1
|
Zhang X, Zhang M, Li Y, Deng P. Identification of Potential Selective PAK4 Inhibitors Through Shape and Protein Conformation Ensemble Screening and Electrostatic-Surface-Matching Optimization. Curr Issues Mol Biol 2025; 47:29. [PMID: 39852144 PMCID: PMC11764389 DOI: 10.3390/cimb47010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
P21-activated kinase 4 (PAK4) plays a crucial role in the proliferation and metastasis of various cancers. However, developing selective PAK4 inhibitors remains challenging due to the high homology within the PAK family. Therefore, developing highly selective PAK4 inhibitors is critical to overcoming the limitations of existing inhibitors. We analyzed the structural differences in the binding pockets of PAK1 and PAK4 by combining cross-docking and molecular dynamics simulations to identify key binding regions and unique structural features of PAK4. We then performed screening using shape and protein conformation ensembles, followed by a re-evaluation of the docking results with deep-learning-driven GNINA to identify the candidate molecule, STOCK7S-56165. Based on this, we applied a fragment-replacement strategy under electrostatic-surface-matching conditions to obtain Compd 26. This optimization significantly improved electrostatic interactions and reduced binding energy, highlighting its potential for selectivity. Our findings provide a novel approach for developing selective PAK4 inhibitors and lay the theoretical foundation for future anticancer drug design.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (X.Z.); (M.Z.); (Y.L.)
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
| | - Meile Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (X.Z.); (M.Z.); (Y.L.)
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
| | - Yihao Li
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (X.Z.); (M.Z.); (Y.L.)
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
| | - Ping Deng
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (X.Z.); (M.Z.); (Y.L.)
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
- Chongqing Key Research Laboratory for Quality Evaluation and Safety Research of APIs, Chongqing 400016, China
| |
Collapse
|
2
|
Ma J, Zhu W, Wang Y, Du H, Ma L, Liu L, Niu C, Li S, Zhang K, Yuan E. Comprehensive multi-omics analysis identifies NUSAP1 as a potential prognostic and immunotherapeutic marker for lung adenocarcinoma. Int J Med Sci 2025; 22:328-340. [PMID: 39781524 PMCID: PMC11704694 DOI: 10.7150/ijms.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/22/2024] [Indexed: 01/12/2025] Open
Abstract
While NUSAP1's association with various tumors is established, its predictive value for prognosis and immunotherapy in lung adenocarcinoma (LUAD) remains unconfirmed. We analyzed Nucleolar Spindle-Associated Protein 1 (NUSAP1) gene expression in TCGA and GTEx datasets and validated it in clinicopathological tissues using qRT-PCR and immunohistochemistry. Additionally, we investigated NUSAP1's relationship with patient prognosis across TCGA and five GEO cohorts. The IMvigor210 cohort was utilized to explore NUSAP1's association with immunotherapy efficacy. Furthermore, single-cell RNA-sequencing data was used to examine the correlation between NUSAP1 and immune cell infiltration. Finally, we analyzed the relationship between NUSAP1 and m6A methylation. NUSAP1 expression was significantly elevated in tumor tissues, correlating with poorer prognosis in LUAD patients. It exhibited a significant correlation with immune cell infiltration in the tumor microenvironment, predominantly expressed in Tprolif cells. LUAD patients with heightened NUSAP1 expression may derive greater benefit from anti-PD-L1 treatment. Additionally, NUSAP1 was tightly linked with m6A methylation. Enrichment analysis revealed its association with key biological functions, including lipid metabolism and cell cycle regulation. Our comprehensive analysis underscores NUSAP1's potential as a prognostic and immunotherapeutic biomarker for LUAD, warranting further investigation.
Collapse
Affiliation(s)
- Jun Ma
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Wenjing Zhu
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Yuan Wang
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Hongmei Du
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Ling Ma
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Lisha Liu
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Chao Niu
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Songlei Li
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Kai Zhang
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| | - Enwu Yuan
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for In vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Wang Y, Minden A. Inhibition of NAMPT by PAK4 Inhibitors. Int J Mol Sci 2024; 25:10138. [PMID: 39337621 PMCID: PMC11431865 DOI: 10.3390/ijms251810138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The serine/threonine kinase PAK4 plays a crucial role in regulating cell proliferation, survival, migration, and invasion. Overexpression of PAK4 correlates with poor prognosis in some cancers. KPT-9274, a PAK4 inhibitor, significantly reduces the growth of triple-negative breast cancer cells and mammary tumors in mouse models, and it also inhibits the growth of several other types of cancer cells. Interestingly, although it was first identified as a PAK4 inhibitor, KPT-9274 was also found to inhibit the enzyme NAMPT (nicotinamide phosphoribosyltransferase), which is crucial for NAD (nicotinamide adenine dinucleotide) synthesis and vital for cellular energy and growth. These results made us question whether growth inhibition in response to KPT-9274 was due to PAK4 inhibition, NAMPT inhibition, or both. To address this, we tested several other PAK4 inhibitors that also inhibit cell growth, to determine whether they also inhibit NAMPT activity. Our findings confirm that multiple PAK4 inhibitors also inhibit NAMPT activity. This was assessed both in cell-free assays and in a breast cancer cell line. Molecular docking studies were also used to help us better understand the mechanism by which PAK4 inhibitors block PAK4 and NAMPT activity, and we identified specific residues on the PAK4 inhibitors that interact with NAMPT and PAK4. Our results suggest that PAK4 inhibitors may have a more complex mechanism of action than previously understood, necessitating further exploration of how they influence cancer cell growth.
Collapse
Affiliation(s)
- Yiling Wang
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
4
|
Ozgencil F, Gunindi HB, Eren G. Dual-targeted NAMPT inhibitors as a progressive strategy for cancer therapy. Bioorg Chem 2024; 149:107509. [PMID: 38824699 DOI: 10.1016/j.bioorg.2024.107509] [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: 03/09/2024] [Revised: 04/29/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024]
Abstract
In mammals, nicotinamide phosphoribosyltransferase (NAMPT) is a crucial enzyme in the nicotinamide adenine dinucleotide (NAD+) synthesis pathway catalyzing the condensation of nicotinamide (NAM) with 5-phosphoribosyl-1-pyrophosphate (PRPP) to produce nicotinamide mononucleotide (NMN). Given the pivotal role of NAD+ in a range of cellular functions, including DNA synthesis, redox reactions, cytokine generation, metabolism, and aging, NAMPT has become a promising target for many diseases, notably cancer. Therefore, various NAMPT inhibitors have been reported and classified as first and second-generation based on their chemical structures and design strategies, dual-targeted being one. However, most NAMPT inhibitors suffer from several limitations, such as dose-dependent toxicity and poor pharmacokinetic properties. Consequently, there is no clinically approved NAMPT inhibitor. Hence, research on discovering more effective and less toxic dual-targeted NAMPT inhibitors with desirable pharmacokinetic properties has drawn attention recently. This review summarizes the previously reported dual-targeted NAMPT inhibitors, focusing on their design strategies and advantages over the single-targeted therapies.
Collapse
Affiliation(s)
- Fikriye Ozgencil
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye
| | - Habibe Beyza Gunindi
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye
| | - Gokcen Eren
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye.
| |
Collapse
|
5
|
Shi X, Xiao B, Feng R. Identification of a glycolysis-related miRNA Signature for Predicting Breast cancer Survival. Mol Biotechnol 2024; 66:1988-2006. [PMID: 37535159 DOI: 10.1007/s12033-023-00837-5] [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: 02/26/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Breast cancer (BC) is a common type of cancer and has a poor prognosis. In this study, we collected the mRNA and miRNA expression profiles of BC patients were obtained from The Cancer Genome Atlas (TCGA) to explore a novel prognostic strategy for BC patients using bioinformatics tools. We found that six glycolysis-related miRNAs (GRmiRs, including hsa-mir-1247, hsa-mir148b, hsa-mir-133a-2, has-mir-1307, hsa-mir-195 and hsa-mir-1258) were correlated with prognosis of BC samples. The risk score model was established based on 6 prognosis-associated GRmiRs. The outcome of high risk group was significantly poorer. Cox regression analysis showed that risk score was an independent prognostic factor. Differentially expressed genes identified between high and low risk groups were mainly enriched in inflammation and immune-related signaling pathways. The proportion of infiltration of 12 kinds of immune cells in high and low risk groups were significantly different. Risk score was closely associated with many immune indexes. Multiple DEGRGs and miRNAs were associated with drugs. In conclusion, glycolysis-related miRNA signature effectively predicts BC prognosis.
Collapse
Affiliation(s)
- Xuejing Shi
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China
| | - Baoqiang Xiao
- Department of General Surgery, Tianjin Hospital, Tianjin, Hexi District, 300211, P.R. China
| | - Rui Feng
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China.
| |
Collapse
|
6
|
Xu S, Ma B, Jian Y, Yao C, Wang Z, Fan Y, Ma J, Chen Y, Feng X, An J, Chen J, Wang K, Xie H, Gao Y, Li L. Development of a PAK4-targeting PROTAC for renal carcinoma therapy: concurrent inhibition of cancer cell proliferation and enhancement of immune cell response. EBioMedicine 2024; 104:105162. [PMID: 38810561 PMCID: PMC11154127 DOI: 10.1016/j.ebiom.2024.105162] [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: 10/10/2023] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Finding the oncogene, which was able to inhibit tumor cells intrinsically and improve the immune answers, will be the future direction for renal cancer combined treatment. Following patient sample analysis and signaling pathway examination, we propose p21-activated kinase 4 (PAK4) as a potential target drug for kidney cancer. PAK4 exhibits high expression levels in patient samples and plays a regulatory role in the immune microenvironment. METHODS Utilizing AI software for peptide drug design, we have engineered a specialized peptide proteolysis targeting chimera (PROTAC) drug with selectivity for PAK4. To address challenges related to drug delivery, we developed a nano-selenium delivery system for efficient transport of the peptide PROTAC drug, termed PpD (PAK4 peptide degrader). FINDINGS We successfully designed a peptide PROTAC drug targeting PAK4. PpD effectively degraded PAK4 with high selectivity, avoiding interference with other homologous proteins. PpD significantly attenuated renal carcinoma proliferation in vitro and in vivo. Notably, PpD demonstrated a significant inhibitory effect on tumor proliferation in a fully immunocompetent mouse model, concomitantly enhancing the immune cell response. Moreover, PpD demonstrated promising tumor growth inhibitory effects in mini-PDX and PDO models, further underscoring its potential for clinical application. INTERPRETATION This PAK4-targeting peptide PROTAC drug not only curtails renal cancer cell proliferation but also improves the immune microenvironment and enhances immune response. Our study paves the way for innovative targeted therapies in the management of renal cancer. FUNDING This work is supported by Research grants from non-profit organizations, as stated in the Acknowledgments.
