1
|
Du Y, Chen X, Chen W, Chen G, Cheng X, Wang H, Guo L, Li C, Yao D. Design, synthesis and biological evaluation of a novel PAK1 degrader for the treatment of triple negative breast cancer. Bioorg Med Chem 2024; 112:117896. [PMID: 39214014 DOI: 10.1016/j.bmc.2024.117896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Triple-negative breast cancer is one of the most malignant subtypes in clinical practice, and it is urgent to find new therapies. The p21-activated kinase I (PAK1) has been considered to be an attractive therapeutic target for TNBC. In this study, we designed and synthesized a series of novel PROTAC PAK1 degraders by conjugating VHL or CRBN ligase ligands to PAK1 inhibitors which are connected by alkyl chains or PEG chains. The most promising compound, 19s, can significantly degrade PAK1 protein at concentrations as low as 0.1 μM, and achieves potent anti-proliferative activity with an IC50 value of 1.27 μM in MDA-MB-231 cells. Additionally, 19s exhibits potent anti-migration activity in vitro and induces rapid tumor regression in vivo. Collectively, these findings document that 19s is a potent and novel PAK1 degrader with promising potential for TNBC treatment.
Collapse
Affiliation(s)
- Yi Du
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xiya Chen
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China
| | - Weiji Chen
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China
| | - Gang Chen
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China
| | - Xiaoling Cheng
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China
| | - Hailing Wang
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China
| | - Ling Guo
- Department of Science and Research, The Affiliated Anning First People's Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan Province, China
| | - Chenyang Li
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| | - Dahong Yao
- School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability & Human Disease Prevention, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518060, China.
| |
Collapse
|
2
|
Somanath PR, Chernoff J, Cummings BS, Prasad SM, Homan HD. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:2236. [PMID: 37190165 PMCID: PMC10137274 DOI: 10.3390/cancers15082236] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (mPCa) has limited therapeutic options and a high mortality rate. The p21-activated kinase (PAK) family of proteins is important in cell survival, proliferation, and motility in physiology, and pathologies such as infectious, inflammatory, vascular, and neurological diseases as well as cancers. Group-I PAKs (PAK1, PAK2, and PAK3) are involved in the regulation of actin dynamics and thus are integral for cell morphology, adhesion to the extracellular matrix, and cell motility. They also play prominent roles in cell survival and proliferation. These properties make group-I PAKs a potentially important target for cancer therapy. In contrast to normal prostate and prostatic epithelial cells, group-I PAKs are highly expressed in mPCA and PCa tissue. Importantly, the expression of group-I PAKs is proportional to the Gleason score of the patients. While several compounds have been identified that target group-I PAKs and these are active in cells and mice, and while some inhibitors have entered human trials, as of yet, none have been FDA-approved. Probable reasons for this lack of translation include issues related to selectivity, specificity, stability, and efficacy resulting in side effects and/or lack of efficacy. In the current review, we describe the pathophysiology and current treatment guidelines of PCa, present group-I PAKs as a potential druggable target to treat mPCa patients, and discuss the various ATP-competitive and allosteric inhibitors of PAKs. We also discuss the development and testing of a nanotechnology-based therapeutic formulation of group-I PAK inhibitors and its significant potential advantages as a novel, selective, stable, and efficacious mPCa therapeutic over other PCa therapeutics in the pipeline.
