Therapies for diseases caused by arthropod-borne parasites

a technology of arthropods and parasites, applied in the field of treating or preventing diseases caused by arthropods, can solve the problems of increased risk of sudden death, heart failure, and/or gastrointestinal disturbance, severe inflammation of the heart or brain, and parasitemia levels that are undetectable by microscopy, etc., to and reduce the transmission of trypanosoma cruzi

Inactive Publication Date: 2016-06-02
RGT UNIV OF CALIFORNIA
View PDF1 Cites 2 Cited by
  • Summary
  • Abstract
  • Description
  • Claims
  • Application Information

AI Technical Summary

Benefits of technology

[0009]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.
[0012]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.

Problems solved by technology

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).
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.
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.
Thus, Chagas disease is a serious illness affecting a significant portion of the population.
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.
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.
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.

Method used

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
View more

Image

Smart Image Click on the blue labels to locate them in the text.
Viewing Examples
Smart Image
  • Therapies for diseases caused by arthropod-borne parasites
  • Therapies for diseases caused by arthropod-borne parasites
  • Therapies for diseases caused by arthropod-borne parasites

Examples

Experimental program
Comparison scheme
Effect test

example 1

Screening Compounds for Effects on Cell Viability, Proliferation and Kinase Activity in Mosquito Cells In Vitro and In Vivo

[0068]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.

[0069]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-Radioactiv...

example 2

Screening Compounds for Effects on Plasmodium Growth, Infectivity and Development in Erythrocytes, Mice and Mosquito Cells

[0073]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.

[0074]P. falciparum growth assay. Aliquots of ring stage P. falci...

example 3

Administration of Kinase Inhibitor(s) to Human Patients to Treat Malaria

[0080]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).

[0081]Primary Objective: to compare the proportions of test and control subjects that are free of parasites in the blood at Day 28 post-treatment.

[0082]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.

[0083]Tertiary Objective(s); compare the responses to treatment of test and control subjects according to the World Health...

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

PUM

PropertyMeasurementUnit
molecular weightaaaaaaaaaa
molecular weightaaaaaaaaaa
molecular weightaaaaaaaaaa
Login to view more

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[0001]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[0002]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[0003]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. Ad...

Claims

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

Application Information

Patent Timeline
no application Login to view more
Patent Type & Authority Applications(United States)
IPC IPC(8): A61K31/541A61K31/404A61K31/473A61K31/5377A61K31/4035A61K31/4178A61K31/52A61K31/4168A61K31/407
CPCA61K31/541A61K31/4168A61K31/404A61K31/473A61K31/5377A61K31/4035A61K31/4178A61K31/52A61K31/407A61K31/416A61K31/4162A61K31/4439A61K45/06A61P33/06G01N33/56905G01N2333/445G01N2333/912Y02A50/30A61K49/0008C12Q1/485G01N33/5008G01N33/5014G01N33/573G01N2440/14G01N2500/04G01N2500/10G01N2500/20
Inventor LUCKHART, SHIRLEYGIULIVI, CECILIA
Owner RGT UNIV OF CALIFORNIA
Who we serve
  • R&D Engineer
  • R&D Manager
  • IP Professional
Why Eureka
  • Industry Leading Data Capabilities
  • Powerful AI technology
  • Patent DNA Extraction
Social media
Try Eureka
PatSnap group products