1
|
Wang X. Clinical and molecular prognostic nomograms for patients with papillary renal cell carcinoma. Discov Oncol 2024; 15:780. [PMID: 39692801 DOI: 10.1007/s12672-024-01669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE To summarize the clinicopathological characteristics and prognostic factors of papillary renal cell carcinoma (pRCC) and to construct clinical and molecular prognostic nomograms using existing databases. METHODS Clinical prognostic models were developed using the Surveillance, Epidemiology, and End Results (SEER) database, while molecular prognostic models were constructed using The Cancer Genome Atlas (TCGA) database. Cox regression and LASSO regression were employed to identify clinicopathological features and molecular markers related to prognosis. The accuracy of the prognostic models was assessed using ROC curves, C-index, decision curve analysis (DCA) curves, and calibration plots. RESULTS In the 2004-2015 SEER cohort, Cox regression analysis revealed that age, grade, AJCC stage, N stage, M stage, and surgery were independent predictors of overall survival (OS) and cancer-specific survival (CSS) in pRCC patients. ROC curves, C-index, and DCA curves indicated that the prognostic nomogram based on clinical independent predictors had better predictive ability than TNM staging and SEER staging. Additionally, in the TCGA cohort, M stage, clinical stage, and the molecular markers IDO1 and PLK1 were identified as independent risk factors. The prognostic nomogram based on molecular independent risk factors effectively predicted the 3-year and 5-year OS and CSS for pRCC patients. CONCLUSIONS The clinical and molecular nomograms constructed in this study provide robust predictive tools for individualized prognosis in pRCC patients, offering better accuracy than traditional staging systems.
Collapse
Affiliation(s)
- Xuhui Wang
- Department of Urology, The Affiliated People's Hospital of Ningbo University, No.251, Baizhang East Road, Yinzhou District, Ningbo, 315040, China.
| |
Collapse
|
2
|
Zeng Y, Ren X, Jin P, Zhang Y, Zhuo M, Wang J. Development of MPS1 Inhibitors: Recent Advances and Perspectives. J Med Chem 2023; 66:16484-16514. [PMID: 38095579 DOI: 10.1021/acs.jmedchem.3c00963] [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: 12/29/2023]
Abstract
Monopolar spindle kinase 1 (MPS1) plays a pivotal role as a dual-specificity kinase governing spindle assembly checkpoint activation and sister chromatid separation in mitosis. Its overexpression has been observed in various human malignancies. MPS1 reduces spindle assembly checkpoint sensitivity, allowing tumor cells with a high degree of aneuploidy to complete mitosis and survive. Thus, MPS1 has emerged as a promising candidate for cancer therapy. Despite the identification of numerous MPS1 inhibitors, only five have advanced to clinical trials with none securing FDA approval for cancer treatment. In this perspective, we provide a concise overview of the structural and functional characteristics of MPS1 by highlighting its relevance to cancer. Additionally, we explore the structure-activity relationships, selectivity, and pharmacokinetics of MPS1 inhibitors featuring diverse scaffolds. Moreover, we review the reported work on enhancing MPS1 inhibitor selectivity, offering valuable insights into the discovery of novel, highly potent small-molecule MPS1 inhibitors.