Collapse
Affiliation(s)
- Shan Xu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Bohan Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Yanlin Jian
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Chen Yao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Zixi Wang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Yizeng Fan
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Jian Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Xiaoyu Feng
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Jiale An
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Jiani Chen
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Hongjun Xie
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Yang Gao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, China.
| |
Collapse
|
7
|
Tang L, Ye H, Chen L, Dong W, Hu X, Yu L. Tumorigenic role of Pak4 in ovarian cancer and its correlation with immune infiltration. BMC Med Genomics 2024; 17:148. [PMID: 38807162 PMCID: PMC11134728 DOI: 10.1186/s12920-024-01917-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Ovarian cancer is the most common cause of gynecological cancer death. Pak4 has been proved to be tumorigenic in many types of cancers, but its role in ovarian cancer is still not clarified. METHODS In this study, we used immunohistochemistry to investigate into Pak4 expression in different histological types of ovarian cancer. TIMER, TISCH2, GEPIA, ualcan, KM plotter, GSCA and GeneMANIA were used to identify the prognostic roles and gene regulation networks of Pak4 in ovarian cancer. Immune infiltration levels were investigated using TIMER database. RESULTS Pak4 was highly expressed in ovarian cancers, regardless of different FIGO stages and histological grades. Single cell sequencing database proved that Pak4 was highly expressed in malignant ovarian cancer cells. Pak4 level was significantly correlated with different histological types of ovarian cancer. Pak4 expression was negatively connected with OS and PFS of ovarian cancer patients. Functions of Pak4 and its interacted genes were mainly involved in protein serine/threonine kinase activity, regulation of actin filament-based process and regulation of cytoskeleton organization. Pak4 level was negatively correlated with immune biomarkers of B cell infiltration (p = 2.39e-05), CD8 + T cell infiltration (p = 1.51e-04), neutrophil (p = 1.74e-06) and dendritic cell (p = 4.41e-08). Close correlation was found between Pak4 expression and T cell exhaustion (p < 0.05). CONCLUSIONS Our results demonstrated the expression level, gene interaction networks and immune infiltration levels of Pak4 in ovarian cancer. And the results revealed role of Pak4 in tumorigenesis and the possibility to be a potential immunotherapeutic target.
Collapse
Affiliation(s)
- Lan Tang
- Department of gynecology, The First College of Medical School, Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei, China
| | - Hong Ye
- Department of gynecology, The First College of Medical School, Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei, China
| | - Li Chen
- Department of gynecology, The First College of Medical School, Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei, China
| | - Weiwei Dong
- Department of gynecology, The First College of Medical School, Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei, China
| | - Xingyan Hu
- Department of gynecology, The First College of Medical School, Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei, China
| | - Lan Yu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Maddeboina K, Yada B, Kumari S, McHale C, Pal D, Durden DL. Recent advances in multitarget-directed ligands via in silico drug discovery. Drug Discov Today 2024; 29:103904. [PMID: 38280625 DOI: 10.1016/j.drudis.2024.103904] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
To combat multifactorial refractory diseases, such as cancer, cardiovascular, and neurodegenerative diseases, multitarget drugs have become an emerging area of research aimed at 'synthetic lethality' (SL) relationships associated with drug-resistance mechanisms. In this review, we discuss the in silico design of dual and triple-targeted ligands, strategies by which specific 'warhead' groups are incorporated into a parent compound or scaffold with primary inhibitory activity against one target to develop one small molecule that inhibits two or three molecular targets in an effort to increase potency against multifactorial diseases. We also discuss the analytical exploration of structure-activity relationships (SARs), physicochemical properties, polypharmacology, scaffold feature extraction of US Food and Drug Administration (FDA)-approved multikinase inhibitors (MKIs), and updates regarding the clinical status of dual-targeted chemotypes.
Collapse
Affiliation(s)
- Krishnaiah Maddeboina
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute/Atrium Health, Charlotte, NC 28204, USA; Department of Biochemistry, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA.
| | - Bharath Yada
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute/Atrium Health, Charlotte, NC 28204, USA
| | - Shikha Kumari
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, CT 06520, USA
| | - Cody McHale
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute/Atrium Health, Charlotte, NC 28204, USA
| | - Dhananjaya Pal
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute/Atrium Health, Charlotte, NC 28204, USA
| | - Donald L Durden
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute/Atrium Health, Charlotte, NC 28204, USA; Department of Biochemistry, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA.
| |
Collapse
|
9
|
Han W, Yang Y, Yu F, Li Q, Liu A, Xu W, Li J, Xue X. Design, synthesis and anticancer activity evaluation of 4-(3-1H-indazolyl)amino quinazoline derivatives as PAK4 inhibitors. Bioorg Med Chem 2023; 95:117501. [PMID: 37864885 DOI: 10.1016/j.bmc.2023.117501] [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: 08/21/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023]
Abstract
A novel series of 4-(3-1H-indazolyl)amino quinazoline derivatives were developed as PAK4 inhibitors based on a scaffold hopping strategy. Compounds 27e, 27g, 27i and 27j were found to exhibit potent inhibitory activity against PAK4 (IC50 = 10, 13, 11 and 9 nM, respectively). Subsequent cellular assay demonstrated that compound 27e possessed the strongest antiproliferative activity against A549 cells with an IC50 value of 0.61 μM, a little bit better than PF-3758309. Further anticancer mechanistic investigation revealed that compound 27e significantly induced apoptosis of A549 cells in a concentration-dependent manner and blocked the cell cycle at phase G0/G1. A docking model between compound 27e and PAK4 was proposed to elucidate its possible binding modes. As a promising PAK4 inhibitor, compound 27e may serve as a candidate for the development of novel PAK4-targeted anticancer drug.
Collapse
Affiliation(s)
- Wei Han
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yusang Yang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Yu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing 211198, China
| | - Qianqian Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Anyao Liu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenbo Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jiabin Li
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaowen Xue
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
10
|
Green JR, Mahalingaiah PKS, Gopalakrishnan SM, Liguori MJ, Mittelstadt SW, Blomme EAG, Van Vleet TR. Off-target pharmacological activity at various kinases: Potential functional and pathological side effects. J Pharmacol Toxicol Methods 2023; 123:107468. [PMID: 37553032 DOI: 10.1016/j.vascn.2023.107468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
In drug discovery, during the lead optimization and candidate characterization stages, novel small molecules are frequently evaluated in a battery of in vitro pharmacology assays to identify potential unintended, off-target interactions with various receptors, transporters, ion channels, and enzymes, including kinases. Furthermore, these screening panels may also provide utility at later stages of development to provide a mechanistic understanding of unexpected safety findings. Here, we present a compendium of the most likely functional and pathological outcomes associated with interaction(s) to a panel of 95 kinases based on an extensive curation of the scientific literature. This panel of kinases was designed by AbbVie based on safety-related data extracted from the literature, as well as from over 20 years of institutional knowledge generated from discovery efforts. For each kinase, the scientific literature was reviewed using online databases and the most often reported functional and pathological effects were summarized. This work should serve as a practical guide for small molecule drug discovery scientists and clinical investigators to predict and/or interpret adverse effects related to pharmacological interactions with these kinases.
Collapse
Affiliation(s)
- Jonathon R Green
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States.
| | | | - Sujatha M Gopalakrishnan
- Drug Discovery Science and Technology, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Michael J Liguori
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Scott W Mittelstadt
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Eric A G Blomme
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Terry R Van Vleet
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| |
Collapse
|
11
|
Tang H, Wang L, Wang T, Yang J, Zheng S, Tong J, Jiang S, Zhang X, Zhang K. Recent advances of targeting nicotinamide phosphoribosyltransferase (NAMPT) for cancer drug discovery. Eur J Med Chem 2023; 258:115607. [PMID: 37413882 DOI: 10.1016/j.ejmech.2023.115607] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme for the biosynthesis of NAD+ in the salvage pathway. NAMPT is overexpressed in various cancers, associating with a poor prognosis and tumor progression. Beyond cancer metabolism, recent evidence unravels additional roles of NAMPT in cancer biology, including DNA repair machinery, crosstalk with oncogenic signaling pathways, cancer cell stemness, and immune responses. NAMPT is a promising therapeutic target for cancer. However, first-generation NAMPT inhibitors exhibited limited efficacy and dose-limiting toxicities in clinical trials. Multiple strategies are being exploited to improve their efficacy and minimize toxic-side effects. This review discusses the biomarkers predictive of response to NAMPT inhibitors, and summarizes the most significant advances in the evolution of structurally distinct NAMPT inhibitors, the manipulation of targeted delivery technologies via antibody-drug conjugates (ADCs), PhotoActivated ChemoTherapy (PACT) and the intratumoral delivery system, as well as the development and pharmacological outcomes of NAMPT degraders. Finally, a discussion of future perspectives and challenges in this area is also included.
