1
|
Tsoplaktsoglou M, Spyratou E, Droulias A, Zachou ME, Efstathopoulos EP. The Contribution of Nanomedicine in Ocular Oncology. Cancers (Basel) 2025; 17:1186. [PMID: 40227824 PMCID: PMC11987995 DOI: 10.3390/cancers17071186] [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: 02/27/2025] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025] Open
Abstract
Nanomedicine is a novel and emerging field that has noted significant progress in both the fields of ophthalmology and cancer treatment. Expanding into ocular oncology, it holds the potential to overcome the limitations of conventional therapies, such as poor drug penetration due to anatomical and physiological ocular barriers and insufficient targeting, which can lead to collateral damage to healthy tissues. By reviewing a series of clinical and preclinical studies, we aim to outline the recent advancements, current trends and future perspectives in nanomedicine for ocular cancer treatment. Beyond improving the existing therapies, like chemotherapy, phototherapies and brachytherapy, nanomedicine enables multimodal applications by combining multiple treatments or integrating imaging for theranostic approaches. Additionally, it paves the way for experimental therapies, such as gene therapy, offering new possibilities for more effective and less invasive treatment strategies in ocular oncology.
Collapse
Affiliation(s)
- Margarita Tsoplaktsoglou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.T.); (A.D.)
| | - Ellas Spyratou
- Department of Applied Medical Physics, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.S.); (M.-E.Z.)
| | - Andreas Droulias
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.T.); (A.D.)
| | - Maria-Eleni Zachou
- Department of Applied Medical Physics, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.S.); (M.-E.Z.)
| | - Efstathios P. Efstathopoulos
- Department of Applied Medical Physics, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.S.); (M.-E.Z.)
| |
Collapse
|
2
|
Li P, Zheng S, Leung HM, Liu LS, Chang TJH, Maryam A, Wang F, Chin YR, Lo PK. TNA-Mediated Antisense Strategy to Knockdown Akt Genes for Triple-Negative Breast Cancer Therapy. SMALL METHODS 2024; 8:e2400291. [PMID: 38779741 PMCID: PMC11579567 DOI: 10.1002/smtd.202400291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Triple-negative breast cancer (TNBC) remains a significant challenge in terms of treatment, with limited efficacy of chemotherapy due to side effects and acquired drug resistance. In this study, a threose nucleic acid (TNA)-mediated antisense approach is employed to target therapeutic Akt genes for TNBC therapy. Specifically, two new TNA strands (anti-Akt2 and anti-Akt3) are designed and synthesized that specifically target Akt2 and Akt3 mRNAs. These TNAs exhibit exceptional enzymatic resistance, high specificity, enhance binding affinity with their target RNA molecules, and improve cellular uptake efficiency compared to natural nucleic acids. In both 2D and 3D TNBC cell models, the TNAs effectively inhibit the expression of their target mRNA and protein, surpassing the effects of scrambled TNAs. Moreover, when administered to TNBC-bearing animals in combination with lipid nanoparticles, the targeted anti-Akt TNAs lead to reduced tumor sizes and decreased target protein expression compared to control groups. Silencing the corresponding Akt genes also promotes apoptotic responses in TNBC and suppresses tumor cell proliferation in vivo. This study introduces a novel approach to TNBC therapy utilizing TNA polymers as antisense materials. Compared to conventional miRNA- and siRNA-based treatments, the TNA system holds promise as a cost-effective and scalable platform for TNBC treatment, owing to its remarkable enzymatic resistance, inexpensive synthetic reagents, and simple production procedures. It is anticipated that this TNA-based polymeric system, which targets anti-apoptotic proteins involved in breast tumor development and progression, can represent a significant advancement in the clinical development of effective antisense materials for TNBC, a cancer type that lacks effective targeted therapy.
