposed to T. cruzi. In some embodiments, the administering commences and concludes before the uninfected individual visits a Chagas disease endemic area. In other embodiments, the administering commences before the uninfected individual visits a Chagas disease endemic area, and continues during the duration of their visit. That is in some embodiments, the administering concludes sometime after the uninfected individual leaves the Chagas disease endemic area (e.g., within thirty days of after last potential exposure). In some embodiments, preventing Chagas disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days. Generally, the present disclosure provides method for treating or preventing Chagas disease comprising: administering a protein kinase inhibitor to a subject infected with Trypanosoma cruzi under conditions effective for treating or preventing Chagas disease. Likewise, the present disclosure provides methods for treating or preventing a parasitic disease comprising: administering a protein kinase inhibitor to a subject infected with an arthropod-borne parasite under conditions effective for treating or preventing a parasitic disease.
  • Furthermore, the present disclosure provides methods of reducing transmission of Trypanosoma cruzi by a Triatominae vector, comprising administering a protein kinase inhibitor to a mammalian subject infected with Trypanosoma cruzi. under conditions effective for reducing transmission of the Trypanosoma cruzi ingested by the Triatominae vector in the subject's blood. The present disclosure also provides methods of reducing transmission of Trypanosoma cruzi by a Triatominae vector, comprising providing a bloodmeal comprising erythrocytes infected with Trypanosoma cruzi to a Triatominae vector in the presence of an effective amount of a protein kinase inhibitor for reducing transmission of the Trypanosoma cruzi ingested by the Triatominae vector in the bloodmeal. In some embodiments, the methods further comprise administering an additional anti-parasitic drug to the mammalian subject. In some embodiments, the bloodmeal is provided to the Triatominae vector in the further presence of an additional antiparasitic drug. In some embodiments, the protein kinase inhibitor comprises a first and a second protein kinase inhibitor. In some embodiments, the protein kinase inhibitor reduces activity of at least one mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some preferred embodiments, the protein kinase inhibitor reduces activity of a Triatominae kinase. In some embodiments, the Triatominae kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms, and p38 MAPK isoforms. In some embodiments, the Triatominae kinase comprises aPKC-zeta. In some embodiments, the subject is infected with Trypanosoma cruzi. In some embodiments, the additional anti-parasitic drug comprises a one or both of benznidazole and nifurtimox.
  • The present disclosure also provides methods of identifying an antiparasitic compound, comprising: measuring activity of a mammalian or insect protein kinase in the presence and absence of a test compound; and identifying the test compound as an antiparasitic compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound. In some embodiments, the methods further comprise growing insect cells in vitro in the presence and absence of the test compound and identifying the test compound as insect-cell safe when viability or doubling time of the insect cells is not significantly reduced in the presence as compared to the absence of the test compound. In some embodiments, the mammalian protein kinase is a human protein kinase. In some embodiments, the human protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some embodiments, the methods are suitable for identifying an anti-Chagas disease compound. In such embodiments, the insect protein kinase is a Triatominae protein kinase, and the insect cells are Triatominae cells. In some embodiments, the Triatominae kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms and p38 MAPK isoforms. In some embodiments, the Triatominae kinase comprises aPKC-zeta. In some embodiments, the Triatominae cells are Rhodnius sp. cells (e.g., Rhodnius prolixus cells). The present disclosure also provides methods of identifying an anti-Chagas disease compound, comprising: comparing development of parasites in Triatominae vector after consumption of a bloodmeal comprising Trypanosoma cruzi in the presence and absence of a test compound; and identifying the test compound as an anti-Chagas disease compound when the development of parasites is reduced in the Triatominae vector in the presence as compared to the absence of the test compound.
  • Additionally the present disclosure provides pharmaceutical compositions comprising a first protein kinase inhibitor, a second protein kinase inhibitor, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitors are present in amounts effective to treat or prevent Chagas disease in a mammalian subject. In further embodiments, the present disclosure provides pharmaceutical compositions comprising a protein kinase inhibitor, an additional antiparasitic agent, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitor and the additional antiparasitic agent are present in amounts effective to treat or prevent Chagas disease in a mammalian subject. In still further embodiments, the present disclosure provides pharmaceutical compositions comprising: a protein kinase inhibitor attached to a mitochondrial-targeting moiety, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the protein kinase inhibitor is present in an amount effective to treat or prevent Chagas disease in a mammalian subject.
  • The present disclosure provides methods for treating or preventing an arthropod-borne parasitic disease comprising: administering a mammalian protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing the arthropod-borne parasitic disease, wherein the parasitic disease is not malaria. In some embodiments, the mammalian protein kinase inhibitor comprises a first kinase inhibitor and a second kinase inhibitor, and the administering reduces activity of a first mammalian protein kinase and a second mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a protein kinase B (PKB) family, and a phosphoinositide 3-kinase (PI3K) family. In some embodiments, the mammalian protein kinase inhibitor is capable of reducing activity of an arthropod kinase. In some embodiments, the methods further comprise administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject. In some embodiments, the subject is infected with an arthropod-borne parasite which is not a Plasmodium sp. parasite. In some embodiments, the subject is acutely infected with the arthropod-borne parasite, while in other embodiments, the subject is chronically infected with the arthropod-borne parasite. In some embodiments, treating the parasitic disease comprises alleviating a symptom of the parasitic disease experienced by the subject. In some embodiments, the methods further comprise administering an effective amount of an additional antiparasitic agent to the mammalian subject. In some embodiments, the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step commences before the visit. In some embodiments, the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step concludes during or after the visit. In some embodiments, preventing the parasitic disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days.
  • Furthermore, the present disclosure provides methods of reducing transmission of an arthropod-borne parasite by an arthropod, comprising administering a protein kinase inhibitor to a mammalian subject infected with an arthropod-borne parasite under conditions effective for reducing transmission of the an arthropod-borne parasite ingested by the arthropod in the subject's blood. The present disclosure also provides methods of reducing transmission of an arthropod-borne parasite by an arthropod vector, comprising providing a bloodmeal comprising erythrocytes infected with an arthropod-borne parasite to an arthropod in the presence of an effective amount of a protein kinase inhibitor for reducing transmission of the an arthropod-borne parasite ingested by the arthropod in the bloodmeal. In some embodiments, the methods further comprise administering an additional anti-parasitic drug to the mammalian subject. In some embodiments, the bloodmeal is provided to the arthropod vector in the further presence of an additional antiparasitic drug. In some embodiments, the protein kinase inhibitor comprises a first and a second protein kinase inhibitor. In some embodiments, the protein kinase inhibitor reduces activity of at least one mammalian protein kinase. In some embodiments, the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family. In some preferred embodiments, the protein kinase inhibitor reduces activity of an arthropod kinase. In some embodiments, the arthropod kinase comprises one or more of the group consisting of cPKC, nPKC-delta, nPKC-epsilon, aPKC-zeta, PKD, PKN, JNK isoforms, and p38 MAPK isoforms. In some embodiments, the arthropod kinase comprises aPKC-zeta. In some embodiments, the subject is infected with an arthropod-borne parasite. In some embodiments, the arthropod-borne parasite is not a species of Plasmodium and the arthropod is not a species of Anopheles.
  • Additionally, the present disclosure provides, methods of identifying an antiparasitic compound, comprising: measuring activity of a mammalian or insect protein kinase in the presence and absence of a test compound; and identifying the test compound as an antiparasitic compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound, and wherein the insect protein kinase is not a Anopheles sp. kinase. In some embodiments, the methods further comprise growing insect cells in vitro in the presence and absence of the test compound and identifying the test compound as insect-cell safe when viability or doubling time of the insect cells is not significantly reduced in the presence as compared to the absence of the test compound, and wherein the insect cells are not Anopheles sp. cells. In some embodiments, the mammalian protein kinase is a human protein kinase. In some embodiments, the insect protein kinase is selected from the group consisting of a sand-fly protein kinase, a tick protein kinase and a tsetse fly protein kinase.
  • Also provided by the present disclosure are pharmaceutical compositions comprising a first protein kinase inhibitor, a second protein kinase inhibitor, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitors are present in amounts effective to treat or prevent an arthropod-borne parasitic disease in a mammalian subject, wherein the parasitic disease is not malaria. In further embodiments, the present disclosure provides pharmaceutical compositions comprising a protein kinase inhibitor, an additional antiparasitic agent, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the kinase inhibitor and the additional antiparasitic agent are present in amounts effective to treat or prevent an arthropod-borne parasitic disease in a mammalian subject, wherein the parasitic disease is not malaria. In still further embodiments, the present disclosure provides pharmaceutical compositions comprising: a protein kinase inhibitor attached to a mitochondrial-targeting moiety, and one or both of a pharmaceutically acceptable excipient and carrier, wherein the protein kinase inhibitor is present in an amount effective to treat or prevent an arthropod-borne parasitic disease in a mammalian subject, wherein the parasitic disease is not malaria. In some embodiments, the parasitic disease is selected from the group consisting of leishmaniasis, babesiosis, and African trypanosomiasis.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A-C provides names and structures of exemplary protein kinase inhibitors.
  • FIG. 2A-2B shows that Anopheles stephensi p38 MAPK can be inhibited by SB203580 (SB) and BIRB 796, although neither inhibitor directly altered Plasmodium falciparum growth in vitro. In FIG. 2A, Anopheles stephensi ASE cells were pre-incubated with 10 μM SB203580 or 10 μM BIRB 796 and 2 h later stimulated with 1 μg/ml LPS. Phosphorylation of MK2, a downstream protein kinase substrate activated by p38 MAPK, was analyzed by western blot. Vehicle treatment was used as control group. Note that p-MK2 levels are undetectable in cells pre-treated with either inhibitor. In FIG. 2B, in vitro culture of P. falciparum was synchronized, treated with 1 μM or 10 μM SB203580 or 1 μM or 10 μM BIRB 796 for 48 h, then fixed and stained with propidium iodide. Parasitemia was determined by flow cytometry. Data were analyzed by Student's t-test (alpha=0.05) and are represented as mean±SEM, n=3. Neither inhibitor significantly altered parasite growth relative to the vehicle control.
  • FIG. 3A-3D shows that inhibition of p38 MAPK signaling reduced Plasmodium falciparum development in Anopheles stephensi. Briefly, 3-5 d old female A. stephensi were fed a bloodmeal with P. falciparum supplemented with SB203580, BIRB 796, or diluent control. Midguts were dissected and oocysts were counted at 10 days following infection. Dots in FIG. 3A and FIG. 3C represent oocyst numbers in one midgut, while percentage positive midguts or percentage of midguts with at least one oocyst are represented as means in FIG. 3B and FIG. 3D. Mean oocysts per midgut (lines, numbers on graph) were analyzed by ANOVA (alpha=0.05) for overall significance and by Student-Neuman-Keuls for pairwise comparisons using Graphpad Prism version 4.0 (San Diego, Calif.). Prevalences of mosquito infection (indicated on graph) were analyzed by chi-square (alpha=0.05). At 10 μM, SB203580 significantly reduced P. falciparum oocysts/midgut and infection prevalence in A. stephensi (*).
  • FIG. 4A-F shows that inhibition of Anopheles stephensi p38 MAPK in vivo increased midgut expression of anti-parasite genes. Briefly, 3-5 d old female A. stephensi were fed with killed lysate of P. falciparum supplemented with BIRB 796 or vehicle. Midgut samples were collected at 1-24 h post-feeding. Expression levels of anti-malarial genes at each time point were analyzed with quantitative PCR relative to the corresponding control group. Each dot represents the fold change in one repeat relative to control levels. Data were analyzed by Student's t-test (alpha=0.05), n=3-5. At 6 h post-feeding immune gene expression levels were significantly different from controls.
