1
|
Sharma D, Dhiman I, Das S, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent Advances in Therapeutic Peptides: Innovations and Applications in Treating Infections and Diseases. ACS OMEGA 2025; 10:17087-17107. [PMID: 40352490 PMCID: PMC12059905 DOI: 10.1021/acsomega.5c02077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Peptides have become a powerful frontier in modern medicine, offering a promising therapeutic solution for various diseases and advancing rapidly in pharmaceutical development. These small amino acid chains, with their innovative design, have attracted significant attention due to their versatility and high receptor specificity, which minimizes off-target effects, along with enhanced therapeutic efficacy, biodegradability, low toxicity, and minimal immunogenicity. They are being explored for use in several clinical domains, like metabolic diseases, immunomodulation, and cancer. Furthermore, antimicrobial peptides (AMPs) have grown to be a promising strategy to combat the worldwide challenge of antibiotic resistance, demonstrating promising results against multidrug-resistant organisms. Both natural and engineered peptides have been discovered and investigated, whereas numerous others are progressing toward clinical trials in a number of therapeutic domains. Recent improvements with surface modification, such as peptide engineering, peptide cyclization, PEGylation, and the utilization of synthetic amino acids to enhance their pharmacokinetic profiles and overcome the inherent disadvantages of these peptides have made it possible for the area to continue to advance. Moreover, their therapeutic potential has been further enhanced by innovative delivery methods, such as self-assembling peptides, nanocarriers, and alternate routes of administration. This Review critically states the potential of peptides as versatile therapeutics along with their modifications and advancements to drive the significant progress to treat infections and chronic diseases, along with their potential benefits and challenges.
Collapse
Affiliation(s)
- Deepshikha Sharma
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Isha Dhiman
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Swarnali Das
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Devlina Das Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Sandeep Kumar Dash
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
- School
of Medicine, University of Leeds, Leeds LS97TF, United Kingdom
| |
Collapse
|
2
|
Wang T, Wang Z. Targeting the "Undruggable": Small-Molecule Inhibitors of Proliferating Cell Nuclear Antigen (PCNA) in the Spotlight in Cancer Therapy. J Med Chem 2025; 68:2058-2088. [PMID: 39904718 DOI: 10.1021/acs.jmedchem.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
PCNA plays multiple roles in cancer development, including cell proliferation regulation, DNA repair, replication, and serving as a widely used biomarker and therapeutic target. Despite its significant role in oncology, PCNA has historically been considered "undruggable" due to the absence of known endogenous small molecule modulators and identifiable ligand binding sites. Unlike other protein-protein interfaces, PCNA lacks explicit binding grooves, featuring a relatively small and shallow surface pocket, which hinders the discovery of traditional small molecule targets. Recent breakthroughs have introduced promising PCNA-targeting candidates, with ATX-101 and AOH1996 entering phase I clinical trials for cancer therapy, garnering academic and industry interest. These achievements provide new evidence for PCNA as a drug target. This article provides insight and perspective on the application of small-molecule PCNA inhibitors in cancer treatment, covering PCNA function, its relationship with cancer, structural modification of small molecule inhibitors, and discovery strategies.
Collapse
Affiliation(s)
- Tiantian Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Zengtao Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
- Jiangxi Provincial Key Laboratory of TCM Female Reproductive Health and Related Diseases Research and Transformation, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
| |
Collapse
|
3
|
Søgaard CK, Otterlei M. Targeting proliferating cell nuclear antigen (PCNA) for cancer therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:209-246. [PMID: 39034053 DOI: 10.1016/bs.apha.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Proliferating cell nuclear antigen (PCNA) is an essential scaffold protein in many cellular processes. It is best known for its role as a DNA sliding clamp and processivity factor during DNA replication, which has been extensively reviewed by others. However, the importance of PCNA extends beyond its DNA-associated functions in DNA replication, chromatin remodelling, DNA repair and DNA damage tolerance (DDT), as new non-canonical roles of PCNA in the cytosol have recently been identified. These include roles in the regulation of immune evasion, apoptosis, metabolism, and cellular signalling. The diverse roles of PCNA are largely mediated by its myriad protein interactions, and its centrality to cellular processes makes PCNA a valid therapeutic anticancer target. PCNA is expressed in all cells and plays an essential role in normal cellular homeostasis; therefore, the main challenge in targeting PCNA is to selectively kill cancer cells while avoiding unacceptable toxicity to healthy cells. This chapter focuses on the stress-related roles of PCNA, and how targeting these PCNA roles can be exploited in cancer therapy.
Collapse
Affiliation(s)
- Caroline K Søgaard
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway; APIM Therapeutics A/S, Trondheim, Norway.
| |
Collapse
|
4
|
Wendel SO, Snow JA, Gu L, Banerjee NS, Malkas L, Wallace NA. The potential of PCNA inhibition as a therapeutic strategy in cervical cancer. J Med Virol 2023; 95:e29244. [PMID: 38010649 PMCID: PMC10683864 DOI: 10.1002/jmv.29244] [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: 08/23/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023]
Abstract
Cervical cancers are the fourth most common and most deadly cancer in women worldwide. Despite being a tremendous public health burden, few novel approaches to improve care for these malignancies have been introduced. We discuss the potential for proliferating cell nuclear antigen (PCNA) inhibition to address this need as well as the advantages and disadvantages for compounds that can therapeutically inhibit PCNA with a specific focus on cervical cancer.