Collapse
Affiliation(s)
- Pan Li
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Shixue Zheng
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Hoi Man Leung
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Ling Sum Liu
- Department of ChemistryMolecular Sciences Research HubImperial College LondonWhite City CampusWood LaneLondonW12 0BZU.K.
| | - Tristan Juin Han Chang
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Alishba Maryam
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Fei Wang
- The Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)Dongguan523059P. R. China
| | - Y. Rebecca Chin
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Pik Kwan Lo
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
- Key Laboratory of Biochip TechnologyBiotechand Health CareShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
3
|
Datta D, Priyanka Bandi S, Colaco V, Dhas N, Siva Reddy DV, Vora LK. Fostering the unleashing potential of nanocarriers-mediated delivery of ocular therapeutics. Int J Pharm 2024; 658:124192. [PMID: 38703931 DOI: 10.1016/j.ijpharm.2024.124192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/21/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Ocular delivery is the most challenging aspect in the field of pharmaceutical research. The major hurdle for the controlled delivery of drugs to the eye includes the physiological static barriers such as the complex layers of the cornea, sclera and retina which restrict the drug from permeating into the anterior and posterior segments of the eye. Recent years have witnessed inventions in the field of conventional and nanocarrier drug delivery which have shown considerable enhancement in delivering small to large molecules across the eye. The dynamic challenges associated with conventional systems include limited drug contact time and inadequate ocular bioavailability resulting from solution drainage, tear turnover, and dilution or lacrimation. To this end, various bioactive-based nanosized carriers including liposomes, ethosomes, niosomes, dendrimer, nanogel, nanofibers, contact lenses, nanoprobes, selenium nanobells, nanosponge, polymeric micelles, silver nanoparticles, and gold nanoparticles among others have been developed to circumvent the limitations associated with the conventional dosage forms. These nanocarriers have been shown to achieve enhanced drug permeation or retention and prolong drug release in the ocular tissue due to their better tissue adherence. The surface charge and the size of nanocarriers (10-1000 nm) are the important key factors to overcome ocular barriers. Various nanocarriers have been shown to deliver active therapeutic molecules including timolol maleate, ampicillin, natamycin, voriconazole, cyclosporine A, dexamethasone, moxifloxacin, and fluconazole among others for the treatment of anterior and posterior eye diseases. Taken together, in a nutshell, this extensive review provides a comprehensive perspective on the numerous facets of ocular drug delivery with a special focus on bioactive nanocarrier-based approaches, including the difficulties and constraints involved in the fabrication of nanocarriers. This also provides the detailed invention, applications, biodistribution and safety-toxicity of nanocarriers-based therapeutcis for the ophthalmic delivery.
Collapse
Affiliation(s)
- Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| | - Sony Priyanka Bandi
- Loka Laboratories Private Limited, Technology Business Incubator, BITS Pilani Hyderabad Campus, Jawahar Nagar, Medchal 500078, Telangana, India.
| | - Viola Colaco
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - D V Siva Reddy
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio TX78227, USA
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| |
Collapse
|
4
|
Tan Y, Pan J, Deng Z, Chen T, Xia J, Liu Z, Zou C, Qin B. Monoacylglycerol lipase regulates macrophage polarization and cancer progression in uveal melanoma and pan-cancer. Front Immunol 2023; 14:1161960. [PMID: 37033945 PMCID: PMC10076602 DOI: 10.3389/fimmu.2023.1161960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Background Although lipid metabolism has been proven to play a key role in the development of cancer, its significance in uveal melanoma (UM) has not yet been elucidated in the available literature. Methods To identify the expression patterns of lipid metabolism in 80 UM patients from the TCGA database, 47 genes involved in lipid metabolism were analyzed. Consensus clustering revealed two distinct molecular groups. ESTIMATE, TIMER, and ssGSEA analyses were done to identify the differences between the two subgroups in tumor microenvironment (TME) and immune state. Using Cox regression and Lasso regression analysis, a risk model based on differentially expressed genes (DEGs) was developed. To validate the expression of monoacylglycerol lipase (MGLL) and immune infiltration in diverse malignancies, a pan-cancer cohort from the UCSC database was utilized. Next, a single-cell sequencing analysis on UM patients from the GEO data was used to characterize the lipid metabolism in TME and the role of MGLL in UM. Finally, in vitro investigations were utilized to study the involvement of MGLL in UM. Results Two molecular subgroups of UM patients have considerably varied survival rates. The majority of DEGs between the two subgroups were associated with immune-related pathways. Low immune scores, high tumor purity, a low number of immune infiltrating cells, and a comparatively low immunological state were associated with a more favorable prognosis. An examination of GO and KEGG data demonstrated that the risk model based on genes involved with lipid metabolism can accurately predict survival in patients with UM. It has been demonstrated that MGLL, a crucial gene in this paradigm, promotes the proliferation, invasion, and migration of UM cells. In addition, we discovered that MGLL is strongly expressed in macrophages, specifically M2 macrophages, which may play a function in the M2 polarization of macrophages and M2 macrophage activation in cancer cells. Conclusion This study demonstrates that the risk model based on lipid metabolism may be useful for predicting the prognosis of patients with UM. By promoting macrophage M2 polarization, MGLL contributes to the evolution of malignancy in UM, suggesting that it may be a therapeutic target for UM.