Collapse
Affiliation(s)
- Yangjie Zeng
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Pengyao Jin
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Yali Zhang
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Ming Zhuo
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Jubo Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
3
|
Yurt F, Özel D, Tunçel A, Gokbayrak O, Aktas S. Synthesis and Optimization of the Docetaxel-Loaded and Durvalumab-Targeted Human Serum Albumin Nanoparticles, In Vitro Characterization on Triple-Negative Breast Cancer Cells. ACS OMEGA 2023; 8:26287-26300. [PMID: 37521641 PMCID: PMC10372957 DOI: 10.1021/acsomega.3c02682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Triple-negative breast cancer (TNBC) tends to behave more aggressively compared to other breast cancer subtypes due to the lack of receptors and its limited targeting therapy. In recent years, nanotechnology advancement has led to the development of various nanoparticle platforms for the targeted treatment of cancers. Especially, HSA-NPs have specific advantages such as biocompatibility, adjustable size during production, and relatively easy synthesis. In this study, HSA-NPs were encapsulated with docetaxel (DTX) and functionalized with polyethylene glycol (PEG), also becoming a targeting nanoplatform modified with durvalumab (DVL), and the whole nanostructure was well characterized. Subsequently, drug release studies and various in vitro cell culture studies such as determining the cytotoxicity and apoptotic levels of the nanoplatforms and PD-L1 using ELISA test were conducted on MDA-MB-468, MDA-MB-231, and MCF-7 cells. According to the results, HSA-DTX@PEG-DVL NPs showed better cytotoxicity compared to DTX in all the three cell lines. In addition, it was observed that the HSA-DTX@PEG-DVL NPs did not lead the cells to late apoptosis but were effective in the early apoptotic stage. Moreover, the ELISA data showed a significantly induced PD-L1 expression due to the presence of DVL in the nanostructure, which indicates that DVL antibodies successfully bind to the HSA-DTX@PEG-DVL nanostructure.
Collapse
Affiliation(s)
- Fatma Yurt
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Derya Özel
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Ayça Tunçel
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Ozde Gokbayrak
- Department
of Basic Oncology, Institute of Oncology, DokuzEylül University, 35340 Izmir, Turkey
| | - Safiye Aktas
- Department
of Basic Oncology, Institute of Oncology, DokuzEylül University, 35340 Izmir, Turkey
| |
Collapse
|
4
|
Phosphorylation-mediated interaction between human E26 transcription factor 1 and specific protein 1 is required for tumor cell migration. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1441-1452. [PMID: 36305724 PMCID: PMC9828152 DOI: 10.3724/abbs.2022148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Transcription factors, human E26 transcription factor 1 (Ets1) and specific protein 1 (Sp1), are known to induce gene expression in tumorigenicity. High Ets1 expression is often associated with colorectal tumorigenesis. In this study, we discover that metastasis and clone formation in SW480 cells mainly depend on the direct interaction between Ets1 and Sp1 instead of high Ets1 expression. The interaction domains are further addressed to be the segment at Sp1(626-708) and the segment at Ets1(244-331). In addition, the phosphorylation inhibition of Ets1 at Tyr283 by either downregulation of Src kinase or Src family inhibitor treatment decreases the interaction between Sp1 and Ets1 and suppresses SW480 migration. Either administration or overexpression of the peptides harboring the interaction segment strongly inhibits the colony formation and migration of SW480 cells. Our findings suggest that the interaction between Ets1 and Sp1 rather than Ets1 alone promotes transformation in SW480 cells and provide new insight into the Ets1 and Sp1 interaction as an antitumour target in SW480 cells.
Collapse
|
5
|
Prajapati R, Somoza Á. Albumin Nanostructures for Nucleic Acid Delivery in Cancer: Current Trend, Emerging Issues, and Possible Solutions. Cancers (Basel) 2021; 13:3454. [PMID: 34298666 PMCID: PMC8304767 DOI: 10.3390/cancers13143454] [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: 05/27/2021] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the major health problems worldwide, and hence, suitable therapies with enhanced efficacy and reduced side effects are desired. Gene therapy, involving plasmids, small interfering RNAs, and antisense oligonucleotides have been showing promising potential in cancer therapy. In recent years, the preparation of various carriers for nucleic acid delivery to the tumor sites is gaining attention since intracellular and extracellular barriers impart major challenges in the delivery of naked nucleic acids. Albumin is a versatile protein being used widely for developing carriers for nucleic acids. It provides biocompatibility, tumor specificity, the possibility for surface modification, and reduces toxicity. In this review, the advantages of using nucleic acids in cancer therapy and the challenges associated with their delivery are presented. The focus of this article is on the different types of albumin nanocarriers, such as nanoparticles, polyplexes, and nanoconjugates, employed to overcome the limitations of the direct use of nucleic acids in vivo. This review also highlights various approaches for the modification of the surface of albumin to enhance its transfection efficiency and targeted delivery in the tumor sites.