Collapse
Affiliation(s)
- He Tang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lin Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianyu Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiamei Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuai Zheng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jun Tong
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangyu Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kuojun Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
12
|
Zhang L, Lin S, Zhang Z, Yan C, Liu F. The role of p21-activated kinase 4 in the progression of oral squamous cell carcinoma by targeting PI3K-AKT signaling pathway. Clin Transl Oncol 2023; 25:739-747. [PMID: 36593383 DOI: 10.1007/s12094-022-02980-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/09/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Oral squamous carcinoma (OSCC), the most common head and neck malignancy, has a strong propensity for malignant proliferation and metastasis, which will decrease the survival of patients. P21-activated kinase 4 (PAK4), a classical serine/threonine protein kinase with multiple cellular functions, has an essential role in cancer cell migration and invasion. Here, we elucidated the function and possible molecular mechanisms of the effect of PAK4 on the biological behaviors of OSCC. METHODS The expression of genes and protein was detected by real-time PCR and western blotting. We used oral squamous carcinoma cell lines, Tca8117, Cal 27, SCC 4, and SCC 9 for validation of our cell function data. Flow cytometry, 3D cultures, and clone formation assay were used to detect proliferation of cells. RNA sequencing and bioinformatic analysis was performed to determine the potential function of PAK4. RESULTS Immunohistochemistry, western blotting and real-time PCR demonstrated that PAK4 expression was up-regulated in OSCC tissues. Overexpression of PAK4 promoted the proliferation, migration and invasion of OSCC cell lines. RNA sequencing (RNA-seq) for the transcriptome-wide analysis of differential gene expression followed by bioinformatic analysis was performed to determine the potential function of PAK4. Based on the KEGG enrichment analysis and GO analysis of differential expression genes (DEGs) we found that PAK4 promotes the cell-cycle machinery, which associated with 44 regulated genes, thereby promoting cancer cell differentiation. CONCLUSIONS This study demonstrates that the PAK4 regulates the biological behaviors of OSCC by PI3K-AKT signaling pathway, and these findings might provide a novel strategy for OSCC treatment.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China.,Nosocomial Infection Management Office, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Shanfeng Lin
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Zeying Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China.,Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Cong Yan
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Fayu Liu
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China.
| |
Collapse
|
13
|
Tang L, Gao Y, Li T. Pan-cancer analysis identifies the immunological and prognostic role of PAK4. Life Sci 2023; 312:121263. [PMID: 36470541 DOI: 10.1016/j.lfs.2022.121263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
AIMS P21-activated kinase 4 (PAK4) belongs to the wider family of Serine/Threonine p21-activated kinases (PAKs) and functions as a hub for signaling pathways in cancer progression. Numerous studies have indicated the significance of PAK4 for tumorigenesis, but no systematic pan-cancer analysis has been performed. MAIN METHODS The current study aimed to investigate the prognostic and immunological functions of PAK4 through bioinformatic analysis of datasets from The Cancer Genome Atlas, UALCAN, GEPIA2, cBioPortal, TIMER2, and Human Protein Atlas. PAK4 expression was correlated with prognosis, DNA methylation, tumor mutational burden, microsatellite instability, and immune cell infiltration. KEY FINDINGS PAK4 was highly expressed in various cancers but showed decreased expression in colon adenocarcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, and thyroid carcinoma. PAK4 was found to have a positive or negative correlation with prognosis of different cancers. PAK4 expression was related to tumor mutational burden in 11 tumor types, and associated with microsatellite instability in 10 tumor types and was correlated with immune infiltration and immune checkpoint genes. SIGNIFICANCE PAK4 could be considered as a prognostic and immunotherapeutic marker for some types of malignant tumor.
Collapse
Affiliation(s)
- Lina Tang
- Advanced Medical Research Center of Zhengzhou University, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China.
| | - Yunling Gao
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, China
| | - Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, China
| |
Collapse
|
14
|
Chetty AK, Ha BH, Boggon TJ. Rho family GTPase signaling through type II p21-activated kinases. Cell Mol Life Sci 2022; 79:598. [PMID: 36401658 PMCID: PMC10105373 DOI: 10.1007/s00018-022-04618-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/07/2022] [Accepted: 10/28/2022] [Indexed: 11/21/2022]
Abstract
Signaling from the Rho family small GTPases controls a wide range of signaling outcomes. Key among the downstream effectors for many of the Rho GTPases are the p21-activated kinases, or PAK group. The PAK family comprises two types, the type I PAKs (PAK1, 2 and 3) and the type II PAKs (PAK4, 5 and 6), which have distinct structures and mechanisms of regulation. In this review, we discuss signal transduction from Rho GTPases with a focus on the type II PAKs. We discuss the role of PAKs in signal transduction pathways and selectivity of Rho GTPases for PAK family members. We consider the less well studied of the Rho GTPases and their PAK-related signaling. We then discuss the molecular basis for kinase domain recognition of substrates and for regulation of signaling. We conclude with a discussion of the role of PAKs in cross talk between Rho family small GTPases and the roles of PAKs in disease.
Collapse
Affiliation(s)
- Ashwin K Chetty
- Yale College, New Haven, CT, 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Byung Hak Ha
- Department of Pharmacology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Titus J Boggon
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Cancer Center, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
15
|
Mozibullah M, Junaid M. Biological Role of the PAK4 Signaling Pathway: A Prospective Therapeutic Target for Multivarious Cancers. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
16
|
Abril-Rodriguez G, Torrejon DY, Karin D, Campbell KM, Medina E, Saco JD, Galvez M, Champhekar AS, Perez-Garcilazo I, Baselga-Carretero I, Singh J, Comin-Anduix B, Puig-Saus C, Ribas A. Remodeling of the tumor microenvironment through PAK4 inhibition sensitizes tumors to immune checkpoint blockade. CANCER RESEARCH COMMUNICATIONS 2022; 2:1214-1228. [PMID: 36588582 PMCID: PMC9799984 DOI: 10.1158/2767-9764.crc-21-0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/14/2022] [Accepted: 09/12/2022] [Indexed: 02/01/2023]
Abstract
PAK4 inhibition can sensitize tumors to immune checkpoint blockade (ICB) therapy, however, the underlying mechanisms remain unclear. We report that PAK4 inhibition reverses immune cell exclusion by increasing the infiltration of CD8 T cells and CD103+ dendritic cells (DCs), a specific type of DCs that excel at cross-presenting tumor antigens and constitute a source of CXCL10. Interestingly, in melanoma clinical datasets, PAK4 expression levels negatively correlate with the presence of CCL21, the ligand for CCR7 expressed in CD103+ DCs. Furthermore, we extensively characterized the transcriptome of PAK4 knock out (KO) tumors, in vitro and in vivo, and established the importance of PAK4 expression in the regulation of the extracellular matrix, which can facilitate immune cell infiltration. Comparison between PAK4 wild type (WT) and KO anti-PD-1 treated tumors revealed how PAK4 deletion sensitizes tumors to ICB from a transcriptomic perspective. In addition, we validated genetically and pharmacologically that inhibition of PAK4 kinase activity is sufficient to improve anti-tumor efficacy of anti-PD-1 blockade in multiple melanoma mouse models. Therefore, this study provides novel insights into the mechanism of action of PAK4 inhibition and provides the foundation for a new treatment strategy that aims to overcome resistance to PD-1 blockade by combining anti-PD-1 with a small molecule PAK4 kinase inhibitor.
Collapse
Affiliation(s)
- Gabriel Abril-Rodriguez
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | - Davis Y. Torrejon
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Daniel Karin
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | - Katie M. Campbell
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Egmidio Medina
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Justin D. Saco
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Mildred Galvez
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Ameya S. Champhekar
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Ivan Perez-Garcilazo
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Ignacio Baselga-Carretero
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| | - Jas Singh
- Arcus Biosciences, Inc., Hayward, California
| | - Begoña Comin-Anduix
- Department of Surgery, Division of Surgical Oncology, UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Cristina Puig-Saus
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Department of Surgery, Division of Surgical Oncology, UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| |
Collapse
|
17
|
Wei Y, Xiang H, Zhang W. Review of various NAMPT inhibitors for the treatment of cancer. Front Pharmacol 2022; 13:970553. [PMID: 36160449 PMCID: PMC9490061 DOI: 10.3389/fphar.2022.970553] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the NAD salvage pathway of mammalian cells and is overexpressed in numerous types of cancers. These include breast cancer, ovarian cancer, prostate cancer, gastric cancer, colorectal cancer, glioma, and b-cell lymphoma. NAMPT is also known to impact the NAD and NADPH pool. Research has demonstrated that NAMPT can be inhibited. NAMPT inhibitors are diverse anticancer medicines with significant anti-tumor efficacy in ex vivo tumor models. A few notable NAMPT specific inhibitors which have been produced include FK866, CHS828, and OT-82. Despite encouraging preclinical evidence of the potential utility of NAMPT inhibitors in cancer models, early clinical trials have yielded only modest results, necessitating the adaptation of additional tactics to boost efficacy. This paper examines a number of cancer treatment methods which target NAMPT, including the usage of individual inhibitors, pharmacological combinations, dual inhibitors, and ADCs, all of which have demonstrated promising experimental or clinical results. We intend to contribute further ideas regarding the usage and development of NAMPT inhibitors in clinical therapy to advance the field of research on this intriguing target.
