1
|
Aquino-Acevedo AN, Orengo-Orengo JA, Cruz-Robles ME, Saavedra HI. Mitotic kinases are emerging therapeutic targets against metastatic breast cancer. Cell Div 2024; 19:21. [PMID: 38886738 PMCID: PMC11184769 DOI: 10.1186/s13008-024-00125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
This review aims to outline mitotic kinase inhibitors' roles as potential therapeutic targets and assess their suitability as a stand-alone clinical therapy or in combination with standard treatments for advanced-stage solid tumors, including triple-negative breast cancer (TNBC). Breast cancer poses a significant global health risk, with TNBC standing out as the most aggressive subtype. Comprehending the role of mitosis is crucial for understanding how TNBC advances from a solid tumor to metastasis. Chemotherapy is the primary treatment used to treat TNBC. Some types of chemotherapeutic agents target cells in mitosis, thus highlighting the need to comprehend the molecular mechanisms governing mitosis in cancer. This understanding is essential for devising targeted therapies to disrupt these mitotic processes, prevent or treat metastasis, and improve patient outcomes. Mitotic kinases like Aurora kinase A, Aurora Kinase B, never in mitosis gene A-related kinase 2, Threonine-Tyrosine kinase, and Polo-kinase 1 significantly impact cell cycle progression by contributing to chromosome separation and centrosome homeostasis. When these kinases go awry, they can trigger chromosome instability, increase cell proliferation, and activate different molecular pathways that culminate in a transition from epithelial to mesenchymal cells. Ongoing clinical trials investigate various mitotic kinase inhibitors as potential biological treatments against advanced solid tumors. While clinical trials against mitotic kinases have shown some promise in the clinic, more investigation is necessary, since they induce severe adverse effects, particularly affecting the hematopoietic system.
Collapse
Affiliation(s)
- Alexandra N Aquino-Acevedo
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Joel A Orengo-Orengo
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Melanie E Cruz-Robles
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Harold I Saavedra
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA.
| |
Collapse
|
2
|
Sinani G, Durgun ME, Cevher E, Özsoy Y. Polymeric-Micelle-Based Delivery Systems for Nucleic Acids. Pharmaceutics 2023; 15:2021. [PMID: 37631235 PMCID: PMC10457940 DOI: 10.3390/pharmaceutics15082021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Nucleic acids can modulate gene expression specifically. They are increasingly being utilized and show huge potential for the prevention or treatment of various diseases. However, the clinical translation of nucleic acids faces many challenges due to their rapid clearance after administration, low stability in physiological fluids and limited cellular uptake, which is associated with an inability to reach the intracellular target site and poor efficacy. For many years, tremendous efforts have been made to design appropriate delivery systems that enable the safe and effective delivery of nucleic acids at the target site to achieve high therapeutic outcomes. Among the different delivery platforms investigated, polymeric micelles have emerged as suitable delivery vehicles due to the versatility of their structures and the possibility to tailor their composition for overcoming extracellular and intracellular barriers, thus enhancing therapeutic efficacy. Many strategies, such as the addition of stimuli-sensitive groups or specific ligands, can be used to facilitate the delivery of various nucleic acids and improve targeting and accumulation at the site of action while protecting nucleic acids from degradation and promoting their cellular uptake. Furthermore, polymeric micelles can be used to deliver both chemotherapeutic drugs and nucleic acid therapeutics simultaneously to achieve synergistic combination treatment. This review focuses on the design approaches and current developments in polymeric micelles for the delivery of nucleic acids. The different preparation methods and characteristic features of polymeric micelles are covered. The current state of the art of polymeric micelles as carriers for nucleic acids is discussed while highlighting the delivery challenges of nucleic acids and how to overcome them and how to improve the safety and efficacy of nucleic acids after local or systemic administration.
Collapse
Affiliation(s)
- Genada Sinani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Altinbas University, 34147 Istanbul, Türkiye;
| | - Meltem Ezgi Durgun
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Erdal Cevher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Yıldız Özsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| |
Collapse
|
3
|
Pereira PA, Serra MES, Serra AC, Coelho JFJ. Application of vinyl polymer-based materials as nucleic acids carriers in cancer therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1820. [PMID: 35637638 DOI: 10.1002/wnan.1820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 04/13/2022] [Accepted: 05/08/2022] [Indexed: 06/15/2023]
Abstract
Nucleic acid-based therapies have changed the paradigm of cancer treatment, where conventional treatment modalities still have several limitations in terms of efficacy and severe side effects. However, these biomolecules have a short half-life in vivo, requiring multiple administrations, resulting in severe suffering, discomfort, and poor patient compliance. In the early days of (nano)biotechnology, these problems caused concern in the medical community, but recently it has been recognized that these challenges can be overcome by developing innovative formulations. This review focuses on the use of vinyl polymer-based materials for the protection and delivery of nucleic acids in cancer. First, an overview of the properties of nucleic acids and their versatility as drugs is provided. Then, key information on the achievements to date, the most effective delivery methods, and the evaluation of functionalization approaches (stimulatory strategies) are critically discussed to highlight the importance of vinyl polymers in the new cancer treatment approaches. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.
Collapse
Affiliation(s)
- Patrícia Alexandra Pereira
- Department of Chemical Engineering, CEMMPRE, University of Coimbra, Rua Sílvio Lima-Pólo II, Coimbra, Portugal
- IPN, Instituto Pedro Nunes, Associação para a Inovação e Desenvolvimento em Ciência e Tecnologia, Rua Pedro Nunes, Coimbra, Portugal
| | | | - Arménio C Serra
- Department of Chemical Engineering, CEMMPRE, University of Coimbra, Rua Sílvio Lima-Pólo II, Coimbra, Portugal
| | - Jorge F J Coelho
- Department of Chemical Engineering, CEMMPRE, University of Coimbra, Rua Sílvio Lima-Pólo II, Coimbra, Portugal
| |
Collapse
|
4
|
Paskeh MDA, Saebfar H, Mahabady MK, Orouei S, Hushmandi K, Entezari M, Hashemi M, Aref AR, Hamblin MR, Ang HL, Kumar AP, Zarrabi A, Samarghandian S. Overcoming doxorubicin resistance in cancer: siRNA-loaded nanoarchitectures for cancer gene therapy. Life Sci 2022; 298:120463. [DOI: 10.1016/j.lfs.2022.120463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023]
|
5
|
Pereira P, Serra AC, Coelho JF. Vinyl Polymer-based technologies towards the efficient delivery of chemotherapeutic drugs. Prog Polym Sci 2021. [DOI: 10.1016/j.progpolymsci.2021.101432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
6
|
Dual Targeting of EGFR with PLK1 Exerts Therapeutic Synergism in Taxane-Resistant Lung Adenocarcinoma by Suppressing ABC Transporters. Cancers (Basel) 2021; 13:cancers13174413. [PMID: 34503223 PMCID: PMC8430738 DOI: 10.3390/cancers13174413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Our previous studies led us to hypothesize that downregulation of PLK1 expression or its activity can overcome the hurdles of taxane resistance by downregulating ABC transporters. Targeting PLK1 with shRNA or non-functional mutants downregulated ABCB1, ABCC9, and ABCG2 in paclitaxel-resistant lung adenocarcinoma (LUADTXR), similar to the downregulation effects from treatment with PLK1 inhibitors. Since EGFR is highly expressed in LUADTXR cells, gefitinib was combined with PLK1 inhibitors. Under these conditions, LUADTXR cells tend to undergo apoptosis more effectively than parental cells, showing a synergistic effect on downregulation of ABC transporters through c-Myc or AP-1. Clinical data provide evidence for the relationship between survival rates and expressions of PLK1 and EGFR in LUAD patients. Taken together, our data suggest that a combination of gefitinib and PLK1 inhibitors exerts strong synergism in LUADTXR, providing a benefit to overcome the limitations associated with taxanes. Abstract To overcome the limitations of chemoresistance, combination therapies using druggable targets have been investigated. Our previous studies led us to hypothesize that the downregulation of PLK1 expression or activity can be one strategy to overcome the hurdles of taxane resistance by the downregulation of ABC transporters. To explore this, various versions of PLK1 including a constitutively active version, kinase-dead form, and polo-box domain mutant were expressed in paclitaxel-resistant lung adenocarcinoma (LUADTXR). Targeting PLK1 using shRNA or non-functional mutants downregulated ABCB1, ABCC9, and ABCG2 in LUADTXR cells, which was similar to the downregulation effects from treatment with PLK1 inhibitors. The high expression of EGFR in LUAD led us to administer gefitinib, showing a markedly reduced EGFR level in LUADTXR cells. When gefitinib and PLK1 inhibitors were combined, LUADTXR cells tended to undergo apoptosis more effectively than parental cells, showing a synergistic effect on the downregulation of ABC transporters through c-Myc and AP-1. Clinical data provide evidence for the relevance between survival rates and expressions of PLK1 and EGFR in LUAD patients. Based on these results, we suggest that a combination of gefitinib and PLK1 inhibitors exerts strong synergism in LUADTXR, which helps to overcome the limitations associated with taxanes.