Collapse
Affiliation(s)
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain;
| |
Collapse
|
6
|
Raab CA, Raab M, Becker S, Strebhardt K. Non-mitotic functions of polo-like kinases in cancer cells. Biochim Biophys Acta Rev Cancer 2021; 1875:188467. [PMID: 33171265 DOI: 10.1016/j.bbcan.2020.188467] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Inhibitors of mitotic protein kinases are currently being developed as non-neurotoxic alternatives of microtubule-targeting agents (taxanes, vinca alkaloids) which provide a substantial survival benefit for patients afflicted with different types of solid tumors. Among the mitotic kinases, the cyclin-dependent kinases, the Aurora kinases, the kinesin spindle protein and Polo-like kinases (PLKs) have emerged as attractive targets of cancer therapeutics. The functions of mammalian PLK1-5 are traditionally linked to the regulation of the cell cycle and to the stress response. Especially the key role of PLK1 and PLK4 in cellular growth and proliferation, their overexpression in multiple types of human cancer and their druggability, make them appealing targets for cancer therapy. Inhibitors for PLK1 and PLK4 are currently being tested in multiple cancer trials. The clinical success of microtubule-targeting agents is attributed not solely to the induction of a mitotic arrest in cancer cells, but also to non-mitotic effects like targeting intracellular trafficking on microtubules. This raises the question whether new cancer targets like PLK1 and PLK4 regulate critical non-mitotic functions in tumor cells. In this article we summarize the important roles of PLK1-5 for the regulation of non-mitotic signaling. Due to these functions it is conceivable that inhibitors for PLK1 or PLK4 can target interphase cells, which underscores their attractive potential as cancer drug targets. Moreover, we also describe the contribution of the tumor-suppressors PLK2, PLK3 and PLK5 to cancer cell signaling outside of mitosis. These observations highlight the urgent need to develop highly specific ATP-competitive inhibitors for PLK4 and for PLK1 like the 3rd generation PLK-inhibitor Onvansertib to prevent the inhibition of tumor-suppressor PLKs in- and outside of mitosis. The remarkable feature of PLKs to encompass a unique druggable domain, the polo-box-domain (PBD) that can be found only in PLKs offers the opportunity for the development of inhibitors that target PLKs exclusively. Beyond the development of mono-specific ATP-competitive PLK inhibitors, the PBD as drug target will support the design of new drugs that eradicate cancer cells based on the mitotic and non-mitotic function of PLK1 and PLK4.
Collapse
Affiliation(s)
| | - Monika Raab
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Sven Becker
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Gynecology, Goethe-University, Frankfurt, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Partner Site Frankfurt am Main, Frankfurt, Germany.
| |
Collapse
|
7
|
Rajitha B, Malla RR, Vadde R, Kasa P, Prasad GLV, Farran B, Kumari S, Pavitra E, Kamal MA, Raju GSR, Peela S, Nagaraju GP. Horizons of nanotechnology applications in female specific cancers. Semin Cancer Biol 2019; 69:376-390. [PMID: 31301361 DOI: 10.1016/j.semcancer.2019.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/23/2019] [Accepted: 07/04/2019] [Indexed: 12/20/2022]
Abstract
Female-specific cancers are the most common cancers in women worldwide. Early detection methods remain unavailable for most of these cancers, signifying that most of them are diagnosed at later stages. Furthermore, current treatment options for most female-specific cancers are surgery, radiation and chemotherapy. Although important milestones in molecularly targeted approaches have been achieved lately, current therapeutic strategies for female-specific cancers remain limited, ineffective and plagued by the emergence of chemoresistance, which aggravates prognosis. Recently, the application of nanotechnology to the medical field has allowed the development of novel nano-based approaches for the management and treatment of cancers, including female-specific cancers. These approaches promise to improve patient survival rates by reducing side effects, enabling selective delivery of drugs to tumor tissues and enhancing the uptake of therapeutic compounds, thus increasing anti-tumor activity. In this review, we focus on the application of nano-based technologies to the design of novel and innovative diagnostic and therapeutic strategies in the context of female-specific cancers, highlighting their potential uses and limitations.