Collapse
Affiliation(s)
| | - Jazmine A Snow
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Long Gu
- Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Nilam Sanjib Banerjee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Linda Malkas
- Beckman Research Institute of City of Hope, Duarte, California, USA
| | | |
Collapse
|
5
|
Gu L, Li M, Li CM, Haratipour P, Lingeman R, Jossart J, Gutova M, Flores L, Hyde C, Kenjić N, Li H, Chung V, Li H, Lomenick B, Von Hoff DD, Synold TW, Aboody KS, Liu Y, Horne D, Hickey RJ, Perry JJP, Malkas LH. Small molecule targeting of transcription-replication conflict for selective chemotherapy. Cell Chem Biol 2023; 30:1235-1247.e6. [PMID: 37531956 PMCID: PMC10592352 DOI: 10.1016/j.chembiol.2023.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 02/12/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
Targeting transcription replication conflicts, a major source of endogenous DNA double-stranded breaks and genomic instability could have important anticancer therapeutic implications. Proliferating cell nuclear antigen (PCNA) is critical to DNA replication and repair processes. Through a rational drug design approach, we identified a small molecule PCNA inhibitor, AOH1996, which selectively kills cancer cells. AOH1996 enhances the interaction between PCNA and the largest subunit of RNA polymerase II, RPB1, and dissociates PCNA from actively transcribed chromatin regions, while inducing DNA double-stranded breaks in a transcription-dependent manner. Attenuation of RPB1 interaction with PCNA, by a point mutation in RPB1's PCNA-binding region, confers resistance to AOH1996. Orally administrable and metabolically stable, AOH1996 suppresses tumor growth as a monotherapy or as a combination treatment but causes no discernable side effects. Inhibitors of transcription replication conflict resolution may provide a new and unique therapeutic avenue for exploiting this cancer-selective vulnerability.
Collapse
Affiliation(s)
- Long Gu
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| | - Min Li
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Caroline M Li
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Pouya Haratipour
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Robert Lingeman
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Jennifer Jossart
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Margarita Gutova
- Department of Developmental & Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Linda Flores
- Department of Developmental & Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Caitlyn Hyde
- Department of Developmental & Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Nikola Kenjić
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA
| | - Haiqing Li
- Department of Genomics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Vincent Chung
- Department of Medical Oncology, City of Hope, Duarte, CA, USA
| | - Hongzhi Li
- Department of Bioinformatics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Brett Lomenick
- Proteome Exploration Laboratory, California Institute of Technology, Pasadena, CA, USA
| | - Daniel D Von Hoff
- Clinical Translational Research Division, Translational Genomics Research Institute, 445N 5th Street, Phoenix, AZ 85004, USA
| | - Timothy W Synold
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Karen S Aboody
- Department of Developmental & Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Yilun Liu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - David Horne
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Robert J Hickey
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - J Jefferson P Perry
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Linda H Malkas
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
6
|
Gu L, Hickey RJ, Malkas LH. Therapeutic Targeting of DNA Replication Stress in Cancer. Genes (Basel) 2023; 14:1346. [PMID: 37510250 PMCID: PMC10378776 DOI: 10.3390/genes14071346] [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: 04/20/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/30/2023] Open
Abstract
This article reviews the currently used therapeutic strategies to target DNA replication stress for cancer treatment in the clinic, highlighting their effectiveness and limitations due to toxicity and drug resistance. Cancer cells experience enhanced spontaneous DNA damage due to compromised DNA replication machinery, elevated levels of reactive oxygen species, loss of tumor suppressor genes, and/or constitutive activation of oncogenes. Consequently, these cells are addicted to DNA damage response signaling pathways and repair machinery to maintain genome stability and support survival and proliferation. Chemotherapeutic drugs exploit this genetic instability by inducing additional DNA damage to overwhelm the repair system in cancer cells. However, the clinical use of DNA-damaging agents is limited by their toxicity and drug resistance often arises. To address these issues, the article discusses a potential strategy to target the cancer-associated isoform of proliferating cell nuclear antigen (caPCNA), which plays a central role in the DNA replication and damage response network. Small molecule and peptide agents that specifically target caPCNA can selectively target cancer cells without significant toxicity to normal cells or experimental animals.
Collapse
Affiliation(s)
- Long Gu
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Robert J Hickey
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Linda H Malkas
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
7
|
Fatima M, Abourehab MAS, Aggarwal G, Jain GK, Sahebkar A, Kesharwani P. Advancement of cell-penetrating peptides in combating triple-negative breast cancer. Drug Discov Today 2022; 27:103353. [PMID: 36099963 DOI: 10.1016/j.drudis.2022.103353] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/23/2022] [Accepted: 09/06/2022] [Indexed: 11/03/2022]
Abstract
Extensive research efforts have been made and are still ongoing in the search for an ideal anti-cancer therapy. Almost all chemotherapeutics require a carrier or vehicle, a drug delivery system that can transport the drug specifically to the targeted cancer cells, sparing normal cells. Cell-penetrating peptides (CPPs) provide an effective and efficient pathway for the intra-cellular transportation of various bioactive molecules in several biomedical therapies. They are now well-recognized as facilitators of intracellular cargo delivery and have excellent potential for targeted anti-cancer therapy. In this review, we explain CPPs, recent progress in the development of new CPPs, and their utilization to transport cargoes such as imaging agents, chemotherapeutics, and short-interfering RNAs (siRNA) into tumor cells, contributing to the advancement of novel tumor-specific delivery systems.
