1
|
Kumar KP, Madhusoodanan M, Pangath M, Menon D. Innovative landscapes in intraperitoneal therapy of ovarian cancer. Drug Deliv Transl Res 2025; 15:1877-1906. [PMID: 39888579 DOI: 10.1007/s13346-024-01765-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Epithelial ovarian cancer is the most prevalent gynecological malignancy, characterized by high mortality rates due to its late-stage diagnosis and frequent recurrence. The current standard of care for ovarian cancer is a combination of debulking surgery followed by the conventional mode of chemotherapy. Despite significant advances in therapeutic modalities, the overall survival rate of EOC continues to be poor, mainly because low concentrations of the chemotherapeutics reach the peritoneum, which is the primary site of ovarian cancer, leading to disease relapse. Here, intraperitoneal chemotherapy gains advantage due to its ability to deliver the drug molecules directly to the peritoneal cavity and provide localized and sustained effects. This is facilitated by the use of diverse kinds of nano or micron sized delivery systems, which help in transporting drugs, vaccines, antibodies and genes appropriately to the peritoneum for its desired function. This review article delves on how intraperitoneal delivery impacts the therapy of epithelial ovarian cancer spanning the conventional therapeutic modes to the recent nanoinnovations in chemotherapy, immunotherapy and gene therapy.
Collapse
Affiliation(s)
- Krishna Pradeep Kumar
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesha Madhusoodanan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Meghna Pangath
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Deepthy Menon
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
2
|
Gao J, Zhai Y, Lu W, Jiang X, Zhou J, Wu L, Du L, Ou C, Zhang X, He H, Zhu J, Zhang Z, Li M, Wu Y, Pan X. ROS-sensitive PD-L1 siRNA cationic selenide nanogels for self-inhibition of autophagy and prevention of immune escape. Bioact Mater 2024; 41:597-610. [PMID: 39280899 PMCID: PMC11393550 DOI: 10.1016/j.bioactmat.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
In the field of cancer therapy, inhibiting autophagy has emerged as a promising strategy. However, pharmacological disruption of autophagy can lead to the upregulation of programmed death-ligand 1 (PD-L1), enabling tumor immune evasion. To address this issue, we developed innovative ROS-responsive cationic poly(ethylene imine) (PEI) nanogels using selenol chemistry-mediated multicomponent reaction (MCR) technology. This procedure involved simple mixing of low-molecular-weight PEI (LMW PEI), γ-selenobutylacetone (γ-SBL), and poly(ethylene glycol) methacrylate (PEGMA). Through high-throughput screening, we constructed a library of AxSeyOz nanogels and identified the optimized A1.8Se3O0.5/siPD-L1 nanogels, which exhibited a size of approximately 200 nm, excellent colloidal stability, and the most effective PD-L1 silencing efficacy. These nanogels demonstrated enhanced uptake by tumor cells, excellent oxidative degradation ability, and inhibited autophagy by alkalinizing lysosomes. The A1.8Se3O0.5/siPD-L1 nanogels significantly downregulated PD-L1 expression and increased the expression of major histocompatibility complex class I (MHC-I), resulting in robust proliferation of specific CD8+ T cells and a decrease in MC38 tumor growth. As a result, the A1.8Se3O0.5/siPD-L1 nanogels inhibited tumor growth through self-inhibition of autophagy, upregulation of MHC-I, and downregulation of PD-L1. Designed with dynamic diselenide bonds, the A1.8Se3O0.5/siPD-L1 nanogels showed synergistic antitumor efficacy through self-inhibition of autophagy and prevention of immune escape.
Collapse
Affiliation(s)
- Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Yonghua Zhai
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Weihong Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xianghe Jiang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jingsheng Zhou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Chunqing Ou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Hanliang He
- The Department of Orthopedic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215028, China
| | - Jian Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Meiyun Li
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiangqiang Pan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
3
|
Lee SH, Ok SH, Park KE, Bae SI, Hwang Y, Ahn SH, Sim G, Bae M, Sohn JT. Epidermal growth factor receptor phosphorylation contributes to levobupivacaine-induced contraction in isolated rat aorta. Eur J Pharmacol 2024; 967:176389. [PMID: 38311282 DOI: 10.1016/j.ejphar.2024.176389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Vasoconstriction induced by levobupivacaine, a local anesthetic, is mediated by increased levels of calcium, tyrosine kinase, c-Jun NH2-terminal kinase (JNK), and phospholipase D, which are associated with prolonged local anesthesia. Epidermal growth factor receptor (EGFR) phosphorylation is associated with vasoconstriction. However, its role in levobupivacaine-induced contractions remains unknown. We determined whether EGFR phosphorylation is associated with levobupivacaine-induced contractions in isolated rat thoracic aortas and identified the underlying cellular signaling pathways. The effects of various inhibitors and a calcium-free solution alone or in combination on levobupivacaine-induced contractions were then assessed. Furthermore, we examined the effects of various inhibitors on levobupivacaine-induced EGFR and JNK phosphorylation and calcium levels in vascular smooth muscle cells (VSMCs) of rat aortas. The EGFR tyrosine kinase inhibitor AG1478, matrix metalloproteinase (MMP) inhibitor GM6001, Src kinase inhibitors PP1 and PP2, and JNK inhibitor SP600125 attenuated levobupivacaine-induced contractions. Moreover, although the calcium-free solution abolished levobupivacaine-induced contractions, calcium reversed this inhibitory effect. The magnitude of the calcium-mediated reversal of abolished levobupivacaine-induced contractions was lower in the combination treatment with calcium-free solution and AG1478 than in the treatment with calcium-free solution alone. Levobupivacaine induced EGFR and JNK phosphorylation. However, AG1478, GM6001, and PP2 attenuated levobupivacaine-induced EGFR and JNK phosphorylation. Moreover, although levobupivacaine induced JNK phosphorylation in control siRNA-transfected VSMCs, EGFR siRNA inhibited levobupivacaine-induced JNK phosphorylation. Furthermore, AG1478 inhibited levobupivacaine-induced calcium increases in VSMCs. Collectively, these findings suggest that levobupivacaine-induced EGFR phosphorylation, which may occur via the Src kinase-MMP pathway, contributes to vasoconstriction via JNK phosphorylation and increased calcium levels.