Collapse
Affiliation(s)
- Payaningal R. Somanath
- Department of Clinical & Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- MetasTx LLC, Basking Ridge, NJ 07920, USA
| | - Jonathan Chernoff
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian S. Cummings
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Sandip M. Prasad
- Morristown Medical Center, Atlantic Health System, Morristown, NJ 07960, USA
| | | |
Collapse
|
3
|
Biswal J, Jayaprakash P, Rayala SK, Venkatraman G, Rangaswamy R, Jeyaraman J. WaterMap and Molecular Dynamic Simulation-Guided Discovery of Potential PAK1 Inhibitors Using Repurposing Approaches. ACS OMEGA 2021; 6:26829-26845. [PMID: 34693105 PMCID: PMC8529594 DOI: 10.1021/acsomega.1c02032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Indexed: 06/13/2023]
Abstract
p21-Activated kinase 1 (PAK1) is positioned at the nexus of several oncogenic signaling pathways. Currently, there are no approved inhibitors for disabling the transfer of phosphate in the active site directly, as they are limited by lower affinity, and poor kinase selectivity. In this work, a repurposing study utilizing FDA-approved drugs from the DrugBank database was pursued with an initial selection of 27 molecules out of ∼2162 drug molecules, based on their docking energies and molecular interaction patterns. From the molecules that were considered for WaterMap analysis, seven molecules, namely, Mitoxantrone, Labetalol, Acalabrutinib, Sacubitril, Flubendazole, Trazodone, and Niraparib, ascertained the ability to overlap with high-energy hydration sites. Considering many other displaced unfavorable water molecules, only Acalabrutinib, Flubendazole, and Trazodone molecules highlighted their prominence in terms of binding affinity gains through ΔΔG that ranges between 6.44 and 2.59 kcal/mol. Even if Mitoxantrone exhibited the highest docking score and greater interaction strength, it did not comply with the WaterMap and molecular dynamics simulation results. Moreover, detailed MD simulation trajectory analyses suggested that the drug molecules Flubendazole, Niraparib, and Acalabrutinib were highly stable, observed from their RMSD values and consistent interaction pattern with Glu315, Glu345, Leu347, and Asp407 including the hydrophobic interactions maintained in the three replicates. However, the drug molecule Trazodone displayed a loss of crucial interaction with Leu347, which was essential to inhibit the kinase activity of PAK1. The molecular orbital and electrostatic potential analyses elucidated the reactivity and strong complementarity potentials of the drug molecules in the binding pocket of PAK1. Therefore, the CADD-based reposition efforts, reported in this work, helped in the successful identification of new PAK1 inhibitors that requires further investigation by in vitro analysis.
Collapse
Affiliation(s)
- Jayashree Biswal
- Structural
Biology and Bio-Computing Laboratory, Department of Bioinformatics,
Science Block, Alagappa University, Karaikudi 630 004, Tamil Nadu, India
| | - Prajisha Jayaprakash
- Structural
Biology and Bio-Computing Laboratory, Department of Bioinformatics,
Science Block, Alagappa University, Karaikudi 630 004, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department
of Biotechnology, Indian Institute of Technology
Madras, Room No. BT 306, Chennai 600 036, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department
of Human Genetics, College of Biomedical Sciences, Sri Ramachandra University, Porur, Chennai 600 116, Tamil Nadu, India
| | - Raghu Rangaswamy
- Structural
Biology and Bio-Computing Laboratory, Department of Bioinformatics,
Science Block, Alagappa University, Karaikudi 630 004, Tamil Nadu, India
| | - Jeyakanthan Jeyaraman
- Structural
Biology and Bio-Computing Laboratory, Department of Bioinformatics,
Science Block, Alagappa University, Karaikudi 630 004, Tamil Nadu, India
| |
Collapse
|
4
|