Collapse
Affiliation(s)
- Yao Tan
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Juan Pan
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
- Department of Clinical Medical Research Center, The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, Guangdong, China
| | - Zhenjun Deng
- Department of Dermatology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tao Chen
- School of Medicine, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Jinquan Xia
- Department of Clinical Medical Research Center, The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, Guangdong, China
| | - Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Chang Zou
- School of Life and Health Sciences, The Chinese University of Kong Hong, Shenzhen, China
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| |
Collapse
|
5
|
Han H, Li S, Xu M, Zhong Y, Fan W, Xu J, Zhou T, Ji J, Ye J, Yao K. Polymer- and lipid-based nanocarriers for ocular drug delivery: Current status and future perspectives. Adv Drug Deliv Rev 2023; 196:114770. [PMID: 36894134 DOI: 10.1016/j.addr.2023.114770] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/21/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Ocular diseases seriously affect patients' vision and life quality, with a global morbidity of over 43 million blindness. However, efficient drug delivery to treat ocular diseases, particularly intraocular disorders, remains a huge challenge due to multiple ocular barriers that significantly affect the ultimate therapeutic efficacy of drugs. Recent advances in nanocarrier technology offer a promising opportunity to overcome these barriers by providing enhanced penetration, increased retention, improved solubility, reduced toxicity, prolonged release, and targeted delivery of the loaded drug to the eyes. This review primarily provides an overview of the progress and contemporary applications of nanocarriers, mainly polymer- and lipid-based nanocarriers, in treating various eye diseases, highlighting their value in achieving efficient ocular drug delivery. Additionally, the review covers the ocular barriers and administration routes, as well as the prospective future developments and challenges in the field of nanocarriers for treating ocular diseases.
Collapse
Affiliation(s)
- Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Su Li
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Mingyu Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Yueyang Zhong
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Wenjie Fan
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Jingwei Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Tinglian Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China.
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China.
| |
Collapse
|
6
|
Intraocular siRNA Delivery Mediated by Penetratin Derivative to Silence Orthotopic Retinoblastoma Gene. Pharmaceutics 2023; 15:pharmaceutics15030745. [PMID: 36986605 PMCID: PMC10053059 DOI: 10.3390/pharmaceutics15030745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Gene therapy brings a ray of hope for inherited ocular diseases that may cause severe vision loss and even blindness. However, due to the dynamic and static absorption barriers, it is challenging to deliver genes to the posterior segment of the eye by topical instillation. To circumvent this limitation, we developed a penetratin derivative (89WP)-modified polyamidoamine polyplex to deliver small interference RNA (siRNA) via eye drops to achieve effective gene silencing in orthotopic retinoblastoma. The polyplex could be spontaneously assembled through electrostatic and hydrophobic interactions, as demonstrated by isothermal titration calorimetry, and enter cells intactly. In vitro cellular internalization revealed that the polyplex possessed higher permeability and safety than the lipoplex composed of commercial cationic liposomes. After the polyplex was instilled in the conjunctival sac of the mice, the distribution of siRNA in the fundus oculi was significantly increased, and the bioluminescence from orthotopic retinoblastoma was effectively inhibited. In this work, an evolved cell-penetrating peptide was employed to modify the siRNA vector in a simple and effective way, and the formed polyplex interfered with intraocular protein expression successfully via noninvasive administration, which showed a promising prospect for gene therapy for inherited ocular diseases.