  • FIG. 5 shows that the midgut pJNK1/3 in A. stephensi is reduced after a P. falciparum-infected bloodmeal containing 10 μM SP600125, relative to controls. NB=no blood, B=blood, P=parasites, SP=SP600125.
  • FIG. 6A-D shows the prevalence (%) and intensity (mean oocysts/midgut) of P. falciparum infection in two separate cohorts of A. stephensi (n=50) is reduced after a P. falciparum-infected bloodmeal containing 10 μM SP600125, relative to controls.
  • FIG. 7 shows the fold change in midgut mRNA expression levels of complement-like immune factors (TEP1, APL1, LRIM1), a leucine-rich repeat gene (LRRD7), defensin (DEF), and nitric oxide synthase (NOS) following a bloodmeal containing 10 μM SP600125, relative to an untreated control bloodmeal (set at 1, dashed line). n=2 biological replicates.
  • DETAILED DESCRIPTION
  • The present disclosure provides methods for treating or preventing diseases caused by arthropod-borne parasites by administration of a protein kinase inhibitor, optionally with one or both of a further protein kinase inhibitor and an antiparasitic agent to a mammalian subject infected with or at risk of exposure to an arthropod-borne parasite. In some aspects, a protein kinase inhibitor is administered to the subject, while in other aspects a protein kinase inhibitor and one or both of a further protein kinase inhibitor and an antiparasitic agent are administered to the subject. In some aspects, the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in arthropods feeding on recipients of the regimens (e.g., treated mammalian subjects). Additionally, the present disclosure provides methods for screening candidate anti-parasitic agents.
  • In one aspect, the present disclosure provides methods for treating or preventing Chagas disease by administration of a protein kinase inhibitor and optionally one or both of a further protein kinase inhibitor and an anti-Chagas disease agent to a mammalian subject infected with or at risk of exposure to Trypanosoma cruzi. In some aspects, the therapeutic and prophylactic regimens of the present disclosure are effective in reducing parasite development in Triatominae (e.g., kissing bugs) feeding on recipients of the regimens (e.g., treated mammalian subjects). Additionally, the present disclosure provides methods for screening candidate anti-Chagas disease agents.
  • Among the 140 triatomine species, Rhodnius prolixus—an important vector of Trypanosoma cruzi, the causative agent of Chagas' disease in Central and South America—has become a model for insect physiology and biochemistry. The genome of this species was recently targeted for sequencing, along with several organ-specific cDNA libraries including the midgut, the insect organ in which T. cruzi grows and develops. As such, R. prolixus, like Anopheles mosquitoes, can be targeted via ingested drugs from the mammalian host during bloodfeeding to inhibit parasite development. Critically, the midgut transcriptome of R. prolixus includes highly conserved proteins of the major signaling pathways initially determined to be of interest in the context of malaria, including the MAPKs (JNK, p38 MAPK), and the PKCs (see, e.g., Ribeiro et al., PLoS Neglected Tropical Diseases, 8: e2594, 2014).
  • The disclosure herein pertaining to malaria, Plasmodium sp. and mosquitoes is provided as an exemplary disclosure of embodiments that are applicable in general to diseases caused by arthropod-borne parasites. In particular, the disclosure herein pertaining to malaria, Plasmodium sp. and mosquitoes is also generally applicable to Chagas disease (American trypanosomiasis), Trypanosoma cruzi and Triatominae.
  • Kinase Inhibitors
  • PKC (protein kinase C) isoenzymes are an important family of serine/threonine protein kinases that contribute to many diverse cellular and tissue functions, as well as human disease pathologies including cancer development and progression (Bosco et al., Mini Rev Med Chem, 11:185-199, 2011; and Rosse et al., Nat Rev Mol Cell Biol, 11:103-112, 2010). The molecular architecture of PKC family members is conserved throughout the cPKC (classical PKC; cPKC α, cPKC β and cPKC γ), nPKC (novel PKC; nPKC δ, nPKC ε, nPKC η and nPKC θ) and aPKC (atypical PKC; a PKC ζ and aPKC ι) isoforms. All PKCs are comprised of a C-terminal serine/threonine protein kinase domain belonging to the AGC kinase (protein kinase A/protein kinase G/PKC family kinase) superfamily, and an N-terminal regulatory domain (Parker and Murray-Rust, J Cell Science, 117:131-132, 2004). The kinase domain has a C-terminal extension unique to AGC kinases that contains essential ‘priming’ phosphorylation sites required for catalytic activation (Pearce et al., Nat Rev Mol Cell Biol, 11:9-22, 2010), whereas in aPKCs, the most C-terminal of these is replaced by a phospho-mimetic Glu residue. The regulatory domain within all PKC isoforms has an inhibitory region (pseudo-substrate motif) and distinctive arrangements of C1, C2 and/or PB1 (Phox/Bem1) domains (the last being exclusive to the aPKCs) that together are crucial for conferring isoform-specific functions.
  • The aPKC isoforms (PKC ζ and PKC ι) diverge from other PKC family members as their regulatory domains are unresponsive to diacylglycerol, phosphatidylserine and Ca2+. Instead they are regulated by protein interactions, for example with polarity proteins Par-6, Par-3 and the Rho family GTPase cdc42 (Suzuki and Ohno, J Cell Sci, 119:979-987, 2003). aPKC has a role in establishing cell polarity and is required for normal cell proliferation, mitotic spindle orientation and migration.
  • There has been considerable interest in aPKCs as drug targets due to their roles in cancer development and progression (Fields and Regala, Pharmacol Res, 55:487-497, 2007). For example, PKC ζ is required for epidermal growth factor-induced migration of human breast and lung cancer cells and for colony-stimulating factor 1 chemotaxis of macrophages, among others. PKC ι promotes nicotine-induced migration and invasion of lung cancer cells via phosphorylation of calpains and is involved in other types of cancer. In addition, aPKC has been shown to interact with bona fide oncogenes, such as Ras and Notch in the fruit fly Drosophila melanogaster to promote an epithelial cell overgrowth that resembles cancer. Recent advances in malaria drug discovery have led to a proposal to use Plasmodium kinases as targets for next generation antimalarials (Zhang et al., Curr Top Med Chem, 12:456-472, 2012; Lin et al., PLoS One, 7, e49040, 2012; Lucet et al., Future Med Chem, 4:2295-2310, 2012; Aaberg et al., Biol Chem, 393:1121-1129, 2012; Biamonte at al., Bioorg Med Chem Lett, 23:2829-2843, 2013; Nag et al., Curr Drug Discovery Technol, 10:85-91, 2013; and Ochocki and Distefano, Med Chem Commm, 4:476-492, 2009). These reports, however, failed to consider manipulating kinase signalling pathways in the human or mosquito hosts to reduce parasitemia and block transmission.
  • Like many protein kinases, aPKC isoenzymes can be inhibited by small-molecule chemicals either through their ATP-binding or allosteric pockets (Knight and Shokat, Chem Biol, 12:621-637, 2005). Only a few selective chemical biology tools inhibit aPKC catalytic activity. These few catalytic inhibitors include the non-selective Gö6983 and Gö6976 compounds and a PKC ζ pseudo-substrate peptide containing a membrane-targeting myristoylation site (Roffey et al., Curr Opin Cell Biol, 21:268-279, 2009). The development of effective ATP-competitive inhibitors against aPKCs, similar to other protein kinases, has been complicated by challenges with both potency and selectivity. In general, such inhibitors acquire selectivity by associating with both the ATP-binding site and with adjacent residues, which are less well conserved. Other approaches include screening for allosteric inhibitors of PKC ζ that target the PIF pocket (Frohner et al., J Med Chem, 54:6714-6723, 2011; and Lopez-Garcia et al., Chem Biol, 18:1463-1473, 2011) or that block PKC ι interaction with Par-6 (Pillai et al., Int J Biochem Cell Biol, 43:784-794, 2011; Erdogran et al., J Biol Chem, 281:28450-18459, 2006; and Regala et al., Cancer Res, 68: 5888-5895, 2008). Recent studies have shown differential effects between PKC ζ and PKC ι for compounds targeting other aPKC-specific pockets outside of the ATP cleft. For instance, the use of CRT0066854 and its derivative compounds are predicted to be useful tools in further differentiating the roles of PKC ζ and PKC ι in establishing cell polarity and growth-factor-stimulated signaling pathways (Kjaer et al., Biochem J, 451:329-342, 2013).
  • PKC ζ plays an important role in the activation of TNF-alpha and NF-kB pathways, the latter of which are critical to the balance between host protection and pathology in malaria (Randall and Engwerda, Exp Parasitol, 126:326-331, 2010). In the context of malaria parasite biology, mammalian PKC activity has recently been shown to be critical for malaria parasite egress from infected red blood cells (Millholland et al., Cell Host Microbe, 13:15-28, 2013). Intriguingly, erythrocyte PKCs are comprised of cPKCα, as well as aPKC ζ and aPKC ι (Govekar and Zingde, Ann Hematol, 80:531-534, 2001). Thus during development of the present disclosure, aPKCs have been determined to be a primary target of PKC inhibitors that can reduce parasitemia. The effect on host RBC PKC activity is further substantiated by an examination of host versus parasite IC50s: the IC50 for PKC ζ is 60 nM (Gschwendt et al., FEBS Lett, 392:77-80. 1996), whereas the IC50 for the distantly (and only) related malaria parasite kinase PfPKB is >1000 nM (Kumar et al., J Biol Chem, 279:24255-24264, 2004). In the mosquito host, conserved PKC activity also controls parasite development: ingestion of PKC inhibitors, at levels consistent with those achievable in human blood, during feeding on an infected meal resulted in significantly reduced P. falciparum development in the mosquito Anopheles stephensi (Pakpour et al., PLoS One, 8(10): e76535, 2013). Further, expression of A. stephensi PKC ζ mRNA was significantly increased (5-fold) in the mosquito midgut upon blood feeding, a change that was consistent with PKC-dependent regulation of epithelial barrier permeability that likely controls resistance to parasite infection.
  • Effective inhibitor targeting of PKC activity in general and for aPKCs in particular is challenging. However, networked kinases, which interact as kinase and kinase substrate, may provide an opportunity for novel inhibitor design. In the mosquito, p38 MAPK inhibition significantly reduces malaria parasite development, observations confirmed with commercially available p38 MAPK inhibitors during development of the present disclosure (FIG. 3A-D). Prior to this work, the p38 MAPK inhibitor SB203580 was reported to reduce P. falciparum replication in human erythrocytes in vitro at concentrations exceeding 5 μM (Brumlik et al., Exp Parasitol, 128:170-175, 2011). However, no such effects of SB203580 up to 10 μM and no activity of a second p38 MAPK inhibitor BIRB 796 up to 10 μM against P. falciparum growth in vitro was observed (FIG. 2B). However, both inhibitors at 10 μM significantly reduced p38 MAPK activity in the mosquito in vivo when provided in the bloodmeal immediately prior to feeding, indicating that mosquito p38 MAPK activity controls parasite development (FIG. 2A). In mammalian cells, the PKC regulatory domain interacts directly with p38 MAPK kinase, which blocks PKC activity by preventing its autophosphorylation on T560 (but not on S113 or S186) and activation (Kim et al., Cell Death Differ, 12:201-212, 2005). Thus strategies to target p38 MAPK can be leveraged to enhance aPKC targeting. Specifically, in some embodiments, a PKC inhibitor (e.g., a molecule that is directed to the PKC PIF pocket) is used in combination with a p38 MAPK inhibitor to inhibit networked p38 MAPK and PKC ζ to enhance control of infection in the human host and transmission via the exposed mosquito. Of note, some p38 MAPK inhibitors have progressed into human clinical trials for not only cancer but also to inflammatory disease and cardiovascular disease with manageable side effects (Hammaker et al., Ann Rheum Dis, 69(S1):77-82, 2010).
  • In addition to inhibitors of aPKC and p38 MAPK, other kinase inhibitors, such as the JNK inhibitor SP600125 has been shown to reduce malaria disease severity, but without an effect on parasitemia (Anand et al., Parasitol Res, 112:1959-1966, 2013). However, during development of the present disclosure, inhibition of mosquito JNK activity was found to alter P. falciparum development in A. stephensi. Hence, in some embodiments, a JNK inhibitor in combination with a PKC inhibitor is used to reduce disease severity and parasitemia. Combinations of small molecule inhibitors (SMIs) of conserved kinases are provided as “next-generation” anti-malarials to control both disease severity and transmission, as well as to reduce risk of development of parasite drug resistance.
  • In some embodiments, SMIs of conserved signaling kinases are delivered as orally available therapeutics to malaria patients in endemic countries. The following list of SMI therapeutic attributes is applicable to both pediatric and adult patients: (1) single dose (or few dose) efficacy with inhibition of parasite growth and resolution of disease following treatment, (2) few to no contraindications and side effects, (3) long-term stability under conditions in endemic countries where refrigeration during storage and distribution are limited at best, (4) parasite transmission blocking activity in mosquitoes that feed on treated patients to counter potential “escape” of parasites from treated patients, (5) well-tolerated by the most sensitive populations, children and pregnant women as well as by G6PDH-deficient patients.