Carroll DJ, Cao Y, Bochner BS, O’Sullivan JA. Siglec-8 Signals Through a Non-Canonical Pathway to Cause Human Eosinophil Death In Vitro. Front Immunol 2021; 12:737988. [PMID: 34721399 PMCID: PMC8549629 DOI: 10.3389/fimmu.2021.737988] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a glycan-binding receptor bearing immunoreceptor tyrosine-based inhibitory and switch motifs (ITIM and ITSM, respectively) that is selectively expressed on eosinophils, mast cells, and, to a lesser extent, basophils. Previous work has shown that engagement of Siglec-8 on IL-5-primed eosinophils causes cell death via CD11b/CD18 integrin-mediated adhesion and NADPH oxidase activity and identified signaling molecules linking adhesion, reactive oxygen species (ROS) production, and cell death. However, the proximal signaling cascade activated directly by Siglec-8 engagement has remained elusive. Most members of the Siglec family possess similar cytoplasmic signaling motifs and recruit the protein tyrosine phosphatases SHP-1/2, consistent with ITIM-mediated signaling, to dampen cellular activation. However, the dependence of Siglec-8 function in eosinophils on these phosphatases has not been studied. Using Siglec-8 antibody engagement and pharmacological inhibition in conjunction with assays to measure cell-surface upregulation and conformational activation of CD11b integrin, ROS production, and cell death, we sought to identify molecules involved in Siglec-8 signaling and determine the stage of the process in which each molecule plays a role. We demonstrate here that the enzymatic activities of Src family kinases (SFKs), Syk, SHIP1, PAK1, MEK1, ERK1/2, PLC, PKC, acid sphingomyelinase/ceramidase, and Btk are all necessary for Siglec-8-induced eosinophil cell death, with no apparent role for SHP-1/2, SHIP2, or c-Raf. While most of these signaling molecules are necessary for Siglec-8-induced upregulation of CD11b integrin at the eosinophil cell surface, Btk is phosphorylated and activated later in the signaling cascade and is instead necessary for CD11b activation. In contrast, SFKs and ERK1/2 are phosphorylated far earlier in the process, consistent with their role in augmenting cell-surface levels of CD11b. In addition, pretreatment of eosinophils with latrunculin B or jasplakinolide revealed that actin filament disassembly is necessary and sufficient for surface CD11b integrin upregulation and that actin polymerization is necessary for downstream ROS production. These results show that Siglec-8 signals through an unanticipated set of signaling molecules in IL-5-primed eosinophils to induce cell death and challenges the expectation that ITIM-bearing Siglecs signal through inhibitory pathways involving protein tyrosine phosphatases to achieve their downstream functions.
Collapse
Affiliation(s)
| | | | | | - Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
5
|
Verma A, Najahi-Missaoui W, Cummings BS, Somanath PR. Sterically stabilized liposomes targeting P21 (RAC1) activated kinase-1 and secreted phospholipase A 2 suppress prostate cancer growth and metastasis. Oncol Lett 2020; 20:179. [PMID: 32934746 PMCID: PMC7471734 DOI: 10.3892/ol.2020.12040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Metastatic prostate cancer (PCa) has a very high mortality rate in men, in Western countries and lacks reliable treatment. The advanced-stage PCa cells overexpress P21 (RAC1) activated kinase-1 (PAK1) and secreted phospholipase A2 (sPLA2) suggesting the potential utility of pharmacologically targeting these molecules to treat metastatic PCa. The small molecule, inhibitor targeting PAK1 activation-3 (IPA3) is a highly specific allosteric inhibitor of PAK1; however, it is metabolically unstable once in the plasma thus, limiting its utility as a chemotherapeutic agent. In the present study, the efficacy and specificity of IPA3 were combined with the stability and the sPLA2-targeted delivery method of two sterically stabilized liposomes [sterically stabilized long-circulating liposomes (SSL)-IPA3 and sPLA2 responsive liposomes (SPRL)-IPA3, respectively] to inhibit PCa growth and metastasis. It was found that twice-a-week administration of either SSL-IPA3 or SPRL-IPA3 for 3 weeks effectively suppressed the growth of PC-3 cell tumor xenografts implanted in athymic nude mice. Both drug formulations also inhibited the metastasis of intravenously administered murine RM1 PCa cells to the lungs of C57BL/6 mice. Whereas the twice-a-week administration of SSL-IPA3 significantly inhibited the spontaneous PCa metastasis to the lungs in Transgenic Adenocarcinoma of the Mouse Prostate mice, the administration of free IPA3 had no significant therapeutic benefit. The results present two novel IPA3 encapsulated liposomes to treat metastatic PCa.