Collapse
Affiliation(s)
- Yichen Wei
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Haotian Xiang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Wenqiu Zhang,
| |
Collapse
|
18
|
NAD/NAMPT and mTOR Pathways in Melanoma: Drivers of Drug Resistance and Prospective Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23179985. [PMID: 36077374 PMCID: PMC9456568 DOI: 10.3390/ijms23179985] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma represents the most fatal skin cancer due to its aggressive behavior and high metastatic potential. The introduction of BRAF/MEK inhibitors and immune-checkpoint inhibitors (ICIs) in the clinic has dramatically improved patient survival over the last decade. However, many patients either display primary (i.e., innate) or develop secondary (i.e., acquired) resistance to systemic treatments. Therapeutic resistance relies on the rewiring of multiple processes, including cancer metabolism, epigenetics, gene expression, and interactions with the tumor microenvironment that are only partially understood. Therefore, reliable biomarkers of resistance or response, capable of facilitating the choice of the best treatment option for each patient, are currently missing. Recently, activation of nicotinamide adenine dinucleotide (NAD) metabolism and, in particular, of its rate-limiting enzyme nicotinamide phosphoribosyltransferase (NAMPT) have been identified as key drivers of targeted therapy resistance and melanoma progression. Another major player in this context is the mammalian target of rapamycin (mTOR) pathway, which plays key roles in the regulation of melanoma cell anabolic functions and energy metabolism at the switch between sensitivity and resistance to targeted therapy. In this review, we summarize known resistance mechanisms to ICIs and targeted therapy, focusing on metabolic adaptation as one main mechanism of drug resistance. In particular, we highlight the roles of NAD/NAMPT and mTOR signaling axes in this context and overview data in support of their inhibition as a promising strategy to overcome treatment resistance.
Collapse
|
19
|
Li Y, Lu Q, Xie C, Yu Y, Zhang A. Recent advances on development of p21-activated kinase 4 inhibitors as anti-tumor agents. Front Pharmacol 2022; 13:956220. [PMID: 36105226 PMCID: PMC9465411 DOI: 10.3389/fphar.2022.956220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
The p21-activated kinase 4 (PAK4) is a member of the PAKs family. It is overexpressed in multiple tumor tissues. Pharmacological inhibition of PAK4 attenuates proliferation, migration, and invasion of cancer cells. Recent studies revealed that inhibition of PAK4 sensitizes immunotherapy which has been extensively exploited as a new strategy to treat cancer. In the past few years, a large number of PAK4 inhibitors have been reported. Of note, the allosteric inhibitor KPT-9274 has been tested in phase Ⅰ clinic trials. Herein, we provide an update on recent research progress on the PAK4 mediated signaling pathway and highlight the development of the PAK4 small molecular inhibitors in recent 5 years. Meanwhile, challenges, limitations, and future developmental directions will be discussed as well.
Collapse
Affiliation(s)
- Yang Li
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Lu
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Chenghu Xie
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yiming Yu
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ao Zhang
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Ao Zhang,
| |
Collapse
|
20
|
Ma Y, Nikfarjam M, He H. The trilogy of P21 activated kinase, autophagy and immune evasion in pancreatic ductal adenocarcinoma. Cancer Lett 2022; 548:215868. [PMID: 36027997 DOI: 10.1016/j.canlet.2022.215868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/22/2022] [Accepted: 08/06/2022] [Indexed: 11/02/2022]
Abstract
Pancreatic Ductal Adenocarcinoma (PDA) is one of the most lethal types of cancer with a dismal prognosis. KRAS mutation is a commonly identified oncogene in PDA tumorigenesis and P21-activated kinases (PAKs) are its downstream mediator. While PAK1 is more well-studied, PAK4 also attracted increasing interest. In PDA, PAK inhibition not only reduces cancer cell viability but also sensitises it to chemotherapy. While PDA remains resistant to existing immunotherapies, PAK inhibition has been shown to increase cancer immunogenicity of melanoma, glioblastoma and PDA. Furthermore, autophagy plays an important role in PDA immune evasion, and accumulating evidence has pointed to a connection between PAK and cancer cell autophagy. In this literature review, we aim to summarize currently available studies that have assessed the potential connection between PAK, autophagy and immune evasion in PDA biology to guide future research.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC, 3084, Australia; Department of Hepatopancreatic-Biliary Surgery, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
21
|
Navas LE, Carnero A. Nicotinamide Adenine Dinucleotide (NAD) Metabolism as a Relevant Target in Cancer. Cells 2022; 11:cells11172627. [PMID: 36078035 PMCID: PMC9454445 DOI: 10.3390/cells11172627] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/25/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
NAD+ is an important metabolite in cell homeostasis that acts as an essential cofactor in oxidation–reduction (redox) reactions in various energy production processes, such as the Krebs cycle, fatty acid oxidation, glycolysis and serine biosynthesis. Furthermore, high NAD+ levels are required since they also participate in many other nonredox molecular processes, such as DNA repair, posttranslational modifications, cell signalling, senescence, inflammatory responses and apoptosis. In these nonredox reactions, NAD+ is an ADP-ribose donor for enzymes such as sirtuins (SIRTs), poly-(ADP-ribose) polymerases (PARPs) and cyclic ADP-ribose (cADPRs). Therefore, to meet both redox and nonredox NAD+ demands, tumour cells must maintain high NAD+ levels, enhancing their synthesis mainly through the salvage pathway. NAMPT, the rate-limiting enzyme of this pathway, has been identified as an oncogene in some cancer types. Thus, NAMPT has been proposed as a suitable target for cancer therapy. NAMPT inhibition causes the depletion of NAD+ content in the cell, leading to the inhibition of ATP synthesis. This effect can cause a decrease in tumour cell proliferation and cell death, mainly by apoptosis. Therefore, in recent years, many specific inhibitors of NAMPT have been developed, and some of them are currently in clinical trials. Here we review the NAD metabolism as a cancer therapy target.
Collapse
Affiliation(s)
- Lola E. Navas
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, 41013 Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, 41013 Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
22
|
Choi S, Lee YR, Kim KM, Choi E, Jeon BH. Dual Function of Secreted APE1/Ref-1 in TNBC Tumorigenesis: An Apoptotic Initiator and a Regulator of Chronic Inflammatory Signaling. Int J Mol Sci 2022; 23:ijms23169021. [PMID: 36012284 PMCID: PMC9409365 DOI: 10.3390/ijms23169021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
The simultaneous regulation of cancer cells and inflammatory immune cells in the tumor microenvironment (TME) can be an effective strategy in treating aggressive breast cancer types, such as triple-negative breast cancer (TNBC). Apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1) is a multi-functional nuclear protein that can be stimulated and then secreted. The extracellular APE1/Ref-1 causes a reduction in disulfide bonds in cytokine receptors, resulting in their conformational changes, thereby inhibiting inflammatory signaling. Furthermore, the secreted APE1/Ref-1 in response to acetylation has been shown to bind to a receptor for the advanced glycation end product (RAGE), initiating the apoptotic cell death of TNBC in vitro and in vivo. This study used PPTLS-APE1/Ref-1 in an adenovirus vector (Ad-PPTLS-APE1/Ref-1) for the constant expression of extracellular APE1/Ref-1, and our results demonstrated its dual function as an apoptotic initiator and inflammation regulator. Injecting MDA-MB 231 orthotopic xenografts with the Ad-PPTLS-APE1/Ref-1 inhibited tumor growth and development in response to acetylation. Moreover, Ad-PPTLS-APE1/Ref-1 generated reactive oxygen species (ROS), and tumor tissues derived from these xenografts exhibited apoptotic bodies. Compared to normal mice, a comparable ratio of anti- and pro-inflammatory cytokines was observed in the plasma of Ad-PPTLS-APE1/Ref-1-injected mice. Mechanistically, the disturbed cytokine receptor by reducing activity of PPTLS-APE1/Ref-1 inhibited inflammatory signaling leading to the inactivation of the p21-activated kinase 1-mediated signal transducer and activator of transcription 3/nuclear factor-κB axis in tumor tissues. These results suggest that the regulation of inflammatory signaling with adenoviral-mediated PPTLS-APE1/Ref-1 in tumors modulates the secretion of pro-inflammatory cytokines in TME, thereby inhibiting aggressive cancer cell progression, and could be considered as a promising and safe therapeutic strategy for treating TNBCs.
Collapse
Affiliation(s)
- Sunga Choi
- Department of Bioinformatics and Biosystems, Seongnam Campus of Korea Polytechnics, Seongnam-si 13122, Korea
- Correspondence: ; Tel.: +82-31-739-4140; Fax: +82-31-739-3375
| | - Yu-Ran Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Ki-Mo Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea
| | - Euna Choi
- Department of Biology, Union University, Jackson, TN 38305, USA
| | - Byeong-Hwa Jeon
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
23
|
Yuan Y, Zhang H, Li D, Li Y, Lin F, Wang Y, Song H, Liu X, Li F, Zhang J. PAK4 in cancer development: Emerging player and therapeutic opportunities. Cancer Lett 2022; 545:215813. [DOI: 10.1016/j.canlet.2022.215813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
|
24
|
CORO1C, a novel PAK4 binding protein, recruits phospho-PAK4 at serine 99 to the leading edge and promotes the migration of gastric cancer cells. Acta Biochim Biophys Sin (Shanghai) 2022; 54:673-685. [PMID: 35593474 PMCID: PMC9827817 DOI: 10.3724/abbs.2022044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Gastric cancer is one of the malignant tumors in the world. PAK4 plays an important role in the occurrence and development of gastric cancer, especially in the process of invasion and metastasis. Here we discover that CORO1C, a member of coronin family that regulates microfilament and lamellipodia formation, recruits cytoplasmic PAK4 to the leading edge of gastric cancer cells by C-terminal extension (CE) domain of CORO1C (353-457 aa). The localization of PAK4 on the leading edge of the cell depends on two necessary conditions: the phosphorylation of PAK4 on serine 99 and the binding to the CE domain of CORO1C. Unphosphorylated PAK4 on serine 99 is closely associated with microtubules by PAK4/GEF-H1/Tctex-1 complex. Once phosphorylated, PAK4 is released from microtubule, and then is recruited by CORO1C to the leading edge and regulates the CORO1C/RCC2 (regulator of chromosome condensation 2) complex, leading to the migration of gastric cancer cells. Our results reveal a new mechanism by which PAK4 regulates the migration potential of gastric cancer cells through microtubule-microfilament cross talk.