Collapse
|
7
|
Giordo R, Wehbe Z, Paliogiannis P, Eid AH, Mangoni AA, Pintus G. Nano-targeting vascular remodeling in cancer: Recent developments and future directions. Semin Cancer Biol 2022; 86:784-804. [DOI: 10.1016/j.semcancer.2022.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/16/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
|
8
|
Zhang J, Dong M, Feng Y, Zhang D, Lin M, Yuan C, Li H, Wang L, Zhang H, Liang C. Egr1/HSP70 Promoter-Driven Activation of Gene Expression for Synergistic Anti-Hepatoma Using PEI-MZF Nanoparticles and Radiation. Int J Nanomedicine 2022; 17:423-441. [PMID: 35115776 PMCID: PMC8801369 DOI: 10.2147/ijn.s334015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/14/2021] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Spatially restricted gene expression circumvents the gene expression and gene vector problem by enabling localized amplification. The objective of this study is to construct a spatially restricted gene expression for liver cancer therapy based upon the MFH-absorbing properties of PEI- Mn0.5zn0.5Fe2o4, gene therapy and radiation. METHODS Mn0.5zn0.5Fe2O4 (MZF) magnetic nanoparticles were prepared by an improved chemical co-precipitation method, modified by polyethylene imine (PEI), and then the structure, modification characters, biocompatibility, temperature rise and control ability and binding efficiency of the plasmid were characterized. Then, the dual-promoter plasmid PCDNA3.1-EGR1-HSP70-HSVTK was constructed. The recombinant vectors were identified by enzyme digestion analysis and DNA sequencing. The TK gene expression level was detected by realtime-PCR assay in HEK293 cells. Also, the HSV-TK gene expression was detected in SMMC7721 cells with the help of PEI-Mn0.5Zn0.5Fe2O4. In vitro anti-tumor experiment, MTT assay and flow cytometry were used to evaluate the therapeutic effects of the cultured SMMC7721 cells treated by different ways. In vivo anti-tumor experiment, the xenografted mice were treated by different ways for three times to detect the antitumor effect. RESULTS The Mn0.5Zn0.5Fe2O4 magnetic nanoparticles could be successfully prepared through improved co-precipitation process and showed good biocompatibility. And PEI had been coated on MZF complex. The modified PEI-MZF presented favorable dispensability, responsibility to magnetism, good loading capability and transfect capability. Also, pCDNA3.1-Egr1-Hsp70-HSVTK plasmid had been constructed successfully and could be induced by heat and irradiation. It would be used for further target gene therapy research. The antitumor results in vitro showed: The therapeutic effects of nanosized PEI-MZF-HSV-TK complex could significantly inhibit the proliferation of cultured liver cancer cells (SMC7721), induce cell apoptosis and had a prominent cell cycle disturbance in the S phase in vitro. The results in vivo showed: The combined therapy induced by PEI-MZF-HSV-TK could inhabit the growth of hepatocellular carcinoma xenografts by killing and inhabiting the proliferation of the tumor cells. CONCLUSION The novel site-directed heat/radiation-inducible expression system based upon the hyperthermia (by MFH) and radiation possessed superior antitumor effect in vivo and in vitro.
Collapse
Affiliation(s)
- Jia Zhang
- Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, People’s Republic of China
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Min Dong
- Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, People’s Republic of China
| | - Yan Feng
- Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, People’s Republic of China
| | - Dongsheng Zhang
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Mei Lin
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Chenyan Yuan
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Hongbo Li
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Ling Wang
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Hao Zhang
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Chen Liang
- Imaging and Nuclear Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
9
|
Zhang Q, Zhang Q, Li H, Zhao X, Zhang H. LiCl induces apoptosis via CHOP/NOXA/Mcl-1 axis in human choroidal melanoma cells. Cancer Cell Int 2021; 21:96. [PMID: 33557839 PMCID: PMC7869481 DOI: 10.1186/s12935-021-01778-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/16/2021] [Indexed: 12/22/2022] Open
Abstract