Collapse
Affiliation(s)
- Soo Hee Lee
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si, Gyeongsangnam-do, Republic of Korea; Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea
| | - Seong-Ho Ok
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si, Gyeongsangnam-do, Republic of Korea; Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea
| | - Kyeong-Eon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Sung Il Bae
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Yeran Hwang
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Seung Hyun Ahn
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Gyujin Sim
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Moonju Bae
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Ju-Tae Sohn
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea.
| |
Collapse
|
4
|
Qin B, Chen X, Zhu J, Kopechek J, Helfield B, Yu F, Cyriac J, Lavery L, Grandis JR, Villanueva FS. Ultrasound enhanced siRNA delivery using cationic liposome-microbubble complexes for the treatment of squamous cell carcinoma. Nanotheranostics 2024; 8:285-297. [PMID: 38577322 PMCID: PMC10988211 DOI: 10.7150/ntno.90516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Rationale: Microbubble (MB) contrast agents combined with ultrasound targeted microbubble cavitation (UTMC) are a promising platform for site-specific therapeutic oligonucleotide delivery. We investigated UTMC-mediated delivery of siRNA directed against epidermal growth factor receptor (EGFR), to squamous cell carcinoma (SCC) via a novel MB-liposome complex (LPX). Methods: LPXs were constructed by conjugation of cationic liposomes to the surface of C4F10 gas-filled lipid MBs using biotin/avidin chemistry, then loaded with siRNA via electrostatic interaction. Luciferase-expressing SCC-VII cells (SCC-VII-Luc) were cultured in Petri dishes. The Petri dishes were filled with media in which LPXs loaded with siRNA against firefly luciferase (Luc siRNA) were suspended. Ultrasound (US) (1 MHz, 100-µs pulse, 10% duty cycle) was delivered to the dishes for 10 sec at varying acoustic pressures and luciferase assay was performed 24 hr later. In vivo siRNA delivery was studied in SCC-VII tumor-bearing mice intravenously infused with a 0.5 mL saline suspension of EGFR siRNA LPX (7×108 LPX, ~30 µg siRNA) for 20 min during concurrent US (1 MHz, 0.5 MPa spatial peak temporal peak negative pressure, five 100-µs pulses every 1 ms; each pulse train repeated every 2 sec to allow reperfusion of LPX into the tumor). Mice were sacrificed 2 days post treatment and tumor EGFR expression was measured (Western blot). Other mice (n=23) received either EGFR siRNA-loaded LPX + UTMC or negative control (NC) siRNA-loaded LPX + UTMC on days 0 and 3, or no treatment ("sham"). Tumor volume was serially measured by high-resolution 3D US imaging. Results: Luc siRNA LPX + UTMC caused significant luciferase knockdown vs. no treatment control, p<0.05) in SCC-VII-Luc cells at acoustic pressures 0.25 MPa to 0.9 MPa, while no significant silencing effect was seen at lower pressure (0.125 MPa). In vivo, EGFR siRNA LPX + UTMC reduced tumor EGFR expression by ~30% and significantly inhibited tumor growth by day 9 (~40% decrease in tumor volume vs. NC siRNA LPX + UTMC, p<0.05). Conclusions: Luc siRNA LPXs + UTMC achieved functional delivery of Luc siRNA to SCC-VII-Luc cells in vitro. EGFR siRNA LPX + UTMC inhibited tumor growth and suppressed EGFR expression in vivo, suggesting that this platform holds promise for non-invasive, image-guided targeted delivery of therapeutic siRNA for cancer treatment.
Collapse
Affiliation(s)
- Bin Qin
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xucai Chen
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhui Zhu
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Kopechek
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandon Helfield
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francois Yu
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jissy Cyriac
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Linda Lavery
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Flordeliza S. Villanueva
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Szymanowska A, Rodriguez-Aguayo C, Lopez-Berestein G, Amero P. Non-Coding RNAs: Foes or Friends for Targeting Tumor Microenvironment. Noncoding RNA 2023; 9:52. [PMID: 37736898 PMCID: PMC10514839 DOI: 10.3390/ncrna9050052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a group of molecules critical for cell development and growth regulation. They are key regulators of important cellular pathways in the tumor microenvironment. To analyze ncRNAs in the tumor microenvironment, the use of RNA sequencing technology has revolutionized the field. The advancement of this technique has broadened our understanding of the molecular biology of cancer, presenting abundant possibilities for the exploration of novel biomarkers for cancer treatment. In this review, we will summarize recent achievements in understanding the complex role of ncRNA in the tumor microenvironment, we will report the latest studies on the tumor microenvironment using RNA sequencing, and we will discuss the potential use of ncRNAs as therapeutics for the treatment of cancer.