Collapse
|
25
|
Nguyen RY, Xiao H, Gong X, Arroyo A, Cabral AT, Fischer TT, Flores KM, Zhang X, Robert ME, Ehrlich BE, Mak M. Cytoskeletal dynamics regulates stromal invasion behavior of distinct liver cancer subtypes. Commun Biol 2022; 5:202. [PMID: 35241781 PMCID: PMC8894393 DOI: 10.1038/s42003-022-03121-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Drug treatment against liver cancer has limited efficacy due to heterogeneous response among liver cancer subtypes. In addition, the functional biophysical phenotypes which arise from this heterogeneity and contribute to aggressive invasive behavior remain poorly understood. This study interrogated how heterogeneity in liver cancer subtypes contributes to differences in invasive phenotypes and drug response. Utilizing histological analysis, quantitative 2D invasion metrics, reconstituted 3D hydrogels, and bioinformatics, our study linked cytoskeletal dynamics to differential invasion profiles and drug resistance in liver cancer subtypes. We investigated cytoskeletal regulation in 2D and 3D culture environments using two liver cancer cell lines, SNU-475 and HepG2, chosen for their distinct cytoskeletal features and invasion profiles. For SNU-475 cells, a model for aggressive liver cancer, many cytoskeletal inhibitors abrogated 2D migration but only some suppressed 3D migration. For HepG2 cells, cytoskeletal inhibition did not significantly affect 3D migration but did affect proliferative capabilities and spheroid core growth. This study highlights cytoskeleton driven phenotypic variation, their consequences and coexistence within the same tumor, as well as efficacy of targeting biophysical phenotypes that may be masked in traditional screens against tumor growth.
Collapse
Affiliation(s)
- Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Alfredo Arroyo
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Aidan T Cabral
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Kaitlin M Flores
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, CT, USA
| | | | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
26
|
p21-Activated kinases as promising therapeutic targets in hematological malignancies. Leukemia 2022; 36:315-326. [PMID: 34697424 DOI: 10.1038/s41375-021-01451-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023]
Abstract
The p21-Activated Kinases (PAKs) are a family of six serine/threonine kinases that were originally identified as downstream effectors of the Rho GTPases Cdc42 and Rac. Since the first PAK was discovered in 1994, studies have revealed their fundamental and biological importance in the development of physiological systems. Within the cell, PAKs also play significant roles in regulating essential cellular processes such as cytoskeletal dynamics, gene expression, cell survival, and cell cycle progression. These processes are often deregulated in numerous cancers when different PAKs are overexpressed or amplified at the chromosomal level. Furthermore, PAKs modulate multiple oncogenic signaling pathways which facilitate apoptosis escape, uncontrolled proliferation, and drug resistance. There is growing insight into the critical roles of PAKs in regulating steady-state hematopoiesis, including the properties of hematopoietic stem cells (HSC), and the initiation and progression of hematological malignancies. This review will focus on the most recent studies that provide experimental evidence showing how specific PAKs regulate the properties of leukemic stem cells (LSCs) and drug-resistant cells to initiate and maintain hematological malignancies. The current understanding of the molecular and cellular mechanisms by which the PAKs operate in specific human leukemia or lymphomas will be discussed. From a translational point of view, PAKs have been suggested to be critical therapeutic targets and potential prognosis markers; thus, this review will also discuss current therapeutic strategies against hematological malignancies using existing small-molecule PAK inhibitors, as well as promising combination treatments, to sensitize drug-resistant cells to conventional therapies. The challenges of toxicity and non-specific targeting associated with some PAK inhibitors, as well as how future approaches for PAK inhibition to overcome these limitations, will also be addressed.
Collapse
|
27
|
Khan HY, Uddin MH, Balasubramanian SK, Sulaiman N, Iqbal M, Chaker M, Aboukameel A, Li Y, Senapedis W, Baloglu E, Mohammad RM, Zonder J, Azmi AS. PAK4 and NAMPT as Novel Therapeutic Targets in Diffuse Large B-Cell Lymphoma, Follicular Lymphoma, and Mantle Cell Lymphoma. Cancers (Basel) 2021; 14:160. [PMID: 35008323 PMCID: PMC8750170 DOI: 10.3390/cancers14010160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), grade 3b follicular lymphoma (FL), and mantle cell lymphoma (MCL) are aggressive non-Hodgkin's lymphomas (NHL). Cure rates are suboptimal and novel treatment strategies are needed to improve outcomes. Here, we show that p21-activated kinase 4 (PAK4) and nicotinamide phosphoribosyl transferase (NAMPT) is critical for lymphoma subsistence. Dual targeting of PAK4-NAMPT by the Phase I small molecule KPT-9274 suppressed cell proliferation in DLBCL, FL, and MCL. Growth inhibition was concurrent with apoptosis induction alongside activation of pro-apoptotic proteins and reduced pro-survival markers. We observed NAD suppression, ATP reduction, and consequent cellular metabolic collapse in lymphoma cells due to KPT-9274 treatment. KPT-9274 in combination with standard-of-care chemotherapeutics led to superior inhibition of cell proliferation. In vivo, KPT-9274 could markedly suppress the growth of WSU-DLCL2 (DLBCL), Z-138, and JeKo-1 (MCL) sub-cutaneous xenografts, and a remarkable increase in host life span was shown, with a 50% cure of a systemic WSU-FSCCL (FL) model. Residual tumor analysis confirmed a reduction in total and phosphorylated PAK4 and activation of the pro-apoptotic cascade. This study, using various preclinical experimental models, demonstrates the therapeutic potential of targeting PAK4-NAMPT in DLBCL, FL, and MCL. The orally bioavailable, safe, and efficacious PAK4-NAMPT dual inhibitor KPT-9274 warrants further clinical investigation.
Collapse
Affiliation(s)
- Husain Yar Khan
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Md. Hafiz Uddin
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Suresh Kumar Balasubramanian
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Noor Sulaiman
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Marium Iqbal
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Mahmoud Chaker
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Amro Aboukameel
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Yiwei Li
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | | | | | - Ramzi M. Mohammad
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Jeffrey Zonder
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| | - Asfar S. Azmi
- Departments of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.Y.K.); (M.H.U.); (S.K.B.); (N.S.); (M.I.); (M.C.); (A.A.); (Y.L.); (R.M.M.); (J.Z.)
| |
Collapse
|
28
|
The Use of Nanomedicine to Target Signaling by the PAK Kinases for Disease Treatment. Cells 2021; 10:cells10123565. [PMID: 34944073 PMCID: PMC8700304 DOI: 10.3390/cells10123565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
P21-activated kinases (PAKs) are serine/threonine kinases involved in the regulation of cell survival, proliferation, inhibition of apoptosis, and the regulation of cell morphology. Some members of the PAK family are highly expressed in several types of cancer, and they have also been implicated in several other medical disorders. They are thus considered to be good targets for treatment of cancer and other diseases. Although there are several inhibitors of the PAKs, the utility of some of these inhibitors is reduced for several reasons, including limited metabolic stability. One way to overcome this problem is the use of nanoparticles, which have the potential to increase drug delivery. The overall goals of this review are to describe the roles for PAK kinases in cell signaling and disease, and to describe how the use of nanomedicine is a promising new method for administering PAK inhibitors for the purpose of disease treatment and research. We discuss some of the basic mechanisms behind nanomedicine technology, and we then describe how these techniques are being used to package and deliver PAK inhibitors.
Collapse
|
29
|
p21-Activated kinase 1 (PAK1) in aging and longevity: An overview. Ageing Res Rev 2021; 71:101443. [PMID: 34390849 DOI: 10.1016/j.arr.2021.101443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023]
Abstract
The p21-activated kinases (PAKs) belong to serine/threonine kinases family, regulated by ∼21 kDa small signaling G proteins RAC1 and CDC42. The mammalian PAK family comprises six members (PAK1-6) that are classified into two groups (I and II) based on their domain architecture and regulatory mechanisms. PAKs are implicated in a wide range of cellular functions. PAK1 has recently attracted increasing attention owing to its involvement in oncogenesis, tumor progression, and metastasis as well as several life-limiting diseases and pathological conditions. In Caenorhabditis elegans, PAK1 functions limit the lifespan under basal conditions by inhibiting forkhead transcription factor DAF-16. Interestingly, PAK depletion extended longevity and attenuated the onset of age-related phenotypes in a premature-aging mouse model and delayed senescence in mammalian fibroblasts. These observations implicate PAKs as not only oncogenic but also aging kinases. Therefore, PAK-targeting genetic and/or pharmacological interventions, particularly PAK1-targeting, could be a viable strategy for developing cancer therapies with relatively no side effects and promoting healthy longevity. This review describes PAK family proteins, their biological functions, and their role in regulating aging and longevity using C. elegans. Moreover, we discuss the effect of small-molecule PAK1 inhibitors on the lifespan and healthspan of C. elegans.