  • Structures of exemplary kinase inhibitors are shown in FIG. 1. Kinase inhibitors for use in the methods of the present disclosure include but are not limited to inhibitors of PKC, JNK, MAPK and other kinases, as well as salts and derivatives thereof. Exemplary PKC inhibitors include: bisindolylmaleimide IV (3,4-bis(1H-indol-3-yl)pyrrole-2,5-dione); chelerythrine chloride (1,2-dimethoxy-12-methyl-[1,3]benzodioxolo[5,6-c]phenanthridin-12-ium; chloride); Go 6976 (5,6,7,13-Tetrahydro-13-methyl-5-oxo-12H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-12-propa-nenitrile); Go 6983 (3-[1-[3-(dimethylamino)propyl]-5-methoxyindol-3-yl]-4-(1H-indol-3-yl)pyrrole-2,5-dione); PKCβII/EGFR Inhibitor (5,6-bis(4-fluoroanilino)isoindole-1,3-dione); PKCβ Inhibitor (3-anilino-4-[1-(3-imidazol-1-ylpropyl)indol-3-yl]pyrrole-2,5-dione); N-benzoyl-staurosporine, staurosporine (Streptomyces sp.); bisindolylmaleimide III, hydrochloride (3-[1-(3-aminopropyl)indol-3-yl]-4-(1H-indol-3-yl)pyrrole-2,5-dione; hydrochloride); Go 7874, hydrochloride; H-8, dihydrochloride (N-[2-(methylamino)ethyl]isoquinoline-5-sulfonamide; dihydrochloride); HA 1004, dihydrochloride (2-[2-(isoquinolin-5-ylsulfonylamino)ethyl]guanidine; dihydrochloride); UCN-01 (7-hydroxystaurosporine); and Ro-31-8220 (methanesulfonic acid; 3-[3-[4-(1-methylindol-3-yl)-2,5-dioxopyrrol-3-yl]indol-1-yl]propyl carbamimidothioate). Exemplary MAPK inhibitors include: p38 MAP Kinase Inhibitor IV (3,4,6-trichloro-2-(2,3,5-trichloro-6-hydroxyphenyl)sulfonylphenol); p38 MAP Kinase Inhibitor VI, JX401 ((4-benzylpiperidin-1-yl)-(2-methoxy-4-methylsulfanylphenyl)methanone); p38 MAP Kinase Inhibitor VII, SD-169 (1H-indole-5-carboxamide); p38 MAP Kinase Inhibitor VIII ([4-(2-amino-4-bromoanilino)-2-chlorophenyl]-(2-methylphenyl)methanone); BIRB 796 (1-[5-tert-butyl-2-(4-methylphenyl)pyrazol-3-yl]-3-[4-(2-morpholin-4-ylethoxy)naphthalen-1-yl]urea); SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine); SB 203580, Sulfone (4-[4-(4-fluorophenyl)-2-(4-methylsulfonylphenyl)-1H-imidazol-5-yl]pyridine); and SB 239063 (4-[4-(4-fluorophenyl)-5-(2-methoxypyrimidin-4-yl)imidazol-1-yl]cyclohexan-1-ol). Exemplary inhibitors of other kinases include: U0126 ((2Z,3Z)-2,3-bis[amino-(2-aminophenyl)sulfanylmethylidene]butanedinitrile); PD98059 (2-(2-amino-3-methoxyphenyl)chromen-4-one); and MEK1/2 Inhibitor II (N-(cyclopropylmethoxy)-3,4,5-trifluoro-2-(4-iodo-2-methylanilino)benzamide).
  • Additional kinase inhibitors for use in the methods and compositions of the present disclosure are listed in Tables I-IV below.
  • TABLE I
    Exemplary PKCepsilon Inhibitors
    PubChem Parameter
    Description CAS No. CID (nM)
    Staurosporine 62996-74-1 5279 IC50 = 0.08
    Phorbol 12,13-dibutyrate 37558-16-0 37783 Kd = 0.81
    SureCN2934634 N/A 45375865 IC50 = 1.6
    GSK690693 937174-76-0 16725726 Kd = 5.3
    Sotrastaurin 425637-18-9 10296883 IC50 = 6.2
    Indolactam V 90365-57-4 105000 Ki = 7.7
    Enzastaurin 170364-57-5 176167 Kd = 8.9
    CHEMBL350335 N/A 9847089 IC50 = 10
    Balanol analog 4 N/A 5327921 IC50 = 10
    Ro-31-8220 138489-18-6 5083 IC50 = 10
    Ruboxistaurin 169939-94-0 153999 Kd = 11
    A674563 552325-73-2 11314340 Kd = 11
    CHEMBL606245 N/A 44160269 IC50 = 14
    Ro-32-0432 151342-35-7 127757 IC50 = 17.2
    Ophiocordin 63590-19-2 5287736 IC50 = 20
    Cediranib 288383-20-0 9933475 IC50 = 24
    Bisindolylmaleimide I 133052-90-1 2396 IC50 < 25
    K-252a 97161-97-2 3813 IC50 = 33
    SureCN2934911 N/A 45375866 IC50 = 36
    Ro-32-0557 N/A 19095896 IC50 = 48
    PKCb Inhibitor 257879-35-9 6419755 IC50 > 50
    Cdk1/2 Inhibitor III 443798-55-8 5330812 IC50 > 50
    N-Benzoylstaurosporine 120685-11-2 56603681 IC50 = 50
    Balanol analog 5 N/A 5327922 IC50 = 50
    CHEMBL103055 N/A 10207821 IC50 = 75
    AT9283 1092788-83-4 24905142 IC50 > 100
    Bisindolylmaleimide XI, HCl N/A 11605551 IC50 = 108
    CHEMBL295806 N/A 44294447 IC50 = 120
    SureCN5757856 N/A 10302405 IC50 = 140
    SB218078 135897-06-2 447446 IC50 > 150
    PKR Inhibitor 608512-97-6 6490494 IC50 > 150
    Go6983 133053-19-7 3499 IC50 > 150
    Lestaurtinib 111358-88-4 126565 Kd = 160
    Bisindolylmaleimide IV 119139-23-0 2399 IC50 = 190
    Y-27632 146986-50-7 448042 IC50 > 250
    Arcyriaflavin A 118458-54-1 5327723 IC50 = 310
    SureCN3774124 N/A 10209082 IC50 = 330
    Alvocidib 131740-09-5 9910986 Kd = 380
    BMS-690514 859853-30-8 11349170 Kd < 400
    BCP9000906 457081-03-7 5494425 IC50 = 500
    H-1152; Glycyl 913844-45-8 16760635 IC50 = 500
    Ruxolitinib 941678-49-5 25126798 Kd = 530
    SureCN2579964 N/A 24948986 IC50 < 750
    PP121 1092788-83-4 24905142 IC50 < 750
    SureCN2505235 N/A 5353854 IC50 = 1000
    WZ3146 1214265-56-1 44607360 Kd > 1000
    WZ4002 213269-23-8 44607530 Kd > 1000
    Icotinib 610798-31-7 22024915 IC50 > 1000
    MK5108 1010085-13-8 24748204 IC50 > 1000
    JAK3 Inhibitor VI 856436-16-3 16760524 IC50 > 1000
  • TABLE II
    Exemplary MAPK14 p38 Inhibitors
    PubChem Parameter
    Description CAS No. CID (nM)
    Doramapimod 285983-48-4 156422 Kd = 0.046
    SPDD01923 N/A 447721 IC50 = 0.11
    Kinome_3519 N/A 16730109 Ki = 0.2
    SCIO-469 309913-83-5 9871074 Kd = 0.48
    VX745 209410-46-8 3038525 IC50 = 0.8
    CHEMBL420047 N/A 23646852 Kd = 1
    CHEMBL383172 N/A 11235063 Ki = 1.6
    SureCN4219451 N/A 9999342 IC50 = 1.8
    SureCN5632345 N/A 44593646 Ki = 1.9
    PH797804 586379-66-0 22049997 IC50 = 2.5
    CHEMBL215652 N/A 23647319 IC50 = 2.5
    AMG-47a 882663-88-9 16086114 IC50 = 3
    SureCN4762364 N/A 10473563 IC50 = 3.2
    VX702 745833-23-2 10341154 Kd = 3.7
    SureCN3936664 N/A 11626920 IC50 = 5
    Aminopyrimidine amide, 13b N/A 16118737 IC50 = 5
    2ofv N/A 15991573 IC50 = 6
    LY2228820 862507-23-1 11570805 IC50 = 7
    CHEMBL213451 N/A 23647330 IC50 = 7.6
    SureCN5495613 913376-83-7 24764449 IC50 = 9
    SureCN6744546 N/A 24856363 Ki = 9
    SureCN5774497 N/A 9948405 IC50 = 9.6
    SB202190 152121-30-7 5353940 Kd = 9.8
    SB203580 152121-47-6 176155 IC50 = 10
    CHEMBL1964275 N/A 57394915 Kd < 10
    SB220025 165806-53-1 5164 IC50 = 19
    Dasatinib 302962-49-8 11153014 Kd = 27
    p38 MAP Kinase Inhibitor 219138-24-6 4665 IC50 = 35
    Sorafenib 284461-73-0 216239 IC50 = 38
    CHEMBL1092754 N/A 44541014 IC50 = 50
    PD169316 152121-53-4 4712 IC50 > 50
    AST-487 630124-46-8 11409972 Kd = 73
    A-83-01 909910-43-6 16218924 IC50 = 100
    SB681323 444606-18-2 10297982 IC50 < 100
    AT9283 1092788-83-4 24905142 IC50 > 100
    Regorafenib 755037-03-7 11167602 IC50 < 100
    7-hydroxystaurosporine 112953-11-4 72271 IC50 > 100
    SKF-86002 72873-74-6 5228 IC50 = 110
    Nilotinib 641571-10-0 644241 IC50 > 150
    ZM336372 208260-29-1 5730 IC50 = 180
    CHEMBL364623 302962-49-8 11153014 IC50 = 202
    FR180204 865362-74-9 11493598 Ki = 310
    Foretinib 849217-64-7 42642645 Kd = 320
    ML3403 581098-48-8 6419739 IC50 = 380
    SureCN4875304 N/A 46871765 IC50 = 540
    AG1478 175178-82-2 2051 IC50 = 560
    HG-9-91-01 N/A N/A IC50 < 600
    LY364947 396129-53-6 447966 IC50 = 740
    Celocoxib 194044-54-7 2662 IC50 = 810
    BX517 N/A 11161844 IC50 > 900
  • TABLE III
    Exemplary JNK1 Inhibitors
    PubChem
    Description CAS No. CID Parameter (nM)
    Kinome_3027 N/A 11640926 Ki = 1 nM
    JNK Inhibitor VIII 894804-07-0 11624601 Ki = 2 nM
    Kinome_3024 N/A 11539329 Ki = 3 nM
    Kinome_3028 N/A 11590363 Ki = 3 nM
    Kinome_2553 N/A 16007116 Ki = 3.8 nM
    Hesperadin 422513-13-1 10142586 Kd < 10 nM
    Lestaurtinib 111358-88-4 126565 Kd = 11 nM
    R406 841290-81-1 11984591 Kd = 38
    JNKIN7 N/A 57340685 IC50 < 40 nM
    JNKIN8 N/A 57340686 IC50 < 40 nM
    TTT-3002 N/A N/A IC50 < 40 nM
    1;9-Pyrazoloanthrone 129-56-6 8515 IC50 = 40 nM
    7-hydroxystaurosporine 112953-11-4 72271 IC50 > 45 nM
    Momelotinib 1056634-68-4 25062766 IC50 < 100 nM
    Go6976 136194-77-9 3501 IC50 = 100 nM
    AT9283 1092788-83-4 24905142 IC50 > 100 nM
    Staurosporine aglycone 99533-80-9 3035817 IC50 < 100 nM
    PP1 Analog II; 1NM-PP1 221244-14-0 5154691 IC50 = 140 nM
    K-252a 97161-97-2 3813 IC50 > 150 nM
    AS601245 345987-15-7 11422035 IC50 = 150 nM
    CHEMBL1788116 N/A 11422034 IC50 = 150 nM
    NVP-TAE684 761439-42-3 16038120 Kd = 160 nM
    GSK1838705A 1116235-97-2 25182616 Kd = 210 nM
    Staurosporine 62996-74-1 5279 Kd = 220 nM
    TG101348 936091-26-8 16722836 Kd = 260 nM
    Nilotinib 641571-10-0 644241 Kd = 450 nM
    AST-487 630124-46-8 11409972 Kd = 460 nM
    NU6140 444723-13-1 10202471 IC50 = 500 nM
    KW2449 1000669-72-6 11427553 Kd = 580 nM
    CZC-25146 330003-04-7 N/A IC50 < 600 nM
    Nintedanib 928326-83-4 9809715 Kd = 630 nM
    KT5720 108068-98-0 3844 IC50 < 800 nM
    BX517 N/A 11161844 IC50 > 900 nM
    A 443654 552325-16-3 10172943 IC50 > 900 nM
    Pyrrolo-pyrimidone; 17 N/A 16119021 IC50 = 960 nM
    WZ3146 1214265-56-1 44607360 Kd > 1000 nM
    WZ4002 213269-23-8 44607530 Kd > 1000 nM
    MK5108 1010085-13-8 24748204 IC50 > 1000 nM
    Silmitasertib 1009820-21-6 24748573 IC50 > 1000 nM
    SNS314 N/A 16047143 IC50 > 1000 nM
    K00596a 873225-46-8 9549298 IC50 = 100 nM
    GSK-3 Inhibitor IX 667463-62-9 5287844 IC50 = 100 nM
    Baricitinib 1187594-09-7 44205240 IC50 > 1000 nM
    (5Z)-7-Oxozeaenol 66018-38-0 N/A IC50 = 1000 nM
    CP673451 343787-29-1 10158940 IC50 > 1000 nM
    SureCN10063060 N/A 52936621 Ki > 1000 nM
    SB203580 152121-47-6 176155 Kd = 1100 nM
    SureCN5632345 N/A 44593646 Kd < 1250 nM
    SureCN7018367 N/A 18792927 Kd < 1250 nM
    IKK-2 Inhibitor IV 507475-17-4 9903786 Kd < 1250 nM
  • TABLE IV
    Exemplary JNK3 Inhibitors
    PubChem
    Description CAS No. CID Parameter (nM)
    Hesperadin 422513-13-1 10142586 Kd < 10 nM
    SureCN7018367 312917-37-6 18792927 Kd < 10 nM
    Lestaurtinib 111358-88-4 126565 Kd = 12 nM
    Kinome_3027 N/A 11640926 Ki = 18 nM
    1;9-Pyrazoloanthrone 129-56-6 8515 Kd = 22 nM
    SB203580 152121-47-6 176155 Kd = 35 nM
    JNKIN7 N/A 57340685 IC50 < 40 nM
    JNKIN8 N/A 57340686 IC50 < 40 nM
    SB202190 152121-30-7 5353940 Kd = 42 nM
    KW2449 1000669-72-6 11427553 Kd = 51 nM
    JNK Inhibitor VIII 894804-07-0 11624601 Ki = 52 nM
    TTT-3002 N/A N/A IC50 < 60 nM
    Doramapimod 285983-48-4 156422 Kd = 62 nM
    NVP-TAE684 761439-42-3 16038120 Kd = 67 nM
    AS601245 345987-15-7 11422035 IC50 = 70 nM
    CHEMBL1788116 N/A 11422034 IC50 = 70 nM
    AT9283 1092788-83-4 24905142 IC50 > 100 nM
    Staurosporine aglycone 99533-80-9 3035817 IC50 < 100 nM
    7-hydroxystaurosporine 112953-11-4 72271 IC50 > 100 nM
    MK5108 1010085-13-8 24748204 IC50 > 100 nM
    Staurosporine 62996-74-1 5279 Kd = 110 nM
    Kinome_2553 N/A 16007116 IC50 = 120 nM
    GSK1838705A 1116235-97-2 25182616 Kd = 130 nM
    AMG-47a 882663-88-9 16086114 IC50 = 145 nM
    JNK Inhibitor IX 312917-14-9 766949 IC50 = 200 nM
    TG101348 936091-26-8 16722836 Kd = 200 nM
    JNJ-28312141 885692-52-4 N/A Kd = 240 nM
    Nintedanib 928326-83-4 9809715 Kd = 270 nM
    Go6976 136194-77-9 3501 IC50 > 300 nM
    AC1NS4N8 N/A 23649240 Kd < 400 nM
    Syk Inhibitor IV 732983-37-8 10200390 IC50 < 400 nM
    PDK1/Akt/Flt Inhibitor 331253-86-2 5113385 IC50 = 500 nM
    CZC-25146 330003-04-7 N/A IC50 < 600 nM
    R406 841290-81-1 11984591 IC50 < 600 nM
    GSK650394A 890842-28-1 25022668 IC50 > 600 nM
    Momelotinib 1056634-68-4 25062766 IC50 < 750 nM
    AST-487 630124-46-8 11409972 Kd = 760 nM
    VX702 745833-23-2 10341154 Kd = 780 nM
    HG-9-91-01 N/A N/A IC50 < 800 nM
    Ruboxistaurin 169939-94-0 153999 IC50 > 900 nM
    GSK-3 Inhibitor IX 667463-62-9 5287844 IC50 > 900 nM
    A 443654 552325-16-3 10172943 IC50 > 900 nM
    HG-10-102-01 1351758-81-0 N/A IC50 = 1000 nM
    WZ3146 1214265-56-1 44607360 Kd > 1000 nM
    WZ4002 213269-23-8 44607530 Kd > 1000 nM
    Syk Inhibitor 622387-85-3 6419747 IC50 = 1000 nM
    SGI-1776 N/A N/A IC50 > 1000 nM
    Silmitasertib 1009820-21-6 24748573 IC50 > 1000 nM
    SNS314 N/A 16047143 IC50 > 1000 nM
    K00596a 873225-46-8 9549298 IC50 = 1000 nM
  • A therapeutically effective amount of a small molecule inhibitor (SMI), such as a protein kinase inhibitor, may vary depending upon the route of administration and dosage form. Effective amounts of SMIs typically fall in the range of about 0.0001 to 1000 mg/kg/day, typically 0.001 to 100 mg/kg/day, and more typically in the range of about 0.01 up to 10 mg/kg/day. In some embodiments, the SMI is administered to a subject in an amount less than any of the following daily doses (mg/kg): 100, 50, 10, 5.0, 1.0, 0.5 or 1. In some embodiments, the SMI is administered to a subject in an amount greater than any of the following daily doses (mg/kg): 0.001, 0.005, 0.01, 0.05, 0.1 or 0.5. That is, the daily dose (mg/kg) can be any of a range of having an upper limit of 100, 50, 10, 5.0, 1.0, 0.5 or 1, and an independently selected lower limit of 0.001, 0.005, 0.01, 0.05, 0.1 or 0.5, wherein the lower limit is less than the upper limit. Thus, the actual amount per day for an adult mammal weighing 70 kg is typically between 0.07 and 7,000 mg, or more typically between 0.7 and 700 mg, where this amount can be administered as a single dose per day or as a series of part-doses (such as, for example, two, three, four, five or six) per day, so that the total daily dose is the same.
  • Typically, the SMI is selected to provide a formulation that exhibits a high therapeutic index. The therapeutic index is the dose ratio between toxic and therapeutic effects, which can be expressed as the ratio between LD50 and ED50. The LD50 is the dose lethal to 50% of the population and the ED50 is the dose therapeutically effective in 50% of the population. The LD50 and ED50 are determined by standard pharmaceutical procedures in animal cell cultures or experimental animals.
  • In some embodiments a SMI, such as a protein kinase inhibitor, is in the form of a pharmaceutically acceptable salt. For instance, when the SMI has a basic group, such as, for example, an amino group, pharmaceutically acceptable salts can be formed with inorganic acids (such as hydrochloric acid, hydroboric acid, nitric acid, sulfuric acid, and phosphoric acid), organic acids (e.g., formic acid, acetic acid, fumaric acid, oxalic acid, tartaric acid, lactic acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, and p-toluenesulfonic acid) or acidic amino acids (such as aspartic acid and glutamic acid). Alternatively, when the SMI has an acidic group, such as for example, a carboxylic acid group, it can form salts with metals, such as alkali and earth alkali metals (e.g., sodium, lithium, potassium, calcium, magnesium, zinc), ammonia, organic amines (e.g., trimethylamine, triethylamine, pyridine, picoline, ethanolamine, diethanolamine, triethanolamine), or basic amino acids (e.g., arginine, lysine and ornithine).