Collapse
Affiliation(s)
- Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (A.S.); (C.R.-A.); (G.L.-B.)
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (A.S.); (C.R.-A.); (G.L.-B.)
- Center for RNA Interference and Non-Coding RNA, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (A.S.); (C.R.-A.); (G.L.-B.)
- Center for RNA Interference and Non-Coding RNA, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (A.S.); (C.R.-A.); (G.L.-B.)
| |
Collapse
|
7
|
Han Y, Shin SH, Lim CG, Heo YH, Choi IY, Kim HH. Synthetic RNA Therapeutics in Cancer. J Pharmacol Exp Ther 2023; 386:212-223. [PMID: 37188531 DOI: 10.1124/jpet.123.001587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
Recent advances in the RNA delivery system have facilitated the development of a separate field of RNA therapeutics, with modalities including mRNA, microRNA (miRNA), antisense oligonucleotide (ASO), small interfering RNA, and circular (circRNA) that have been incorporated into oncology research. The main advantages of the RNA-based modalities are high flexibility in designing RNA and rapid production for clinical screening. It is challenging to eliminate tumors by tackling a single target in cancer. In the era of precision medicine, RNA-based therapeutic approaches potentially constitute suitable platforms for targeting heterogeneous tumors that possess multiple sub-clonal cancer cell populations. In this review, we discussed how synthetic coding and non-coding RNAs, such as mRNA, miRNA, ASO, and circRNA, can be applied in the development of therapeutics. SIGNIFICANCE STATEMENT: With development of vaccines against coronavirus, RNA-based therapeutics have received attention. Here, the authors discuss different types of RNA-based therapeutics potentially effective against tumor that are highly heterogeneous giving rise to resistance and relapses to the conventional therapeutics. Moreover, this study summarized recent findings suggesting combination approaches of RNA therapeutics and cancer immunotherapy.
Collapse
Affiliation(s)
- Youngjin Han
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| | - Seung-Hyun Shin
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| | - Chang Gyu Lim
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| | - Yong Ho Heo
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| | - In Young Choi
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| | - Ha Hyung Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., Gyeonggi-do, Republic of Korea (Y.H., S.-H.S., C.G.L., Y.H.H., I.Y.C.); and Biotherapeutics and Glycomics Laboratory, College of Pharmacy (Y.H.H., H.H.K.) and Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea (H.H.K.)
| |
Collapse
|
8
|
Li X, Xu X, Xu M, Geng Z, Ji P, Liu Y. Hydrogel systems for targeted cancer therapy. Front Bioeng Biotechnol 2023; 11:1140436. [PMID: 36873346 PMCID: PMC9977812 DOI: 10.3389/fbioe.2023.1140436] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
When hydrogel materials with excellent biocompatibility and biodegradability are used as excellent new drug carriers in the treatment of cancer, they confer the following three advantages. First, hydrogel materials can be used as a precise and controlled drug release systems, which can continuously and sequentially release chemotherapeutic drugs, radionuclides, immunosuppressants, hyperthermia agents, phototherapy agents and other substances and are widely used in the treatment of cancer through radiotherapy, chemotherapy, immunotherapy, hyperthermia, photodynamic therapy and photothermal therapy. Second, hydrogel materials have multiple sizes and multiple delivery routes, which can be targeted to different locations and types of cancer. This greatly improves the targeting of drugs, thereby reducing the dose of drugs and improving treatment effectiveness. Finally, hydrogel can intelligently respond to environmental changes according to internal and external environmental stimuli so that anti-cancer active substances can be remotely controlled and released on demand. Combining the abovementioned advantages, hydrogel materials have transformed into a hit in the field of cancer treatment, bringing hope to further increase the survival rate and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Xinlin Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xinyi Xu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Mengfei Xu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Zhaoli Geng
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Ping Ji
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yi Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| |
Collapse
|
9
|
EGFR-Targeted Cellular Delivery of Therapeutic Nucleic Acids Mediated by Boron Clusters. Int J Mol Sci 2022; 23:ijms232314793. [PMID: 36499115 PMCID: PMC9740766 DOI: 10.3390/ijms232314793] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
New boron carriers with high boron content and targeted cancer-cell delivery are considered the first choice for boron neutron capture therapy (BNCT) for cancer treatment. Previously, we have shown that composites of antisense oligonucleotide and boron clusters are functional nanoparticles for the downregulation of expression of epidermal growth factor receptor (EGFR) and can be loaded into EGFR-overexpressing cancer cells without a transfection factor. In this study, we hypothesize that free cellular uptake is mediated by binding and activation of the EGFR by boron clusters. Proteomic analysis of proteins pulled-down from various EGFR-overexpressing cancer cells using short oligonucleotide probes, conjugated to 1,2-dicarba-closo-dodecaborane (1,2-DCDDB, [C2B10H12]) and [(3,3'-Iron-1,2,1',2'-dicarbollide)-] (FESAN, [Fe(C2B9H11)2]-), evidenced that boron cage binds to EGFR subdomains. Moreover, inductively coupled plasma mass spectrometry (ICP MS) and fluorescence microscopy analyses confirmed that FESANs-highly decorated B-ASOs were efficiently delivered and internalized by EGFR-overexpressing cells. Antisense reduction of EGFR in A431 and U87-MG cells resulted in decreased boron accumulation compared to control cells, indicating that cellular uptake of B-ASOs is related to EGFR-dependent internalization. The data obtained suggest that EGFR-mediated cellular uptake of B-ASO represents a novel strategy for cellular delivery of therapeutic nucleic acids (and possibly other medicines) conjugated to boron clusters.