Background Choroidal melanoma is the most common primary intraocular malignancy that occurs in adults. Lithium Chloride Promotes Apoptosis in Human Leukemia NB4 Cells by Inhibiting Glycogen Synthase Kinase-3 Beta. In this study, we aimed to understand whether LiCl exerts anticancer effects on choroidal melanoma cells and elucidate the underlying molecular mechanisms. Methods Human choroidal melanoma cells were treated with LiCl, and cell survival was assessed with MTT assays. Cell reproductive viability was measured by plate colony formation assays. Cell apoptosis was evaluated using flow cytometry, and proteins were detected using western blotting. A human choroidal melanoma xenograft model was established to demonstrate the effect of LiCl on human choroidal melanoma in vivo. Results We found that LiCl inhibited cell survival and clonogenic potential and induced apoptosis in human choroidal melanoma cells. LiCl also reduced the proliferation of choroidal melanoma cells in vivo. Moreover, the upregulation of NOXA and downregulation of Mcl-1 were responsible for LiCl-induced apoptosis. Mcl-1 overexpression obviously impaired LiCl-induced apoptosis and cleavage of caspase8, caspase9, caspase3 and PARP. Moreover, the protein expression of endoplasmic reticulum stress markers, including IRE1α, Bip, p-eIF2α, ATF4 and CHOP, were upregulated following treatment with LiCl. When CHOP expression was knocked down and cells were treated with LiCl, the protein level of NOXA was partially increased, and Mcl-1 expression was increased, while the cleavage of caspase8, caspase9, caspase3 and PARP that was induced by the LiCl was reduced compared with the vehicle treated group. Prolonged ER stress results in the activation of the apoptotic pathway. Conclusions In summary, LiCl induced an endoplasmic reticulum stress response while activating intrinsic apoptosis. Furthermore, the CHOP/NOXA/Mcl-1 axis contributed to LiCl-induced apoptosis both in vitro and in vivo. The present study provides important mechanistic insight into potential cancer treatments involving LiCl and enhances the understanding of human choroidal melanoma.
Collapse
Affiliation(s)
- Qiuqiu Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.,Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China, 277100
| | - Qianwei Zhang
- Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China, 277100
| | - Huiyuan Li
- Department of Ophthalmology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, China
| | - Xiaofei Zhao
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.
| | - Han Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.
| |
Collapse
|
10
|
Wei C, Zhao X, Wang L, Zhang H. TRIP suppresses cell proliferation and invasion in choroidal melanoma via promoting the proteasomal degradation of Twist1. FEBS Lett 2020; 594:3170-3181. [PMID: 32640040 DOI: 10.1002/1873-3468.13882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Choroidal melanoma (CM) remains the most prevalent form of intraocular malignancy, and the prognosis of affected patients is poor. While the E3 ubiquitin ligase TRAF-interacting protein (TRIP) is known to play key regulatory roles in multiple diseases, its relevance in CM remains uncertain. In the present study, we found that TRIP overexpression is sufficient to inhibit the proliferation, invasion, and epithelial-mesenchymal transition (EMT) of CM cells in vitro, whereas the opposite phenotypes are observed following TRIP knockdown. We further determined that TRIP is able to promote the K48-polyubiquitination of EMT-associated transcription factor Twist-related protein 1, thereby suppressing EMT progression. Together, our results suggest that TRIP plays an important role in regulating the progression of CM and that it may therefore be an important therapeutic target for the treatment of this disease.
Collapse
Affiliation(s)
- Chao Wei
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Zhao
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Wang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Han Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
11
|
Sun H, Wang X, Zhai S. The Rational Design and Biological Mechanisms of Nanoradiosensitizers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E504. [PMID: 32168899 PMCID: PMC7153263 DOI: 10.3390/nano10030504] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy (RT) has been widely used for cancer treatment. However, the intrinsic drawbacks of RT, such as radiotoxicity in normal tissues and tumor radioresistance, promoted the development of radiosensitizers. To date, various kinds of nanoparticles have been found to act as radiosensitizers in cancer radiotherapy. This review focuses on the current state of nanoradiosensitizers, especially the related biological mechanisms, and the key design strategies for generating nanoradiosensitizers. The regulation of oxidative stress, DNA damage, the cell cycle, autophagy and apoptosis by nanoradiosensitizers in vitro and in vivo is highlighted, which may guide the rational design of therapeutics for tumor radiosensitization.