Collapse
|
7
|
Shi H, Sun S, Xu H, Zhao Z, Han Z, Jia J, Wu D, Lu J, Liu H, Yu R. Combined Delivery of Temozolomide and siPLK1 Using Targeted Nanoparticles to Enhance Temozolomide Sensitivity in Glioma. Int J Nanomedicine 2020; 15:3347-3362. [PMID: 32494134 PMCID: PMC7229804 DOI: 10.2147/ijn.s243878] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/15/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Temozolomide (TMZ) is the first-line chemotherapeutic option to treat glioma; however, its efficacy and clinical application are limited by its drug resistance properties. Polo-like kinase 1 (PLK1)-targeted therapy causes G2/M arrest and increases the sensitivity of glioma to TMZ. Therefore, to limit TMZ resistance in glioma, an angiopep-2 (A2)-modified polymeric micelle (A2PEC) embedded with TMZ and a small interfering RNA (siRNA) targeting PLK1 (siPLK1) was developed (TMZ-A2PEC/siPLK). MATERIALS AND METHODS TMZ was encapsulated by A2-PEG-PEI-PCL (A2PEC) through the hydrophobic interaction, and siPLK1 was complexed with the TMZ-A2PEC through electrostatic interaction. Then, an angiopep-2 (A2) modified polymeric micelle (A2PEC) embedding TMZ and siRNA targeting polo-like kinase 1 (siPLK1) was developed (TMZ-A2PEC/siPLK). RESULTS In vitro experiments indicated that TMZ-A2PEC/siPLK effectively enhanced the cellular uptake of TMZ and siPLK1 and resulted in significant cell apoptosis and cytotoxicity of glioma cells. In vivo experiments showed that glioma growth was inhibited, and the survival time of the animals was prolonged remarkably after TMZ-A2PEC/siPLK1 was injected via their tail vein. DISCUSSION The results demonstrate that the combination of TMZ and siPLK1 in A2PEC could enhance the efficacy of TMZ in treating glioma.
Collapse
Affiliation(s)
- Hui Shi
- Clinical Medical College, Nanjing Medical University, Nanjing, People’s Republic of China
- The Second People’s Hospital of Lianyungang, Lianyungang, People’s Republic of China
| | - Shuo Sun
- Clinical Medical College, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Haoyue Xu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Zongren Zhao
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Zhengzhong Han
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jun Jia
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Dongmei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, People’s Republic of China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, People’s Republic of China
| | - Hongmei Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Rutong Yu
- Clinical Medical College, Nanjing Medical University, Nanjing, People’s Republic of China
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
8
|
Chu PY, Tsai SC, Ko HY, Wu CC, Lin YH. Co-Delivery of Natural Compounds with a Dual-Targeted Nanoparticle Delivery System for Improving Synergistic Therapy in an Orthotopic Tumor Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23880-23892. [PMID: 31192580 DOI: 10.1021/acsami.9b06155] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Various natural compounds including epigallocatechin gallate (EGCG) and curcumin (CU) have potential in developing anticancer therapy. However, their clinical use is commonly limited by instability and low tissue distribution. EGCG and CU combined treatment can improve the efficacy with synergistic effects. To improve the synergistic effect and overcome the limitations of low tissue distribution, we applied a dual cancer-targeted nanoparticle system to co-deliver EGCG and CU. Nanoparticles were composed of hyaluronic acid, fucoidan, and poly(ethylene glycol)-gelatin to encapsulate EGCG and CU. Furthermore, a dual targeting system was established with hyaluronic acid and fucoidan, which were used as agents for targeting CD44 on prostate cancer cells and P-selectin in tumor vasculature, respectively. Their effect and efficacy were investigated in prostate cancer cells and a orthotopic prostate tumor model. The EGCG/CU-loaded nanoparticles bound to prostate cancer cells, which were uptaken more into cells, leading to a better anticancer efficiency compared to the EGCG/CU combination solution. In addition, the releases of EGCG and CU were regulated by their pH value that avoided the premature release. In mice, treatment of the cancer-targeted EGCG/CU-loaded nanoparticles significantly attenuated the orthotopic tumor growth without inducing organ injuries. Overall, the dual-targeted nanoparticle system for the co-delivery of EGCG and CU greatly improved its synergistic effect in cancer therapy, indicating its great potential in developing treatments for prostate cancer therapy.
Collapse
|
9
|
Shakil S, Baig MH, Tabrez S, Rizvi SMD, Zaidi SK, Ashraf GM, Ansari SA, Khan AAP, Al-Qahtani MH, Abuzenadah AM, Chaudhary AG. Molecular and enzoinformatics perspectives of targeting Polo-like kinase 1 in cancer therapy. Semin Cancer Biol 2019; 56:47-55. [PMID: 29122685 DOI: 10.1016/j.semcancer.2017.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/22/2017] [Accepted: 11/04/2017] [Indexed: 12/25/2022]
Abstract
Cancer is a disease that has been the focus of scientific research and discovery and continues to remain so. Polo-like kinases (PLKs) are basically serine/threonine kinase enzymes that control cell cycle from yeast to humans. PLK-1 stands for 'Polo-like kinase-1'. It is the most investigated protein among PLKs. It is crucial for intracellular processes, hence a 'hot' anticancer drug-target. Accelerating innovations in Enzoinformatics and associated molecular visualization tools have made it possible to literally perform a 'molecular level walk' traversing through and observing the minutest contours of the active site of relevant enzymes. PLK-1 as a protein consists of a kinase domain at the protein N-terminal and a Polo Box Domain (PBD) at the C-terminal connected by a short inter-domain linking region. PBD has two Polo-Boxes. PBD of PLK-1 gives the impression of "a small clamp sandwiched between two clips", where the two Polo Boxes are the 'clips' and the 'phosphopeptide' is the small 'clamp'. Broadly, two major sites of PLK-1 can be potential targets: one is the adenosine-5'-triphosphate (ATP)-binding site in the kinase domain and the other is PBD (more preferred due to specificity). Targeting PLK-1 RNA and the interaction of PLK-1 with a key binding partner can also be approached. However, the list of potent small molecule inhibitors targeting the PBD site of PLK-1 is still not long enough and needs due input from the scientific community. Recently, eminent scientists have proposed targeting the 'Y'-shaped pocket of PLK-1-PBD and encouraged design of ligands that should be able to concurrently bind to two or more modules of the 'Y' pocket. Hence, it is suggested that during molecular interaction analyses, particular focus should be kept on the moiety in each ligand/drug candidate which directly interacts with the amino acid residue(s) that belong(s) to one of the three binding modules which together create this Y-shaped cavity. This obviously includes (but it is not limited to) the 'shallow cleft'-forming residues i.e. Trp414, H538 and K540, as significance of these binding residues has been consistently highlighted by many studies. The present article attempts to give a concise yet critically updated overview of targeting PLK-1 for cancer therapy.
Collapse
Affiliation(s)
- Shazi Shakil
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohammad H Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Syed M Danish Rizvi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Syed K Zaidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shakeel A Ansari
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aftab Aslam Parwaz Khan
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad H Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M Abuzenadah
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adeel G Chaudhary
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Ovarian solid tumors: Current treatment and recent developments using stimuli-responsive polymers: A systemic review. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
11
|
Shin SB, Woo SU, Yim H. Cotargeting Plk1 and androgen receptor enhances the therapeutic sensitivity of paclitaxel-resistant prostate cancer. Ther Adv Med Oncol 2019; 11:1758835919846375. [PMID: 31156720 PMCID: PMC6515847 DOI: 10.1177/1758835919846375] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022] Open
Abstract
Backgrounds: Despite the clinical success of taxanes, they still have limitations, such as chemoresistance. To overcome the limitations of paclitaxel, genetic alterations and targeting effects of altered genes were observed in paclitaxel-resistant cancer. Because paclitaxel-resistant cancer shows high levels of Plk1, a promising target in chemotherapy, the effectiveness of Plk1 inhibitors in paclitaxel-resistant cancer cells has been investigated. Methods: Paclitaxel-resistant cancer cells were developed by exposure of stepwise escalating levels of paclitaxel. Genetic alterations were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunoblotting. Using a cell viability assay, combined targeting effects for Plk1 and androgen receptor (AR) were determined. Clinical data were analyzed to understand the relationship between Plk1 and AR in prostate cancer patients. Results: Treatment with Plk1 inhibitors markedly reduced the expression of MDR1, MRP1, and Plk1 in the paclitaxel-resistant cancer. Among Plk1 inhibitors, genistein, recently found as a direct Plk1 inhibitor, tended to be more effective in the paclitaxel-resistant prostate cancer than the parental cancer cells, which was related to the suppression of the AR, as well as inhibition of Plk1 activity. A combination of Plk1 inhibitors and AR antagonist bicalutamide exhibited a synergistic effect in LNCaPTXR, as well as LNCaP cells, by inhibiting Plk1 and AR. Analysis of clinical data provides evidence for the relevance between Plk1 and AR in prostate cancer patients, showing that Plk1 and AR are strong predictors of poor survival rates. Conclusions: We suggest that cotargeting Plk1 and AR would be effective in advanced chemoresistant prostate cancer cells to overcome the limitations associated with paclitaxel.