Collapse
Affiliation(s)
- Mahak Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110 062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia 61519, Egypt
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110 062, India.
| |
Collapse
|
8
|
Keane S, de Weerd HA, Ejeskär K. DLG2 impairs dsDNA break repair and maintains genome integrity in neuroblastoma. DNA Repair (Amst) 2022; 112:103302. [DOI: 10.1016/j.dnarep.2022.103302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/03/2022]
|
9
|
Novel Peptide Therapeutic Approaches for Cancer Treatment. Cells 2021; 10:cells10112908. [PMID: 34831131 PMCID: PMC8616177 DOI: 10.3390/cells10112908] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
Peptides are increasingly being developed for use as therapeutics to treat many ailments, including cancer. Therapeutic peptides have the advantages of target specificity and low toxicity. The anticancer effects of a peptide can be the direct result of the peptide binding its intended target, or the peptide may be conjugated to a chemotherapy drug or radionuclide and used to target the agent to cancer cells. Peptides can be targeted to proteins on the cell surface, where the peptide–protein interaction can initiate internalization of the complex, or the peptide can be designed to directly cross the cell membrane. Peptides can induce cell death by numerous mechanisms including membrane disruption and subsequent necrosis, apoptosis, tumor angiogenesis inhibition, immune regulation, disruption of cell signaling pathways, cell cycle regulation, DNA repair pathways, or cell death pathways. Although using peptides as therapeutics has many advantages, peptides have the disadvantage of being easily degraded by proteases once administered and, depending on the mode of administration, often have difficulty being adsorbed into the blood stream. In this review, we discuss strategies recently developed to overcome these obstacles of peptide delivery and bioavailability. In addition, we present many examples of peptides developed to fight cancer.
Collapse
|
10
|
Yang Y, Ma Y, Yuan M, Peng Y, Fang Z, Wang J. Identifying the biomarkers and pathways associated with hepatocellular carcinoma based on an integrated analysis approach. Liver Int 2021; 41:2485-2498. [PMID: 34033190 DOI: 10.1111/liv.14972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 05/11/2021] [Accepted: 05/19/2021] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. The molecular mechanism underlying HCC is still unclear. In this study, we conducted a comprehensive analysis to explore the genes, pathways and their interactions involved in HCC. METHODS We analysed the gene expression datasets corresponding to 488 samples from 10 studies on HCC and identified the genes differentially expressed in HCC samples. Then, the genes were compared against Phenolyzer and GeneCards to screen those potentially associated with HCC. The features of the selected genes were explored by mapping them onto the human protein-protein interaction network, and a subnetwork related to HCC was constructed. Hub genes in this HCC specific subnetwork were identified, and their relevance with HCC was investigated by survival analysis. RESULTS We identified 444 differentially expressed genes (177 upregulated and 267 downregulated) related to HCC. Functional enrichment analysis revealed that pathways like p53 signalling and chemical carcinogenesis were eriched in HCC genes. In the subnetwork related to HCC, five disease modules were detected. Further analysis identified six hub genes from the HCC specific subnetwork. Survival analysis showed that the expression levels of these genes were negatively correlated with survival rate of HCC patients. CONCLUSIONS Based on a systems biology framework, we identified the genes, pathways, as well as the disease specific network related to HCC. We also found novel biomarkers whose expression patterns were correlated with progression of HCC, and they could be candidates for further investigation.
Collapse
Affiliation(s)
- Yichen Yang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China.,Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yuequn Ma
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Meng Yuan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Yonglin Peng
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Zhonghai Fang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Chang HR, Jung E, Cho S, Jeon YJ, Kim Y. Targeting Non-Oncogene Addiction for Cancer Therapy. Biomolecules 2021; 11:129. [PMID: 33498235 PMCID: PMC7909239 DOI: 10.3390/biom11020129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
While Next-Generation Sequencing (NGS) and technological advances have been useful in identifying genetic profiles of tumorigenesis, novel target proteins and various clinical biomarkers, cancer continues to be a major global health threat. DNA replication, DNA damage response (DDR) and repair, and cell cycle regulation continue to be essential systems in targeted cancer therapies. Although many genes involved in DDR are known to be tumor suppressor genes, cancer cells are often dependent and addicted to these genes, making them excellent therapeutic targets. In this review, genes implicated in DNA replication, DDR, DNA repair, cell cycle regulation are discussed with reference to peptide or small molecule inhibitors which may prove therapeutic in cancer patients. Additionally, the potential of utilizing novel synthetic lethal genes in these pathways is examined, providing possible new targets for future therapeutics. Specifically, we evaluate the potential of TONSL as a novel gene for targeted therapy. Although it is a scaffold protein with no known enzymatic activity, the strategy used for developing PCNA inhibitors can also be utilized to target TONSL. This review summarizes current knowledge on non-oncogene addiction, and the utilization of synthetic lethality for developing novel inhibitors targeting non-oncogenic addiction for cancer therapy.