Collapse
|
30
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
31
|
Naїja A, Merhi M, Inchakalody V, Fernandes Q, Mestiri S, Prabhu KS, Uddin S, Dermime S. The role of PAK4 in the immune system and its potential implication in cancer immunotherapy. Cell Immunol 2021; 367:104408. [PMID: 34246086 DOI: 10.1016/j.cellimm.2021.104408] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 01/06/2023]
Abstract
The p21 activated kinases (PAKs) are known to play a role in the regulation of cell morphology and functions. Among the various members of PAKs family, only the PAK4 protein has been shown to be overexpressed in cancer cells and its upregulation was associated with tumor development. Indeed, several studies have shown that PAK4 overexpression is implicated in carcinogenesis by different mechanisms including promotion of cell proliferation, invasion and migration, protection of cells from apoptosis, stimulation of the tumor-specific anchorage-independent cell growth and regulation of the cytoskeletal organisation and adhesion. Moreover, high PAK4 protein levels have been observed in several solid tumors and have been shown able to enhance cancer cell resistance to many treatments especially chemotherapy. Interestingly, it has been recently demonstrated that PAK4 downregulation can inhibit the PD-1/PD-L1 immune regulatory pathway. Taken together, these findings not only implicate PAK4 in oncogenic transformation and in prediction of tumor response to treatment but also suggest its role as an attractive target for immunotherapy. In the current review we will summarize the different mechanisms of PAK4 implication in tumor development, describe its role as a regulator of the immune response and as a potential novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Azza Naїja
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Inchakalody
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Queenie Fernandes
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Sarra Mestiri
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic health system, Hamad medical Corporation, Doha, Qatar
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
32
|
Wang H, Song P, Gao Y, Shen L, Xu H, Wang J, Cheng M. Drug discovery targeting p21-activated kinase 4 (PAK4): a patent review. Expert Opin Ther Pat 2021; 31:977-987. [PMID: 34369844 DOI: 10.1080/13543776.2021.1944100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: The Ser/Thr protein kinase PAK4 is a downstream regulator of Cdc42, mediating cytoskeleton remodeling, and cell motility, and inhibiting apoptosis and transcriptional regulation. Nowadays, efforts in PAK4 inhibitor development are focusing on improving inhibitory selectivity, cellular potency, and in vivo pharmacokinetic properties, and identifying the feasibility of immunotherapy combination in oncology therapy.Areas covered: This review summarized the development of PAK4 inhibitors that reported on patents in the past two decades. According to their binding features, these inhibitors were classified into type I, type I 1/2, and PAMs. Their designing ideas and SAR were elucidated in this review. Moreover, synergistic therapy of PAK4 inhibitors with PD-1/PD-L1 or CAR-T were also summarized .Expert opinion: In the past years, preclinical and clinical studies of PAK4 inhibitors ended in failure due to poor selectivity, cellular activity, or pharmacokinetic issues. There are researchers questioning the reliability of PAK4 as a drug target, particularly PAK4-related therapy is concerned with the distinguishment of the non-kinase functions and catalytic functions triggered by PAK4 phosphorylation. Meanwhile, synergistic effects of PAK4 inhibitors with PD-1/PD-L1 and CAR-T immunotherapy shed light for the development of PAK4 inhibitors.
Collapse
Affiliation(s)
- Hanxun Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Peilu Song
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Yinli Gao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Lanlan Shen
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Hanqin Xu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Jian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Maosheng Cheng
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
33
|
Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-Lowering Agents for Cancer Treatment. Nutrients 2021; 13:1665. [PMID: 34068917 PMCID: PMC8156468 DOI: 10.3390/nu13051665] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
34
|
p21-activated kinases as viable therapeutic targets for the treatment of high-risk Ewing sarcoma. Oncogene 2021; 40:1176-1190. [PMID: 33414491 DOI: 10.1038/s41388-020-01600-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 01/04/2023]
Abstract
Ewing sarcoma (ES) is the second most common bone tumor in children and young adults. Unfortunately, there have been minimal recent advancements in improving patient outcomes, especially in metastatic and recurrent diseases. In this study, we investigated the biological role of p21-activated kinases (PAKs) in ES, and the ability to therapeutically target them in high-risk disease. Via informatics analysis, we established the inverse association of PAK1 and PAK4 expression with clinical stage and outcome in ES patients. Through expression knockdown and small-molecule inhibition of PAKs, utilizing FRAX-597, KPT-9274, and PF-3758309 in multiple ES cell lines and patient-derived xenograft models, we further explored the role of PAKs in ES tumor growth and metastatic capabilities. In vitro studies in several ES cell lines indicated that diminishing PAK1 and PAK4 expression reduces tumor cell viability, migratory, and invasive properties. In vivo studies using PAK4 inhibitors, KPT-9274 and PF-3758309 demonstrated significant inhibition of primary and metastatic tumor formation, while transcriptomic analysis of PAK4-inhibitor-treated tumors identified concomitant suppression of Notch, β-catenin, and hypoxia-mediated signatures. In addition, the analysis showed enrichment of anti-tumor immune regulatory mechanisms, including interferon (IFN)-ɣ and IFN-α responses. Altogether, our molecular and pre-clinical studies are the first to establish a critical role for PAKs in ES development and progression, and consequently as viable therapeutic targets for the treatment of high-risk ES in the near future.
Collapse
|
35
|
Navas LE, Carnero A. NAD + metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther 2021; 6:2. [PMID: 33384409 PMCID: PMC7775471 DOI: 10.1038/s41392-020-00354-w] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/11/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
NAD+ was discovered during yeast fermentation, and since its discovery, its important roles in redox metabolism, aging, and longevity, the immune system and DNA repair have been highlighted. A deregulation of the NAD+ levels has been associated with metabolic diseases and aging-related diseases, including neurodegeneration, defective immune responses, and cancer. NAD+ acts as a cofactor through its interplay with NADH, playing an essential role in many enzymatic reactions of energy metabolism, such as glycolysis, oxidative phosphorylation, fatty acid oxidation, and the TCA cycle. NAD+ also plays a role in deacetylation by sirtuins and ADP ribosylation during DNA damage/repair by PARP proteins. Finally, different NAD hydrolase proteins also consume NAD+ while converting it into ADP-ribose or its cyclic counterpart. Some of these proteins, such as CD38, seem to be extensively involved in the immune response. Since NAD cannot be taken directly from food, NAD metabolism is essential, and NAMPT is the key enzyme recovering NAD from nicotinamide and generating most of the NAD cellular pools. Because of the complex network of pathways in which NAD+ is essential, the important role of NAD+ and its key generating enzyme, NAMPT, in cancer is understandable. In the present work, we review the role of NAD+ and NAMPT in the ways that they may influence cancer metabolism, the immune system, stemness, aging, and cancer. Finally, we review some ongoing research on therapeutic approaches.
Collapse
Affiliation(s)
- Lola E Navas
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cancer, Sevilla, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain. .,CIBER de Cancer, Sevilla, Spain.
| |
Collapse
|
36
|
Islam R, Lam KW. Recent progress in small molecule agents for the targeted therapy of triple-negative breast cancer. Eur J Med Chem 2020; 207:112812. [DOI: 10.1016/j.ejmech.2020.112812] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
|
37
|
Mpilla GB, Philip PA, El-Rayes B, Azmi AS. Pancreatic neuroendocrine tumors: Therapeutic challenges and research limitations. World J Gastroenterol 2020; 26:4036-4054. [PMID: 32821069 PMCID: PMC7403797 DOI: 10.3748/wjg.v26.i28.4036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/10/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are known to be the second most common epithelial malignancy of the pancreas. PNETs can be listed among the slowest growing as well as the fastest growing human cancers. The prevalence of PNETs is deceptively low; however, its incidence has significantly increased over the past decades. According to the American Cancer Society's estimate, about 4032 (> 7% of all pancreatic malignancies) individuals will be diagnosed with PNETs in 2020. PNETs often cause severe morbidity due to excessive secretion of hormones (such as serotonin) and/or overall tumor mass. Patients can live for many years (except for those patients with poorly differentiated G3 neuroendocrine tumors); thus, the prevalence of the tumors that is the number of patients actually dealing with the disease at any given time is fairly high because the survival is much longer than pancreatic ductal adenocarcinoma. Due to significant heterogeneity, the management of PNETs is very complex and remains an unmet clinical challenge. In terms of research studies, modest improvements have been made over the past decades in the identification of potential oncogenic drivers in order to enhance the quality of life and increase survival for this growing population of patients. Unfortunately, the majority of systematic therapies approved for the management of advanced stage PNETs lack objective response or at most result in modest benefits in survival. In this review, we aim to discuss the broad challenges associated with the management and the study of PNETs.
Collapse
Affiliation(s)
- Gabriel Benyomo Mpilla
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Philip Agop Philip
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Bassel El-Rayes
- Department of Hematology Oncology, Emory Winship Institute, Atlanta, GA 30322, United States
| | - Asfar Sohail Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| |
Collapse
|
38
|
Trott JF, Abu Aboud O, McLaughlin B, Anderson KL, Modiano JF, Kim K, Jen KY, Senapedis W, Chang H, Landesman Y, Baloglu E, Pili R, Weiss RH. Anti-Cancer Activity of PAK4/NAMPT Inhibitor and Programmed Cell Death Protein-1 Antibody in Kidney Cancer. KIDNEY360 2020; 1:376-388. [PMID: 35224510 PMCID: PMC8809296 DOI: 10.34067/kid.0000282019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/12/2020] [Indexed: 06/14/2023]
Abstract
BACKGROUND Kidney cancer (or renal cell carcinoma, RCC) is the sixth most common malignancy in the United States and is increasing in incidence. Despite new therapies, including targeted therapies and immunotherapies, most RCCs are resistant to treatment. Thus, several laboratories have been evaluating new approaches to therapy, both with single agents as well as combinations. Although we have previously shown efficacy of the dual PAK4/nicotinamide phosphoribosyltransferase (NAMPT) inhibitor KPT-9274, and the immune checkpoint inhibitors (CPI) have shown utility in the clinic, there has been no evaluation of this combination either clinically or in an immunocompetent animal model of kidney cancer. METHODS In this study, we use the renal cell adenocarcinoma (RENCA) model of spontaneous murine kidney cancer. Male BALB/cJ mice were injected subcutaneously with RENCA cells and, after tumors were palpable, they were treated with KPT-9274 and/or anti-programmed cell death 1 (PDCD1; PD1) antibody for 21 days. Tumors were measured and then removed at animal euthanasia for subsequent studies. RESULTS We demonstrate a significant decrease in allograft growth with the combination treatment of KPT-9274 and anti-PD1 antibody without significant weight loss by the animals. This is associated with decreased (MOUSE) Naprt expression, indicating dependence of these tumors on NAMPT in parallel to what we have observed in human RCC. Histology of the tumors showed substantial necrosis regardless of treatment condition, and flow cytometry of antibody-stained tumor cells revealed that the enhanced therapeutic effect of KPT-9274 and anti-PD1 antibody was not driven by infiltration of T cells into tumors. CONCLUSIONS This study highlights the potential of the RENCA model for evaluating immunologic responses to KPT-9274 and checkpoint inhibitor (CPI) and suggests that therapy with this combination could improve efficacy in RCC beyond what is achievable with CPI alone.