  • In a preferred embodiment, a SMI, such as a protein kinase inhibitor, is contained within a pharmaceutical composition. A preferred pharmaceutical composition is a single dosage form for oral administration, such as a tablet or capsule. A pharmaceutical composition in the form of a tablet suitable for oral administration can comprise one or more pharmaceutically acceptable carriers and/or excipients suitable (e.g., inactive ingredients). Exemplary carriers include but are not limited to lactose, cellulose (for example microcrystalline cellulose), and mannitol. Exemplary excipients include but are not limited to binding agents such as hydroxypropylmethylcellulose or povidone (polyvinylpyrollidone), lubricants such as magnesium stearate, and disintegrants such as sodium starch glycollate, croscarmellose sodium, or crospovidone (cross-linked polyvinylpyrollidone).
  • In some embodiments, kinase inhibitor formulations are comprised of at least two SMIs having high bioavailability (>20%) are administered to a population of parasite-infected patients. In the case of malaria, particularly preferred SMIs have an EC50<10 nM in vitro and a single-digit mg/kg activity ED90 in a human erythrocyte-engrafted SCID mouse model. The kinase inhibitor formulations are either single-drug formulations comprising an SMI as the only active ingredient, or as a multi-drug formulation comprising a first SMI and a second SMI. The patient population is comprised of both children (over six months of age) and adults with confirmed and uncomplicated Plasmodium infection who are able to swallow oral medication. The patient pool does not exclude G6PD-deficient patients.
  • Patients receive the orally-available kinase formulations at an effective dose over the course of the study, during which the adequate clinical and parasitological response (ACPR) and the clinical parasite reduction ratio (PRR) are monitored. The study may range from 28 up to or over 48 days, with patients exhibiting a clinical efficacy at day 28 from a single dose (or a few doses) of the kinase formulation, measured as a PRR of over 50%.
  • The formulations described herein are also suitable for administration as prophylactic treatments in uninfected individuals traveling to a parasitic disease endemic area (e.g., malaria-endemic area).
  • Modified Kinase Inhibitors
  • Cell signaling connects immunity and basic intermediary metabolism at the mitochondrial level (Arnoult et al. EMBO REP 12:901-10, 2011). The links between mitochondria and host defense include modulation of bioenergetics by several protein kinases (PKs) (e.g., PKC, ERK, JNK, and p38 MAPK) (Horbinski and Chu, Free Radic Biol Med, 38:2-11, 2005) following PK activation/translocation to mitochondria in response to immune stimuli. As determined during development of the present disclosure, inhibition of mosquito p38 MAPK significantly reduces malaria parasite development. Without being bound by theory, the inhibition of parasite development is thought to be in response to p38 MAPK-regulated changes in mosquito midgut mitochondrial bioenergetics.
  • Some of the phosphorylation-dephosphorylation events that cells undergo when challenged with immune stimuli are not necessarily related to mitochondria-driven apoptosis, but to bioenergetics. These latter effects have been ascribed to the translocation and/or activation of critical PK within mitochondria. Thus, it would be advantageous to modify SMIs so as to redirect their localization to the mitochondrial compartment. The modified SMIs may result in less drug resistance, catabolism, and off-target effects relative to unmodified SMIs that localize to the cytosolic space (Mourtada et al., PLoS One, 8:e60253, 2013).
  • Suitable modifications to direct delivery of a SMI to a mitochondria include attachment of the SMI to a mitochondrial-targeting moiety, including but are not limited to lipophilic cations, mitochondrial-penetrating peptides, mitochondrial-targeting dyes, liposomes, and nanoparticles (see, e.g., Rin Jean et al., ACS Chem Biol, 9:323-333, 2014). Exemplary lipophilic cations include but are not limited to triphenylphosphonium (TPP)(Ross et al., Biochemistry, 70:222-230, 2005), AAI (Sun et al., Cancer Res, 54:1465-1471, 1994), and MKT-077 (Weisberg et al., Cancer Res, 56:551-555, 1996). Exemplary mitochondrial-penetrating peptides include but are not limited to SS-31 (Zhao et al., J Biol Chem, 279: 34682-34690, 2004), MPP (Horton et al., Chem Biol 15: 375-382, 2008), and those including 8-15 hydroxyl-containing (Serine, Threonine), positively charged amino acids (Arginine, Lysine) or hydrophobic natural and synthetic amino acids (phenylalanine, cyclohexylalanine)(Horton et al., Chem Biol, 15:375-82, 2008; and Yousif et al., Chembiochem, 10:1939-50, 2009).. Exemplary mitochondrial-targeting dyes include but are not limited to rhodamine and the dyes listed in Table V.
  • TABLE V
    Mitochondrial Targeting Dyes
    Dye Description
    4-Di-1-ASP (DASPMI) 4-(4-(Dimethylamino)-styryl)-N-methylpyridinium iodide
    DASPEI 2-(4-(dimethylamino)styryl)-N-ethylpyridinium iodide
    Dihydrorhodamine 123 Reduced form of Rhodamine 123
    Dihydrorhodamine 123, Reduced form of Rhodamine 123
    dihydrochloride
    JC-1 chloride Membrane potential-dependent mitochondrial stain
    JC-1 iodide Membrane potential-dependent mitochondrial stain
    MitoGreen Green-fluorescent mitochondrial dye
    MitoRed Red-fluorescent mitochondrial dye
    Nonyl Acridine Orange (NAO) Membrane potential-independent mitochondrial stain
    Rhodamine 123 Green-fluorescent mitochondria stain
    Tetrabromorhodamine 123, Green-fluorescent mitochondria stain
    bromide
    TMRE Tetramethylrhodamine ethyl ester, perchlorate; membrane
    potential-sensitive dye
    TMRM Tetramethylrhodamine methyl ester, perchlorate; membrane
    potential-sensitive dye
  • Another strategy for modification of SMIs for use in the methods and compositions of the present disclosure involve the use of bifunctional inhibitors. These inhibitors tend to be more effective than those solely directed towards either the ATP binding site (Hu et al., Drug Discovery Today, 1:438-447, 1996) or the substrate-binding site (Lee et al., J Am Chem Soc, 126:3394-3395, 2004) because they are directed to both the ATP and substrate binding sites (Ricouart et al., J Med Chem, 34:73-78, 1991). The use of bisubstrate analog inhibitors has been described and applied to several kinases (Parang et al., Nature Structural Biology, 8:37-41, 2001; Parang and Cole, Pharmacology & Therapeutics, 93:145-157, 2002; Hines et al., Bioorganicchemistry, 33:285-297, 2005; Broom, J Med Chem, 32:2-7, 1989; and Lee et al., Chembiochem, 9:507-509, 2008), including PKCs (van Wandelen et al., ACS Chemical Biology, 8:1479-1487, 2013; van Ameijde et al., Organic & Biomolecular Chemistry, 8:1629-1639, 2010; and Poot et al., Chembiochem, 10:2042-2051, 2009), but never targeted specifically to mitochondria. A recent review cited lack of PKC isozyme selectivity and problematic delivery as reasons why, even after over 20 years of development, the full therapeutic potential of PKC has not yet been realized (Villalba and Altman, Current Cancer Drug Targets, 2:125-137, 2002). In some embodiments, bifunctional protein kinase inhibitors are targeted to mitochondria.
  • Combination Therapies
  • The present disclosure further provides methods for treating or preventing malaria comprising administering at least one kinase inhibitor and an additional antimalarial (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing malaria. In some embodiments, the kinase inhibitor and the antimalarial are present in a single formulation, while in other embodiments the kinase inhibitor and the antimalarial are present in separate formulations. In some embodiments, the additional antimalarial comprises one or more compounds of the antimalarial classes selected from the group consisting of amino alcohols, aminoquinolines, antibiotics, antifolates, endoperoxides, sulfonamides, and others (see, e.g., Delves et al., PLoS Medicine, 9(2) e1001169, 2012).
  • In particular, the additional antimalarial compound(s) may be aminoquinolines (including, but not limited to, amodiaquine, naphthoquine, AQ-13, tert-butyl isoquine, hydroxycholoquine, pyronaridine, diethylprimaquine, bulaquine, primaquine, tafenoquine, piperaquine, NPC-1161B, and chloroquine), antibiotics (including, but not limited to, azithromycin, trimethoprim, trimethoprim-sulphamethoxazole, tetracycline, mirincamycin, doxycycline, thiostrepton, fosmidomycin, and clindamycin), endoperoxides (including, but not limited to, artemeter, artesunate, OZ439, OZ277, artemisinin, artemisone, and dihydroartemisinin), antifolates (such as pyrimethamine, proguanil, dapsone, cycloguanil, P218, and chlorproguanil), sulfonamides (such as sulfadoxine, sulfadiazine, and sulfmethoxazole), amino alcohols (such as lumefantrine, halofantrine, mefloquine, and quinine), or other antimalarial drugs (including, but not limited to, DSM1, DSM265, P218, BCX4945, synthetic peroxides, methylene blue, riboflavin, pentamidine, DHEA, cycloheximide, atovaquone, deferoxamine, and N-acetyl-D-penicillamine). In some embodiments, the combination therapy includes a fixed-dose artemisinin combination therapy. In some aspects, the artemisinin combination therapy is comprised of one or more kinase inhibitor as well as one or more of artemether, artesunate, dihydroartemisinin, artemisone, and artemisinin. In some embodiments, the additional antimalarial counteracts the malarial parasite at the same life stage(s) as the kinase inhibitor. In other embodiments, the additional antimalarial counteracts the malarial parasite at a different life stage from the kinase inhibitor.
  • The present disclosure also provides methods for treating or preventing other parasitic diseases comprising administering at least one kinase inhibitor and an additional antiparasitic agent (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing the parasitic disease. In some embodiments, the kinase inhibitor and the antiparasitic agent are present in a single formulation, while in other embodiments the kinase inhibitor and the antiparasitic agent are present in separate formulations. In some embodiments, the parasitic disease is not malaria.
  • In some preferred embodiments, present disclosure provides methods for treating or preventing Chagas disease comprising administering at least one kinase inhibitor and an additional antiparasitic agent (e.g., a compound that is not a kinase inhibitor) to a subject in need thereof under conditions suitable for treating or preventing the parasitic disease. In some embodiments, the kinase inhibitor and the antiparasitic agent are present in a single formulation, while in other embodiments the kinase inhibitor and the antiparasitic agent are present in separate formulations. In some embodiments, the additional antiparasitic agent comprises an azole- or nitro-derivative, such as benznidazole, or nifurtimox.
  • Treatment of Infections by Other Protozoan Parasites
  • In other embodiments, the use of SMIs directed to mammalian kinase pathways is applied to diseases caused by other intracellular parasitic protozoa. A large number of protozoal pathogens spend a portion of their life cycle in a mammalian host, (e.g., human). Diseases caused by infection with other intracellular parasitic protozoa are also treated or prevented by mammalian kinase inhibitors. In particular, the present disclosure provides kinase inhibitors and methods for use thereof in mammalian subjects infected with a protozoan parasite selected from the group consisting of Giardia lamblia, Cryptosporidium parvum, Cyclospora cayetanensis, Entamoeba histolytica, Trichomonas tenax, Trichomonas hominis, Trichomonas vaginalis, Trypanosoma brucei gambiense, Trypanosoma brucei rhodesiense, Trypanosoma cruzi, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Toxoplasma gondii, Theileria lawrenci, and Theileria parva. In preferred embodiments, the protozoan parasite is vectored by an arthropod (e.g., sand fly-transmitted Leishmania, tick-borne Babesia microti, kissing bug-transmitted Trypanosoma cruzi, tsetse-transmitted Trypanosoma brucei), and the SMI is directed to mammalian and arthropod kinase pathways. In some embodiments, the disease is Chagas disease, caused by infection with Trypanosoma cruzi. In general, the present disclosure provides methods for use of kinase inhibitors in hosts infected with a protozoan parasite.
  • General Techniques
  • The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989); Oligonucleotide Synthesis (Gait, ed., 1984); Animal Cell Culture (Freshney, ed., 1987); Handbook of Experimental Immunology (Weir & Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (Miller & Calos, eds., 1987); Current Protocols in Molecular Biology (Ausubel et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (Coligan et al., eds., 1991); The Immunoassay Handbook (Wild ed., Stockton Press NY, 1994); Bioconjugate Techniques (Hermanson, ed., Academic Press, 1996); and Methods of Immunological Analysis (Masseyeff, Albert, and Staines, eds., Weinheim: VCH Verlags gesellschaft mbH, 1993).
  • DEFINITIONS
  • As used herein, the singular form “a”, “an”, and “the” includes plural references unless indicated otherwise. For example, “an” excipient includes one or more excipients.
  • The phrase “comprising” as used herein is open-ended, indicating that such embodiments may include additional elements. In contrast, the phrase “consisting of” is closed, indicating that such embodiments do not include additional elements (except for trace impurities). The phrase “consisting essentially of” is partially closed, indicating that such embodiments may further comprise elements that do not materially change the basic characteristics of such embodiments. It is understood that aspects and embodiments described herein as “comprising” include “consisting” and/or “consisting essentially of” aspects and embodiments.