Collapse
Affiliation(s)
- Balney Rajitha
- Department of Pathology, WellStar Hospital, Marietta, GA, 30060, USA
| | - Rama Rao Malla
- Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, AP, 530045, India
| | - Ramakrishna Vadde
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, AP, 516003, India
| | - Prameswari Kasa
- Dr. LV Prasad Diagnostics and Research Laboratory, Khairtabad, Hyderabad, TS, 500004, India
| | | | - Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Seema Kumari
- Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, AP, 530045, India
| | - Eluri Pavitra
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100, Inha-ro, Incheon 22212, Republic of Korea
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia; Novel Global Community Educational Foundation, Australia
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea
| | - Sujatha Peela
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam, AP, 532410, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Song R, Hou G, Yang J, Yuan J, Wang C, Chai T, Liu Z. Effects of PLK1 on proliferation, invasion and metastasis of gastric cancer cells through epithelial-mesenchymal transition. Oncol Lett 2018; 16:5739-5744. [PMID: 30405751 PMCID: PMC6202541 DOI: 10.3892/ol.2018.9406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/31/2018] [Indexed: 12/26/2022] Open
Abstract
Effects of polo-like kinase (PLK1) on proliferation, migration and invasion capacities of gastric cancer cells through epithelial-mesenchymal transition (EMT) were investigated. Small-interfering ribonucleic acid (siRNA) with targeted interference in PLK1 gene was designed and transfected into gastric cancer MGC-803 cells via Lipofectamine to inhibit the expression of PLK1 gene in MGC-803 cells. The proliferation of MGC-803 cells was detected via methyl thiazolyl tetrazolium (MTT) assay. The mRNA and protein expression of PLK1 and EMT-related marker (E-cadherin) was detected via real-time polymerase chain reaction and western blot analysis, respectively. The effects of interference in PLK1 gene on migration and invasion of MGC-803 cells were studied via wound healing assay and Transwell chamber assay, respectively. Results of MTT assay showed that compared with that in control group, the cell proliferation in PLK1 siRNA group was significantly inhibited (p<0.01). Compared with those in control group, the mRNA and protein expression of PLK1 in PLK1 siRNA group was significantly decreased (p<0.01), but the mRNA and protein expression of E-cadherin was obviously upregulated (p<0.01). Results of wound healing assay and invasion assay showed that the capacity of migration and invasion of MGC-803 cells in PLK1 siRNA group was significantly inhibited compared with those in control group (p<0.01). In conclusion, PLK1 enhances the proliferation, migration and invasion of gastric cancer MGC-803 cells through affecting EMT.
Collapse
Affiliation(s)
- Rui Song
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Ge Hou
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Jun Yang
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Jinjin Yuan
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Cheng Wang
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Ting Chai
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Zongwen Liu
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| |
Collapse
|
9
|
Modifying plasmid-loaded HSA-nanoparticles with cell penetrating peptides - Cellular uptake and enhanced gene delivery. Int J Pharm 2017; 522:198-209. [PMID: 28279738 DOI: 10.1016/j.ijpharm.2017.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/02/2017] [Accepted: 03/05/2017] [Indexed: 01/07/2023]
Abstract
Gene therapy bears great potential for the cure of a multitude of human diseases. Research efforts focussed on the use of viral delivery vectors in the past decades, neglecting non-viral gene therapies of physical or chemical origin due to low transfection efficiency. However, side effects such as activation of oncogenes and inflammatory reactions upon immune cell activation are major obstacles impeding the clinical applicability of viral gene therapy vectors. The aim of this study was the development of a non-viral gene delivery system based on plasmid-loaded human serum albumin nanoparticles, which are biocompatible, biodegradable, and non-toxic in relevant concentrations. The surface of said nanoparticles was modified with different cell penetrating peptides, namely Tat, nona-arginine R9, and the penetratin analogue EB1. We hypothesise that the surface modified nanoparticles can effectively enter HEK 293T cells based on the cell penetrating properties of the different peptides attached. A variety of inhibitors were used targeting distinct uptake pathways in an effort to understand the mechanisms utilized by the various cell penetrating peptides on the surface of the nanoparticles. A significant increase in transfection efficiency compared to free DNA or polyplexes was seen for these novel delivery vectors.