Collapse
Affiliation(s)
- Sol-Bi Shin
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, Korea
| | - Sang-Uk Woo
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea
| |
Collapse
|
12
|
Gao Y, Jia L, Wang Q, Hu H, Zhao X, Chen D, Qiao M. pH/Redox Dual-Responsive Polyplex with Effective Endosomal Escape for Codelivery of siRNA and Doxorubicin against Drug-Resistant Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:16296-16310. [PMID: 30997984 DOI: 10.1021/acsami.9b02016] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The enhanced endo-lysosomal sequestration still remains a big challenge in overcoming multidrug resistance (MDR). Herein, a dual-responsive polyplex with effective endo-lysosomal escape based on methoxypoly(ethylene glycol)-polylactide-polyhistidine-ss-oligoethylenimine (mPEG- b-PLA-PHis-ssOEI) was developed for codelivering MDR1 siRNA and doxorubicin (DOX). The polyplex showed good encapsulation of DOX and siRNA as well as triggered payload release in response to pH/redox stimuli because of the PHis protonation and the disulfide bond cleavage. The polyplex at an N/P ratio of 7 demonstrated a much higher payload delivery efficiency, MDR1 gene silence efficiency, cytotoxicity against MCF-7/ADR cell, and stronger MCF-7/ADR tumor growth inhibition than the polyplexes at higher N/P ratios. This was attributed to the stronger electrostatic attraction between siRNA and OEIs at higher N/P ratios that suppressed the release of MDR1 siRNA and OEIs, which played a dominant role in overcoming payload endo-lysosomal sequestration by the OEI-induced membrane permeabilization effect. Consequently, the polyplex with effective endo-lysosomal escape provides a rational approach for codelivery of siRNAs and chemotherapy agents for MDR reversal.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry
- ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/administration & dosage
- Doxorubicin/chemistry
- Drug Carriers/administration & dosage
- Drug Resistance, Neoplasm/drug effects
- Endosomes/chemistry
- Humans
- Hydrogen-Ion Concentration
- Lipids/chemistry
- MCF-7 Cells
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Neoplasms/drug therapy
- Neoplasms/pathology
- Polyesters/chemistry
- Polyethylene Glycols/chemistry
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
Collapse
Affiliation(s)
| | - Li Jia
- Department of Pharmacy , Heze Medical College , Heze 274000 , P. R. China
| | | | | | | | | | | |
Collapse
|
13
|
Varghese JJ, Schmale IL, Wang Y, Hansen ME, Newlands SD, Ovitt CE, Benoit DSW. Retroductal Nanoparticle Injection to the Murine Submandibular Gland. J Vis Exp 2018. [PMID: 29781991 DOI: 10.3791/57521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Two common goals of salivary gland therapeutics are prevention and cure of tissue dysfunction following either autoimmune or radiation injury. By locally delivering bioactive compounds to the salivary glands, greater tissue concentrations can be safely achieved versus systemic administration. Furthermore, off target tissue effects from extra-glandular accumulation of material can be dramatically reduced. In this regard, retroductal injection is a widely used method for investigating both salivary gland biology and pathophysiology. Retroductal administration of growth factors, primary cells, adenoviral vectors, and small molecule drugs has been shown to support gland function in the setting of injury. We have previously shown the efficacy of a retroductally injected nanoparticle-siRNA strategy to maintain gland function following irradiation. Here, a highly effective and reproducible method to administer nanomaterials to the murine submandibular gland through Wharton's duct is detailed (Figure 1). We describe accessing the oral cavity and outline the steps necessary to cannulate Wharton's duct, with further observations serving as quality checks throughout the procedure.
Collapse
Affiliation(s)
- Jomy J Varghese
- Department of Biomedical Engineering, University of Rochester;
| | - Isaac L Schmale
- Department of Otolaryngology Head and Neck Surgery, University of Rochester Medical Center
| | - Yuchen Wang
- Department of Biomedical Engineering, University of Rochester
| | | | - Shawn D Newlands
- Department of Otolaryngology Head and Neck Surgery, University of Rochester Medical Center
| | | | | |
Collapse
|
14
|
Serpine1 Knockdown Enhances MMP Activity after Flexor Tendon Injury in Mice: Implications for Adhesions Therapy. Sci Rep 2018; 8:5810. [PMID: 29643421 PMCID: PMC5895578 DOI: 10.1038/s41598-018-24144-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/27/2018] [Indexed: 12/12/2022] Open
Abstract
Injuries to flexor tendons can be complicated by fibrotic adhesions, which severely impair the function of the hand. Adhesions have been associated with TGF-β1, which causes upregulation of PAI-1, a master suppressor of protease activity, including matrix metalloproteinases (MMP). In the present study, the effects of inhibiting PAI-1 in murine zone II flexor tendon injury were evaluated utilizing knockout (KO) mice and local nanoparticle-mediated siRNA delivery. In the PAI-1 KO murine model, reduced adherence of injured tendon to surrounding subcutaneous tissue and accelerated recovery of normal biomechanical properties compared to wild type controls were observed. Furthermore, MMP activity was significantly increased in the injured tendons of the PAI-1 KO mice, which could explain their reduced adhesions and accelerated remodeling. These data demonstrate that PAI-1 mediates fibrotic adhesions in injured flexor tendons by suppressing MMP activity. In vitro siRNA delivery to silence Serpine1 expression after treatment with TGF-β1 increased MMP activity. Nanoparticle-mediated delivery of siRNA targeting Serpine1 in injured flexor tendons significantly reduced target gene expression and subsequently increased MMP activity. Collectively, the data demonstrate that PAI-1 can be a druggable target for treating adhesions and accelerating the remodeling of flexor tendon injuries.
Collapse
|
15
|
Morey M, Pandit A. Responsive triggering systems for delivery in chronic wound healing. Adv Drug Deliv Rev 2018; 129:169-193. [PMID: 29501700 DOI: 10.1016/j.addr.2018.02.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/27/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022]
Abstract
Non-communicable diseases including cancer, cardiovascular disease, diabetes, and neuropathy are chronic in nature. Treatment of these diseases with traditional delivery systems is limited due to lack of site-specificity, non-spatiotemporal release and insufficient doses. Numerous responsive delivery systems which respond to both physiological and external stimuli have been reported in the literature. However, effective strategies incorporating a multifactorial approach are required to control these complex wounds. This can be achieved by fabricating spatiotemporal release systems, multimodal systems or dual/multi-stimuli responsive delivery systems loaded with one or more bioactive components. Critically, these next generation stimuli responsive delivery systems that are at present not feasible are required to treat chronic wounds. This review provides a critical assessment of recent developments in the field of responsive delivery systems, highlighting their limitations and providing a perspective on how these challenges can be overcome.
Collapse
Affiliation(s)
- Mangesh Morey
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
16
|
Wang Y, Newman MR, Benoit DSW. Development of controlled drug delivery systems for bone fracture-targeted therapeutic delivery: A review. Eur J Pharm Biopharm 2018; 127:223-236. [PMID: 29471078 DOI: 10.1016/j.ejpb.2018.02.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/17/2018] [Accepted: 02/17/2018] [Indexed: 01/09/2023]
Abstract
Impaired fracture healing is a major clinical problem that can lead to patient disability, prolonged hospitalization, and significant financial burden. Although the majority of fractures heal using standard clinical practices, approximately 10% suffer from delayed unions or non-unions. A wide range of factors contribute to the risk for nonunions including internal factors, such as patient age, gender, and comorbidities, and external factors, such as the location and extent of injury. Current clinical approaches to treat nonunions include bone grafts and low-intensity pulsed ultrasound (LIPUS), which realizes clinical success only to select patients due to limitations including donor morbidities (grafts) and necessity of fracture reduction (LIPUS), respectively. To date, therapeutic approaches for bone regeneration rely heavily on protein-based growth factors such as INFUSE, an FDA-approved scaffold for delivery of bone morphogenetic protein 2 (BMP-2). Small molecule modulators and RNAi therapeutics are under development to circumvent challenges associated with traditional growth factors. While preclinical studies has shown promise, drug delivery has become a major hurdle stalling clinical translation. Therefore, this review overviews current therapies employed to stimulate fracture healing pre-clinically and clinically, including a focus on drug delivery systems for growth factors, parathyroid hormone (PTH), small molecules, and RNAi therapeutics, as well as recent advances and future promise of fracture-targeted drug delivery.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Maureen R Newman
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Chemical Engineering, 4517 Wegmans Hall, University of Rochester, Rochester, NY 14627, USA; Department of Orthopaedics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Department of Biomedical Genetics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Center for Oral Biology, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
17
|
Wang M, Wang J, Li B, Meng L, Tian Z. Recent advances in mechanism-based chemotherapy drug-siRNA pairs in co-delivery systems for cancer: A review. Colloids Surf B Biointerfaces 2017; 157:297-308. [DOI: 10.1016/j.colsurfb.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
|
18
|
Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing. Biomaterials 2017; 139:127-138. [PMID: 28601703 DOI: 10.1016/j.biomaterials.2017.06.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/16/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023]
Abstract
Despite great potential, delivery remains as the most significant barrier to the widespread use of siRNA therapeutics. siRNA has delivery limitations due to susceptibility to RNase degradation, low cellular uptake, and poor tissue-specific localization. Here, we report the development of a hybrid nanoparticle (NP)/hydrogel system that overcomes these challenges. Hydrogels provide localized and sustained delivery via controlled release of entrapped siRNA/NP complexes while NPs protect and enable efficient cytosolic accumulation of siRNA. To demonstrate therapeutic efficacy, regenerative siRNA against WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1) complexed with NP were entrapped within poly(ethylene glycol) (PEG)-based hydrogels and implanted at sites of murine mid-diaphyseal femur fractures. Results showed localization of hydrogels and controlled release of siRNA/NPs at fractures for 28 days, a timeframe over which fracture healing occurs. siRNA/NP sustained delivery from hydrogels resulted in significant Wwp1 silencing at fracture callus compared to untreated controls. Fractures treated with siRNA/NP hydrogels exhibited accelerated bone formation and significantly increased biomechanical strength. This NP/hydrogel siRNA delivery system has outstanding therapeutic promise to augment fracture healing. Owing to the structural similarities of siRNA, the development of the hydrogel platform for in vivo siRNA delivery has myriad therapeutic possibilities in orthopaedics and beyond.