  • The terms “small molecule inhibitor” and “SMI” refer to an organic compound (e.g., JNK inhibitor SP600125) of low molecular weight (e.g., less than 900 Daltons, preferably less than 500 Daltons, unless bound to a mitochondrial-targeting moiety in which case the size could be >2 kDa but less than 5 kDa) that inhibits a biological process. Biopolymers such as nucleic acids, proteins, and polysaccharides of greater than 10 kDa are not small molecules, although the nucleotide, amino acid, and saccharide constituents are small molecules.
  • The term “mitochondrial-targeting moiety” refers to a functional group of a compound that directs the compound to the mitochondria or to a specific compartment therein. Examples of mitochondrial-targeting moieties include but are not limited to lipophilic cations, mitochondrial-penetrating peptides, mitochondrial-targeting dyes, liposomes and nanoparticles.
  • Administration a protein kinase inhibitor in combination with an additional protein kinase inhibitor or an additional anti-parasite drug includes simultaneous (concurrent) and consecutive administration in any order.
  • An “effective amount of” or “under conditions effective for” refers to administration of a protein kinase inhibitor in an amount sufficient to carry out a specifically stated purpose. An “effective amount” may be determined empirically and in a routine manner, in relation to the stated purpose.
  • The term “therapeutically effective amount” refers to an amount of an agent (e.g., protein kinase inhibitor) effective to “treat” a disease or disorder in a subject (e.g., a mammal such as a human). In the case of Chagas disease, the therapeutically effective amount of the agent reduces a sign or symptom of Chagas disease.
  • The terms “treating” or “treatment” of a disease refer to executing a protocol, which may include administering one or more drugs to an individual (human or otherwise), in an effort to alleviate signs or symptoms of the disease. Thus, “treating” or “treatment” does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols that have a measurable effect on the individual.
  • As used herein, and as well-understood in the art, “treatment” is an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • The terms “reduce” and “reduction” as used in reference to biological function (e.g., kinase activity, parasite development, etc.) refer to a measurable decrease in the function by preferably at least 50%, more preferably at least 75% and most preferably at least 90%. Depending upon the function, the reduction may be from 10-fold to 1,000,000-fold, or from 10, 100 or 1000-fold to 10,000, 100,000 or 1,000,000-fold.
  • EXAMPLES
  • Abbreviations: ACPR (adequate clinical and parasitological response); AMAS (amastigote); HTS (high throughput screen); JNK (c-Jun N-terminal kinase); MAPK (mitogen-activated protein kinase); PI3K (phosphoinositide 3-kinase); PKB (protein kinase B); PKC (protein kinase C); SMI (small molecule inhibitor); TRYP (trypomastigote).
  • Example 1 Screening Compounds for Effects on Cell Viability, Proliferation and Kinase Activity in Mosquito Cells In Vitro and In Vivo
  • Testing with immortalized Anopheles cells. Multiple cell lines created from minced mosquito embryos or larvae are available from Anopheles stephensi (ASE, MSQ43) and from Anopheles gambiae (4a3B, SUA). These cells are not derived from specific tissues, but rather were selected because they grew continuously under culture conditions after isolation. In general, these cells are phagocyte-like and immune-responsive, but cellular physiology is more muscle cell-like (Giulivi et al., Biochem J, 415:309-16, 2008). Small molecule inhibitors (SMIs) are analyzed using these cells first to establish concentration ranges that inhibit target protein kinases without toxic effects on mosquito cells.
  • Cell death assay. SMIs are diluted over a log-range for toxicity analyses. Assays against mosquito cells are performed in 96 well plates using CytoTox 96® Non-Radioactive Cytotoxicity Assay (Promega).
  • Cell doubling time assay. SMIs are diluted over a log-range to assay effects on mosquito cell doubling time (about 22 h). Assays are performed in 96 well plates using CellTiter 96® Aqueous Non-Radioactive Cell Proliferation Assay (Promega).
  • In vitro kinase inhibition assays. HTSs (high-throughput screens) of candidate SMIs are performed against Anopheles sp. recombinant kinases (e.g., PKCs, JNK, and p38 MAPK). IMAP immobilized metal ion-affinity fluorescence polarization (Molecular Devices) is used to screen 10,000 kinase-focused collection compounds (Cancer Research Technology) at 30 μM. An exemplary HTS uses 10 pM of the kinase, 100 nM 5-FAM (fluorescein-amidite)-labeled peptide, and 30 μM ATP in a buffer consisting of 20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM DTT and 0.25 mM EGTA as described (Kjaer et al., Biochem J, 451:329-342, 2013). SMIs with low IC50 (<2 μM) are further tested using the Millipore KinaseProfiler™ service (Millipore) or Kinomescan™ (Discoverx) to determine the selectivity of key compounds against a panel of kinases (human and predicted mosquito). A preferred SMI targets overlapping but diverse kinase-mediated pathways.
  • Cellular kinase inhibition assays. Conserved PKCs (Pakpour et al., PLoS One, PLoS ONE 8(10): e76535, 2013) and MAPKs control malaria parasite development in A. stephensi (ERK: Surachetpong et al. 2009 PLoS Pathog. 5(4):e1000366; p38 MAPK and JNK, unpublished data). The present disclosure is based on the understanding that SMIs against conserved kinases ingested in a blood meal containing malaria parasites from a treated human host will alter mosquito signaling to kill ingested parasites. SMI dilutions that are not toxic are assayed against mosquito cells in 96 well plates. Synthetic peptide targets (MARCKS peptide MO2-58 for cPKC, PKC-delta, PKC-epsilon; sc-3108 peptide for PKC-zeta; sc-3106 for PKC-mu or PKD) are used to distinguish effects of SMIs on PKC activity in Anopheles cells. Synthetic peptide targets (ERKtide, p38 substrate P03-58 or MK-2, and JNK substrate c-Jun) are used to distinguish effects of SMIs on MAPK activity in Anopheles cells. Briefly, cells are treated with SMIs, lysed and prepared for dot blot analysis of kinase activity analyses through detection of phosphorylation of appropriate substrates. GENBANK Accession Nos. of representative Anopheles gambiae kinase sequences are provided in Table 1-1.
  • TABLE 1-1
    Mosquito Kinase Sequences
    Kinase Type Accession No.
    cPKC conventional PKC EAA0266.4
    nPKC-delta novel PKC EDO64332.2
    nPKC-epsilon novel PKC EAA07888.5
    aPKC-zeta atypical PKC EAA00497.3
    EAA00702.3
    PKD protein kinase D EAA06222.6
    PKN PKC-like EAA03911.6
    JNKa c-Jun N terminal kinase EAA03630.4
    GI: 157020897
    JNKb c-Jun N terminal kinase EAA05905.3
    GI: 116131141
    p38 MAPK mitogen-activated protein kinase EAA00194.4
    GI: 157013175
  • Example 2 Screening Compounds for Effects on Plasmodium Growth, Infectivity and Development in Erythrocytes, Mice and Mosquito Cells
  • Plasmodium growth and mammalian infection assays. In order to attribute SMI activity to effects on mosquito signaling proteins specifically, SMIs are routinely screened for growth effects on P. falciparum parasites maintained in human erythrocyte culture in vitro. Because parasites are ingested by mosquitoes in the formed of infected erythrocytes, testing SMIs against parasite-infected erythrocytes in vitro provides insight as to whether an SMI of interest affects only the mosquito host or affects both the mosquito host and malaria parasite biology. While SMI effects on the mosquito can all be beneficial in terms of transmission reduction, beneficial effects of SMIs for the malaria patient would necessarily depend on the demonstration of an effect of the SMI on parasite growth in erythrocytes.
  • P. falciparum growth assay. Aliquots of ring stage P. falciparum NF54 culture are synchronized using sorbitol 48 h prior to the assay and then plated in 96-well flat-bottom plates in complete RPMI 1640 with HEPES, hypoxanthine, and 10% heat-inactivated human serum. Parasites are treated with an SMI of interest or with an equivalent volume of dimethyl sulfoxide (DMSO) diluent for 50 h in a candle jar in a 37° C. incubator. The assays are terminated by replacing the culture medium with RPMI 1640 with 10% formaldehyde in PBS. Erythrocytes are stained with 10 μg of propidium iodide/ml in PBS for 1-2 h at room temperature. Infected RBCs are counted with a flow cytometer. The levels of parasite growth in response to treatment are normalized to DMSO controls, which are set to 100%. (see e.g., Pakpour et al., Infect Immun, 80:2141-9, 2012; and Drexler et al., J Exp Biol, 216:208-17, 2013). For SMIs or combinations of SMIs with an effect on parasite growth, analyses of target erythrocyte kinase(s) are completed (see, e.g., Millholland et al., Cell Host Microbe, 13:15-28, 2013).
  • Plasmodium yoelii yoelii 17XNL infection assay in mice. An established murine malaria model is used to assess effects of SMIs on P. y. yoelii in vivo (Luckhart et al., PLoS Pathog, 9(2):e1003180, 2013; and Chau et al., Infect Immun, 2013 Epub ahead of print). Parasitemias predictably follow a temporal rise that can reach high levels (10-15% infected of total erythrocytes) with significant detectable pathology to a variety of organs. With GFP-tagged P. y. yoelii 17XNL, parasite densities in mouse blood are analyzed using a fluorescence plate reader (96 well plate). Although GFP emission of parasites through live tissue and skin is low, parasite loads can be quantified as described in sampled blood from live mice and in the mouse body following infection with P. y. yoelii 17XNL labeled with GFP-luciferase (obtained from Dr. S. Kappe and described in Miller et al., PLoS ONE, 8(4): e60820, 2013). The bioluminescent signal in live mice infected with P. y. yoelii-GFP-luciferase blood stage parasites is intense in the spleen and the lungs, perhaps due to parasite sequestration in these tissues. Quantification of blood parasitemia using a fluorescence plate reader and of parasite burden in live mice using whole body imaging facilitates high throughput analyses of parasite infection over time using various doses of SMIs.
  • High throughput studies are initiated in the mouse model with IP injection of known SMI doses. To assess treatment, SMI formulations are administered daily by IP injection for 1-4 days to Plasmodium-infected mice. To assess prophylaxis, SMI formulations are administered daily by IP injection for 3-4 days to mice prior to Plasmodium infection. The effects of SMIs on parasitemia and disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established. The same types of studies of are performed with orally delivered SMIs or control compounds to establish efficacy against infection and disease, as well as to determine pharmacokinetics in blood by high-performance chromatography and/or mass spectrometry of parent compounds.
  • Plasmodium growth and development in A. stephensi. A strain of P. falciparum that expresses GFP only in sexual stage parasites is used to monitor gametocyte ingestion and development within the first 24-32 hours following feeding by A. stephensi (Luckhart et al., PLoS Pathog, 9(2):e1003180, 2013). In a similar way, development of oocysts (3-12 days after infection) and sporozoites (12-16 days after infection) of GFP-tagged P. y. yoelii 17XNL is monitored in A. stephensi after feeding on infected mice. The presence of sporozoites is key to understanding risk of transmission, so detection of this stage is critical to monitor transmission-blocking efficacy of SMIs. Hence, high-throughput analyses can be performed to detect and quantify infection of A. stephensi with both P. falciparum and P. y. yoelii 17XNL. The ability to detect (i) P. falciparum infection following ingestion of SMI-treated parasite culture and (ii) following feeding on SMI-treated, parasite-infected mice provides key information on SMI transmission-blocking efficacy using the best models for human and mouse parasites currently available. Following high-throughput screening for SMIs that reduce transmission, quantitative analyses of gametocytogenesis in SMI-treated P. falciparum culture and in SMI-treated, infected mice is used to determine if transmission-blocking effects of SMIs are due to effects on sexual stage parasite development, to SMI effects on the mosquito host signaling kinases, or both. To understand SMI effects on the mosquito host, SMI action against conserved kinase targets in mosquito tissues is examined (see, e.g., Pakpour et al., Infect Immun, 80:2141-2149, 2012; and Surachetpong et al., PLoS Pathog. 5(4):e1000366, 2009).
  • Screening kinase inhibitor activity in A. stephensi. Addition of 10 μM SP600125 to a bloodmeal with P. falciparum notably decreased phosphorylation of JNK1/3 in the midgut of A. stephensi 3 hr after feeding relative to an identical untreated bloodmeal (FIG. 5). By comparison, provision of 1 μM SP600125 via bloodmeal resulted in a 15% reduction in midgut pJNK1/3 levels relative to controls, suggesting that relatively low levels of a JNK SMI are biologically active in A. stephensi.
  • When 1 μM or 10 μM SP600125 was provided to either A. freeborni or A. stephensi daily in water or once weekly via bloodmeal (with oviposition), no negative impact on lifespan was recorded for either mosquito species. In A. stephensi provided with a single dose of 10 μM SP600125 in a P. falciparum-infected bloodmeal, parasite infection prevalence in each of two cohorts (n=50), as well as infection intensity (mean oocysts in mosquitoes with at least one oocyst) was reduced (FIG. 6). The JNK inhibition of P. falciparum infection in A. stephensi was associated with enhanced expression of an array of immune genes in the mosquito midgut (FIG. 7).
  • Example 3 Administration of Kinase Inhibitor(s) to Human Patients to Treat Malaria
  • This example provides a description of a clinical study among malaria-infected individuals. The clinical and parasitological response (CPR) in test subjects receiving treatment with candidate kinase inhibitor(s) (test formulation) is compared to the CPR of control subjects receiving treatment with a control formulation (placebo or comparator).