Collapse
Affiliation(s)
- Hae Ryung Chang
- Department of Biological Sciences and Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea; (E.J.); (S.C.)
| | - Eunyoung Jung
- Department of Biological Sciences and Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea; (E.J.); (S.C.)
| | - Soobin Cho
- Department of Biological Sciences and Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea; (E.J.); (S.C.)
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea;
| | - Yonghwan Kim
- Department of Biological Sciences and Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea; (E.J.); (S.C.)
| |
Collapse
|
12
|
Cardano M, Tribioli C, Prosperi E. Targeting Proliferating Cell Nuclear Antigen (PCNA) as an Effective Strategy to Inhibit Tumor Cell Proliferation. Curr Cancer Drug Targets 2020; 20:240-252. [PMID: 31951183 DOI: 10.2174/1568009620666200115162814] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022]
Abstract
Targeting highly proliferating cells is an important issue for many types of aggressive tumors. Proliferating Cell Nuclear Antigen (PCNA) is an essential protein that participates in a variety of processes of DNA metabolism, including DNA replication and repair, chromatin organization and transcription and sister chromatid cohesion. In addition, PCNA is involved in cell survival, and possibly in pathways of energy metabolism, such as glycolysis. Thus, the possibility of targeting this protein for chemotherapy against highly proliferating malignancies is under active investigation. Currently, approaches to treat cells with agents targeting PCNA rely on the use of small molecules or on peptides that either bind to PCNA, or act as a competitor of interacting partners. Here, we describe the status of the art in the development of agents targeting PCNA and discuss their application in different types of tumor cell lines and in animal model systems.
Collapse
Affiliation(s)
- Miriana Cardano
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Carla Tribioli
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Ennio Prosperi
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| |
Collapse
|
13
|
King D, Li XD, Almeida GS, Kwok C, Gravells P, Harrison D, Burke S, Hallsworth A, Jamin Y, George S, Robinson SP, Lord CJ, Poon E, Yeomanson D, Chesler L, Bryant HE. MYCN expression induces replication stress and sensitivity to PARP inhibition in neuroblastoma. Oncotarget 2020; 11:2141-2159. [PMID: 32577161 PMCID: PMC7289530 DOI: 10.18632/oncotarget.27329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022] Open
Abstract
This study investigates the influence expression of the MYCN oncogene has on the DNA damage response, replication fork progression and sensitivity to PARP inhibition in neuroblastoma. In a panel of neuroblastoma cell lines, MYCN amplification or MYCN expression resulted in increased cell death in response to a range of PARP inhibitors (niraparib, veliparib, talazoparib and olaparib) compared to the response seen in non-expressing/amplified cells. MYCN expression slowed replication fork speed and increased replication fork stalling, an effect that was amplified by PARP inhibition or PARP1 depletion. Increased DNA damage seen was specifically induced in S-phase cells. Importantly, PARP inhibition caused a significant increase in the survival of mice bearing MYCN expressing tumours in a transgenic murine model of MYCN expressing neuroblastoma. Olaparib also sensitized MYCN expressing cells to camptothecin- and temozolomide-induced cell death to a greater degree than non-expressing cells. In summary, MYCN expression leads to increased replication stress in neuroblastoma cells. This effect is exaggerated by inhibition of PARP, resulting in S-phase specific DNA damage and ultimately increased tumour cell death. PARP inhibition alone or in combination with classical chemotherapeutics is therefore a potential therapeutic strategy for neuroblastoma and may be more effective in MYCN expressing tumours.
Collapse
Affiliation(s)
- David King
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Xiao Dun Li
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
- Present address: Medical Research Council Cancer Unit, Hutchison/Medical Research Council Research Centre, University of Cambridge, Cambridge, UK
| | - Gilberto S. Almeida
- Divisions of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, UK
| | - Colin Kwok
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Polly Gravells
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Daniel Harrison
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Saoirse Burke
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Albert Hallsworth
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Yann Jamin
- Divisions of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, UK
| | - Sally George
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Simon P. Robinson
- Divisions of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, UK
| | - Christopher J. Lord
- CRUK Gene Function Laboratory and Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - Evon Poon
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | | | - Louis Chesler
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Helen E. Bryant
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| |
Collapse
|
14
|
Smith SJ, Li CM, Lingeman RG, Hickey RJ, Liu Y, Malkas LH, Raoof M. Molecular Targeting of Cancer-Associated PCNA Interactions in Pancreatic Ductal Adenocarcinoma Using a Cell-Penetrating Peptide. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:250-256. [PMID: 32368614 PMCID: PMC7190754 DOI: 10.1016/j.omto.2020.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma is a particularly difficult cancer to treat due to a lack of effective screening or treatment. Pancreatic cancer cells exhibit high proliferating cell nuclear antigen (PCNA) expression, which is associated with poor prognosis. PCNA, an important nuclear DNA replication and repair protein, regulates a myriad of proteins via the interdomain connector loop. Within this region, amino acids 126–133 are critical for PCNA interactions in cancer cells. Here, we investigate the ability of a decoy cell-penetrating peptide, R9-caPeptide, that mimics the interdomain connector loop region of PCNA to disrupt PCNA-protein interactions in pancreatic cancer cells. Our data suggest that R9-caPeptide causes dose-dependent toxicity in a panel of pancreatic cancer cell lines by inhibiting DNA replication fork progression and PCNA-regulated DNA repair, ultimately causing lethal DNA damage. Overall, these studies lay the foundation for novel therapeutic strategies that target PCNA in pancreatic cancer.
