Polo-like kinase 1 (Plk1) is one of the most attractive targets for anti-cancer therapy. Efforts to generate Plk1-specific inhibitors by targeting the catalytic activity of Plk1 have proven to be difficult due to similarities with the catalytic domains of other structurally related kinases. Here, we propose to develop a new class of mono-specific Plk1 inhibitors by employing a novel approach of targeting the non-catalytic, but functionally essential, PBD of Plk1. To this end, we have been taking advantage of the crystal structures of the Plk1 PBD in complex with a highly specific ligand, PLHSpT.
The first aim of the project is to develop PLHSpT-derived templates for drug design and subsequent modifications. Since the function of the N-terminal Pro-4 (number indicates the relative position of the residue from pT) residue of PLHSpT can be substituted by hydrophobic moieties and the side chain of the Leu-3 residue is not involved in interactions with surrounding PBD residues, we first generated N-substituted glycine (Nsg)-containing LHSpT or HSpT peptoidpeptide hybrids and their respective cyclic forms. Then, the Nsg residue of the hybrids was modified by covalently conjugating them with site-specifically synthesized hydrophobic moieties to fill in the intramolecular cavity present between the compound and the PBD. Since the His-2, Ser-1 and p-Thr residues are critical for the specificity and high affinity binding, these residues will not be modified. Peptide-derived inhibitors are commonly associated with problems in stability, lipophilicity, and transcellular permeability. Hence, the second aim of the project is to enhance the stability, membrane permeability, and tumor-specific targeting of the above compounds by generating innovative prodrugs. Selected compounds with a high PBD-binding affinity and specificity were converted to phosphatase-insensitive, non-hydrolyzable, p-Thr mimetic (Pmab) forms and then further modified to generate Ala- or Val-ester-conjugated phosphonic diamide prodrugs. The latter modification is not only to eliminate the electronegativity of the dianionic phosphonic acid moiety for better transcellular permeation but also to target the compound to the highly active PepT1 transporter for efficient cellular uptake. For the compounds that exhibit anti-Plk1 PBD activities at the cultured cell level, we plan to investigate whether the addition of a tumor-targeting RGD motif facilitates tumor-specific delivery of the compounds. As the third aim of the project, we will determine the potency and selectivity of the resulting compounds in the inhibition of Plk1-dependent cell proliferation activity in mouse tumor models. Because of the harsh chemical and enzymatic conditions of the gastrointestinal tract, intravenous injection into mouse-tail vein will be the choice of compound administration. Since peptide-derived inhibitors often exhibit physicochemical drawbacks, we also propose to take complementary approaches to isolate PBD-inhibitory compounds or moieties by screening the NCI natural products extract repository or by carrying out in silico screening. Structural analyses and computer modeling of the isolated small molecule compounds together with the above PLHSpT-derived inhibitors will allow us to perform site-specific replacements and modifications of the latter inhibitors to achieve enhanced in vivo stability and bioavailability. To this end, we will bring the expertise of our collaborators ranging from in vitro high throughput screening and cell-based assays, in silico screening and computer modeling, and X-ray crystallography. The ultimate goal of this multifaceted approach is to generate a new class of mono-specific anti-Plk1 therapeutic agents that have potential to treat various cancers in humans.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC010681-08
Application #
8552776
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
8
Fiscal Year
2012
Total Cost
$331,824
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Park, Jung-Eun; Hymel, David; Burke Jr, Terrence R et al. (2017) Current progress and future perspectives in the development of anti-polo-like kinase 1 therapeutic agents. F1000Res 6:1024
Lee, Kyung S; Burke Jr, Terrence R; Park, Jung-Eun et al. (2015) Recent Advances and New Strategies in Targeting Plk1 for Anticancer Therapy. Trends Pharmacol Sci 36:858-877
Kim, Ju Hee; Ku, Bonsu; Lee, Kyung S et al. (2015) Structural analysis of the polo-box domain of human Polo-like kinase 2. Proteins :
Jia, Jia-Lin; Han, Young-Hyun; Kim, Hak-Cheol et al. (2015) Structural basis for recognition of Emi2 by Polo-like kinase 1 and development of peptidomimetics blocking oocyte maturation and fertilization. Sci Rep 5:14626
Park, Jung-Eun; Kim, Tae-Sung; Kim, Bo Yeon et al. (2015) Selective blockade of cancer cell proliferation and anchorage-independent growth by Plk1 activity-dependent suicidal inhibition of its polo-box domain. Cell Cycle 14:3624-34
Qian, Wen-Jian; Park, Jung-Eun; Grant, Robert et al. (2015) Neighbor-directed histidine N (?)-alkylation: A route to imidazolium-containing phosphopeptide macrocycles. Biopolymers 104:663-73
Qian, Wen-Jian; Park, Jung-Eun; Lim, Dan et al. (2014) Mono-anionic phosphopeptides produced by unexpected histidine alkylation exhibit high Plk1 polo-box domain-binding affinities and enhanced antiproliferative effects in HeLa cells. Biopolymers 102:444-55
Srinivasrao, Ganipisetti; Park, Jung-Eun; Kim, Sungmin et al. (2014) Design and synthesis of a cell-permeable, drug-like small molecule inhibitor targeting the polo-box domain of polo-like kinase 1. PLoS One 9:e107432
Kim, Sun-Ok; Sakchaisri, Krisada; Thimmegowda, N R et al. (2013) STK295900, a dual inhibitor of topoisomerase 1 and 2, induces G(2) arrest in the absence of DNA damage. PLoS One 8:e53908
Qian, Wenjian; Park, Jung-Eun; Liu, Fa et al. (2013) Effects on polo-like kinase 1 polo-box domain binding affinities of peptides incurred by structural variation at the phosphoamino acid position. Bioorg Med Chem 21:3996-4003

Showing the most recent 10 out of 18 publications