Collapse
|
10
|
Abstract
Viruses are intracellular pathogen that exploit host cellular machinery for their propagation. Extensive research on virus-host interaction have shed light on an alternative antiviral strategy that targets host cell factors. Epidermal growth factor receptor (EGFR) is a versatile signal transducer that is involved in a range of cellular processes. Numerous studies have revealed how viruses exploit the function of EGFR in different stages of viral life cycle. In general, viruses attach onto the host cell surface and interacts with EGFR to facilitate viral entry, viral replication and spread as well as evasion from host immunosurveillance. Moreover, virus-induced activation of EGFR signalling is associated with mucin expression, tissue damage and carcinogenesis that contribute to serious complications. Herein, we review our current understanding of roles of EGFR in viral infection and its potential as therapeutic target in managing viral infection. We also discuss the available EGFR-targeted therapies and their limitations.
Collapse
Affiliation(s)
- Kah Man Lai
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
11
|
Lee SH, Kwon SC, Ok SH, Ahn SH, Bae SI, Kim JY, Hwang Y, Park KE, Kim M, Sohn JT. Dexmedetomidine-Induced Aortic Contraction Involves Transactivation of the Epidermal Growth Factor Receptor in Rats. Int J Mol Sci 2022; 23:ijms23084320. [PMID: 35457136 PMCID: PMC9024600 DOI: 10.3390/ijms23084320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 01/27/2023] Open
Abstract
In this study, we examined whether aortic contraction, induced by the alpha-2 adrenoceptor agonist dexmedetomidine, is involved in the transactivation of the epidermal growth factor receptor (EGFR) in isolated endothelium-denuded rat aortas. Additionally, we aimed to elucidate the associated underlying cellular mechanisms. The effects of the alpha-2 adrenoceptor inhibitor rauwolscine, EGFR tyrosine kinase inhibitor AG1478, Src kinase inhibitors PP1 and PP2, and matrix metalloproteinase inhibitor GM6001 on EGFR tyrosine phosphorylation and c-Jun NH2-terminal kinase (JNK) phosphorylation induced by dexmedetomidine in rat aortic smooth muscles were examined. In addition, the effects of these inhibitors on dexmedetomidine-induced contraction in isolated endothelium-denuded rat aorta were examined. Dexmedetomidine-induced contraction was inhibited by the alpha-1 adrenoceptor inhibitor prazosin, rauwolscine, AG1478, PP1, PP2, and GM6001 alone or by a combined treatment with prazosin and AG1478. AG1478 (3 × 10−6 M) inhibited dexmedetomidine-induced contraction in isolated endothelium-denuded rat aortas pretreated with rauwolscine. Dexmedetomidine-induced EGFR tyrosine and JNK phosphorylation were inhibited by rauwolscine, PP1, PP2, GM6001, and AG1478. Furthermore, dexmedetomidine-induced JNK phosphorylation reduced upon EGFR siRNA treatment. Therefore, these results suggested that the transactivation of EGFR associated with dexmedetomidine-induced contraction, mediated by the alpha-2 adrenoceptor, Src kinase, and matrix metalloproteinase, caused JNK phosphorylation and increased calcium levels.
Collapse
Affiliation(s)
- Soo Hee Lee
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital 11, Samjeongja-ro, Seongsan-gu, Changwon-si 51472, Gyeongsangnam-do, Korea; (S.H.L.); (S.-H.O.)
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea
- Institute of Health Sciences, Gyeongsang National University, Jinju-si 52727, Gyeongsangnam-do, Korea
| | - Seong-Chun Kwon
- Department of Physiology, Institute of Clinical and Translational Research, Catholic Kwandong University, College of Medicine, Gangneung 25601, Korea;
| | - Seong-Ho Ok
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital 11, Samjeongja-ro, Seongsan-gu, Changwon-si 51472, Gyeongsangnam-do, Korea; (S.H.L.); (S.-H.O.)