  • Primary Objective: to compare the proportions of test and control subjects that are free of parasites in the blood at Day 28 post-treatment.
  • Secondary Objective(s): to compare the proportions of test and control subjects free of parasites in the blood at Day 7 post-treatment; to compare the time of clearance of fever in test and control subjects; and to compare the time to clearance of parasites in the blood in test and control subjects.
  • Tertiary Objective(s); compare the responses to treatment of test and control subjects according to the World Health Organization System (see WHO 2009 Methods for Surveillance of Antimalarial Drug Efficacy), described below.
  • Normal, Infected Subjects. This study is a subject- and observer-blinded, randomized, controlled study of malaria-infected, but otherwise healthy, adult and children subjects (ages 6 months to 55 years) to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • G6PD Deficient Subjects. This study is a subject- and observer-blinded, randomized, controlled study of G6PD-deficient, malaria-infected adult and children subjects (ages 6 months to 55 years) to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • Normal and G6PD-deficient subjects are divided into groups and receive either a once-daily or twice-daily dose, given on study Day 0. Subjects receiving either the once-daily or twice-daily dosing are further divided into groups receiving dosing on Day 0, Days 0-1, Days 0-2, Days 0-3, Days 0-4, Days 0-5, Days 0-6, or Days 0-7. All subjects are asked to return at Days 1, 2, 3, 7, 14, 21, 28 and 48 for clinical evaluation and parasitemia analysis.
  • Study Population. Subjects are selected from among malaria-infected male and female volunteers. Inclusion and exclusion criteria met by study participants are as follows. Inclusion Criteria: infection with Plasmodium (including P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi), ability to swallow oral medication, willingness to comply with the study protocol and ability to adhere to strict dosing requirements. Exclusion Criteria: clinically debilitating illness; signs or symptoms of severe or complicated malaria; and treatment with antimalarials within the previous 7 days.
  • WHO Classification of Responses to Treatment. For the purpose of this analysis, possible responses to treatment include early treatment failure, late clinical failure, late parasitological failure, and adequate clinical and parasitological response (ACPR) as described (see WHO 2009 Methods for Surveillance of Antimalarial Drug Efficacy). ACPR is defined as absence of parasitemia at Day 28, in patients who did not previously meet any of the criteria of early treatment failure, late clinical failure or late parasitological failure. Early treatment failure (ETF) includes patients exhibiting severe malaria between Days 1-3 in the presence of parasitemia, parasitemia on Day 2 higher than Day 0, parasitemia on Day 3 with a temperature of greater than or equal to 37.5° C., and parasitemia on Day 3 less than or equal to 25% of that on Day 0. Late clinical failure (LCF) includes patients not exhibiting ETF who have danger signs or severe malaria in the presence of parasitemia on any day between Day 4 and Day 28, and presence of parasitemia on any day between Day 4 and Day 28 with a temperature of greater than or equal to 37.5° C. Late parasitological failure (LPF) includes patients not exhibiting ETF or LCF who have parasitemia on any day between Day 7 and Day 28 in the absence of fever (less than 37.5° C.).
  • Preferred test formulations have a clinical efficacy (ACPR) on Day 7 of at least 90%, more preferably at least 95% and most preferably at least 99%. In some aspects, preferred test formulations have clinical efficacy (ACPR) of at least 50% on Day 28, more preferably at least 75% and most preferably at least 95%. Additional efficacy targets are described in Burrows et al., Malaria J, 12:187, 2013.
  • Statistical Analyses. All statistical tests comparing demographic, patient characteristic and safety data are conducted using the Kaplan-Meier method. ACPR is measured by blood smear or polymerase chain reaction.
  • Example 4 Administration of Kinase Inhibitor(s) to Human Subjects to Prevent Malaria
  • This example provides a description of a clinical study among healthy individuals traveling in malaria-endemic areas, which compares protection from malaria resulting from prophylactic treatment with candidate kinase inhibitor(s) (test formulation) to protection from malaria in control subjects receiving treatment with a control formulation (placebo or comparator).
  • Primary Objective: to compare the proportions of test and control subjects free of parasites in the blood at Day 28 post-treatment.
  • Secondary Objective(s): to compare the proportions of test and control subjects who have parasitemia in the absence of fever (less than 37.5° C.) at Day 28 post-treatment.
  • Normal, Non-infected Subjects. This study is a subject- and observer-blinded, randomized, controlled study of healthy adult and children subjects (ages 6 months to 55 years) at risk of contracting malaria by traveling to an endemic area to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • G6PD Deficient Subjects. This study is a subject- and observer-blinded, randomized, controlled study of G6PD-deficient, but otherwise healthy, adult and children subjects (ages 6 months to 55 years) at risk of contracting malaria by traveling to an endemic area to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age prior to randomization.
  • Subjects are divided into groups and receive either a once-daily or twice-daily dose, given on study Day 0 (1 or 2 days before entering the endemic area). All subjects are asked to obtain a parasitemia analysis and clinical evaluation at Day 28 post-treatment.
  • Study Population. Subjects are selected from among healthy male and female volunteers. Inclusion and exclusion criteria met by study participants are as follows. Inclusion Criteria: at risk of contracting malaria by traveling to a malaria endemic area, ability to swallow oral medication, willingness to comply with the study protocol and ability to adhere to strict dosing requirements, age over six months. Exclusion Criteria: clinically debilitating illness; signs or symptoms of severe or complicated malaria; and treatment with antimalarials within the previous 7 days.
  • Classification of Responses to Treatment. For the purpose of this analysis, possible responses to treatment include prophylaxis or treatment failure. Prophylaxis is defined as absence of parasitemia at Day 28 post-treatment. Treatment failure is defined as the presence of parasitemia at or before Day 28.
  • Preferred test formulations have a clinical efficacy (ACPR) of at least 50% on Day 28, more preferably at least 75% and most preferably at least 95%. Additional efficacy targets are described in Burrows et al., Malaria J, 12:187, 2013.
  • Statistical Analyses. All statistical tests comparing demographic, patient characteristic and safety data are conducted using the Kaplan-Meier method. Parasitemia is measured by blood smear or polymerase chain reaction.
  • Example 5 Screening Compounds for Effects on Trypanosoma Growth and Development In Vitro
  • Amastigote Growth Assay. SMIs are screened for growth effects on T. cruzi parasites maintained in Vero cells in vitro. Vero cells (1×105/well) seeded on round coverslips into 24-well plates are infected with bloodstream T. cruzi trypomastigotes (TRYP). After 24 hours of parasite-host cell interaction (10:1 parasite:cell ratio), the infected cultures are washed to remove free parasites and incubated for another 72 hours with a test SMI (1 to 10 micromolar), a placebo, or control comparator. Cell cultures are set up in triplicate and maintained at 37° C. in 5% CO2. Uninfected treated cultures exposed to vehicle (1% DMSO) are used as controls. The method for determining the rate of infection of host cells by T. cruzi has previously been described in detail (Nakajima-Shimada et al., Antimicrob Agents and Chemo, 40:2455-2458, 1996). Briefly, host cells attached to the coverslip seeded in 24-well plate are gently rinsed with phosphate-buffered saline, air-dried, fixed in absolute methanol and stained with Giemsa. The coverslip is then transferred to a glass slide where the mounted cells are finally observed under a light microscope. The percentage of host cells with more than one amastigote (AMAS) in the cytoplasm are counted and the mean number of AMAS per infected cell is determined by analyzing a total of 400 host cells distributed in randomly chosen microscopic fields.
  • Trypomastigote Growth Assay. Bloodstream TRYP are subjected to treatment by incubation of 1.5×105 TRYP with 1-10 micromolar of a test SMI, a placebo, or control comparator. Parasites are diluted in RPMI 1640 medium supplemented with 10% FCS, seeded in 96-well microplates, and incubated at 37° C. in a 5% CO2 atmosphere. After 24 hours, the remaining live parasites are counted in a Neubauer chamber as previously described (Fernandez et al., Experimental Parasitology, 124:172-180, 2010). Trypanocydal effect is determined by counting remaining TRYP at each concentration of drug with respect to a negative control group. Each assay is performed in triplicate.
  • Example 6 Screening Compounds for Effects on Trypanosoma Growth, Infectivity and Development in Mice
  • At least two T. cruzi strains are used for the infection assay: one lethal at the acute phase and the other non-lethal at the acute phase. Use of two strains in this matter facilitates evaluation of parasitemia reduction and cure in the acute and chronic phases of infection.
  • Parasitemia Reduction
  • Groups: N=5 (6 to 8 week old mice).
  • Infection: Lethal strain (100 blood TRYP) injected intradermally into the hind foot pad or alternatively or by intraperitoneal inoculation.
  • Treatment: SMI formulations are administered daily by IP injection for 5 days to Trypanosoma-infected mice, starting at parasitemia levels above 104 TRYP/ml of blood. Control formulations, a placebo or comparator, are tested in parallel to the SMI formulations. The effects of SMIs on parasitemia (e.g., twice weekly) and disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established. The same types of studies are performed with orally delivered SMIs.
  • Cure at the Acute Phase
  • Groups: N=5 (6 to 8 week old mice).
  • Infection: Lethal strain (100 blood TRYP) injected intradermally into the hind footpad or alternatively by intraperitoneal inoculation.
  • Treatment: SMI formulations are administered daily by IP injection for 20 to 30 days to Trypanosoma-infected mice, starting at parasitemia levels above 104 TRYP/ml of blood. Control formulations, a placebo or comparator, are tested in parallel to the SMI formulations. The effects of SMIs on parasitemia and disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established. The same types of studies are performed with orally delivered SMIs. PCR may also be employed to detect AMAS in mouse tissues.
  • Cure at the Chronic Phase.
  • Groups: N=5 (6 to 8 week old mice).
  • Infection: Non-lethal strain injected intradermally into the hind footpad or alternatively by intraperitoneal inoculation. In particular, a fluorescently-labeled non-lethal K98 strain may be used.
  • Treatment: SMI formulations are administered daily by IP injection for 30 days to Trypanosoma-infected mice, starting at parasitemia levels above 104 TRYP/ml of blood. Control formulations, a placebo or comparator, are tested in parallel to the SMI formulations. The effects of SMIs on parasitemia and disease severity over time is then assessed. If a SMI is bioactive against parasite infection, oral bioavailability is established. The same types of studies are performed with orally delivered SMIs. PCR may also be employed to detect AMAS in mouse tissues. If fluorescently labeled parasites of the non-lethal K98 strain are used, parasitemia measurements may be performed by microscopy or automated fluorescent cell counting.
  • Example 7 Administration of Kinase Inhibitor(s) to Human Patients to Treat Chagas Disease
  • This example provides a description of a clinical study among Trypanosoma cruzi-infected individuals. The clinical and parasitological response (CPR) in test subjects receiving treatment with candidate kinase inhibitor(s) (test formulation) is compared to the CPR of control subjects receiving treatment with a control formulation (placebo or comparator).
  • Primary Objective: to compare the proportions of test and control subjects that are free of parasites in the blood at 12 months post-treatment.
  • Secondary Objective(s): to compare the proportions of test and control subjects free of parasites in the blood at Weeks 8, 16, 24, and 40 post-treatment; to compare the safety and tolerability of the candidate kinase inhibitor(s) in test and control subjects; and to compare the time to clearance of parasites in the blood in test and control subjects.
  • Normal, Infected Subjects. This study is a subject- and observer-blinded, randomized, controlled study of Trypanosoma cruzi-infected, but otherwise healthy, adult subjects (ages 18 years and older) to receive oral dosing with either the test formulation or the control formulation. Subjects are stratified by age, sex and pregnancy status (where applicable) prior to randomization.
  • Study Population. Subjects are selected from among Trypanosoma cruzi-infected male and female volunteers. Inclusion and exclusion criteria met by study participants are as follows. Inclusion Criteria: infection with Trypanosoma cruzi, ability to swallow oral medication, willingness to comply with the study protocol and ability to adhere to strict dosing requirements. Exclusion Criteria: clinically debilitating illness; and signs or symptoms of severe or complicated Chagas disease.
  • Classification of Responses to Treatment. For the purpose of this analysis, a positive response to treatment (parasitological cure) is indicated by an absence of parasites in the blood, as measured by real time polymerase chain reaction.
  • Statistical Analyses. All statistical tests comparing demographic, patient characteristic and safety data are conducted using the Kaplan-Meier method. Parasitological cure is measured by real time polymerase chain reaction.