Collapse
|
19
|
Mathew AP, Cho KH, Uthaman S, Cho CS, Park IK. Stimuli-Regulated Smart Polymeric Systems for Gene Therapy. Polymers (Basel) 2017; 9:E152. [PMID: 30970831 PMCID: PMC6432211 DOI: 10.3390/polym9040152] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/02/2023] Open
Abstract
The physiological condition of the human body is a composite of different environments, each with its own parameters that may differ under normal, as well as diseased conditions. These environmental conditions include factors, such as pH, temperature and enzymes that are specific to a type of cell, tissue or organ or a pathological state, such as inflammation, cancer or infection. These conditions can act as specific triggers or stimuli for the efficient release of therapeutics at their destination by overcoming many physiological and biological barriers. The efficacy of conventional treatment modalities can be enhanced, side effects decreased and patient compliance improved by using stimuli-responsive material that respond to these triggers at the target site. These stimuli or triggers can be physical, chemical or biological and can be internal or external in nature. Many smart/intelligent stimuli-responsive therapeutic gene carriers have been developed that can respond to either internal stimuli, which may be normally present, overexpressed or present in decreased levels, owing to a disease, or to stimuli that are applied externally, such as magnetic fields. This review focuses on the effects of various internal stimuli, such as temperature, pH, redox potential, enzymes, osmotic activity and other biomolecules that are present in the body, on modulating gene expression by using stimuli-regulated smart polymeric carriers.
Collapse
Affiliation(s)
- Ansuja Pulickal Mathew
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Ki-Hyun Cho
- Department of Plastic Surgery, Institute of Dermatology and Plastic Surgery, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA.
| | - Saji Uthaman
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| |
Collapse
|
20
|
Liu Z, Sun Q, Wang X. PLK1, A Potential Target for Cancer Therapy. Transl Oncol 2016; 10:22-32. [PMID: 27888710 PMCID: PMC5124362 DOI: 10.1016/j.tranon.2016.10.003] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
Polo-like kinase 1 (PLK1) plays an important role in the initiation, maintenance, and completion of mitosis. Dysfunction of PLK1 may promote cancerous transformation and drive its progression. PLK1 overexpression has been found in a variety of human cancers and was associated with poor prognoses in cancers. Many studies have showed that inhibition of PLK1 could lead to death of cancer cells by interfering with multiple stages of mitosis. Thus, PLK1 is expected to be a potential target for cancer therapy. In this article, we examined PLK1’s structural characteristics, its regulatory roles in cell mitosis, PLK1 expression, and its association with survival prognoses of cancer patients in a wide variety of cancer types, PLK1 interaction networks, and PLK1 inhibitors under investigation. Finally, we discussed the key issues in the development of PLK1-targeted cancer therapy.
Collapse
Affiliation(s)
- Zhixian Liu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingrong Sun
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
21
|
Malcolm DW, Sorrells JE, Van Twisk D, Thakar J, Benoit DSW. Evaluating side effects of nanoparticle-mediated siRNA delivery to mesenchymal stem cells using next generation sequencing and enrichment analysis. Bioeng Transl Med 2016; 1:193-206. [PMID: 27981244 PMCID: PMC5125403 DOI: 10.1002/btm2.10035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
RNA interference has immense potential to modulate cell functions. However, effective delivery of small interfering RNA (siRNA) while avoiding deleterious side effects has proven challenging. This study investigates both intended and unintended effects of diblock copolymer nanoparticle (NP) delivery of siRNA delivery to human mesenchymal stem cells (hMSC). Specifically, siRNA delivery was investigated at a range of NP‐siRNA:hMSC ratios with a focus on the effects of NP‐siRNA treatment on hMSC functions. Additionally, next generation RNA sequencing (RNAseq) was used with enrichment analysis to observe side effects in hMSC gene expression. Results show NP‐siRNA delivery is negatively correlated with hMSC density. However, higher NP‐siRNA:hMSC ratios increased cytotoxicity and decreased metabolic activity. hMSC proliferation was largely unaffected by NP‐siRNA treatment, except for a threefold reduction in hMSCs seeded at 4,000 cells/cm2. Flow cytometry reveals that apoptosis is a function of NP‐siRNA treatment time and seeding density; ∼14% of the treated hMSCs seeded at 8,000 cells/cm2 were annexin V+‐siRNA+ 24 hr after treatment, while 11% of the treated population was annexin V+‐siRNA−. RNAseq shows that NP‐siRNA treatment results in transcriptomic changes in hMSCs, while pathway analysis shows upregulation of apoptosis signaling and downregulation of metabolism, cell cycle, and DNA replication pathways, as corroborated by apoptosis, metabolism, and proliferation assays. Additionally, multiple innate immune signaling pathways such as toll‐like receptor, RIG‐I‐like receptor, and nuclear factor‐κB signaling pathways are upregulated. Furthermore, and consistent with traditional siRNA immune activation, cytokine–cytokine receptor signaling was also upregulated. Overall, this study provides insight into NP‐siRNA:hMSC ratios that are favorable for siRNA delivery. Moreover, NP‐siRNA delivery results in side effects across the hMSC transcriptome that suggest activation of the innate immunity that could alter MSC functions associated with their therapeutic potential.
Collapse
Affiliation(s)
- Dominic W Malcolm
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY14642
| | - Janet E Sorrells
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627
| | - Daniel Van Twisk
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627
| | - Juilee Thakar
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627; Dept. of Biostatistics and Computational Biology University of Rochester Rochester NY 14642
| | - Danielle S W Benoit
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY 14642; Dept. of Chemical Engineering University of Rochester Rochester NY 14627
| |
Collapse
|
22
|
Fan Q, Cai Q, Xu Y. FOXM1 is a downstream target of LPA and YAP oncogenic signaling pathways in high grade serous ovarian cancer. Oncotarget 2016; 6:27688-99. [PMID: 26299613 PMCID: PMC4695018 DOI: 10.18632/oncotarget.4280] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/01/2015] [Indexed: 01/21/2023] Open
Abstract
Lysophosphatidic acid (LPA), a prototypical ligand for G protein coupled receptors, and Forkhead box protein M1 (FOXM1), a transcription factor that regulates expression of a wide array of genes involved in cancer initiation and progression, are two important oncogenic signaling molecules in human epithelial ovarian cancers (EOC). We conducted in vitro mechanistic studies using pharmacological inhibitors, genetic forms of the signaling molecules, and RNAi-mediated gene knock-down to uncover the molecular mechanisms of how these two molecules interact in EOC cells. Additionally, in vivo mouse studies were performed to confirm the functional involvement of FOXM1 in EOC tumor formation and progression. We show for the first time that LPA up-regulates expression of active FOXM1 splice variants in a time- and dose-dependent manner in the human EOC cell lines OVCA433, CAOV3, and OVCAR5. Gi-PI3K-AKT and G12/13-Rho-YAP signaling pathways were both involved in the LPA receptor (LPA1-3) mediated up-regulation of FOXM1 at the transcriptional level. In addition, down-regulation of FOXM1 in CAOV3 xenografts significantly reduced tumor and ascites formation, metastasis, and expression of FOXM1 target genes involved in cell proliferation, migration, or invasion. Collectively, our data link the oncolipid LPA, the oncogene YAP, and the central regulator of cell proliferation/mutagenesis FOXM1 in EOC cells. Moreover, these results provide further support for the importance of these pathways as potential therapeutic targets in EOC.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
23
|
Zhou J, Horev B, Hwang G, Klein MI, Koo H, Benoit DSW. Characterization and optimization of pH-responsive polymer nanoparticles for drug delivery to oral biofilms. J Mater Chem B 2015; 4:3075-3085. [PMID: 27429754 DOI: 10.1039/c5tb02054a] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We previously reported on cationic, pH-responsive p(DMAEMA)-b-p(DMAEMA-co-BMA-co-PAA) block copolymer micelles with high affinity for dental and biofilm surfaces and efficient anti-bacterial drug release in response to acidic pH, characteristic of cariogenic (tooth-decay causing) biofilm microenvironments. Here, we show that micelle pH-responsive behaviors can be enhanced through alterations in corona:core molecular weight ratios (CCR). Although similarly stable at physiological pH, upon exposure to acidic pH, micelles with CCR of 4.1 were less stable than other CCR examined. Specifically, a ~1.5-fold increase in critical micelle concentration (CMC) and ~50% decrease in micelle diameters were observed for micelles with CCR of 4.1, compared to no changes in micelles with CCR of 0.8. While high CCR was shown to enhance pH-responsive drug release, it did not alter drug loading and dental surface binding of micelles. Diblocks were shown to encapsulate the antibacterial drug, farnesol, at maximal loading capacities of up to ~27 wt% and at >94% efficiencies, independent of CCR or core size, resulting in micelle diameter increases due to contributions of drug volume. Additionally, micelles with small diameters (~17 nm) show high binding capacity to hydroxyapatite and dental pellicle emulating surfaces based on Langmuir fit analyses of binding data. Finally, micelles with high CCR that have enhanced pH-responsive drug release and binding were shown to exhibit greater antibiofilm efficacy in situ. Overall, these data demonstrate how factors essential for nanoparticle carrier (NPC)-mediated drug deliverycan be enhanced via modification of diblock characteristics, resulting in greater antibiofilm efficacy in situ.