Collapse
|
10
|
Look J, Wilhelm N, von Briesen H, Noske N, Günther C, Langer K, Gorjup E. Ligand-Modified Human Serum Albumin Nanoparticles for Enhanced Gene Delivery. Mol Pharm 2015. [PMID: 26218774 DOI: 10.1021/acs.molpharmaceut.5b00153] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of nonviral gene delivery systems is a great challenge to enable safe gene therapy. In this study, ligand-modified nanoparticles based on human serum albumin (HSA) were developed and optimized for an efficient gene therapy. Different glutaraldehyde cross-linking degrees were investigated to optimize the HSA nanoparticles for gene delivery. The peptide sequence arginine-glycine-aspartate (RGD) and the HIV-1 transactivator of transduction sequence (Tat) are well-known as promising targeting ligands. Plasmid DNA loaded HSA nanoparticles were covalently modified on their surface with these different ligands. The transfection potential of the obtained plasmid DNA loaded RGD- and Tat-modified nanoparticles was investigated in vitro, and optimal incubation conditions for these preparations were studied. It turned out that Tat-modified HSA nanoparticles with the lowest cross-linking degree of 20% showed the highest transfection potential. Taken together, ligand-functionalized HSA nanoparticles represent promising tools for efficient and safe gene therapy.
Collapse
Affiliation(s)
- Jennifer Look
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster , Corrensstraße 48, Muenster 48149, Germany
| | - Nadine Wilhelm
- Fraunhofer Institute for Biomedical Engineering , Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Hagen von Briesen
- Fraunhofer Institute for Biomedical Engineering , Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Nadja Noske
- apceth GmbH & Co. KG , Max-Lebsche-Platz 30, 81377 Munich, Germany
| | | | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster , Corrensstraße 48, Muenster 48149, Germany
| | - Erwin Gorjup
- Fraunhofer Institute for Biomedical Engineering , Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| |
Collapse
|
11
|
Bergs JWJ, Wacker MG, Hehlgans S, Piiper A, Multhoff G, Rödel C, Rödel F. The role of recent nanotechnology in enhancing the efficacy of radiation therapy. Biochim Biophys Acta Rev Cancer 2015; 1856:130-43. [PMID: 26142869 DOI: 10.1016/j.bbcan.2015.06.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/29/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
Abstract
Radiation therapy is one of the most commonly used non-surgical interventions in tumor treatment and is often combined with other modalities to enhance its efficacy. Despite recent advances in radiation oncology, treatment responses, however, vary considerably between individual patients. A variety of approaches have been developed to enhance radiation response or to counteract resistance to ionizing radiation. Among them, a relatively novel class of radiation sensitizers comprises nanoparticles (NPs) which are highly efficient and selective systems in the nanometer range. NPs can either encapsulate radiation sensitizing agents, thereby protecting them from degradation, or sensitize cancer cells to ionizing radiation via their physicochemical properties, e.g. high Z number. Moreover, they can be chemically modified for active molecular targeting and the imaging of tumors. In this review we will focus on recent developments in nanotechnology, different classes and modifications of NPs and their radiation sensitizing properties.