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea
- Institute of Health Sciences, Gyeongsang National University, Jinju-si 52727, Gyeongsangnam-do, Korea
| | - Seung Hyun Ahn
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Sung Il Bae
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Yeran Hwang
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Kyeong-Eon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Mingu Kim
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea; (S.H.A.); (S.I.B.); (J.-Y.K.); (Y.H.); (K.-E.P.); (M.K.)
| | - Ju-Tae Sohn
- Institute of Health Sciences, Gyeongsang National University, Jinju-si 52727, Gyeongsangnam-do, Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 beon-gil, Jinju-si 52727, Gyeongsangnam-do, Korea
- Correspondence: ; Tel.: +82-55-750-8586
| |
Collapse
|
12
|
Bose CK. siRNA and Ovarian Cancer. INDIAN JOURNAL OF GYNECOLOGIC ONCOLOGY 2021. [DOI: 10.1007/s40944-021-00583-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
13
|
Vetvicka D, Sivak L, Jogdeo CM, Kumar R, Khan R, Hang Y, Oupický D. Gene silencing delivery systems for the treatment of pancreatic cancer: Where and what to target next? J Control Release 2021; 331:246-259. [PMID: 33482273 DOI: 10.1016/j.jconrel.2021.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
Despite intensive research efforts and development of numerous new anticancer drugs and treatment strategies over the past decades, there has been only very limited improvement in overall patient survival and in effective treatment options for pancreatic cancer. Current chemotherapy improves survival in terms of months and death rates in pancreatic cancer patients are almost equivalent to incidence rates. It is imperative to develop new therapeutic approaches. Among them, gene silencing shows promise of effectiveness in both tumor cells and stromal cells by inhibiting tumor-promoting genes. This review summarizes potential targets for gene silencing in both pancreatic cancer cells and abundant stromal cells focusing on non-viral delivery systems for small RNAs and discusses the potential immunological implications. The review concludes with the importance of multifactorial therapy of pancreatic cancer.
Collapse
Affiliation(s)
- David Vetvicka
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States; Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovska 1, Prague 2 12000, Czech Republic
| | - Ladislav Sivak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno CZ-61300, Czech Republic
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Rubayat Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
14
|
Diaz-Dussan D, Peng YY, Kumar P, Narain R. Oncogenic Epidermal Growth Factor Receptor Silencing in Cervical Carcinoma Mediated by Dynamic Sugar-Benzoxaborole Polyplexes. ACS Macro Lett 2020; 9:1464-1470. [PMID: 35653664 DOI: 10.1021/acsmacrolett.0c00599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although, various types of pharmaceuticals have been developed for cervical carcinomas, treatment with these drugs often results in a number of undesirable side effects, toxicity and multidrug resistance. Here, we aimed at modifying the genetic profiling of cancer cells by silencing the expression of the epidermal growth factor receptor (EGFR) gene. We have synthesized two kinds of RAFT-made, biocompatible, and cationic polymers for the encapsulation of silencing RNA (siRNA). This vector has a dual capability: it contains a cationic segment to complex with the siRNA and an omega-end modified with an oxaborole group via thiol-ene click chemistry that responds to the acidic tumor microenvironment. This structural innovation enables this macromolecule to interact with multiple polyplexes and release the siRNA in a mild acidic environment. A strategy that has shown enhanced gene silencing without elevating the cytotoxicity of the system, as determined by Western blot analysis. The success of this approach has afforded further interest in utilizing boron-carbohydrate interaction in the development of nonviral vectors for gene therapy.
Collapse
Affiliation(s)
- Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Piyush Kumar
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| |
Collapse
|
15
|
Sun Z, Song C, Wang C, Hu Y, Wu J. Hydrogel-Based Controlled Drug Delivery for Cancer Treatment: A Review. Mol Pharm 2020; 17:373-391. [PMID: 31877054 DOI: 10.1021/acs.molpharmaceut.9b01020] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As an emerging drug carrier, hydrogels have been widely used for tumor drug delivery. A hydrogel drug carrier can cause less severe side effects than systemic chemotherapy and can achieve sustained delivery of a drug at tumor sites. In addition, hydrogels have excellent biocompatibility and biodegradability and lower toxicity than nanoparticle carriers. Smart hydrogels can respond to stimuli in the environment (e.g., heat, pH, light, and ultrasound), enabling in situ gelation and controlled drug release, which greatly enhance the convenience and efficiency of drug delivery. Here, we summarize the different sizes of hydrogels used for cancer treatment and their related delivery routes, discuss the design strategies for stimuli-responsive hydrogels, and review the research concerning smart hydrogels reported in the past few years.
Collapse
Affiliation(s)
- Zhaoyi Sun
- School of Chemistry and Chemical Engineering , Nanjing University , 210046 Nanjing , China
| | - Chengjun Song
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China
| | - Chao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China.,Jiangsu Key Laboratory for Nano Technology , Nanjing University , 210093 Nanjing , China.,Institute of Drug R&D , Medical School of Nanjing University , 210093 Nanjing , China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China.,Jiangsu Key Laboratory for Nano Technology , Nanjing University , 210093 Nanjing , China.,Institute of Drug R&D , Medical School of Nanjing University , 210093 Nanjing , China
| |
Collapse
|
16
|
Dong Q, Li F, Xu Y, Xiao J, Xu Y, Shang D, Zhang C, Yang H, Tian Z, Mi K, Li X, Zhang Y. RNAactDrug: a comprehensive database of RNAs associated with drug sensitivity from multi-omics data. Brief Bioinform 2019; 21:2167-2174. [PMID: 31799597 DOI: 10.1093/bib/bbz142] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/30/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
Drug sensitivity has always been at the core of individualized cancer chemotherapy. However, we have been overwhelmed by large-scale pharmacogenomic data in the era of next-generation sequencing technology, which makes it increasingly challenging for researchers, especially those without bioinformatic experience, to perform data integration, exploration and analysis. To bridge this gap, we developed RNAactDrug, a comprehensive database of RNAs associated with drug sensitivity from multi-omics data, which allows users to explore drug sensitivity and RNA molecule associations directly. It provides association data between drug sensitivity and RNA molecules including mRNAs, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) at four molecular levels (expression, copy number variation, mutation and methylation) from integrated analysis of three large-scale pharmacogenomic databases (GDSC, CellMiner and CCLE). RNAactDrug currently stores more than 4 924 200 associations of RNA molecules and drug sensitivity at four molecular levels covering more than 19 770 mRNAs, 11 119 lncRNAs, 438 miRNAs and 4155 drugs. A user-friendly interface enriched with various browsing sections augmented with advance search facility for querying the database is offered for users retrieving. RNAactDrug provides a comprehensive resource for RNA molecules acting in drug sensitivity, and it could be used to prioritize drug sensitivity-related RNA molecules, further promoting the identification of clinically actionable biomarkers in drug sensitivity and drug development more cost-efficiently by making this knowledge accessible to both basic researchers and clinical practitioners. Database URL: http://bio-bigdata.hrbmu.edu.cn/RNAactDrug.