Collapse
Affiliation(s)
- Shanna J Smith
- Department of Molecular and Cellular Biology, Beckman Research Institute at City of Hope, Duarte, CA 91010, USA
| | - Caroline M Li
- Department of Molecular and Cellular Biology, Beckman Research Institute at City of Hope, Duarte, CA 91010, USA
| | - Robert G Lingeman
- Department of Molecular and Cellular Biology, Beckman Research Institute at City of Hope, Duarte, CA 91010, USA
| | - Robert J Hickey
- Department of Molecular Pharmacology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yilun Liu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Linda H Malkas
- Department of Molecular and Cellular Biology, Beckman Research Institute at City of Hope, Duarte, CA 91010, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Palrasu M, Knapinska AM, Diez J, Smith L, LaVoi T, Giulianotti M, Houghten RA, Fields GB, Minond D. A Novel Probe for Spliceosomal Proteins that Induces Autophagy and Death of Melanoma Cells Reveals New Targets for Melanoma Drug Discovery. Cell Physiol Biochem 2019; 53:656-686. [PMID: 31573152 PMCID: PMC6990463 DOI: 10.33594/000000164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 09/25/2019] [Indexed: 12/24/2022] Open
Abstract
Background/Aims: Despite recent advances in melanoma drug discovery, the average overall survival of patients with late stage metastatic melanoma is approximately 3 years, suggesting a need for approaches that identify new melanoma targets. We have previously reported a discovery of novel anti-melanoma compound 2155–14 (Onwuha-Ekpete et al., J Med Chem. 2014 Feb 27; 57(4):1599–608). In the report presented herein we aim to identify its target(s) and mechanism of action. Methods: We utilized biotinylated analog of 2155–14 to pull down its targets from melanoma cells. Proteomics in combination with western blot were used to identify the targets. Mechanism of action of 2155–14 was determined using flow cytometry, RT-PCR, microscopy, western blot, and enzymatic activity assays. Where applicable, one-way analysis of variance (ANOVA) was used followed by Dunnett post hoc test. Results: In the present study, we identified ATP-dependent RNA helicase DDX1 and heterogeneous nuclear ribonucleoproteins (hnRNPs) H1, H2 and A2/B1 as targets of anti-melanoma compound 2155–14. To the best of our knowledge, this is a first report suggesting that these proteins could be targeted for melanoma therapy. Mechanistic investigations showed that 2155–14 induces ER stress leading to potentiation of basal autophagy resulting in melanoma cell death in BRAF and NRAS mutated melanoma cells. Conclusion: Identification of mode of action of 2155–14 may provide insight into novel therapies against a broad range of melanoma subtypes. These studies were enabled by the novel probe derived from a mixture-based library, an important class of chemical biology tools for discovering novel targets.
Collapse
Affiliation(s)
- Manikandan Palrasu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Anna M Knapinska
- Department of Chemistry & Biochemistry, Center for Molecular Biology & Biotechnology, Florida Atlantic University, Jupiter, FL, USA
| | - Juan Diez
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Lyndsay Smith
- Department of Chemistry & Biochemistry, Center for Molecular Biology & Biotechnology, Florida Atlantic University, Jupiter, FL, USA
| | - Travis LaVoi
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, USA
| | - Marc Giulianotti
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, USA
| | | | - Gregg B Fields
- Department of Chemistry & Biochemistry, Center for Molecular Biology & Biotechnology, Florida Atlantic University, Jupiter, FL, USA
| | - Dmitriy Minond
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL, USA.,Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA,
| |
Collapse
|
16
|
Perumal SK, Xu X, Yan C, Ivanov I, Benkovic SJ. Recognition of a Key Anchor Residue by a Conserved Hydrophobic Pocket Ensures Subunit Interface Integrity in DNA Clamps. J Mol Biol 2019; 431:2493-2510. [PMID: 31051173 DOI: 10.1016/j.jmb.2019.04.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/22/2019] [Accepted: 04/22/2019] [Indexed: 10/26/2022]
Abstract
Sliding clamp proteins encircle duplex DNA and are involved in processive DNA replication and the DNA damage response. Clamp proteins are ring-shaped oligomers (dimers or trimers) and are loaded onto DNA by an ATP-dependent clamp loader complex that ruptures the interface between two adjacent subunits. Here we measured the solution dynamics of the human clamp protein, proliferating cell nuclear antigen, by monitoring the change in the fluorescence of a site-specifically labeled. To unravel the origins of clamp subunit interface stability, we carried out comprehensive comparative analysis of the interfaces of seven sliding clamps. We used computational modeling (molecular dynamic simulations and MM/GBSA binding energy decomposition analyses) to identify conserved networks of hydrophobic residues critical for clamp stability and ring-opening dynamics. The hydrophobic network is shared among clamp proteins and exhibits a "key in a keyhole" pattern where a bulky aromatic residue from one clamp subunit is anchored into a hydrophobic pocket of the opposing subunit. Bioinformatics and dynamic network analyses showed that this oligomeric latch is conserved across DNA sliding clamps from all domains of life and dictates the dynamics of clamp opening and closing.