Collapse
Affiliation(s)
- Hainan Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
- Shandong Vocational College of Light Industry, Zibo 255300, Shandong, China
| | - Xiaoling Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| |
Collapse
|
12
|
Liu S, Song W, Liu F, Zhang J, Zhu S. Antitumor efficacy of VP22-CD/5-FC suicide gene system mediated by lentivirus in a murine uveal melanoma model. Exp Eye Res 2018; 172:144-151. [DOI: 10.1016/j.exer.2018.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/15/2018] [Accepted: 04/12/2018] [Indexed: 12/22/2022]
|
13
|
Hirani A, Grover A, Lee YW, Pathak Y, Sutariya V. Nanotechnology for Omics-Based Ocular Drug Delivery. Ophthalmology 2018. [DOI: 10.4018/978-1-5225-5195-9.ch017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Millions of people suffer from ocular diseases that impair vision and can lead to blindness. Advances in genomics and proteomics have revealed a number of different molecular markers specific for different ocular diseases, thereby optimizing the processes of drug development and discovery. Nanotechnology can increase the throughput of data obtained in omics-based studies and allows for more sensitive diagnostic techniques as more efficient drug delivery systems. Biocompatible and biodegradable nanomaterials developed through omics-based research are able to target reported molecular markers for different ocular diseases and offer novel alternatives to conventional drug therapy. In this chapter, the authors review the pathophysiology, current genomic and proteomic information, and current nanomaterial-based therapies of four ocular diseases: glaucoma, uveal melanoma, age-related macular degeneration, and diabetic retinopathy. Omics-based research can be used to elucidate specific genes and proteins and develop novel nanomedicine formulations to prevent, halt, or cure ocular diseases at the transcriptional or translational level.
Collapse
Affiliation(s)
- Anjali Hirani
- University of South Florida, USA & Virginia Tech-Wake Forest University, USA
| | | | | | | | | |
Collapse
|
14
|
c-FLIP and the NOXA/Mcl-1 axis participate in the synergistic effect of pemetrexed plus cisplatin in human choroidal melanoma cells. PLoS One 2017; 12:e0184135. [PMID: 28863158 PMCID: PMC5581197 DOI: 10.1371/journal.pone.0184135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 08/18/2017] [Indexed: 01/03/2023] Open
Abstract
Choroidal melanoma is the most common primary malignant intraocular tumor, and very few effective therapies are available to treat it. Our study aimed to understand whether pemetrexed plus cisplatin exerts a beneficial synergistic effect in human choroidal melanoma cells and to delineate the underlying molecular mechanism. To accomplish these aims, we treated choroidal melanoma cells with pemetrexed and cisplatin and assessed cell survival with SRB and MTT assays. Proteins were detected using western blotting analysis. NOXA and CHOP were knocked down with siRNA. We found that pemetrexed or cisplatin alone inhibited survival and induced apoptosis in human choroidal melanoma cells. Furthermore, the expression levels of c-FLIP, an anti-apoptotic protein in the extrinsic apoptosis pathway, and Mcl-1, an anti-apoptotic protein in the intrinsic apoptosis pathway, were decreased by pemetrexed or cisplatin respectively, while the expression of a pro-apoptotic protein in the intrinsic apoptosis pathway, NOXA, was up-regulated. Moreover, pemetrexed or cisplatin alone increased the protein expression of the endoplasmic reticulum stress markers IRE1α, Bip and CHOP. Silencing CHOP expression reduced NOXA expression. These findings suggest that the pemetrexed or cisplatin induced intrinsic apoptosis via activation of the ER stress response. Importantly, combining the two compounds more strongly induced apoptosis. Following the cotreatment, CHOP and NOXA expression increased, while c-FLIP and Mcl-1 expression decreased, and these effects were more pronounced than when using either compound alone. This result suggests that pemetrexed and cisplatin synergistically activate ER stress response-induced apoptosis in choroidal melanoma cells. To summarize, the c-FLIP and NOXA/Mcl-1 axis participated in the synergistic effect of pemetrexed plus cisplatin in human choroidal melanoma cells. Intrinsic apoptosis was induced via activation of the ER stress response. Our study provides important mechanistic insights into potential cancer treatment with pemetrexed plus cisplatin and enriches our understanding of human choroidal melanoma.