Collapse
Affiliation(s)
- Judith W J Bergs
- Department of Radiotherapy and Oncology, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt, Germany
| | - Matthias G Wacker
- Fraunhofer-Institute for Molecular Biology and Applied Ecology, Department of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Albrecht Piiper
- Department of Medicine I, Goethe-University, Frankfurt am Main, Germany
| | - Gabriele Multhoff
- German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt, Germany; Department of Radiation Oncology, Technische Universität München, Ismaninger Str. 22, D-81675 Munich, Germany; Clinical Cooperation Group (CCG) "Innate Immunity in Tumor Biology", Helmholtz Zentrum München, German Research Center for Environmental Health Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| |
Collapse
|
12
|
Kazazian K, Brashavitskaya O, Zih FSW, Berger-Richardson D, Xu RSZ, Pacholczyk K, Macmillan J, Swallow CJ. Polo-Like Kinases in Colorectal Cancer: Potential for Targeted Therapy. CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0275-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
13
|
Langiu M, Dadparvar M, Kreuter J, Ruonala MO. Human serum albumin-based nanoparticle-mediated in vitro gene delivery. PLoS One 2014; 9:e107603. [PMID: 25229502 PMCID: PMC4168126 DOI: 10.1371/journal.pone.0107603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 08/21/2014] [Indexed: 11/19/2022] Open
Abstract
The genetic treatment of neurodegenerative diseases still remains a challenging task since many approaches fail to deliver the therapeutic material in relevant concentrations into the brain. As viral vectors comprise the risk of immune and inflammatory responses, human serum albumin (HSA) nanoparticles were found to represent a safer and more convenient alternative. Their ability to cross the blood-brain barrier (BBB) and deliver drugs into the brain in order to enhance gene-based therapy has been previously demonstrated. The present study deals with the development of pGL3-PEI-coated HSA nanoparticles and subsequent in vitro testing in cerebellar granular and HeLa cells. The luciferase control vector pGL3 was chosen as reporter plasmid encoding for the firefly luciferase protein, linear polyethylenimine (22 kDa) as endosomolytic agent for enhancing the cells’ transfection. Studies on particle characteristics, their cellular uptake into aforementioned cell lines and on subcellular localisation, and transfection efficiency in the cerebellar cells proved the feasibility of nanoparticle-based gene delivery.
Collapse
Affiliation(s)
- Monica Langiu
- Center for Membrane Proteomics, Goethe University, Frankfurt am Main, Germany
| | - Miriam Dadparvar
- Center for Membrane Proteomics, Goethe University, Frankfurt am Main, Germany
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Jörg Kreuter
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Mika O. Ruonala
- Center for Membrane Proteomics, Goethe University, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
14
|
Gaca S, Reichert S, Multhoff G, Wacker M, Hehlgans S, Botzler C, Gehrmann M, Rödel C, Kreuter J, Rödel F. Targeting by cmHsp70.1-antibody coated and survivin miRNA plasmid loaded nanoparticles to radiosensitize glioblastoma cells. J Control Release 2013; 172:201-206. [DOI: 10.1016/j.jconrel.2013.08.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/14/2013] [Accepted: 08/17/2013] [Indexed: 10/26/2022]
|
15
|
Systemic siRNA Delivery via Peptide-Tagged Polymeric Nanoparticles, Targeting PLK1 Gene in a Mouse Xenograft Model of Colorectal Cancer. Int J Biomater 2013; 2013:252531. [PMID: 24159333 PMCID: PMC3789392 DOI: 10.1155/2013/252531] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/07/2013] [Indexed: 12/12/2022] Open
Abstract
Polymeric nanoparticles were developed from a series of chemical reactions using chitosan, polyethylene glycol, and a cell-targeting peptide (CP15). The nanoparticles were complexed with PLK1-siRNA. The optimal siRNA loading was achieved at an N : P ratio of 129.2 yielding a nanoparticle size of >200 nm. These nanoparticles were delivered intraperitoneally and tested for efficient delivery, cytotoxicity, and biodistribution in a mouse xenograft model of colorectal cancer. Both unmodified and modified chitosan nanoparticles showed enhanced accumulation at the tumor site. However, the modified chitosan nanoparticles showed considerably, less distribution in other organs. The relative gene expression as evaluated showed efficient delivery of PLK1-siRNA (0.5 mg/kg) with 50.7 ± 19.5% knockdown (P = 0.031) of PLK1 gene. The in vivo data reveals no systemic toxicity in the animals, when tested for systemic inflammation and liver toxicity. These results indicate a potential of using peptide-tagged nanoparticles for systemic delivery of siRNA at the targeted tumor site.