Collapse
Affiliation(s)
- Arti Verma
- Program in Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Brian S. Cummings
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
- Interdisciplinary Toxicology Program, University of Georgia, Augusta, GA 30602, USA
| | - Payaningal R. Somanath
- Program in Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA
- Department of Medicine and Cancer Center, Augusta University, Augusta, GA 30602, USA
| |
Collapse
|
6
|
Zhang M, Fang X, Wang C, Hua Y, Huang C, Wang M, Zhu L, Wang Z, Gao Y, Zhang T, Liu H, Zhang Y, Lu S, Lu T, Chen Y, Li H. Design and synthesis of 1H-indazole-3-carboxamide derivatives as potent and selective PAK1 inhibitors with anti-tumour migration and invasion activities. Eur J Med Chem 2020; 203:112517. [DOI: 10.1016/j.ejmech.2020.112517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022]
|
7
|
Verma A, Artham S, Alwhaibi A, Adil MS, Cummings BS, Somanath PR. PAK1 inhibitor IPA-3 mitigates metastatic prostate cancer-induced bone remodeling. Biochem Pharmacol 2020; 177:113943. [PMID: 32240651 DOI: 10.1016/j.bcp.2020.113943] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Metastatic prostate cancer (PCa) has high mortality and a poor 5-year survival rate primarily due to the lack of effective treatments. Bone is the primary site of PCa metastasis in humans and the development of reliable therapeutic options for bone metastatic PCa will make a huge impact in reducing the mortality among these patients. Although P21 activated kinases (PAKs) have been studied in the past for their role in cancer, the efficacy of targeting PAKs to treat lung and bone metastatic PCa has not been tested yet. In the current study, we report that targeting PAK1 using IPA-3, an allosteric inhibitor of PAK1 kinase activity, significantly inhibits the murine metastatic PCa (RM1) cell proliferation and motility in vitro, and metastasis to the lungs in vivo. More importantly, we demonstrate for the first time that treatment with IPA-3 can blunt metastatic PCa-induced bone remodeling in vivo as analyzed by the 3-dimensional microcomputer tomography analysis. Our study has identified IPA-3 as a potential drug to treat bone metastatic PCa.
Collapse
Affiliation(s)
- Arti Verma
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Abdulrahman Alwhaibi
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Mir S Adil
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States; Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, United States.
| |
Collapse
|
8
|
Phosphorylation-dependent activity-based conformational changes in P21-activated kinase family members and screening of novel ATP competitive inhibitors. PLoS One 2019; 14:e0225132. [PMID: 31738805 PMCID: PMC6860928 DOI: 10.1371/journal.pone.0225132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/29/2019] [Indexed: 12/04/2022] Open
Abstract
P21-activated kinases (PAKs) are serine/threonine protein kinases that are subdivided into two groups on the basis of their domain architecture: group-I (PAK1–3) and group-II (PAK4–6). PAKs are considered as attractive drug targets that play vital role in cell proliferation, survival, motility, angiogenesis and cytoskeletal dynamics. In current study, molecular dynamics simulation-based comparative residual contributions and differential transitions were monitored in both active and inactive states of human PAK homologs for therapeutic intervention. Due to their involvement in cancer, infectious diseases, and neurological disorders, it is inevitable to develop novel therapeutic strategies that specifically target PAKs on the basis of their activity pattern. In order to isolate novel inhibitors that are able to bind at the active sites of PAK1 and PAK4, high throughput structure-based virtual screening was performed. Multiple lead compounds were proposed on the basis of their binding potential and targeting region either phosphorylated (active) or unphosphorylated PAK isoform (inactive). Thus, ATP-competitive inhibitors may prove ideal therapeutic choice against PAK family members. The detailed conformational readjustements occurring in the PAKs upon phosphorylation-dephosphorylation events may serve as starting point for devising novel drug molecules that are able to target on activity basis. Overall, the observations of current study may add valuable contribution in the inventory of novel inhibitors that may serve as attractive lead compounds for targeting PAK family members on the basis of activity-based conformational changes.