Collapse
Affiliation(s)
- Senthil K Perumal
- Department of Chemistry, 414 Wartik Laboratory, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiaojun Xu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA
| | - Chunli Yan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA
| | - Ivaylo Ivanov
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA.
| | - Stephen J Benkovic
- Department of Chemistry, 414 Wartik Laboratory, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
17
|
Kucherlapati M. Examining transcriptional changes to DNA replication and repair factors over uveal melanoma subtypes. BMC Cancer 2018; 18:818. [PMID: 30107825 PMCID: PMC6092802 DOI: 10.1186/s12885-018-4705-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
Background Uncontrolled replication is a process common to all cancers facilitated by the summation of changes accumulated as tumors progress. The aim of this study was to examine small groups of genes with known biology in replication and repair at the transcriptional and genomic levels, correlating alterations with survival in uveal melanoma tumor progression. Selected components of Pre-Replication, Pre-Initiation, and Replisome Complexes, DNA Damage Response and Mismatch Repair have been observed. Methods Two groups have been generated for selected genes above and below the average alteration level and compared for expression and survival across The Cancer Genome Atlas uveal melanoma subtypes. Significant differences in expression between subtypes monosomic or disomic for chromosome 3 have been identified by Fisher’s exact test. Kaplan Meier survival distribution based on disease specific survival has been compared by Log-rank test. Results Genes with significant alteration include MCM2, MCM4, MCM5, CDC45, MCM10, CIZ1, PCNA, FEN1, LIG1, POLD1, POLE, HUS1, CHECK1, ATRIP, MLH3, and MSH6. Exon 4 skipping in CIZ1 previously identified as a cancer variant, and reportedly used as an early serum biomarker in lung cancer was found. Mismatch Repair protein MLH3 was found to have splicing variations with deletions to both Exon 5 and Exon 7 simultaneously. PCNA, FEN1, and LIG1 had increased relative expression levels not due to mutation or to copy number variation. Conclusion The current study proposes changes in relative and differential expression to replication and repair genes that support the concept their products are causally involved in uveal melanoma. Specific avenues for early biomarker identification and therapeutic approach are suggested.
Collapse
Affiliation(s)
- Melanie Kucherlapati
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA. .,Department of Medicine, Division of Genetics, Brigham and Women's Hospital, 77 Avenue Louis Pasteur NRB 160B, Boston, 02115, MA, USA.
| |
Collapse
|
18
|
Gu L, Lingeman R, Yakushijin F, Sun E, Cui Q, Chao J, Hu W, Li H, Hickey RJ, Stark JM, Yuan YC, Chen Y, Vonderfecht SL, Synold TW, Shi Y, Reckamp KL, Horne D, Malkas LH. The Anticancer Activity of a First-in-class Small-molecule Targeting PCNA. Clin Cancer Res 2018; 24:6053-6065. [PMID: 29967249 DOI: 10.1158/1078-0432.ccr-18-0592] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/30/2018] [Accepted: 06/26/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Proliferating cell nuclear antigen (PCNA) plays an essential role in regulating DNA synthesis and repair and is indispensable to cancer cell growth and survival. We previously reported a novel cancer associated PCNA isoform (dubbed caPCNA), which was ubiquitously expressed in a broad range of cancer cells and tumor tissues, but not significantly in nonmalignant cells. We found the L126-Y133 region of caPCNA is structurally altered and more accessible to protein-protein interaction. A cell-permeable peptide harboring the L126-Y133 sequence blocked PCNA interaction in cancer cells and selectively kills cancer cells and xenograft tumors. On the basis of these findings, we sought small molecules targeting this peptide region as potential broad-spectrum anticancer agents. EXPERIMENTAL DESIGN By computer modeling and medicinal chemistry targeting a surface pocket partly delineated by the L126-Y133 region of PCNA, we identified a potent PCNA inhibitor (AOH1160) and characterized its therapeutic properties and potential toxicity. RESULTS AOH1160 selectively kills many types of cancer cells at below micromolar concentrations without causing significant toxicity to a broad range of nonmalignant cells. Mechanistically, AOH1160 interferes with DNA replication, blocks homologous recombination-mediated DNA repair, and causes cell-cycle arrest. It induces apoptosis in cancer cells and sensitizes them to cisplatin treatment. AOH1160 is orally available to animals and suppresses tumor growth in a dosage form compatible to clinical applications. Importantly, it does not cause significant toxicity at 2.5 times of an effective dose. CONCLUSIONS These results demonstrated the favorable therapeutic properties and the potential of AOH1160 as a broad-spectrum therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Long Gu
- Department of Molecular & Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California.