Collapse
|
15
|
Hirani A, Grover A, Lee YW, Pathak Y, Sutariya V. Nanotechnology for Omics-Based Ocular Drug Delivery. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Millions of people suffer from ocular diseases that impair vision and can lead to blindness. Advances in genomics and proteomics have revealed a number of different molecular markers specific for different ocular diseases, thereby optimizing the processes of drug development and discovery. Nanotechnology can increase the throughput of data obtained in omics-based studies and allows for more sensitive diagnostic techniques as more efficient drug delivery systems. Biocompatible and biodegradable nanomaterials developed through omics-based research are able to target reported molecular markers for different ocular diseases and offer novel alternatives to conventional drug therapy. In this chapter, the authors review the pathophysiology, current genomic and proteomic information, and current nanomaterial-based therapies of four ocular diseases: glaucoma, uveal melanoma, age-related macular degeneration, and diabetic retinopathy. Omics-based research can be used to elucidate specific genes and proteins and develop novel nanomedicine formulations to prevent, halt, or cure ocular diseases at the transcriptional or translational level.
Collapse
Affiliation(s)
- Anjali Hirani
- University of South Florida, USA & Virginia Tech-Wake Forest University, USA
| | | | | | | | | |
Collapse
|
16
|
Zhang W, Liu C, Hai B, Du G, Wang H, Leng H, Xu Y, Song C. A Convenient In Vivo Model Using Small Interfering RNA Silencing to Rapidly Assess Skeletal Gene Function. PLoS One 2016; 11:e0167222. [PMID: 27893850 PMCID: PMC5125699 DOI: 10.1371/journal.pone.0167222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
It is difficult to study bone in vitro because it contains various cell types that engage in cross-talk. Bone biologically links various organs, and it has thus become increasingly evident that skeletal physiology must be studied in an integrative manner in an intact animal. We developed a model using local intraosseous small interfering RNA (siRNA) injection to rapidly assess the effects of a target gene on the local skeletal environment. In this model, 160-g male Sprague-Dawley rats were treated for 1-2 weeks. The left tibia received intraosseous injection of a parathyroid hormone 1 receptor (Pth1r) or insulin-like growth factor 1 receptor (Igf-1r) siRNA transfection complex loaded in poloxamer 407 hydrogel, and the right tibia received the same volume of control siRNA. All the tibias received an intraosseous injection of recombinant human parathyroid hormone (1-34) (rhPTH (1-34)) or insulin-like growth factor-1 (IGF-1). Calcein green and alizarin red were injected 6 and 2 days before euthanasia, respectively. IGF-1R and PTH1R expression levels were detected via RT-PCR assays and immunohistochemistry. Bone mineral density (BMD), microstructure, mineral apposition rates (MARs), and strength were determined by dual-energy X-ray absorptiometry, micro-CT, histology and biomechanical tests. The RT-PCR and immunohistochemistry results revealed that IGF-1R and PTH1R expression levels were dramatically diminished in the siRNA-treated left tibias compared to the right tibias (both p<0.05). Using poloxamer 407 hydrogel as a controlled-release system prolonged the silencing effect of a single dose of siRNA; the mRNA expression levels of IGF-1R were lower at two weeks than at one week (p<0.01). The BMD, bone microstructure parameters, MAR and bone strength were significantly decreased in the left tibias compared to the right tibias (all p<0.05). This simple and convenient local intraosseous siRNA injection model achieved gene silencing with very small quantities of siRNA over a short treatment period (≤7 days).
Collapse
Affiliation(s)
- Wen Zhang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Can Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Bao Hai
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Guohong Du
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Hong Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- * E-mail:
| |
Collapse
|
17
|
Liko F, Hindré F, Fernandez-Megia E. Dendrimers as Innovative Radiopharmaceuticals in Cancer Radionanotherapy. Biomacromolecules 2016; 17:3103-3114. [PMID: 27608327 DOI: 10.1021/acs.biomac.6b00929] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Radiotherapy is one of the most commonly used cancer treatments, with an estimate of 40% success that could be improved further if more efficient targeting and retention of radiation at the tumor site were achieved. This review focuses on the use of dendrimers in radionanotherapy, an emerging technology aimed to improve the efficiency of radiotherapy by implementing nanovectorization, an already established praxis in drug delivery and diagnosis. The labeling of dendrimers with radionuclides also aims to reduce the dose of radiolabeled materials and, hence, their toxicity and tumor resistance. Examples of radiolabeled dendrimers with alpha, beta, and Auger electron emitters are commented, along with the use of dendrimers in boron neutron capture therapy (BNCT). The conjugation of radiolabeled dendrimers to monoclonal antibodies for a more efficient targeting and the application of dendrimers in gene delivery radiotherapy are also covered.