Collapse
|
16
|
Gaca S, Reichert S, Rödel C, Rödel F, Kreuter J. Survivin-miRNA-loaded nanoparticles as auxiliary tools for radiation therapy: preparation, characterisation, drug release, cytotoxicity and therapeutic effect on colorectal cancer cells. J Microencapsul 2012; 29:685-94. [DOI: 10.3109/02652048.2012.680511] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Sun W, Liu BL, Chen AS, Cao XK, Su Q. Small interfering RNA-mediated knockdown of polo-like kinase 1 promotes apoptosis in human hepatocellular carcinoma cell line BCL-7402. Shijie Huaren Xiaohua Zazhi 2011; 19:2822-2828. [DOI: 10.11569/wcjd.v19.i27.2822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of small interfering RNA (siRNA)-mediated Polo-like kinase 1 (Plk1) gene silencing on p53 expression and cell apoptosis in human hepatocellular carcinoma cell line BCL-7402, and to explore the feasibility of targeting the human Plk1 gene as a therapeutic strategy for hepatocellular carcinoma.
METHODS: Two siRNA sequences (siRNA1 and siRNA2) targeting the human Plk1 gene were designed and synthesized. BCL-7402 cells were transfected with blank control, negative control, siRNA1 or siRNA2 via lipofection. After transfection, reverse transcription-polymerase chain reaction (RT- PCR) was used to examine the expression of Plk1 mRNA , and Western blot was used to examine the expression of Plk1 and P53 proteins in transfected BCL-7402 cells. Cell cycle distribution and apoptosis of transfected cells were monitored by flow cytometry (FCM). The ultrastructural changes of transfected BCL-7402 cells were observed by transmission electron microscopy (TEM).
RESULTS: BCL-7402 cells transfected with low doses of siRNAs targeting the Plk1 gene showed greatly decreased levels of Plk1 mRNA and protein. In the siRNA1 group, Plk1 mRNA expression was reduced by 51% and 62% and Plk1 protein expression by 65% and 81% 24 and 48 h after transfection (all P < 0.01). In the siRNA2 group, Plk1 mRNA expression was reduced by 42% and 56% and Plk1 protein expression by 51% and 65% 24 and 48 h after transfection (all P < 0.01). P53 protein levels increased obviously with the decrease in Plk1 protein levels (P < 0.01). The percentage of cells at G2/M phase increased obviously 24 h after transfection (P < 0.01). Apoptosis rate increased remarkably and apoptotic phenotypes could be seen by TEM. in cells 48 h after transfection.
CONCLUSION: SiRNAs targeting the human Plk1 gene remarkably inhibited Plk1 expression, increased p53 gene expression, and promoted apoptosis, suggesting that the Plk1 gene plays important roles in cell cycle control and apoptosis of BCL-7402 cells.
Collapse
|
18
|
Strebhardt K. Multifaceted polo-like kinases: drug targets and antitargets for cancer therapy. Nat Rev Drug Discov 2010; 9:643-60. [PMID: 20671765 DOI: 10.1038/nrd3184] [Citation(s) in RCA: 547] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The polo-like kinase 1 (PLK1) acts in concert with cyclin-dependent kinase 1-cyclin B1 and Aurora kinases to orchestrate a wide range of critical cell cycle events. Because PLK1 has been preclinically validated as a cancer target, small-molecule inhibitors of PLK1 have become attractive candidates for anticancer drug development. Although the roles of the closely related PLK2, PLK3 and PLK4 in cancer are less well understood, there is evidence showing that PLK2 and PLK3 act as tumour suppressors through their functions in the p53 signalling network, which guards the cell against various stress signals. In this article, recent insights into the biology of PLKs will be reviewed, with an emphasis on their role in malignant transformation, and progress in the development of small-molecule PLK1 inhibitors will be examined.
Collapse
Affiliation(s)
- Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|