Collapse
|
9
|
Solution structures and biophysical analysis of full-length group A PAKs reveal they are monomeric and auto-inhibited in cis. Biochem J 2019; 476:1037-1051. [PMID: 30858169 PMCID: PMC6448136 DOI: 10.1042/bcj20180867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022]
Abstract
The group A p21-activated kinases (PAKs) exist in an auto-inhibited form until activated by GTPase binding and auto-phosphorylation. In the auto-inhibited form, a regulatory domain binds to the kinase domain (KD) blocking the binding of substrates, and CDC42 or Rac binding to the regulatory domain relieves this auto-inhibition allowing auto-phosphorylation on the KD activation loop. We have determined the crystal structure of the PAK3 catalytic domain and by small angle X-ray scattering, the solution-phase structures of full-length inactive PAK1 and PAK3. The structures reveal a compact but elongated molecular shape that demonstrates that, together with multiple independent biophysical measurements and in contrast with previous assumptions, group A PAKs are monomeric both before and after activation, consistent with an activation mechanism of cis-auto-inhibition and initial cis-auto-phosphorylation, followed by transient dimerisation to allow trans-auto-phosphorylation for full activation, yielding a monomeric active PAK protein.
Collapse
|
10
|
Biswal J, Jayaprakash P, Suresh Kumar R, Venkatraman G, Poopandi S, Rangasamy R, Jeyaraman J. Identification of Pak1 inhibitors using water thermodynamic analysis. J Biomol Struct Dyn 2019; 38:13-31. [PMID: 30661460 DOI: 10.1080/07391102.2019.1567393] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
p21-activated kinases (Paks) play an integral component in various cellular diverse processes. The full activation of Pak is dependent upon several serine residues present in the N-terminal region, a threonine present at the activation loop, and finally the phosphorylation of these residues ensure the complete activation of Pak1. The present study deals with the identification of novel potent candidates of Pak1 using computational methods as anti-cancer compounds. A diverse energy based pharmacophore (e-pharmacophore) was developed using four co-crystal inhibitors of Pak1 having pharmacophore features of 5 (DRDRR), 6 (DRHADR), and 7 (RRARDRP and DRRDADH) hypotheses. These models were used for rigorous screening against e-molecule database. The obtained hits were filtered using ADME/T and molecular docking to identify the high affinity binders. These hits were subjected to hierarchical clustering using dendritic fingerprint inorder to identify structurally diverse molecules. The diverse hits were scored against generated water maps to obtain WM/MM ΔG binding energy. Furthermore, molecular dynamics simulation and density functional theory calculations were performed on the final hits to understand the stability of the complexes. Five structurally diverse novel Pak1 inhibitors (4835785, 32198676, 32407813, 76038049, and 32945545) were obtained from virtual screening, water thermodynamics and WM/MM ΔG binding energy. All hits revealed similar mode of binding pattern with the hinge region residues replacing the unstable water molecules in the binding site. The obtained novel hits could be used as a platform to design potent drugs that could be experimentally tested against cancer patients having increased Pak1 expression.
Collapse
Affiliation(s)
- Jayashree Biswal
- Department of Bioinformatics, Science Block Alagappa University, Karaikudi Tamil Nadu, India
| | - Prajisha Jayaprakash
- Department of Bioinformatics, Science Block Alagappa University, Karaikudi Tamil Nadu, India
| | - Rayala Suresh Kumar
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Human Genetics College of Biomedical Sciences, Sri Ramachandra University, Porur, Chennai, Tamil Nadu, India
| | - Saritha Poopandi
- Department of Bioinformatics, Science Block Alagappa University, Karaikudi Tamil Nadu, India
| | - Raghu Rangasamy
- Department of Bioinformatics, Science Block Alagappa University, Karaikudi Tamil Nadu, India
| | - Jeyakanthan Jeyaraman
- Department of Bioinformatics, Science Block Alagappa University, Karaikudi Tamil Nadu, India
| |
Collapse
|
11
|
Kamal R, Kumar R, Kumar V, Kumar V, Bansal KK, Sharma PC. Synthesis, Anthelmintic and Antimicrobial Evaluation of New 2‐Arylidene‐1‐(4‐methyl‐6‐phenylpyrimidin‐2‐yl)hydrazines. ChemistrySelect 2019. [DOI: 10.1002/slct.201802822] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Raj Kamal
- Department of ChemistryKurukshetra University, Kurukshetra, Haryana India)- 136119
| | - Ravinder Kumar
- Department of ChemistryKurukshetra University, Kurukshetra, Haryana India)- 136119
| | - Vipan Kumar
- Department of ChemistryKurukshetra University, Kurukshetra, Haryana India)- 136119
| | - Vikas Kumar
- Department of BiotechnologyMaharishi Markandeshwar (Deemed to be University), Mullana, Haryana India)- 133207
| | - Kushal K. Bansal
- Department of Pharmaceutical SciencesKurukshetra University, Kurukshetra, Haryana India)- 136119
| | - Prabodh C. Sharma
- Department of Pharmaceutical SciencesKurukshetra University, Kurukshetra, Haryana India)- 136119
| |
Collapse
|
12
|
Degorce SL, Bodnarchuk MS, Cumming IA, Scott JS. Lowering Lipophilicity by Adding Carbon: One-Carbon Bridges of Morpholines and Piperazines. J Med Chem 2018; 61:8934-8943. [PMID: 30189136 DOI: 10.1021/acs.jmedchem.8b01148] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this article, we report our investigation of a phenomenon by which bridging morpholines across the ring with one-carbon tethers leads to a counterintuitive reduction in lipophilicity. This effect was also found to occur in piperazines and piperidines and lowered the measured log D7.4 of the bridged molecules by as much as -0.8 relative to their unbridged counterparts. As lowering lipophilicity without introducing additional heteroatoms can be desirable, we believe this potentially provides a useful tactic to improve the drug-like properties of molecules containing morpholine-, piperazine-, and piperidine-like motifs.
Collapse
Affiliation(s)
- Sébastien L Degorce
- Medicinal Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, Unit 310 Darwin Building , Cambridge CB4 0WG , United Kingdom
| | - Michael S Bodnarchuk
- Medicinal Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, Unit 310 Darwin Building , Cambridge CB4 0WG , United Kingdom
| | - Iain A Cumming
- Medicinal Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, Unit 310 Darwin Building , Cambridge CB4 0WG , United Kingdom
| | - James S Scott
- Medicinal Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, Unit 310 Darwin Building , Cambridge CB4 0WG , United Kingdom
| |
Collapse
|
13
|
|
14
|
Regulating Cdc42 and Its Signaling Pathways in Cancer: Small Molecules and MicroRNA as New Treatment Candidates. Molecules 2018; 23:molecules23040787. [PMID: 29596304 PMCID: PMC6017947 DOI: 10.3390/molecules23040787] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/19/2018] [Accepted: 03/24/2018] [Indexed: 12/13/2022] Open
Abstract
Despite great improvements in the diagnosis and treatment of neoplasms, metastatic disease is still the leading cause of death in cancer patients, with mortality rates still rising. Given this background, new ways to treat cancer will be important for development of improved cancer control strategies. Cdc42 is a member of the Rho GTPase family and plays an important role in cell-to-cell adhesion, formation of cytoskeletal structures, and cell cycle regulation. It thus influences cellular proliferation, transformation, and homeostasis, as well as the cellular migration and invasion processes underlying tumor formation. Cdc42 acts as a collection point for signal transduction and regulates multiple signaling pathways. Moreover, recent studies show that in most human cancers Cdc42 is abnormally expressed and promoting neoplastic growth and metastasis. Regarding possible new treatments for cancer, miRNA and small molecules targeting Cdc42 and related pathways have been recently found to be effective on cancer. In this review, we analyze the newly recognized regulation mechanisms for Cdc42 and Cdc42-related signal pathways, and particularly new treatments using small molecules and miRNAs to inhibit the abnormal overexpression of Cdc42 that may slow down the metastasis process, improve cancer therapy and lead to novel strategies for development of antineoplastic drugs.