| | - Robert Lingeman
- Department of Molecular & Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Fumiko Yakushijin
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California
| | - Emily Sun
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Qi Cui
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Jianfei Chao
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Weidong Hu
- Department of Immunology, Beckman Research Institute of City of Hope, Duarte, California
| | - Hongzhi Li
- Department of Bioinformatics, Beckman Research Institute of City of Hope, Duarte, California
| | - Robert J Hickey
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California.,Translational Biomarker Discovery Core, Beckman Research Institute of City of Hope, Duarte, California
| | - Jeremy M Stark
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, California
| | - Yate-Ching Yuan
- Department of Bioinformatics, Beckman Research Institute of City of Hope, Duarte, California
| | - Yuan Chen
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California
| | - Steven L Vonderfecht
- Center for Comparative Medicine, Beckman Research Institute of City of Hope, Duarte, California
| | - Timothy W Synold
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Yanhong Shi
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Karen L Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California
| | - Linda H Malkas
- Department of Molecular & Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California
| |
Collapse
|
19
|
Shemesh A, Kundu K, Peleg R, Yossef R, Kaplanov I, Ghosh S, Khrapunsky Y, Gershoni-Yahalom O, Rabinski T, Cerwenka A, Atlas R, Porgador A. NKp44-Derived Peptide Binds Proliferating Cell Nuclear Antigen and Mediates Tumor Cell Death. Front Immunol 2018; 9:1114. [PMID: 29875773 PMCID: PMC5974751 DOI: 10.3389/fimmu.2018.01114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/03/2018] [Indexed: 11/21/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is considered as a hub protein and is a key regulator of DNA replication, repair, cell cycle control, and apoptosis. PCNA is overexpressed in many cancer types, and PCNA overexpression is correlated with cancer virulence. Membrane-associated PCNA is a ligand for the NKp44 (NCR2) innate immune receptor. The purpose of this study was to characterize the PCNA-binding site within NKp44. We have identified NKp44-derived linear peptide (pep8), which can specifically interact with PCNA and partly block the NKp44–PCNA interaction. We then tested whether NKp44-derived pep8 (NKp44-pep8) fused to cell-penetrating peptides (CPPs) can be employed for targeting the intracellular PCNA for the purpose of anticancer therapy. Treatment of tumor cells with NKp44-pep8, fused to R11-NLS cell-penetrating peptide (R11-NLS-pep8), reduced cell viability and promoted cell death, in various murine and human cancer cell lines. Administration of R11-NLS-pep8 to tumor-bearing mice suppressed tumor growth in the 4T1 breast cancer and the B16 melanoma in vivo models. We therefore identified the NKp44 binding site to PCNA and further developed an NKp44-peptide-based agent that can inhibit tumor growth through interfering with the function of intracellular PCNA in the tumor cell.
Collapse
Affiliation(s)
- Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kiran Kundu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Refael Peleg
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rami Yossef
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Irena Kaplanov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Susmita Ghosh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yana Khrapunsky
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Orly Gershoni-Yahalom
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tatiana Rabinski
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Roee Atlas
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
20
|
Smith SJ, Hickey RJ, Malkas LH. Validating the disruption of proliferating cell nuclear antigen interactions in the development of targeted cancer therapeutics. Cancer Biol Ther 2016; 17:310-9. [PMID: 26889573 DOI: 10.1080/15384047.2016.1139247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human DNA replication and repair is a highly coordinated process involving the specifically timed actions of numerous proteins and enzymes. Many of these proteins require interaction with proliferating cell nuclear antigen (PCNA) for activation within the process. The interdomain connector loop (IDCL) of PCNA provides a docking site for many of those proteins, suggesting that this region is critically important in the regulation of cellular function. Previous work in this laboratory has demonstrated that a peptide mimicking a specific region of the IDCL (caPeptide) has the ability to disrupt key protein-protein interactions between PCNA and its binding partners, thereby inhibiting DNA replication within the cells. In this study, we confirm the ability of the caPeptide to disrupt DNA replication function using both intact cell and in vitro DNA replication assays. Further, we were able to demonstrate that treatment with caPeptide results in a decrease of polymerase δ activity that correlates with the observed decrease in DNA replication. We have also successfully developed a surface plasmon resonance (SPR) assay to validate the disruption of the PCNA-pol δ interaction with caPeptide.