Collapse
Affiliation(s)
- Jiayi Zhou
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Benjamin Horev
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Geelsu Hwang
- Biofilm Research Lab, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marlise I Klein
- Department of Dental Materials and Prosthodontics, Araraquara Dental School, Univ Estadual Paulista, UNESP, Sao Paulo, Brazil
| | - Hyun Koo
- Biofilm Research Lab, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthodontics and Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Horev B, Klein MI, Hwang G, Li Y, Kim D, Koo H, Benoit DS. pH-activated nanoparticles for controlled topical delivery of farnesol to disrupt oral biofilm virulence. ACS NANO 2015; 9:2390-404. [PMID: 25661192 PMCID: PMC4395463 DOI: 10.1021/nn507170s] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Development of effective therapies to control oral biofilms is challenging, as topically introduced agents must avoid rapid clearance from biofilm-tooth interfaces while targeting biofilm microenvironments. Additionally, exopolysaccharides-matrix and acidification of biofilm microenvironments are associated with cariogenic (caries-producing) biofilm virulence. Thus, nanoparticle carriers capable of binding to hydroxyapatite (HA), saliva-coated HA (sHA), and exopolysaccharides with enhanced drug release at acidic pH were developed. Nanoparticles are formed from diblock copolymers composed of 2-(dimethylamino)ethyl methacrylate (DMAEMA), butyl methacrylate (BMA), and 2-propylacrylic acid (PAA) (p(DMAEMA)-b-p(DMAEMA-co-BMA-co-PAA)) that self-assemble into ∼21 nm cationic nanoparticles. Nanoparticles exhibit outstanding adsorption affinities (∼244 L-mmol(-1)) to negatively charged HA, sHA, and exopolysaccharide-coated sHA due to strong electrostatic interactions via multivalent tertiary amines of p(DMAEMA). Owing to hydrophobic cores, nanoparticles load farnesol, a hydrophobic antibacterial drug, at ∼22 wt %. Farnesol release is pH-dependent with t1/2 = 7 and 15 h for release at pH 4.5 and 7.2, as nanoparticles undergo core destabilization at acidic pH, characteristic of cariogenic biofilm microenvironments. Importantly, topical applications of farnesol-loaded nanoparticles disrupted Streptococcus mutans biofilms 4-fold more effectively than free farnesol. Mechanical stability of biofilms treated with drug-loaded nanoparticles was compromised, resulting in >2-fold enhancement in biofilm removal under shear stress compared to free farnesol and controls. Farnesol-loaded nanoparticles effectively attenuated biofilm virulence in vivo using a clinically relevant topical treatment regimen (2×/day) in a rodent dental caries disease model. Strikingly, treatment with farnesol-loaded nanoparticles reduced both the number and severity of carious lesions, while free farnesol had no effect. Nanoparticle carriers have great potential to enhance the efficacy of antibiofilm agents through multitargeted binding and pH-responsive drug release due to microenvironmental triggers.
Collapse
Affiliation(s)
- Benjamin Horev
- Department of Biomedical Engineering, University of Rochester, NY 14627, United States
| | - Marlise I. Klein
- Center for Oral Biology, University of Rochester, NY 14627, United States
| | - Geelsu Hwang
- Biofilm Research Lab, Levy Center for Oral Health, University of Pennsylvania, PA 19104, United States
| | - Yong Li
- Biofilm Research Lab, Levy Center for Oral Health, University of Pennsylvania, PA 19104, United States
| | - Dongyeop Kim
- Biofilm Research Lab, Levy Center for Oral Health, University of Pennsylvania, PA 19104, United States
| | - Hyun Koo
- Center for Oral Biology, University of Rochester, NY 14627, United States
- Biofilm Research Lab, Levy Center for Oral Health, University of Pennsylvania, PA 19104, United States
- Department of Orthodontics and Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, PA 19104, United States
- Address correspondence to: ;
| | - Danielle S.W. Benoit
- Department of Biomedical Engineering, University of Rochester, NY 14627, United States
- Department of Chemical Engineering, University of Rochester, NY 14627, United States
- Center of Musculoskeletal Research, University of Rochester, NY 14627, United States
| |
Collapse
|
25
|
Qian J, Berkland C. pH-sensitive triblock copolymers for efficient siRNA encapsulation and delivery. Polym Chem 2015. [DOI: 10.1039/c5py00219b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A pH-sensitive triblock copolymer was synthesized for efficient siRNA encapsulation by double emulsion and the formed nanocapsules showed successful delivery of siRNA in vitro.
Collapse
Affiliation(s)
- Jian Qian
- Department of Pharmaceutical Chemistry
- The University of Kansas
- Lawrence
- USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry
- The University of Kansas
- Lawrence
- USA
- Department of Chemical and Petroleum Engineering
| |
Collapse
|
26
|
Chen M, Zhu X, Yan D. A controlled release system for simultaneous promotion of gene transfection and antitumor effects. RSC Adv 2014. [DOI: 10.1039/c4ra10447a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
27
|
Li J, Zhang Y, Gao Y, Cui Y, Liu H, Li M, Tian Y. Downregulation of HNF1 homeobox B is associated with drug resistance in ovarian cancer. Oncol Rep 2014; 32:979-88. [PMID: 24968817 DOI: 10.3892/or.2014.3297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 05/29/2014] [Indexed: 11/05/2022] Open
Abstract
The expression of HNF1 homeobox B (HNF1B) is associated with cancer risk in several tumors, including ovarian cancer, and its decreased expression play roles in cancer development. However, the study of HNF1B and cancer is limited, and its association with drug resistance in cancer has never been reported. On the basis of array data retrieved from Oncomine and Gene Expression Omnibus (GEO) online database, we found that the mRNA expression of HNF1B in 586 ovarian serous cystadenocarcinomas and in platinum-resistant A2780 epithelial ovarian cancer cells was significantly decreased, indicating a potential role of HNF1B in drug resistance in ovarian cancer. Based on this finding, comprehensive bioinformatics analyses, including protein/gene interaction, protein-small molecule/chemical interaction, biological process annotation, gene co-occurrence and pathway enrichment analysis and microRNA-mRNA interaction, were performed to illustrate the association of HNF1B with drug resistance in ovarian cancer. We found that among the proteins/genes, small molecules/chemicals and microRNAs which directly interacted with HNF1B, the majority was associated with drug resistance in cancer, particularly in ovarian cancer. Biological process annotation revealed that HNF1B closely related to 24 biological processes which were all notably associated with ovarian cancer and drug resistance. These results indicated that the downregulation of HNF1B may contribute to drug resistance in ovarian cancer, via its direct interactions with these drug resistance-related proteins/genes, small molecules/chemicals and microRNAs, and via its regulations on the drug resistance-related biological processes. Pathway enrichment analysis of 36 genes which co-occurred with HNF1B, ovarian cancer and drug resistance indicated that the HNF1B may perform its drug resistance-related functions through 4 pathways including ErbB signaling, focal adhesion, apoptosis and p53 signaling. Collectively, in this study, we illustrated for the first time that HNF1B may contribute to drug resistance in ovarian cancer, potentially through the 4 pathways. The present study may pave the way for further investigation of the drug resistance-related functions of HNF1B in ovarian cancer.
Collapse
Affiliation(s)
- Jianchao Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Yonghong Zhang
- Department of Obstetrics and Gynecology, Muping Traditional Chinese Medicine Hospital, Yantai, Shandong, P.R. China
| | - Yutao Gao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Affiliated to Capital Medical University, Beijing, P.R. China
| | - Yuqian Cui
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, P.R. China
| | - Mi Li
- Department of Nursing, Shandong College of Traditional Chinese Medicine, Yantai, Shandong, P.R. China
| | - Yongjie Tian
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
28
|
Jhaveri AM, Torchilin VP. Multifunctional polymeric micelles for delivery of drugs and siRNA. Front Pharmacol 2014; 5:77. [PMID: 24795633 PMCID: PMC4007015 DOI: 10.3389/fphar.2014.00077] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 03/31/2014] [Indexed: 12/18/2022] Open
Abstract
Polymeric micelles, self-assembling nano-constructs of amphiphilic copolymers with a core-shell structure have been used as versatile carriers for delivery of drugs as well as nucleic acids. They have gained immense popularity owing to a host of favorable properties including their capacity to effectively solubilize a variety of poorly soluble pharmaceutical agents, biocompatibility, longevity, high stability in vitro and in vivo and the ability to accumulate in pathological areas with compromised vasculature. Moreover, additional functions can be imparted to these micelles by engineering their surface with various ligands and cell-penetrating moieties to allow for specific targeting and intracellular accumulation, respectively, to load them with contrast agents to confer imaging capabilities, and incorporating stimuli-sensitive groups that allow drug release in response to small changes in the environment. Recently, there has been an increasing trend toward designing polymeric micelles which integrate a number of the above functions into a single carrier to give rise to “smart,” multifunctional polymeric micelles. Such multifunctional micelles can be envisaged as key to improving the efficacy of current treatments which have seen a steady increase not only in hydrophobic small molecules, but also in biologics including therapeutic genes, antibodies and small interfering RNA (siRNA). The purpose of this review is to highlight recent advances in the development of multifunctional polymeric micelles specifically for delivery of drugs and siRNA. In spite of the tremendous potential of siRNA, its translation into clinics has been a significant challenge because of physiological barriers to its effective delivery and the lack of safe, effective and clinically suitable vehicles. To that end, we also discuss the potential and suitability of multifunctional polymeric micelles, including lipid-based micelles, as promising vehicles for both siRNA and drugs.