Collapse
Affiliation(s)
- Flonja Liko
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', and Plateforme de Radiobiologie et d'IMagerie EXpérimentale, PRIMEX, SFR ICAT 4208, Université Angers, UMR-S1066, 49933 Angers, Cedex 9, France.,Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela , Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - François Hindré
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', and Plateforme de Radiobiologie et d'IMagerie EXpérimentale, PRIMEX, SFR ICAT 4208, Université Angers, UMR-S1066, 49933 Angers, Cedex 9, France
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela , Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
18
|
You S, Luo J, Grossniklaus HE, Gou ML, Meng K, Zhang Q. Nanomedicine in the application of uveal melanoma. Int J Ophthalmol 2016; 9:1215-25. [PMID: 27588278 DOI: 10.18240/ijo.2016.08.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/16/2016] [Indexed: 12/28/2022] Open
Abstract
Rapid advances in nanomedicine have significantly changed many aspects of nanoparticle application to the eye including areas of diagnosis, imaging and more importantly drug delivery. The nanoparticle-based drug delivery systems has provided a solution to various drug solubility-related problems in ophthalmology treatment. Nanostructured compounds could be used to achieve local ocular delivery with minimal unwanted systematic side effects produced by taking advantage of the phagocyte system. In addition, the in vivo control release by nanomaterials encapsulated drugs provides prolong exposure of the compound in the body. Furthermore, certain nanoparticles can overcome important body barriers including the blood-retinal barrier as well as the corneal-retinal barrier of the eye for effective delivery of the drug. In summary, the nanotechnology based drug delivery system may serve as an important tool for uveal melanoma treatment.
Collapse
Affiliation(s)
- Shuo You
- Department of Endocrinology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China; Winship Cancer Institute, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Jing Luo
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Hans E Grossniklaus
- Winship Cancer Institute, School of Medicine, Emory University, Atlanta, Georgia 30322, USA; Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA; Department of Pathology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Ma-Ling Gou
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ke Meng
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qing Zhang
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China; Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
19
|
Buckel L, Savariar EN, Crisp JL, Jones KA, Hicks AM, Scanderbeg DJ, Nguyen QT, Sicklick JK, Lowy AM, Tsien RY, Advani SJ. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery. Cancer Res 2015; 75:1376-1387. [PMID: 25681274 DOI: 10.1158/0008-5472.can-14-1931] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022]
Abstract
Intrinsic tumor resistance to radiotherapy limits the efficacy of ionizing radiation (IR). Sensitizing cancer cells specifically to IR would improve tumor control and decrease normal tissue toxicity. The development of tumor-targeting technologies allows for developing potent radiosensitizing drugs. We hypothesized that the anti-tubulin agent monomethyl auristatin E (MMAE), a component of a clinically approved antibody-directed conjugate, could function as a potent radiosensitizer and be selectively delivered to tumors using an activatable cell-penetrating peptide targeting matrix metalloproteinases and RGD-binding integrins (ACPP-cRGD-MMAE). We evaluated the ability of MMAE to radiosensitize both established cancer cells and a low-passage cultured human pancreatic tumor cell line using clonogenic and DNA damage assays. MMAE sensitized colorectal and pancreatic cancer cells to IR in a schedule- and dose-dependent manner, correlating with mitotic arrest. Radiosensitization was evidenced by decreased clonogenic survival and increased DNA double-strand breaks in irradiated cells treated with MMAE. MMAE in combination with IR resulted in increased DNA damage signaling and activation of CHK1. To test a therapeutic strategy of MMAE and IR, PANC-1 or HCT-116 murine tumor xenografts were treated with nontargeted free MMAE or tumor-targeted MMAE (ACPP-cRGD-MMAE). While free MMAE in combination with IR resulted in tumor growth delay, tumor-targeted ACPP-cRGD-MMAE with IR produced a more robust and significantly prolonged tumor regression in xenograft models. Our studies identify MMAE as a potent radiosensitizer. Importantly, MMAE radiosensitization can be localized to tumors by targeted activatable cell-penetrating peptides.
Collapse
Affiliation(s)
- Lisa Buckel
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | - Angel M Hicks
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | | | - Roger Y Tsien
- Department of Pharmacology.,Howard Hughes Medical Institute
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences.,Center for Advanced Radiotherapy Technologies University of California San Diego
| |
Collapse
|