Collapse
Affiliation(s)
- Qun Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jing Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Desi Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Haixiu Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zihan Tian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Thomas R, Weihua Z. Rethink of EGFR in Cancer With Its Kinase Independent Function on Board. Front Oncol 2019; 9:800. [PMID: 31508364 PMCID: PMC6716122 DOI: 10.3389/fonc.2019.00800] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/06/2019] [Indexed: 12/23/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is one of most potent oncogenes that are commonly altered in cancers. As a receptor tyrosine kinase, EGFR's kinase activity has been serving as the primary target for developing cancer therapeutics, namely the EGFR inhibitors including small molecules targeting its ATP binding pocket and monoclonal antibodies targeting its ligand binding domains. EGFR inhibitors have produced impressive therapeutic benefits to responsive types of cancers. However, acquired and innate resistances have precluded current anti-EGFR agents from offering sustainable benefits to initially responsive cancers and benefits to EGFR-positive cancers that are innately resistant. Recent years have witnessed a realization that EGFR possesses kinase-independent (KID) pro-survival functions in cancer cells. This new knowledge has offered a different angle of understanding of EGFR in cancer and opened a new avenue of targeting EGFR for cancer therapy. There are already many excellent reviews on the role of EGFR with a focus on its kinase-dependent functions and mechanisms of resistance to EGFR targeted therapies. The present opinion aims to initiate a fresh discussion about the function of EGFR in cancer cells by laying out some unanswered questions pertaining to EGFR in cancer cells, by rethinking the unmet therapeutic challenges from a view of EGFR's KID function, and by proposing novel approaches to target the KID functions of EGFR for cancer treatment.
Collapse
Affiliation(s)
- Rintu Thomas
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| | - Zhang Weihua
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| |
Collapse
|
18
|
Gorshkov K, Sima N, Sun W, Lu B, Huang W, Travers J, Klumpp-Thomas C, Michael SG, Xu T, Huang R, Lee EM, Cheng X, Zheng W. Quantitative Chemotherapeutic Profiling of Gynecologic Cancer Cell Lines Using Approved Drugs and Bioactive Compounds. Transl Oncol 2019; 12:441-452. [PMID: 30576957 PMCID: PMC6302136 DOI: 10.1016/j.tranon.2018.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Heterogeneous response to chemotherapy is a major issue for the treatment of cancer. For most gynecologic cancers including ovarian, cervical, and placental, the list of available small molecule therapies is relatively small compared to options for other cancers. While overall cancer mortality rates have decreased in the United States as early diagnoses and cancer therapies have become more effective, ovarian cancer still has low survival rates due to the lack of effective treatment options, drug resistance, and late diagnosis. To understand chemotherapeutic diversity in gynecologic cancers, we have screened 7914 approved drugs and bioactive compounds in 11 gynecologic cancer cell lines to profile their chemotherapeutic sensitivity. We identified two HDAC inhibitors, mocetinostat and entinostat, as pan-gynecologic cancer suppressors with IC50 values within an order of magnitude of their human plasma concentrations. In addition, many active compounds identified, including the non-anticancer drugs and other compounds, diversely inhibited the growth of three gynecologic cancer cell groups and individual cancer cell lines. These newly identified compounds are valuable for further studies of new therapeutics development, synergistic drug combinations, and new target identification for gynecologic cancers. The results also provide a rationale for the personalized chemotherapeutic testing of anticancer drugs in treatment of gynecologic cancer.
Collapse
Affiliation(s)
- Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Ni Sima
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA; Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, PR China
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Billy Lu
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Wei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA; Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, PR China
| | - Jameson Travers
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Samuel G Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Tuan Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Emily M Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, PR China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Halbur C, Choudhury N, Chen M, Kim JH, Chung EJ. siRNA-Conjugated Nanoparticles to Treat Ovarian Cancer. SLAS Technol 2019; 24:137-150. [PMID: 30616494 DOI: 10.1177/2472630318816668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ovarian cancer is the fifth-most lethal cancer among women due to a lack of early detection and late-stage treatment options, and it is responsible for more than 14,000 deaths each year in the United States. Recently, there have been advances in RNA interference therapy, specifically with small interfering RNA (siRNA), to reduce tumor burden for ovarian cancer via gene down-regulation. However, delivery of siRNA poses its own challenges, as siRNA is unstable in circulation, is unable to be effectively internalized by cells, and may cause toxicity in off-target sites. To address such challenges, nanoparticle carriers have emerged as delivery platforms for the biocompatible, targeted delivery of siRNA-based therapies. Several preclinical studies have shown the promising effects of siRNA therapy to reduce chemotherapy resistance and proliferation of ovarian cancer cells. This review evaluates the recent advances, clinical applications, and future potential of nanoparticle-mediated delivery of siRNA therapeutics to target genes implicated in ovarian cancer.