Collapse
Affiliation(s)
- Josephine F. Trott
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Omran Abu Aboud
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Bridget McLaughlin
- Comprehensive Cancer Center, University of California, Davis, California
| | - Katie L. Anderson
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California
| | - Kuang-Yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis, California
| | - William Senapedis
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Hua Chang
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Yosef Landesman
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Erkan Baloglu
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Roberto Pili
- Simon Cancer Center, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Robert H. Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
- Comprehensive Cancer Center, University of California, Davis, California
- Medical Service, Veterans Affairs Northern California Health Care System, Sacramento, California
| |
Collapse
|
39
|
Clayton NS, Ridley AJ. Targeting Rho GTPase Signaling Networks in Cancer. Front Cell Dev Biol 2020; 8:222. [PMID: 32309283 PMCID: PMC7145979 DOI: 10.3389/fcell.2020.00222] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
As key regulators of cytoskeletal dynamics, Rho GTPases coordinate a wide range of cellular processes, including cell polarity, cell migration, and cell cycle progression. The adoption of a pro-migratory phenotype enables cancer cells to invade the stroma surrounding the primary tumor and move toward and enter blood or lymphatic vessels. Targeting these early events could reduce the progression to metastatic disease, the leading cause of cancer-related deaths. Rho GTPases play a key role in the formation of dynamic actin-rich membrane protrusions and the turnover of cell-cell and cell-extracellular matrix adhesions required for efficient cancer cell invasion. Here, we discuss the roles of Rho GTPases in cancer, their validation as therapeutic targets and the challenges of developing clinically viable Rho GTPase inhibitors. We review other therapeutic targets in the wider Rho GTPase signaling network and focus on the four best characterized effector families: p21-activated kinases (PAKs), Rho-associated protein kinases (ROCKs), atypical protein kinase Cs (aPKCs), and myotonic dystrophy kinase-related Cdc42-binding kinases (MRCKs).
Collapse
Affiliation(s)
- Natasha S Clayton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Anne J Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
40
|
Selective targeting of NAMPT by KPT-9274 in acute myeloid leukemia. Blood Adv 2020; 3:242-255. [PMID: 30692102 DOI: 10.1182/bloodadvances.2018024182] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Treatment options for acute myeloid leukemia (AML) remain extremely limited and associated with significant toxicity. Nicotinamide phosphoribosyltransferase (NAMPT) is involved in the generation of NAD+ and a potential therapeutic target in AML. We evaluated the effect of KPT-9274, a p21-activated kinase 4/NAMPT inhibitor that possesses a unique NAMPT-binding profile based on in silico modeling compared with earlier compounds pursued against this target. KPT-9274 elicited loss of mitochondrial respiration and glycolysis and induced apoptosis in AML subtypes independent of mutations and genomic abnormalities. These actions occurred mainly through the depletion of NAD+, whereas genetic knockdown of p21-activated kinase 4 did not induce cytotoxicity in AML cell lines or influence the cytotoxic effect of KPT-9274. KPT-9274 exposure reduced colony formation, increased blast differentiation, and diminished the frequency of leukemia-initiating cells from primary AML samples; KPT-9274 was minimally cytotoxic toward normal hematopoietic or immune cells. In addition, KPT-9274 improved overall survival in vivo in 2 different mouse models of AML and reduced tumor development in a patient-derived xenograft model of AML. Overall, KPT-9274 exhibited broad preclinical activity across a variety of AML subtypes and warrants further investigation as a potential therapeutic agent for AML.
Collapse
|
41
|
Li Y, Jia S, Dai W. Fisetin Modulates Human Oral Squamous Cell Carcinoma Proliferation by Blocking PAK4 Signaling Pathways. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:773-782. [PMID: 32158195 PMCID: PMC7049269 DOI: 10.2147/dddt.s229270] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/21/2020] [Indexed: 01/20/2023]
Abstract
Objective Human oral squamous cell carcinoma (OSCC) is a major cause of mortality and morbidity worldwide. There is an urgent need to identify bioactive molecules and potential target genes that could inhibit carcinogenesis for OSCC therapy. Fisetin (3,7,3′,4′-tetrahydroxyflavone), a naturally occurring flavonoid, has been previously shown to have anti-proliferative activities in OSCC; however, its molecular mechanism is unknown. Methods Colony formation, cell viability, Boyden chamber, wound healing, and tumor xenograft assays were used to detect the impact of fisetin on OSCC cells in vitro and in vivo. Western blot analysis was used to examine the corresponding protein expression. Results Fisetin treatment significantly inhibited proliferation and promoted apoptosis by repressing PAK4 expression. Moreover, fisetin treatment attenuated cell migration by blocking PAK4 signaling pathways. In addition, the tumor xenograft showed anti-tumor growth effects of fisetin exposure in vivo. Conclusion Fisetin may represent a potential therapeutic strategy for human OSCC by targeting PAK4 signaling pathways.
Collapse
Affiliation(s)
- Yanshu Li
- Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning, People's Republic of China.,Department of Cell Biology, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Shiheng Jia
- Department of Cell Biology, China Medical University, Shenyang, Liaoning, People's Republic of China.,Department of Clinical Medicine, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wei Dai
- Department of Oromaxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning, People's Republic of China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
42
|
Cordover E, Wei J, Patel C, Shan NL, Gionco J, Sargsyan D, Wu R, Cai L, Kong AN, Jacinto E, Minden A. KPT-9274, an Inhibitor of PAK4 and NAMPT, Leads to Downregulation of mTORC2 in Triple Negative Breast Cancer Cells. Chem Res Toxicol 2020; 33:482-491. [PMID: 31876149 DOI: 10.1021/acs.chemrestox.9b00376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Triple negative breast cancer (TNBC) is difficult to treat due to lack of druggable targets. We have found that treatment with the small molecule inhibitor KPT-9274 inhibits growth of TNBC cells and eventually leads to cell death. KPT-9274 is a dual specific inhibitor of PAK4 and Nicotinamide Phosphoribosyltransferase (NAMPT). The PAK4 protein kinase is often highly expressed in TNBC cells and has important roles in cell growth, survival, and migration. Previously we have found that inhibition of PAK4 leads to growth inhibition of TNBC cells both in vitro and in vivo. Likewise, NAMPT has been shown to be dysregulated in cancer due to its role in cell metabolism. In order to understand better how treating cells with KPT-9274 abrogates TNBC cell growth, we carried out an RNA sequencing of TNBC cells treated with KPT-9274. As a result, we identified Rictor as an important target that is inhibited in the KPT-9274 treated cells. Conversely, we found that Rictor is predicted to be activated when PAK4 is overexpressed in cells, which suggests a role for PAK4 in the regulation of Rictor. Rictor is a component of mTORC2, one of the complexes formed by the serine/threonine kinase mTOR. mTOR is important for the control of cell growth and metabolism. Our results suggest a new mechanism by which the KPT-9274 compound may block the growth of breast cancer cells, which is via inhibition of mTORC2 signaling. Consistent with this, sequencing analysis of PAK4 overexpressing cells indicates that PAK4 has a role in activation of the mTOR pathway.
Collapse
Affiliation(s)
- Emma Cordover
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Janet Wei
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Chadni Patel
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Naing Lin Shan
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - John Gionco
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Li Cai
- Department of Biomedical Engineering , Rutgers, The State University of New Jersey , 599 Taylor Road , Piscataway , New Jersey 08854 , United States
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
43
|
Abril-Rodriguez G, Torrejon DY, Liu W, Zaretsky JM, Nowicki TS, Tsoi J, Puig-Saus C, Baselga-Carretero I, Medina E, Quist MJ, Garcia AJ, Senapedis W, Baloglu E, Kalbasi A, Cheung-Lau G, Berent-Maoz B, Comin-Anduix B, Hu-Lieskovan S, Wang CY, Grasso CS, Ribas A. PAK4 inhibition improves PD-1 blockade immunotherapy. NATURE CANCER 2019; 1:46-58. [PMID: 34368780 PMCID: PMC8340852 DOI: 10.1038/s43018-019-0003-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023]
Abstract
Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.
Collapse
Affiliation(s)
- Gabriel Abril-Rodriguez
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Davis Y Torrejon
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wei Liu
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jesse M Zaretsky
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Theodore S Nowicki
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of California, Los Angeles, Los Angeles, USA
| | - Jennifer Tsoi
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cristina Puig-Saus
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ignacio Baselga-Carretero
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Egmidio Medina
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael J Quist
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alejandro J Garcia
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Anusha Kalbasi
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Gardenia Cheung-Lau
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Beata Berent-Maoz
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Begoña Comin-Anduix
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Siwen Hu-Lieskovan
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Catherine S Grasso
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
44
|
Ge X, Zhao Y, Dong L, Seng J, Zhang X, Dou D. NAMPT regulates PKM2 nuclear location through 14-3-3ζ: Conferring resistance to tamoxifen in breast cancer. J Cell Physiol 2019; 234:23409-23420. [PMID: 31141164 DOI: 10.1002/jcp.28910] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 12/25/2022]
Abstract
The resistance against tamoxifen therapy has become one of the major obstacles in the clinical treatment of breast cancer. Nicotinamide phosphoribosyltransferase (NAMPT) is an essential enzyme catalyzing nicotinamide adenine dinucleotide biosynthesis and is important for tumor metabolism. The study here sought to explore the effect of NAMPT on breast cancer survival with tamoxifen conditioning. We found that NAMPT was highly expressed in breast cancer cells compared with normal mammary epithelial cells. Inhibition of NAMPT by FK866 inhibited cell viability and aggravated apoptosis in cancer cells treated with 4-hydroxytamoxifen. NAMPT overexpression upregulated 14-3-3ζ expression. Knockdown of 14-3-3ζ reduced cell survival and promoted apoptosis. Activation of Akt signaling, rather than ERK1/2 pathway, is responsible for 14-3-3ζ regulation by NAMPT overexpression. Furthermore, NAMPT overexpression led to PKM2 accumulation in the cell nucleus and could be dampened by 14-3-3ζ inhibition. In addition, NAMPT overexpression promoted xenografted tumor growth and apoptosis in nude mice, while 14-3-3ζ inhibition attenuated its effect. Collectively, our data demonstrate that NAMPT contributes to tamoxifen resistance through regulation of 14-3-3ζ expression and PKM2 translocation.
Collapse
Affiliation(s)
- Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingling Dong
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Seng
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangyu Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongwei Dou
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
45
|
Mpilla G, Aboukameel A, Muqbil I, Kim S, Beydoun R, Philip PA, Mohammad RM, Kamgar M, Shidham V, Senapedis W, Baloglu E, Li J, Dyson G, Xue Y, El-Rayes B, Azmi AS. PAK4-NAMPT Dual Inhibition as a Novel Strategy for Therapy Resistant Pancreatic Neuroendocrine Tumors. Cancers (Basel) 2019; 11:1902. [PMID: 31795447 PMCID: PMC6966587 DOI: 10.3390/cancers11121902] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PNET) remain an unmet clinical need. In this study, we show that targeting both nicotinamide phosphoribosyltransferase (NAMPT) and p21-activated kinase 4 (PAK4) could become a synthetic lethal strategy for PNET. The expression of PAK4 and NAMPT was found to be higher in PNET tissue compared to normal cells. PAK4-NAMPT dual RNAi suppressed proliferation of PNET cell lines. Treatment with KPT-9274 (currently in a Phase I trial or analogs, PF3758309 (the PAK4 selective inhibitor) or FK866 (the NAMPT inhibitor)) suppressed the growth of PNET cell lines and synergized with the mammalian target of rapamycin (mTOR) inhibitors everolimus and INK-128. Molecular analysis of the combination treatment showed down-regulation of known everolimus resistance drivers. KPT-9274 suppressed NAD pool and ATP levels in PNET cell lines. Metabolomic profiling showed a statistically significant alteration in cellular energetic pathways. KPT-9274 given orally at 150 mg/kg 5 days/week for 4 weeks dramatically reduced PNET sub-cutaneous tumor growth. Residual tumor analysis demonstrated target engagement in vivo and recapitulated in vitro results. Our investigations demonstrate that PAK4 and NAMPT are two viable therapeutic targets in the difficult to treat PNET that warrant further clinical investigation.
Collapse
Affiliation(s)
- Gabriel Mpilla
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Amro Aboukameel
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Irfana Muqbil
- University of Detroit Mercy, Detroit, MI 48201, USA;
| | - Steve Kim
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Rafic Beydoun
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (R.B.); (V.S.)
| | - Philip A. Philip
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Ramzi M. Mohammad
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Mandana Kamgar
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Vinod Shidham
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (R.B.); (V.S.)
| | | | - Erkan Baloglu
- Karyopharm Therapeutics Inc., Newton, MA 02459, USA; (W.S.); (E.B.)
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Gregory Dyson
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Yue Xue
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA; (Y.X.); (B.E.-R.)
| | - Bassel El-Rayes
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA; (Y.X.); (B.E.-R.)
| | - Asfar S. Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| |
Collapse
|
46
|
Wang H, Gao Z, Song P, Hu B, Wang J, Cheng M. Molecular dynamics simulation and QM/MM calculation reveal the selectivity mechanism of type I 1/2 kinase inhibitors: the effect of intramolecular H-bonds and conformational restriction for improved selectivity. Phys Chem Chem Phys 2019; 21:24147-24164. [PMID: 31657381 DOI: 10.1039/c9cp04353e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Understanding the selectivity mechanisms of inhibitors towards highly similar proteins is extremely important work on the way to a new drug. Here, we aim to reveal the selectivity mechanisms of type I 1/2 kinase inhibitors towards p21-activated kinase (PAK4) and mitogen-activated protein kinase kinase kinase 14 (MAP3K14, NIK). PAK4, belonging to the serine/threonine protein kinases, is involved in cell signaling pathways and controls cellular functions and has received attention as an attractive drug target. The high sequence identity between PAK4 and NIK makes it challenging to design selective PAK4 inhibitors. In this work, computational methods including protein comparison, molecular docking, QM/MM, molecular dynamics simulations, and density functional theory (DFT) calculation were employed to explore the binding mechanisms of selective inhibitors against NIK and PAK4. The simulation results revealed the crucial factors accounting for selective inhibition of PAK4 over NIK, including different protein-ligand interactions, the positions and conformations of key residues, and the ligands flexibilities. This study will shed light on understanding the selectivity mechanisms of PAK4 and NIK inhibitors.
Collapse
Affiliation(s)
- Hanxun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Santiago-Gómez A, Kedward T, Simões BM, Dragoni I, NicAmhlaoibh R, Trivier E, Sabin V, Gee JM, Sims AH, Howell SJ, Clarke RB. PAK4 regulates stemness and progression in endocrine resistant ER-positive metastatic breast cancer. Cancer Lett 2019; 458:66-75. [DOI: 10.1016/j.canlet.2019.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/29/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
|
48
|
P21 activated kinase signaling in cancer. Semin Cancer Biol 2019; 54:40-49. [DOI: 10.1016/j.semcancer.2018.01.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 12/29/2022]
|
49
|
Li N, Lopez MA, Linares M, Kumar S, Oliva S, Martinez-Lopez J, Xu L, Xu Y, Perini T, Senapedis W, Baloglu E, Shammas MA, Hunter Z, Anderson KC, Treon SP, Munshi NC, Fulciniti M. Dual PAK4-NAMPT Inhibition Impacts Growth and Survival, and Increases Sensitivity to DNA-Damaging Agents in Waldenström Macroglobulinemia. Clin Cancer Res 2018; 25:369-377. [PMID: 30206161 DOI: 10.1158/1078-0432.ccr-18-1776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE p21-activated kinase 4 (PAK4) plays a significant biological and functional role in a number of malignancies, including multiple myeloma (MM). On the basis of our promising findings in MM, we here characterize PAK4 expression and role in WM cells, as well effect of dual PAK4-NAMPT inhibitor (KPT-9274) against WM cell growth and viability. EXPERIMENTAL DESIGN We have analyzed mRNA and protein expression levels of PAK4 in WM cells, and used loss-of-function approach to investigate its contribution to WM cell viability. We have further tested the in vitro and in vivo effect of KPT-9274 against WM cell growth and viability. RESULTS We report here high-level expression and functional role of PAK4 in WM, as demonstrated by shRNA-mediated knockdown; and significant impact of KPT-9274 on WM cell growth and viability. The growth inhibitory effect of KPT-9274 was associated with decreased PAK4 expression and NAMPT activity, as well as induction of apoptosis. Interestingly, in WM cell lines treated with KPT-9274, we detected a significant impact on DNA damage and repair genes. Moreover, we observed that apart from inducing DNA damage, KPT-9274 specifically decreased RAD51 and the double-strand break repair by the homologous recombination pathway. As a result, when combined with a DNA alkylating agents bendamustine and melphalan, KPT-9274 provided a synergistic inhibition of cell viability in WM cell lines and primary patient WM cells in vitro and in vivo. CONCLUSIONS These results support the clinical investigation of KPT-9274 in combination with DNA-damaging agent for treatment of WM.
Collapse
Affiliation(s)
- Na Li
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Liaoning, China
| | - Michael A Lopez
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Maria Linares
- Hospital Universitario 12 de Octubre, Complutense School of Medicine, Spanish National Cancer Research Centre, Madrid, Spain
| | - Subodh Kumar
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Stefania Oliva
- Myeloma Unit, Division of Hematology, University of Torino, Torino, Italy
| | - Joaquin Martinez-Lopez
- Hospital Universitario 12 de Octubre, Complutense School of Medicine, Spanish National Cancer Research Centre, Madrid, Spain
| | - Lian Xu
- Bing Center for Waldenstrom's macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Yan Xu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Tommaso Perini
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | - Masood A Shammas
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
| | - Zachary Hunter
- Bing Center for Waldenstrom's macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Steven P Treon
- Bing Center for Waldenstrom's macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Nikhil C Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- VA Boston Healthcare System, Boston, Massachusetts
| | - Mariateresa Fulciniti
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
50
|
|