Claims (45)

1. A method for treating or preventing Chagas disease comprising: administering a mammalian protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing Chagas disease.
2. The method of claim 1, wherein the mammalian protein kinase inhibitor comprises a first kinase inhibitor and a second kinase inhibitor, and the administering reduces activity of a first mammalian protein kinase and a second mammalian protein kinase.
3. The method of claim 1, wherein the mammalian protein kinase comprises a member of a protein kinase C (PKC) family.
4. The method of claim 1, wherein the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family.
5. The method of claim 1, wherein the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a protein kinase B (PKB) family, and a phosphoinositide 3-kinase (PI3K) family.
6. The method of claim 1, wherein the mammalian protein kinase inhibitor is capable of reducing activity of a Triatominae insect kinase.
7. The method of claim 1, further comprising administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject.
8. The method of claim 1, wherein the subject is infected with Trypanosoma cruzi.
9. The method of claim 8, wherein the subject is acutely infected with T. cruzi.
10. The method of claim 8, wherein the subject is chronically infected with T. cruzi.
11. The method of claim 8, wherein treating Chagas disease comprises alleviating a symptom of Chagas disease experienced by the subject.
12. The method of claim 11, further comprising administering an effective amount of an additional antiparasitic agent to the mammalian subject.
13. The method of claim 12, wherein the additional antiparasitic agent comprises one or both of benznidazole and nifurtimox.
14. The method of claim 1, wherein the subject is an uninfected individual planning to visit a Chagas disease endemic area after the administering step has commenced.
15. The method of claim 14, wherein preventing Chagas disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days.
16. A method of identifying an anti-Chagas disease compound, comprising:
(a) measuring activity of a mammalian or Triatominae insect protein kinase in the presence and absence of a test compound; and
(b) identifying the test compound as an anti-Chagas disease compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound.
17. The method of claim 16, further comprising growing insect cells in vitro in the presence and absence of the test compound and identifying the test compound as insect-cell safe when viability or doubling time of the insect cells is not significantly reduced in the presence as compared to the absence of the test compound.
18. The method of claim 16, wherein the mammalian protein kinase is a human protein kinase.
19. The method of claim 16, wherein the Triatominae insect protein kinase is a Rhodnius prolixus protein kinase.
20. (canceled)
21. (canceled)
22. A pharmaceutical composition comprising:
a protein kinase inhibitor attached to a mitochondrial-targeting moiety, and
one or both of a pharmaceutically acceptable excipient and carrier,
wherein the protein kinase inhibitor is present in an amount effective to treat or prevent Chagas disease in a mammalian subject.
23. A method for treating or preventing an arthropod-borne parasitic disease comprising: administering a mammalian protein kinase inhibitor to a mammalian subject in need thereof under conditions effective for treating or preventing the arthropod-borne parasitic disease, wherein the parasitic disease is not malaria.
24. The method of claim 23, wherein the mammalian protein kinase inhibitor comprises a first kinase inhibitor and a second kinase inhibitor, and the administering reduces activity of a first mammalian protein kinase and a second mammalian protein kinase.
25. The method of claim 23, wherein the mammalian protein kinase comprises a member of a protein kinase C (PKC) family.
26. The method of claim 23, wherein the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a c-Jun N-terminal kinase (JNK) family, and a p38 mitogen activated protein kinase (MAPK) family.
27. The method of claim 23, wherein the mammalian protein kinase comprises a member of one or more families selected from the group consisting of a protein kinase C (PKC) family, a protein kinase B (PKB) family, and a phosphoinositide 3-kinase (PI3K) family.
28. The method of claim 23, wherein the mammalian protein kinase inhibitor is capable of reducing activity of an arthropod kinase.
29. The method of claim 23, further comprising administering an effective amount of a further mammalian enzyme inhibitor to the mammalian subject.
30. The method of claim 23, wherein the subject is infected with an arthropod-borne parasite which is not a Plasmodium sp. parasite.
31. The method of claim 30, wherein the subject is acutely infected with the arthropod-borne parasite.
32. The method of claim 30, wherein the subject is chronically infected with the arthropod-borne parasite.
33. The method of claim 30, wherein treating the parasitic disease comprises alleviating a symptom of the parasitic disease experienced by the subject.
34. The method of claim 30, further comprising administering an effective amount of an additional antiparasitic agent to the mammalian subject.
35. The method of claim 23, wherein the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step commences before the visit.
36. The method of claim 35, wherein the subject is an uninfected individual planning to visit a parasitic disease endemic area, and the administering step concludes during or after the visit.
37. The method of claim 36, wherein preventing the parasitic disease comprises protecting the subject from developing parasitemia during their visit to the endemic area for a period of up to thirty days.
38. A method of identifying antiparasitic compound, comprising:
(a) measuring activity of a mammalian or insect protein kinase in the presence and absence of a test compound; and
(b) identifying the test compound as an antiparasitic compound when the activity of the protein kinase is reduced in the presence as compared to the absence of the test compound, and wherein the insect protein kinase is not a Anopheles sp. kinase.
39. The method of claim 38, further comprising growing insect cells in vitro in the presence and absence of the test compound and identifying the test compound as insect-cell safe when viability or doubling time of the insect cells is not significantly reduced in the presence as compared to the absence of the test compound, and wherein the insect cells are not Anopheles sp. cells.
40. The method of claim 38, wherein the mammalian protein kinase is a human protein kinase.
41. The method of claim 38, wherein the insect protein kinase is selected from the group consisting of a sand-fly protein kinase, a tick protein kinase and a tsetse fly protein kinase.
42. (canceled)
43. (canceled)
44. A pharmaceutical composition comprising:
a protein kinase inhibitor attached to a mitochondrial-targeting moiety, and
one or both of a pharmaceutically acceptable excipient and carrier,
wherein the protein kinase inhibitor is present in an amount effective to treat or prevent an arthropod-borne parasitic disease in a mammalian subject, wherein the parasitic disease is not malaria.
45. The pharmaceutical composition of claim 44, wherein the parasitic disease is selected from the group consisting of leishmaniasis, babesiosis, and African trypanosomiasis.
US14/904,686 2013-07-12 2014-07-11 Therapies for diseases caused by arthropod-borne parasites Abandoned US20160151380A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/904,686 US20160151380A1 (en) 2013-07-12 2014-07-11 Therapies for diseases caused by arthropod-borne parasites