Collapse
Affiliation(s)
- Christopher Halbur
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Niharika Choudhury
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Michael Chen
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jun Hyuk Kim
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.,2 Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA.,3 Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA.,4 Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.,5 Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
20
|
Sima N, Sun W, Gorshkov K, Shen M, Huang W, Zhu W, Xie X, Zheng W, Cheng X. Small Molecules Identified from a Quantitative Drug Combinational Screen Resensitize Cisplatin's Response in Drug-Resistant Ovarian Cancer Cells. Transl Oncol 2018; 11:1053-1064. [PMID: 29982103 PMCID: PMC6034569 DOI: 10.1016/j.tranon.2018.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Drug resistance to chemotherapy occurs in many ovarian cancer patients resulting in failure of treatment. Exploration of drug resistance mechanisms and identification of new therapeutics that overcome the drug resistance can improve patient prognosis. Following a quantitative combination screen of 6060 approved drugs and bioactive compounds in a cisplatin-resistant A2780-cis ovarian cancer cell line, 38 active compounds with IC50s under 1 μM suppressed the growth of cisplatin-resistant ovarian cancer cells. Among these confirmed compounds, CUDC-101, OSU-03012, oligomycin A, VE-821, or Torin2 in a combination with cisplatin restored cisplatin's apoptotic response in the A2780-cis cells, while SR-3306, GSK-923295, SNX-5422, AT-13387, and PF-05212384 directly suppressed the growth of A2780-cis cells. One of the mechanisms for overcoming cisplatin resistance in these cells is mediated by the inhibition of epidermal growth factor receptor (EGFR), though not all the EGFR inhibitors are equally active. The increased levels of total EGFR and phosphorylated-EGFR (p-EGFR) in the A2780-cis cells were reduced after the combined treatment of cisplatin with EGFR inhibitors. In addition, a knockdown of EGFR mRNA reduced cisplatin resistance in the A2780-cis cells. Therefore, the top active compounds identified in this work can be studied further as potential treatments for cisplatin-resistant ovarian cancer. The quantitative combinational screening approach is a useful method for identifying effective compounds and drug combinations against drug-resistant cancer cells.
Collapse
Affiliation(s)
- Ni Sima
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Huang
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Biology, The George Washington University Medical School, Washington, DC
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA.
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
21
|
Lili LN, Huang AD, Zhang M, Wang L, McDonald LD, Matyunina LV, Satpathy M, McDonald JF. Time-course analysis of microRNA-induced mesenchymal-to-epithelial transition underscores the complexity of the underlying molecular processes. Cancer Lett 2018; 428:184-191. [PMID: 29733963 DOI: 10.1016/j.canlet.2018.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 12/13/2022]
Abstract
Expression levels of the miR-200 family of miRNAs are significantly reduced during the epithelial-to-mesenchymal transition (EMT) and consequent metastasis of ovarian and other cancers. Consistently, ectopic over-expression of miR-200 family miRNAs in mesenchymal-like cells reverses the process by converting treated cells to an epithelial phenotype, thereby reducing invasiveness and increasing sensitivity to chemotherapeutic drugs. To better understand the dynamics and molecular processes underlying miRNA-induced mesenchymal-to mesenchymal transition (MET), a time-course study was conducted where miRNA-induced morphological and molecular changes associated with MET were monitored over a period of 144 h. Morphological transition from an elongated mesenchymal-like to a cuboidal epithelial-like phenotype is maximized at 48 h with cells returning to the elongated phenotype by 144 h. Changes in the expression of >3000 genes, including many previously associated with epithelial-to-mesenchymal transition (EMT), are most pronounced at 48 h, and approach starting levels of expression by 144 h. The majority of these genes are not direct targets of miR-429. Targeted (siRNA) inhibition of key miR-429 regulated genes previously implicated as drivers of EMT/MET, do not recapitulate miR-429 induced MET indicating that the underlying molecular processes are complex.