Collapse
Affiliation(s)
- Aditi M Jhaveri
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University Boston, MA, USA
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University Boston, MA, USA
| |
Collapse
|
29
|
Chen CK, Wang Q, Jones CH, Yu Y, Zhang H, Law WC, Lai CK, Zeng Q, Prasad PN, Pfeifer BA, Cheng C. Synthesis of pH-responsive chitosan nanocapsules for the controlled delivery of doxorubicin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:4111-4119. [PMID: 24665861 DOI: 10.1021/la4040485] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Well-defined chitosan nanocapsules (CSNCs) with tunable sizes were synthesized through the interfacial cross-linking of N-maleoyl-functionalized chitosan (MCS) in miniemulsions, and their application in the delivery of doxorubicin (Dox) was investigated. MCS was prepared by the amidation reaction of CS with maleic anhydride in water/DMSO at 65 °C for 20 h. Subsequently, thiol-ene cross-linking was conducted in oil-in-water miniemulsions at room temperature under UV irradiation for 1 h, using MCS as both a surfactant and precursor polymer, 1,4-butanediol bis(3-mercapto-propionate) as a cross-linker, and D-α-tocopheryl poly(ethylene glycol) 1000 succinate as a cosurfactant. With the increase in cosurfactant concentration in the reaction systems, the sizes of the resulting CSNCs decreased steadily. Dox-loaded CSNCs were readily prepared by in situ encapsulation of Dox during miniemulsion cross-linking. With acid-labile β-thiopropionate cross-linkages, the Dox-loaded CSNCs demonstrated a faster release rate under acidic conditions. Relative to free Dox, Dox-loaded CSNCs exhibited enhanced cytotoxicity toward MCF-7 breast cancer cells without any noticeable cytotoxicity from empty CSNCs. The effective delivery of Dox to MCF-7 breast cancer cells via Dox-loaded CSNCs was also observed.
Collapse
Affiliation(s)
- Chih-Kuang Chen
- Department of Chemical and Biological Engineering and ‡Institute for Lasers, Photonics and Biophotonics, University at Buffalo, The State University of New York , Buffalo, New York 14260, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Guidry EN, Farand J, Soheili A, Parish CA, Kevin NJ, Pipik B, Calati KB, Ikemoto N, Waldman JH, Latham AH, Howell BJ, Leone A, Garbaccio RM, Barrett SE, Parmar RG, Truong QT, Mao B, Davies IW, Colletti SL, Sepp-Lorenzino L. Improving the in vivo therapeutic index of siRNA polymer conjugates through increasing pH responsiveness. Bioconjug Chem 2014; 25:296-307. [PMID: 24409989 DOI: 10.1021/bc400442p] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Polymer based carriers that aid in endosomal escape have proven to be efficacious siRNA delivery agents in vitro and in vivo; however, most suffer from cytotoxicity due in part to a lack of selectivity for endosomal versus cell membrane lysis. For polymer based carriers to move beyond the laboratory and into the clinic, it is critical to find carriers that are not only efficacious, but also have margins that are clinically relevant. In this paper we report three distinct categories of polymer conjugates that improve the selectivity of endosomal membrane lysis by relying on the change in pH associated with endosomal trafficking, including incorporation of low pKa heterocycles, acid cleavable amino side chains, or carboxylic acid pH sensitive charge switches. Additionally, we determine the therapeutic index of our polymer conjugates in vivo and demonstrate that the incorporation of pH responsive elements dramatically expands the therapeutic index to 10-15, beyond that of the therapeutic index (less than 3), for polymer conjugates previously reported.
Collapse
Affiliation(s)
- Erin N Guidry
- Department of Process Chemistry and ∥Department of Analytical Chemistry, Merck & Co. Inc. , , Rahway, New Jersey, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jeong UH, Garripelli V, Jo S, Myung CS, Hwang SJ, Kim JK, Park JS. Potential of pH-Sensitive Polymer-Anchored Cationic Liposomes for Combinatorial Anticancer Therapy with Doxorubicin and siRNA. J Drug Deliv Sci Technol 2014. [DOI: 10.1016/s1773-2247(14)50004-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
Meng Q, Yin Q, Li Y. Nanocarriers for siRNA delivery to overcome cancer multidrug resistance. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-6030-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Yin Q, Shen J, Zhang Z, Yu H, Li Y. Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor. Adv Drug Deliv Rev 2013; 65:1699-715. [PMID: 23611952 DOI: 10.1016/j.addr.2013.04.011] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/01/2013] [Accepted: 04/13/2013] [Indexed: 12/15/2022]
Abstract
Multidrug resistance (MDR) is a major obstacle to successful cancer therapy, especially for chemotherapy. The new drug delivery system (DDS) provides promising approaches to reverse MDR, for which the poor cellular uptake and insufficient intracellular drug release remain rate-limiting steps for reaching the drug concentration level within the therapeutic window. Stimulus-coupled drug delivery can control the drug-releasing pattern temporally and spatially, and improve the accumulation of chemotherapeutic agents at targeting sites. In this review, the applications of DDS which is responsive to different types of stimuli in MDR cancer therapy is introduced, and the design, construction, stimuli-sensitivity and the effect to reverse MDR of the stimuli-responsive DDS are discussed.
Collapse
|
34
|
Gomes-da-Silva LC, Ramalho JS, Pedroso de Lima MC, Simões S, Moreira JN. Impact of anti-PLK1 siRNA-containing F3-targeted liposomes on the viability of both cancer and endothelial cells. Eur J Pharm Biopharm 2013; 85:356-64. [PMID: 23659854 DOI: 10.1016/j.ejpb.2013.04.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 04/03/2013] [Accepted: 04/09/2013] [Indexed: 01/31/2023]
Abstract
We have previously described the development of novel sterically stabilized F3-targeted pH-sensitive liposomes, which exhibited the ability to target both cancer and endothelial cells. Herein, the therapeutic potential of those liposomes was assessed upon encapsulation of a siRNA against a well-validated molecular target, PLK1. Treatment of prostate cancer (PC3) and angiogenic endothelial (HMEC-1) cells with F3-targeted liposomes containing anti-PLK1 siRNA resulted in a significant decrease in cell viability, which was mediated by a marked PLK1 silencing, both at the mRNA and protein levels. Furthermore, pre-treatment of PC3 cells with F3-targeted liposomes containing anti-PLK1 siRNA enabled a 3-fold reduction of paclitaxel IC50 and a 2.5-fold augment of the percentage of cancer cells in G2/mitosis arrest, which ultimately culminated in cell death. Overall, the F3-targeted nanocarrier containing an anti-PLK1 siRNA might constitute a valuable system for prostate cancer treatment, either applied in a single schedule or combined with conventional chemotherapy.
Collapse
Affiliation(s)
- Lígia C Gomes-da-Silva
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Portugal
| | | | | | | | | |
Collapse
|
35
|
Huang X, Sevimli SI, Bulmus V. pH-labile sheddable block copolymers by RAFT polymerization: Synthesis and potential use as siRNA conjugates. Eur Polym J 2013. [DOI: 10.1016/j.eurpolymj.2013.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
|
37
|
Ma P, Mumper RJ. Anthracycline Nano-Delivery Systems to Overcome Multiple Drug Resistance: A Comprehensive Review. NANO TODAY 2013; 8:313-331. [PMID: 23888183 PMCID: PMC3718073 DOI: 10.1016/j.nantod.2013.04.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Anthracyclines (doxorubicin, daunorubicin, and idarubicin) are very effective chemotherapeutic drugs to treat many cancers; however, the development of multiple drug resistance (MDR) is one of the major limitations for their clinical applications. Nano-delivery systems have emerged as the novel cancer therapeutics to overcome MDR. Up until now, many anthracycline nano-delivery systems have been developed and reported to effectively circumvent MDR both in-vitro and in-vivo, and some of these systems have even advanced to clinical trials, such as the HPMA-doxorubicin (HPMA-DOX) conjugate. Doxil, a DOX PEGylated liposome formulation, was developed and approved by FDA in 1995. Unfortunately, this formulation does not address the MDR problem. In this comprehensive review, more than ten types of developed anthracycline nano-delivery systems to overcome MDR and their proposed mechanisms are covered and discussed, including liposomes; polymeric micelles, conjugate and nanoparticles; peptide/protein conjugates; solid-lipid, magnetic, gold, silica, and cyclodextrin nanoparticles; and carbon nanotubes.