Collapse
|
15
|
Abstract
Malignant carcinomas are often characterized by metastasis, the movement of carcinoma cells from a primary site to colonize distant organs. For metastasis to occur, carcinoma cells first must adopt a pro-migratory phenotype and move through the surrounding stroma towards a blood or lymphatic vessel. Currently, there are very limited possibilities to target these processes therapeutically. The family of Rho GTPases is an ubiquitously expressed division of GTP-binding proteins involved in the regulation of cytoskeletal dynamics and intracellular signaling. The best characterized members of the Rho family GTPases are RhoA, Rac1 and Cdc42. Abnormalities in Rho GTPase function have major consequences for cancer progression. Rho GTPase activation is driven by cell surface receptors that activate GTP exchange factors (GEFs) and GTPase-activating proteins (GAPs). In this review, we summarize our current knowledge on Rho GTPase function in the regulation of metastasis. We will focus on key discoveries in the regulation of epithelial-mesenchymal-transition (EMT), cell-cell junctions, formation of membrane protrusions, plasticity of cell migration and adaptation to a hypoxic environment. In addition, we will emphasize on crosstalk between Rho GTPase family members and other important oncogenic pathways, such as cyclic AMP-mediated signaling, canonical Wnt/β-catenin, Yes-associated protein (YAP) and hypoxia inducible factor 1α (Hif1α) and provide an overview of the advancements and challenges in developing pharmacological tools to target Rho GTPase and the aforementioned crosstalk in the context of cancer therapeutics.
Collapse
|
16
|
From bench (laboratory) to bed (hospital/home): How to explore effective natural and synthetic PAK1-blockers/longevity-promoters for cancer therapy. Eur J Med Chem 2017; 142:229-243. [PMID: 28814374 DOI: 10.1016/j.ejmech.2017.07.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022]
Abstract
PAK family kinases are RAC/CDC42-activated kinases that were first found in a soil amoeba 4 decades ago, and 2 decades later, were discovered in mammals as well. Since then at least 6 members of this family have been identified in mammals. One of them called PAK1 has been best studied so far, mainly because it is essential not only for malignant cell growth and metastasis, but also for many other diseases/disorders such as diabetes (type 2), AD (Alzheimer's disease), hypertension, and a variety of inflammatory or infectious diseases, which definitely shorten our lifespan. Moreover, PAK1-deficient mutant of C. elegans lives longer than the wild-type by 60%, clearly indicating that PAK1 is not only an oncogenic but also ageing kinase. Thus, in theory, both anti-oncogenic and longevity-promoting activities are among the "intrinsic" properties or criteria of "clinically useful" PAK1-blockers. There are a variety of PAK1-blocking natural products such as propolis and curcumin which indeed extend the healthy lifespan of small animals such as C. elegans by inducing the autophagy. Recently, we managed to synthesize a series of potent water-soluble and highly cell-permeable triazolyl esters of COOH-bearing PAK1-blockers such as Ketorolac, ARC (artepillin C) and CA (caffeic acid) via "Click Chemistry" that boosts their anti-cancer activity over 500-fold, mainly by increasing their cell-permeability, and one of them called 15K indeed extends the lifespan of C. elegans. In this mini-review we shall discuss both synthetic and natural PAK1-blockers, some of which would be potentially useful for cancer therapy with least side effect (rather promoting the longevity as well).
Collapse
|
17
|
Abstract
p21-Activated kinase 1 (PAK1) has attracted much attention as a potential therapeutic target due to its central role in many oncogenic signaling pathways, its frequent dysregulation in cancers and neurological disorders, and its tractability as a target for small-molecule inhibition. To date, several PAK1-targeting compounds have been developed as preclinical agents, including one that has been evaluated in a clinical trial. A series of ATP-competitive inhibitors, allosteric inhibitors and peptide inhibitors with distinct biochemical and pharmacokinetic properties represent useful laboratory tools for studies on the role of PAK1 in biology and in disease contexts, and could lead to promising therapeutic agents. Given the central role of PAK1 in vital signaling pathways, future clinical development of PAK1 inhibitors will require careful investigation of their safety and efficacy.
Collapse
|