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361845920P 2013-07-12 2013-07-12
US201461923405P 2014-01-03 2014-01-03
US14/904,686 US20160151380A1 (en) 2013-07-12 2014-07-11 Therapies for diseases caused by arthropod-borne parasites
PCT/US2014/046441 WO2015006753A2 (en) 2013-07-12 2014-07-11 Therapies for diseases caused by arthropod-borne parasites

Publications (1)

Publication Number Publication Date
US20160151380A1 true US20160151380A1 (en) 2016-06-02

Family

ID=52280656

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/904,673 Expired - Fee Related US9918989B2 (en) 2013-07-12 2014-07-11 Combination therapies for malaria
US14/904,686 Abandoned US20160151380A1 (en) 2013-07-12 2014-07-11 Therapies for diseases caused by arthropod-borne parasites

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/904,673 Expired - Fee Related US9918989B2 (en) 2013-07-12 2014-07-11 Combination therapies for malaria

Country Status (2)

Country Link
US (2) US9918989B2 (en)
WO (2) WO2015006753A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9918989B2 (en) 2013-07-12 2018-03-20 The Regents Of The University Of California Combination therapies for malaria
US20210322323A1 (en) * 2020-04-14 2021-10-21 Janssen Biotech, Inc. Pharmaceutical composition for oral administration comprising aminopyrimidine derivative or pharmaceutically acceptable salt, hydrate, or solvate thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9782417B2 (en) 2011-06-16 2017-10-10 Presidents And Fellows Of Harvard College Methods of increasing satellite cell proliferation with kinase inhibitors
AU2017213796A1 (en) * 2016-02-01 2018-09-20 President And Fellows Of Harvard College Small molecules for mouse satellite cell proliferation
EP3442520A4 (en) * 2016-04-11 2020-04-22 Middle Tennessee State University Therapeutic aurones
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
CA3128468A1 (en) 2017-10-05 2019-04-11 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce dux4 and downstream gene expression for the treatment of fshd
CN109750109A (en) * 2019-02-01 2019-05-14 中山大学 It is a kind of for identifying the molecular specificity marker object and method for identifying molecules of triatoma rubrofasciata

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7670631B2 (en) 2003-03-12 2010-03-02 ALFAMA—Investigação e Desenvolvimento de Produtos Farmacêuticos, Lda. Method for the prevention of malaria infection of humans by hepatocyte growth factor antagonists
SI1663978T1 (en) * 2003-07-23 2008-02-29 Bayer Pharmaceuticals Corp Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
WO2005055812A2 (en) * 2003-12-05 2005-06-23 Ciphergen Biosystems, Inc. Serum biomarkers for chagas disease
US8329411B2 (en) * 2004-04-23 2012-12-11 University Of Georgia Research Foundation, Inc. Diagnostic assay for Trypanosoma cruzi infection
BRPI0603871B8 (en) * 2006-08-24 2021-05-25 Fundacao Univ De Brasilia pharmaceutical compositions for the treatment of trypanosomiasis and chagas disease
JP2010516774A (en) * 2007-01-26 2010-05-20 アイアールエム・リミテッド・ライアビリティ・カンパニー Purine compounds and compositions as kinase inhibitors for treating Plasmodium related diseases
WO2010049173A1 (en) * 2008-10-31 2010-05-06 Cenix Bioscience Gmbh Use of inhibitors of host kinases for the treatment of infectious diseases
WO2011115819A2 (en) 2010-03-15 2011-09-22 Genus Oncology, Llc Small molecule inhibitors of muc1 and methods of identifying the same
US9918989B2 (en) 2013-07-12 2018-03-20 The Regents Of The University Of California Combination therapies for malaria

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9918989B2 (en) 2013-07-12 2018-03-20 The Regents Of The University Of California Combination therapies for malaria
US20210322323A1 (en) * 2020-04-14 2021-10-21 Janssen Biotech, Inc. Pharmaceutical composition for oral administration comprising aminopyrimidine derivative or pharmaceutically acceptable salt, hydrate, or solvate thereof

Also Published As

Publication number Publication date
US9918989B2 (en) 2018-03-20
WO2015006752A1 (en) 2015-01-15
WO2015006753A2 (en) 2015-01-15
WO2015006753A3 (en) 2015-04-09
US20160151379A1 (en) 2016-06-02

Similar Documents

Publication Publication Date Title
US20160151380A1 (en) Therapies for diseases caused by arthropod-borne parasites
Antczak et al. Human toxoplasmosis–Searching for novel chemotherapeutics
Cavalli et al. Neglected tropical diseases: multi-target-directed ligands in the search for novel lead candidates against Trypanosoma and Leishmania
Naula et al. Protein kinases as drug targets in trypanosomes and Leishmania
Deng et al. Recent progress on anti-Toxoplasma drugs discovery: Design, synthesis and screening
Tibon et al. Current progress in antimalarial pharmacotherapy and multi-target drug discovery
Beckmann et al. Protein kinases as potential targets for novel anti-schistosomal strategies
Ke et al. Primary cilium: an elaborate structure that blocks cell division?
McFarland et al. Review of experimental compounds demonstrating anti-Toxoplasma activity
CN108992446A (en) With TOR kinase inhibitor for treating cancer
Berninger et al. Novel lead compounds in pre-clinical development against African sleeping sickness
BR112013024211B1 (en) TREATMENT OF SOLID TUMORS
Petrilli et al. A chemical biology approach identified PI3K as a potential therapeutic target for neurofibromatosis type 2
Avery et al. The fight against drug-resistant malaria: novel plasmodial targets and antimalarial drugs
Wu et al. CDK9 inhibitors reactivate p53 by downregulating iASPP
Bhattacharjee et al. Drug resistance in malaria-in a nutshell
Wittlin et al. From magic bullet to magic bomb: reductive bioactivation of antiparasitic agents
Morawietz et al. Targeting kinases in Fasciola hepatica: anthelminthic effects and tissue distribution of selected kinase inhibitors
WO2022082306A1 (en) Use of n-myristoyl transferase (nmt) inhibitors in the treatment of cancer, autoimmune disorders, and inflammatory disorders
De Lima et al. Growth arrest and morphological changes triggered by emodin on Trypanosoma cruzi epimastigotes cultivated in axenic medium
US11147807B2 (en) Pharmaceutical composition containing DUSP1 inhibitor
KR20180037975A (en) MDM2 inhibitor for treating uveal melanoma
Sudhakar et al. Bazedoxifene, a postmenopausal drug, acts as an antimalarial and inhibits hemozoin formation
EP2520297A1 (en) Rubreserine and its derivates with antiparasitic activity
Jia et al. Phosphatase inhibitors BVT-948 and alexidine dihydrochloride inhibit sexual development of the malaria parasite Plasmodium berghei

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUCKHART, SHIRLEY;GIULIVI, CECILIA;SIGNING DATES FROM 20160505 TO 20160516;REEL/FRAME:039182/0234

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION