Human African Trypanosomiasis (HAT), also known as sleeping sickness, is a vector-borne parasitic disease caused by Trypanosoma brucei infections. It threatens over 60 million people in 36 countries of sub-Saharan Africa where medical resources are very limited. There are no satisfactory drugs for the treatment thus far. Trypanosome infections are now referred to as ?most neglected diseases?, and need more research efforts. The dynamics of microtubules is essential for T. brucei cell division and its flagellar motility. Therefore, tubulin is a promising target of anti-trypanosome agents, since compounds interfering with microtubule dynamics can suppress both cell division and movement. Importantly, trypanosomal and mammalian tubulins have structural differences, allowing development of selective inhibitors with minimal toxicity. On the other hand, the tubulin structures in all disease-causing Kinetoplastids are nearly identical, suggesting that our tubulin inhibitors have the potential to suppress other Kinetoplastid infections. We have identified several drug candidates that selectively decreased trypanosome cell viability. Via oral gavage, these compounds significantly inhibited trypanosomal replication in infected mice. However, the potency of the compounds was not strong enough to clear up the infection completely. Nevertheless, the compounds are good leads for further optimization to generate more effective anti-trypanosome agents. We hypothesize that introducing functional groups that can be recognized by the P2 transporter on trypanosome cell surface will enhance uptake of tubulin inhibitors, which will improve their efficacy and selectivity. To test this hypothesis, we will focus on three research aims listed below: 1) Structurally optimize the tubulin inhibitors to improve the trypanosome cell uptake. We will introduce purine mimetic and imidamide moieties into our tubulin inhibitors to improve their cell uptake, as trypanosomes depends on their mammalian host for purines. We will also test if our compounds can inhibit T. cruzi growth due to the great similarity among tubulin homologues of all trypanosomes. 2) Investigate the mechanisms and in vivo potency of the selected compounds. We expect our tubulin inhibitors block trypanosomal tubulin polymerization, which will be tested. In addition, we will determine if our new compounds show improved in vivo activities, since the uptake of these compounds by trypanosomes is enhanced. 3) Determine if our drug candidates can cross the blood-brain barrier. Our drug candidates showed relatively high hydrophobicity and no ionization in normal physiological condition, suggesting that these compounds are likely able to cross the blood-brain barrier. We will determine the drug concentrations in blood and brain tissue after administering to trypanosome-infected mice.

Public Health Relevance

Human African trypanosomiasis (HAT) is a vector-borne parasitic disease caused by Trypanosoma brucei. It threatens over 60 million people in 36 countries of sub-Saharan Africa. Currently, there are no effective vaccines and oral active drugs for the treatments, and all the chemotherapeutic agents used for the disease have to be administered via injection, which minimizes their application due to the limited medical resources in trypanosome endemic areas. The development of orally active drugs for the disease is urgently needed. Trypanosome infections are now referred as the ?most neglected diseases?, and need more research. We focus our efforts on developing orally active and selective tubulin inhibitors for this orphan disease. Due to the tubulin similarity among all the Kinetoplastids, our compounds have great potential to be used for the treatment of other Kinetoplastids infection as well.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Academic Research Enhancement Awards (AREA) (R15)
Project #
2R15AI103889-02
Application #
9171018
Study Section
Special Emphasis Panel (ZRG1-IDM-S (81)A)
Program Officer
O'Neil, Michael T
Project Start
2016-06-17
Project End
2019-05-31
Budget Start
2016-06-17
Budget End
2019-05-31
Support Year
2
Fiscal Year
2016
Total Cost
$436,500
Indirect Cost
$136,500
Name
Cleveland State University
Department
Chemistry
Type
Schools of Arts and Sciences
DUNS #
010841617
City
Cleveland
State
OH
Country
United States
Zip Code
44115
Petty, Aaron; Idippily, Nethrie; Bobba, Viharika et al. (2018) Design and synthesis of small molecule agonists of EphA2 receptor. Eur J Med Chem 143:1261-1276
Bobba, Viharika; Nanavaty, Vishal; Idippily, Nethrie D et al. (2017) Synthesis and biological evaluation of selective tubulin inhibitors as anti-trypanosomal agents. Bioorg Med Chem 25:3215-3222
Nanavaty, Vishal; Lama, Rati; Sandhu, Ranjodh et al. (2016) Orally Active and Selective Tubulin Inhibitors as Anti-Trypanosome Agents. PLoS One 11:e0146289
Voggu, Ramakrishna R; Zhou, Xiang; Su, Bin et al. (2015) A Simple and Rapid LC-MS/MS Method for the Determination of BMCL26 a Novel Anti-Parasitic Agent in Rat Plasma. J Anal Bioanal Tech 6:
Zhong, Bo; Lama, Rati; Kulman, Daniel G et al. (2014) Lead optimization of dual tubulin and Hsp27 inhibitors. Eur J Med Chem 80:243-53
Zhong, Bo; Chennamaneni, Snigdha; Lama, Rati et al. (2013) Synthesis and anticancer mechanism investigation of dual Hsp27 and tubulin inhibitors. J Med Chem 56:5306-20