Collapse
Affiliation(s)
- Loukia N Lili
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - Andrew D Huang
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - Mengnan Zhang
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - Lijuan Wang
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - L DeEtte McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - Lilya V Matyunina
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - Minati Satpathy
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| | - John F McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30309, USA.
| |
Collapse
|
22
|
Singhsa P, Diaz-Dussan D, Manuspiya H, Narain R. Well-Defined Cationic N-[3-(Dimethylamino)propyl]methacrylamide Hydrochloride-Based (Co)polymers for siRNA Delivery. Biomacromolecules 2017; 19:209-221. [PMID: 29195038 DOI: 10.1021/acs.biomac.7b01475] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cationic glycopolymers have shown to be excellent candidates for the fabrication of gene delivery devices due to their ability to electrostatically interact with negatively charged nucleic acids and the carbohydrate residues ensure enhanced stability and low toxicity of the polyplexes. The ability to engineer the polymers for optimized compositions, molecular weights, and architectures is critical in the design of effective gene delivery vehicles. Therefore, in this study, the aqueous reversible addition-fragmentation chain transfer polymerization (RAFT) was used to synthesize well-defined cationic glycopolymers with various cationic segments. For the preparation of cationic parts, N-[3-(dimethylamino)propyl]methacrylamide hydrochloride (DMAPMA·HCl), water-soluble methacrylamide monomer containing tertiary amine, was polymerized to produce DMAPMA·HCl homopolymer, which was then used as macroCTA in the block copolymerization with two other methacrylamide monomers containing different pendant groups, namely, 2-aminoethyl methacrylamide hydrochloride (AEMA) (with primary amine) and N-(3-aminopropyl) morpholine methacrylamide (MPMA) (with morpholine ring). In addition, statistical copolymers of DMAPMA.HCl with either AEMA or MPMA were also synthesized. All resulting cationic polymers were utilized as macroCTA for the RAFT copolymerization with 2-lactobionamidoethyl methacrylamide (LAEMA), which consists of the pendent galactose residues to achieve DMAPMA·HCl-based glycopolymers. From the in vitro cytotoxicity study, the cationic glycopolymers showed better cell viabilities than the corresponding cationic homopolymers. Furthermore, complexation of the cationic polymers with siRNA, cellular uptake of the resulting polyplexes, and gene knockdown efficiencies were evaluated. All cationic polymers/glycopolymers demonstrated good complexation ability with siRNA at low weight ratios. Among these cationic polymer-siRNA polyplexes, the polyplexes prepared from the two glycopolymers, P(DMAPMA65-b-LAEMA15) and P[(DMAPMA65-b-MPMA63)-b-LAEMA16], showed outstanding results in the cellular uptake, high EGFR knockdown, and low post-transfection toxicity, suggesting the great potential in siRNA delivery of these novel glycopolymers.
Collapse
Affiliation(s)
- Pratyawadee Singhsa
- The Petroleum and Petrochemical College, Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University , Soi Chulalongkorn 12, Pathumwan, Bangkok 10330, Thailand.,Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| | - Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| | - Hathaikarn Manuspiya
- The Petroleum and Petrochemical College, Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University , Soi Chulalongkorn 12, Pathumwan, Bangkok 10330, Thailand
| | - Ravin Narain
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering , 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada
| |
Collapse
|
23
|
Diaz-Dussan D, Nakagawa Y, Peng YY, C LVS, Ebara M, Kumar P, Narain R. Effective and Specific Gene Silencing of Epidermal Growth Factor Receptors Mediated by Conjugated Oxaborole and Galactose-Based Polymers. ACS Macro Lett 2017; 6:768-774. [PMID: 35650860 DOI: 10.1021/acsmacrolett.7b00388] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxaborole-based polymers are stimuli-responsive materials that can reversibly interact with diols at pH values higher than their pKa. The strong binding of the oxaborole with cis-hydroxyl groups allow rapid cross-linking of the polymer chains. In this study, we exploited this phenomenon to develop a novel delivery system for the complexation, protection, and delivery of epidermal growth factor receptors (EGFR) siRNA (small interfering RNA). Galactose and oxaborole polymers were first synthesized by the reversible addition-fragmentation chain transfer (RAFT) process, and they were found to show a robust interaction with each other via the oxaborole-diol effect, which allowed the formation of stable polyplexes with siRNA. Although complexes were successfully formed between the neutral galactose and oxaborole-based polymers, these complexes were insufficient in the protection of the siRNA. Therefore, cationic glycopolymers and oxaborole polymers were investigated showing superior complexation with siRNA and exhibiting effective gene silencing in HeLa (cervical) cancer cells, while showing low toxicity. Gene silencing of up to 60% was achieved with these new complexes in the presence and absence of serum. The excellent stability of the complexes under physiological conditions and the observed low cytotoxicity 48 h post-transfection demonstrated the high potential of this new system for gene silencing therapy application in clinics.
Collapse
Affiliation(s)
- Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| | - Yasuhiro Nakagawa
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan and Graduate School of Pure and Applied Science, University of Tsukuba, Ibaraki, Japan
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| | - Leslie V. Sanchez C
- Department of Chemical and Environmental Engineering, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Mitsuhiro Ebara
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan and Graduate School of Pure and Applied Science, University of Tsukuba, Ibaraki, Japan
| | - Piyush Kumar
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| |
Collapse
|
24
|
Zhang Z, Wang X, Luo C, Zhu C, Wang K, Zhang C, Guo Z. Dinuclear Platinum(II) Complexes with Bone-Targeting Groups as Potential Anti-Osteosarcoma Agents. Chem Asian J 2017; 12:1659-1667. [DOI: 10.1002/asia.201700577] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/26/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Zhenqin Zhang
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
- School of Pharmacy; Nanjing Medical University; Nanjing 211166 China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210023 China
| | - Cheng Luo
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
| | - Chengcheng Zhu
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
| | - Kun Wang
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
| | - Changli Zhang
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 China
| |
Collapse
|