Collapse
Affiliation(s)
- Ping Ma
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Russell J. Mumper
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| |
Collapse
|
38
|
|
39
|
Abstract
RNA interference (RNAi) drugs have significant therapeutic potential, but delivery systems with appropriate efficacy and toxicity profiles are still needed. Here, we describe a neutral, ampholytic polymeric delivery system based on conjugatable diblock polymer micelles. The diblock copolymer contains a hydrophilic poly[N-(2-hydroxypropyl)methacrylamide-co-N-(2-(pyridin-2-yldisulfanyl)ethyl)methacrylamide) (poly[HPMA-co-PDSMA]) segment to promote aqueous stability and facilitate thiol-disulfide exchange reactions and a second ampholytic block composed of propylacrylic acid (PAA), dimethylaminoethyl methacrylate (DMAEMA), and butyl methacrylate (BMA). The poly[(HPMA-co-PDSMA)-b-(PAA-co-DMAEMA-co-BMA)] was synthesized using reversible addition-fragmentation chain transfer (RAFT) polymerization with an overall molecular weight of 22 000 g/mol and a PDI of 1.88. Dynamic light scattering and fluorescence measurements indicated that the diblock copolymers self-assemble under aqueous conditions to form polymeric micelles with a hydrodynamic radius and critical micelle concentration of 25 nm and 25 μg/mL, respectively. Red blood cell hemolysis experiments show that the neutral hydrophilic micelles have potent membrane destabilizing activity at endosomal pH values. Thiolated siRNA targeting glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was directly conjugated to the polymeric micelles via thiol exchange reactions with the pyridal disulfide groups present in the micelle corona. Maximum silencing activity in HeLa cells was observed at a 1:10 molar ratio of siRNA to polymer following a 48 h incubation period. Under these conditions 90% mRNA knockdown and 65% protein knockdown at 48 h was achieved with negligible toxicity. In contrast the polymeric micelles lacking a pH-responsive endosomalytic segment demonstrated negligible mRNA and protein knockdown under these conditions. The potent mRNA knockdown and excellent biocompatibility of the neutral siRNA conjugates demonstrate the potential utility of this carrier design for delivering therapeutic siRNA drugs.
Collapse
Affiliation(s)
- Brittany B. Lundy
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Anthony Convertine
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Martina Miteva
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Patrick S. Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| |
Collapse
|
40
|
Nanoparticle-mediated gene silencing confers radioprotection to salivary glands in vivo. Mol Ther 2013; 21:1182-94. [PMID: 23511246 DOI: 10.1038/mt.2013.42] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Radiation treatment of head and neck cancers causes irreversible damage of the salivary glands (SG). Here, we introduce a preclinical mouse model for small-interfering RNA (siRNA)-based gene silencing to provide protection of SG from radiation-induced apoptosis. Novel, pH-responsive nanoparticles complexed with siRNAs were introduced into mouse submandibular glands (SMG) by retroductal injection to modulate gene expression in vivo. To validate this approach, we first targeted Nkcc1, an ion transporter that is essential for saliva secretion. Nkcc1 siRNA delivery resulted in efficient knockdown, as quantified at the mRNA and the protein levels, and the functional result of Nkcc1 knockdown phenocopied the severe decrease in saliva secretion, characteristic of the systemic Nkcc1 gene knockout. To establish a strategy to prevent apoptotic cell loss due to radiation damage, siRNAs targeting the proapoptotic Pkcδ gene were administered into SMG before ionizing radiation. Knockdown of Pkcδ not only reduced the number of apoptotic cells during the acute phase of radiation damage, but also markedly improved saliva secretion at 3 months in irradiated animals, indicating that this treatment confers protection from hyposalivation. These results demonstrate that nanoparticle delivery of siRNAs targeting a proapoptotic gene is a localized, nonviral, and effective means of conferring radioprotection to the SGs.
Collapse
|
41
|
Yu SS, Lau CM, Barham WJ, Onishko HM, Nelson CE, Li H, Smith CA, Yull FE, Duvall CL, Giorgio TD. Macrophage-specific RNA interference targeting via "click", mannosylated polymeric micelles. Mol Pharm 2013; 10:975-87. [PMID: 23331322 DOI: 10.1021/mp300434e] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophages represent an important therapeutic target, because their activity has been implicated in the progression of debilitating diseases such as cancer and atherosclerosis. In this work, we designed and characterized pH-responsive polymeric micelles that were mannosylated using "click" chemistry to achieve CD206 (mannose receptor)-targeted siRNA delivery. CD206 is primarily expressed on macrophages and dendritic cells and upregulated in tumor-associated macrophages, a potentially useful target for cancer therapy. The mannosylated nanoparticles improved the delivery of siRNA into primary macrophages by 4-fold relative to the delivery of a nontargeted version of the same carrier (p < 0.01). Further, treatment for 24 h with the mannose-targeted siRNA carriers achieved 87 ± 10% knockdown of a model gene in primary macrophages, a cell type that is typically difficult to transfect. Finally, these nanoparticles were also avidly recognized and internalized by human macrophages and facilitated the delivery of 13-fold more siRNA into these cells than into model breast cancer cell lines. We anticipate that these mannose receptor-targeted, endosomolytic siRNA delivery nanoparticles will become an enabling technology for targeting macrophage activity in various diseases, especially those in which CD206 is upregulated in macrophages present within the pathologic site. This work also establishes a generalizable platform that could be applied for "click" functionalization with other targeting ligands to direct siRNA delivery.
Collapse
Affiliation(s)
- Shann S Yu
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37235, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yin F, Liu X, Li D, Wang Q, Zhang W, Li L. Bioinformatic analysis of chemokine (C-C motif) ligand 21 and SPARC-like protein 1 revealing their associations with drug resistance in ovarian cancer. Int J Oncol 2013; 42:1305-16. [PMID: 23404140 DOI: 10.3892/ijo.2013.1819] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/14/2013] [Indexed: 11/06/2022] Open
Abstract
Chemokine (C-C motif) ligand 21 (CCL21) and SPARC-like protein 1 (SPARCL1/MAST9/hevin/SC-1) are associated with various biological behavior in the development of cancers. Although the expression of CCL21 and SPARCL1 is downregulated in many solid tumors, their roles in ovarian cancer and their associations with drug resistance have rarely been studied. We performed a comprehensive bioinformatic analysis consisting of motif analysis, literature co-occurrence, gene/protein-gene/protein interaction network, protein-small molecule interaction network, and microRNAs enrichments which revealed that CCL21 and SPARCL1 directly or indirectly interact with a number of genes, proteins, small molecules and pathways associated with drug resistance in ovarian and other cancers. These results suggested that CCL21 and SPARCL1 may contribute to drug resistance in ovarian cancer. This study provided important information for further investigation of drug resistance-related functions of CCL21 and SPARCL1 in ovarian cancer.
Collapse
Affiliation(s)
- Fuqiang Yin
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | | | | | | | | | | |
Collapse
|
43
|
Yaşayan G, Magnusson JP, Sicilia G, Spain SG, Allen S, Davies MC, Alexander C. Multi-modal switching in responsive DNA block co-polymer conjugates. Phys Chem Chem Phys 2013; 15:16263-74. [DOI: 10.1039/c3cp52243a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
44
|
Lee JS, Kim HY, Jeong NY, Lee SY, Yoon YG, Choi YH, Yan C, Chu IS, Koh H, Park HT, Yoo YH. Expression of αB-crystallin overrides the anti-apoptotic activity of XIAP. Neuro Oncol 2012; 14:1332-45. [PMID: 23074197 PMCID: PMC3480270 DOI: 10.1093/neuonc/nos247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 08/15/2012] [Indexed: 11/14/2022] Open
Abstract
Although crystallins are major structural proteins in the lens, α-crystallins perform non-lens functions, and αB-crystallin has been shown to act as an anti-apoptotic mediator in various cells. The present study was undertaken to examine whether αB-crystallin expressed in human malignant glioma cells exerts anti-apoptotic activity. In addition, we sought to elucidate the mechanism underlying any observed anti-apoptotic function of αB-crystallin in these cells. Three glioma cell lines, U373MG, U118MG, and T98G, were used. We observed that only the U373MG cell line expresses αB-crystallin, whereas the other 2 glioma cell lines, U118MG and T98G, demonstrated no endogenous expression of αB-crystallin. We next observed that the silencing of αB-crystallin sensitized U373MG cells to suberoylanilide hydroxamic acid (SAHA)-induced apoptosis and that αB-crystallin associates with caspase-3 and XIAP. Because XIAP is the most potent suppressor of mammalian apoptosis through the direct binding with caspases, we assessed whether XIAP also plays an anti-apoptotic role in SAHA-induced apoptosis in αB-crystallin-expressing U373MG cells. Of note, the silencing of XIAP did not alter the amount of cell death induced by SAHA, indicating that XIAP does not exert an anti-apoptotic activity in U373MG cells. We then determined whether the ectopic expression of αB-crystallin in glioma cells caused a loss of the anti-apoptotic activity of XIAP. Accordingly, we established 2 αB-crystallin over-expressing glioma cell lines, U118MG and T98G, and found that the silencing of XIAP did not sensitize these cells to SAHA-induced apoptosis. These findings suggest that αB-crystallin expressed in glioma cells overrides the anti-apoptotic activity exerted by XIAP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Young Hyun Yoo
- Department of Anatomy and Cell Biology and Mitochondria Hub Regulation Center, College of Medicine, Dong-A University, Busan, Republic of Korea (J.S.L., H.Y.K., N.Y.J, Y.G.Y., Y.H.Y.); Department of Rheumatology, College of Medicine, Dong-A University, Busan, Republic ofKorea (S.Y.L.); Department of Biochemistry and Research Institute of Oriental Medicine, Dongeui University College of Oriental Medicine, Busan, Republic of Korea (Y.H.C.); Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang, China (C.Y.); Korean Bioinformation Center, KRIBB, Daejeon, Republic ofKorea (I.C.); Department of Pharmacology and Mitochondria Hub Regulation Center, College of Medicine, Dong-A University, Busan, Republic of Korea (H.K.); Department of Physiology, Medical Science Research Institute, College of Medicine, Dong-A University, Busan, South Korea (H.T.P.)
| |
Collapse
|
45
|
Cao P, Bae Y. Polymer nanoparticulate drug delivery and combination cancer therapy. Future Oncol 2012; 8:1471-80. [DOI: 10.2217/fon.12.139] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This review describes the scientific background, current achievement and future perspective of combination therapy using polymer nanoparticle drug carriers in cancer treatment. Nanotechnology-based drug delivery is expected to dramatically change combination cancer therapy by controlling accumulation and distribution patterns of multiple drugs selectively in disease sites. Rationally designed polymer materials can produce functional nanoparticulate drug carriers that can be used in various biomedical applications. In comparison with conventional drug combination approaches, using polymer nanoparticle drug carriers appears to suppress tumor growth more efficiently, potentially overcoming multidrug resistance in many cancers. It also provides versatile combination options for a variety of therapeutic agents, molecular targeting agents and nucleotide drugs.