Collapse
Affiliation(s)
- Shanna J Smith
- a Beckman Research Institute at City of Hope , Department of Molecular and Cellular Biology , Duarte , CA , USA
| | - Robert J Hickey
- b Beckman Research Institute at City of Hope , Department of Molecular Pharmacology , Duarte , CA , USA
| | - Linda H Malkas
- a Beckman Research Institute at City of Hope , Department of Molecular and Cellular Biology , Duarte , CA , USA
| |
Collapse
|
21
|
Bielinsky AK. Penetrating enemy territory: Soluble PCNA-peptides stress out MYCN-overexpressing neuroblastomas. EBioMedicine 2016; 2:1844-5. [PMID: 26844252 PMCID: PMC4703722 DOI: 10.1016/j.ebiom.2015.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
22
|
Gu L, Chu P, Lingeman R, McDaniel H, Kechichian S, Hickey RJ, Liu Z, Yuan YC, Sandoval JA, Fields GB, Malkas LH. The Mechanism by Which MYCN Amplification Confers an Enhanced Sensitivity to a PCNA-Derived Cell Permeable Peptide in Neuroblastoma Cells. EBioMedicine 2015; 2:1923-31. [PMID: 26844271 PMCID: PMC4703743 DOI: 10.1016/j.ebiom.2015.11.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 11/01/2022] Open
Abstract
Dysregulated expression of MYC family genes is a hallmark of many malignancies. Unfortunately, these proteins are not amenable to blockade by small molecules or protein-based therapeutic agents. Therefore, we must find alternative approaches to target MYC-driven cancers. Amplification of MYCN, a MYC family member, predicts high-risk neuroblastoma (NB) disease. We have shown that R9-caPep blocks the interaction of PCNA with its binding partners and selectively kills human NB cells, especially those with MYCN amplification, and we now show the mechanism. We found elevated levels of DNA replication stress in MYCN-amplified NB cells. R9-caPep exacerbated DNA replication stress in MYCN-amplified NB cells and NB cells with an augmented level of MYC by interfering with DNA replication fork extension, leading to Chk1 dependence and susceptibility to Chk1 inhibition. We describe how these effects may be exploited for treating NB.
Collapse
Affiliation(s)
- Long Gu
- Department of Molecular & Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Peiguo Chu
- Department of Pathology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Robert Lingeman
- Department of Molecular & Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Heather McDaniel
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| | - Steven Kechichian
- Department of Molecular & Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Robert J Hickey
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Zheng Liu
- Bioinformatic Core, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - Yate-Ching Yuan
- Bioinformatic Core, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| | - John A Sandoval
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, United States of America
| | - Gregg B Fields
- Florida Atlantic University and The Scripps Research Institute/Scripps Florida, Jupiter, FL 33458, United States of America
| | - Linda H Malkas
- Department of Molecular & Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States of America
| |
Collapse
|
23
|
Kroker AJ, Bruning JB. p21 Exploits Residue Tyr151 as a Tether for High-Affinity PCNA Binding. Biochemistry 2015; 54:3483-93. [PMID: 25972089 DOI: 10.1021/acs.biochem.5b00241] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proliferating cell nuclear antigen (PCNA, processivity factor, sliding clamp) is a ring-shaped protein that tethers proteins to DNA in processes, including DNA replication, DNA repair, and cell-cycle control. Often used as a marker for cell proliferation, PCNA is overexpressed in cancer cells, making it an appealing pharmaceutical target. PCNA interacts with proteins through a PCNA interacting protein (PIP)-box, an eight-amino acid consensus sequence; different binding partners display a wide range of affinities based on function. Of all biological PIP-boxes, p21 has the highest known affinity for PCNA, allowing for inhibition of DNA replication and cell growth under cellular stress. As p21 is one of the few PIP-box sequences to contain a tyrosine rather than a phenylalanine in the eighth conserved position, we probed the significance of the hydroxyl group at this position using a mutational approach. Here we present the cocrystal structure of PCNA bound to a mutant p21 PIP-box peptide, p21Tyr151Phe, with associated isothermal titration calorimetry data. The p21Tyr151Phe peptide showed a 3-fold difference in affinity, as well as differences in entropy and enthalpy of binding. These differences can be attributed to a loss of hydrogen bonding capacity, as well as structural plasticity in the PCNA interdomain connector loop and the hydrophobic cavity of PCNA to which p21 binds. Thus, the hydroxyl group of Tyr151 in p21 acts as a tethering point for ideal packing and surface recognition of the peptide interface, increasing the binding affinity of p21 for PCNA.
Collapse
Affiliation(s)
- Alice J Kroker
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - John B Bruning
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
24
|
Lingeman RG, Hickey RJ, Malkas LH. Expression of a novel peptide derived from PCNA damages DNA and reverses cisplatin resistance. Cancer Chemother Pharmacol 2014; 74:981-93. [PMID: 25190177 DOI: 10.1007/s00280-014-2574-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/21/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND An 8 amino acid peptide sequence derived from proliferating cell nuclear antigen (PCNA) has been shown to effectively kill several breast cancer and neuroblastoma cell lines when added exogenously to cell cultures. METHODS In this study, the expression of the 8 amino acid peptide sequence (caPeptide) was placed under control of a tetracycline responsive promoter in MDA-MB-231 cells. RESULTS Endogenous expression of the peptide resulted in an increase in genomic DNA damage. CaPeptide induction combined with treatment of sublethal doses of cisplatin resulted in a marked increase in death of the cisplatin-resistant MDA-MB-231 cell line. CaPeptide was found to interact with POLD3, one of the subunits of DNA polymerase delta necessary for binding to PCNA. CONCLUSION These results suggest an important line of inquiry into the possible role that caPeptide might play in the reversal of cisplatin resistance in breast and other cancers. This is of particular interest in those cancers where cisplatin is the first line of chemotherapy and where the acquisition of resistance is a common malady.
Collapse
Affiliation(s)
- Robert G Lingeman
- Department of Molecular Biology, Beckman Research Institute of the City of Hope, 1450 E. Duarte Rd., Duarte, CA, 91010, USA,
| | | | | |
Collapse
|