Collapse
Affiliation(s)
- Pengxiao Cao
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Younsoo Bae
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
46
|
Schallon A, Synatschke CV, Jérôme V, Müller AHE, Freitag R. Nanoparticulate nonviral agent for the effective delivery of pDNA and siRNA to differentiated cells and primary human T lymphocytes. Biomacromolecules 2012; 13:3463-74. [PMID: 23020076 DOI: 10.1021/bm3012055] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Delivery of polynucleotides such as plasmid DNA (pDNA) and siRNA to nondividing and primary cells by nonviral vectors presents a considerable challenge. In this contribution, we introduce a novel type of PDMAEMA-based star-shaped nanoparticles that (i) are efficient transfection agents in clinically relevant and difficult-to-transfect human cells (Jurkat T cells, primary T lymphocytes) and (ii) can efficiently deliver siRNA to human primary T lymphocytes resulting to more than 40% silencing of the targeted gene. Transfection efficiencies achieved by the new vectors in serum-free medium are generally high and only slightly reduced in the presence of serum, while cytotoxicity and cell membrane disruptive potential at physiological pH are low. Therefore, these novel agents are expected to be promising carriers for nonviral gene transfer. Moreover, we propose a general design principle for the construction of polycationic nanoparticles capable of delivering nucleic acids to the above-mentioned cells.
Collapse
Affiliation(s)
- Anja Schallon
- Process Biotechnology, University of Bayreuth, 95440 Bayreuth, Germany
| | | | | | | | | |
Collapse
|
47
|
Benoit DSW, Boutin ME. Controlling mesenchymal stem cell gene expression using polymer-mediated delivery of siRNA. Biomacromolecules 2012; 13:3841-9. [PMID: 23020123 DOI: 10.1021/bm301294n] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
siRNA treatment has great promise to specifically control gene expression and select cell behaviors but has delivery challenges limiting its use. Particularly for applications in regenerative medicine, uniform and consistent delivery of siRNA to control gene expression and subsequent stem cell functions, such as differentiation, is paramount. Therefore, a diblock copolymer was examined for its ability to effectively deliver siRNA to mesenchymal stem cells (MSCs). The diblock copolymers, which are composed of cationic blocks for siRNA complexation, protection, and uptake and pH-responsive blocks for endosomal escape, were shown to facilitate nearly 100% MSC uptake of siRNA. This is vastly superior to a commercially available control, DharmaFECT, which resulted in only ~60% siRNA positive MSCs. Moreover, the diblock copolymer, at conditions that result in excellent knockdown (down to ~10% of control gene expression), was cytocompatible, causing no negative effects on MSC survivability. In contrast, DharmaFECT/siRNA treatment resulted in only ~60% survivability of MSCs. Longitudinal knockdown after siRNA treatment was examined and protein knockdown persists for ~6 days regardless of delivery system (diblock copolymer or DharmaFECT). Finally, MSC phenotype and differentiation capacity was examined after treatment with control siRNA. There was no statistically significant differences on cell surface markers of diblock copolymer/siRNA or DharmaFECT/siRNA-treated or cells measured 2 weeks after siRNA delivery compared to untreated cells. Upon differentiation with typical media/culture conditions to adipogenic, chondrogenic, and osteogenic lineages and examination of histological staining markers, there was no discernible differences between treated and untreated cells, regardless of delivery mechanism. Thus, diblock copolymers examined herein facilitated uniform siRNA treatment of MSCs, inducing siRNA-specific gene and protein knockdown without adversely affecting MSC survival or differentiation capacity and therefore show great promise for use within regenerative medicine applications.
Collapse
Affiliation(s)
- Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester Medical Center, University of Rochester, Rochester, NY 14627, USA.
| | | |
Collapse
|
48
|
Arany S, Xu Q, Hernady E, Benoit DSW, Dewhurst S, Ovitt CE. Pro-apoptotic gene knockdown mediated by nanocomplexed siRNA reduces radiation damage in primary salivary gland cultures. J Cell Biochem 2012; 113:1955-65. [PMID: 22253051 DOI: 10.1002/jcb.24064] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A critical issue in the management of head and neck tumors is radioprotection of the salivary glands. We have investigated whether siRNA-mediated gene knock down of pro-apoptotic mediators can reduce radiation-induced cellular apoptosis in salivary gland cells in vitro. We used novel, pH-responsive nanoparticles to deliver functionally active siRNAs into cultures of salivary gland cells. The nanoparticle molecules are comprised of cationic micelles that electrostatically interact with the siRNA, protecting it from nuclease attack, and also include pH-responsive endosomolytic constituents that promote release of the siRNA into the target cell cytoplasm. Transfection controls with Cy3-tagged siRNA/nanoparticle complexes showed efficiently internalized siRNAs in more than 70% of the submandibular gland cells. We found that introduction of siRNAs specifically targeting the Pkcδ or Bax genes significantly blocked the induction of these pro-apoptotic proteins that normally occurs after radiation in cultured salivary gland cells. Furthermore, the level of cell death from subsequent radiation, as measured by caspase-3, TUNEL, and mitochondrial disruption assays, was significantly decreased. Thus, we have successfully demonstrated that the siRNA/nanoparticle-mediated knock down of pro-apoptotic genes can prevent radiation-induced damage in submandibular gland primary cell cultures.
Collapse
Affiliation(s)
- Szilvia Arany
- Center for Oral Biology, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Sevimli S, Inci F, Zareie HM, Bulmus V. Well-Defined Cholesterol Polymers with pH-Controlled Membrane Switching Activity. Biomacromolecules 2012; 13:3064-75. [DOI: 10.1021/bm300846e] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
| | - Fatih Inci
- Department of Molecular
Biology-Genetics and Biotechnology Program (MOBGAM), Istanbul Technical University, Istanbul 34469, Turkey
| | - Hadi M. Zareie
- Department of Electrical-Electronics
Engineering and Nanotechnology Graduate Program, Gediz University, Izmir 35665, Turkey
- Microstructural
Analysis Unit, School of Physics and Advanced Materials, University of Technology, Sydney, Altimo NSW 2007,
Australia
| | | |
Collapse
|
50
|
Chiang CS, Fu SY, Wang SC, Yu CF, Chen FH, Lin CM, Hong JH. Irradiation promotes an m2 macrophage phenotype in tumor hypoxia. Front Oncol 2012; 2:89. [PMID: 22888475 PMCID: PMC3412458 DOI: 10.3389/fonc.2012.00089] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/19/2012] [Indexed: 11/16/2022] Open
Abstract
Macrophages display different phenotypes with distinct functions and can rapidly respond to environmental changes. Previous studies on TRAMP-C1 tumor model have shown that irradiation has a strong impact on tumor microenvironments. The major changes include the decrease of microvascular density, the increase of avascular hypoxia, and the aggregation of tumor-associated macrophages in avascular hypoxic regions. Similar changes were observed no matter the irradiation was given to tissue bed before tumor implantation (pre-IR tumors), or to established tumors (IR tumors). Recent results on three murine tumors, TRAMP-C1 prostate adenocarcinoma, ALTS1C1 astrocytoma, and GL261 glioma, further demonstrate that different phenotypes of inflammatory cells are spatially distributed into different microenvironments in both IR and pre-IR tumors. Regions with avascular hypoxia and central necrosis have CD11bhigh/Gr-1+ neutrophils in the center of the necrotic area. Next to them are CD11blow/F4/80+ macrophages that sit at the junctions between central necrotic and surrounding hypoxic regions. The majority of cells in the hypoxic regions are CD11blow/CD68+ macrophages. These inflammatory cell populations express different levels of Arg I. This distribution pattern, except for neutrophils, is not observed in tumors receiving chemotherapy or an anti-angiogenesis agent which also lead to avascular hypoxia. This unique distribution pattern of inflammatory cells in IR tumor sites is interfered with by targeting the expression of a chemokine protein, SDF-1α, by tumor cells, and this also increases radiation-induced tumor growth delay. This indicates that irradiated-hypoxia tissues have distinct tumor microenvironments that favor the development of M2 macrophages and that is affected by the levels of tumor-secreted SDF-1α.
Collapse
Affiliation(s)
- Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University Hsinchu, Taiwan
| | | | | | | | | | | | | |
Collapse
|