251
|
Doroudian M, MacLoughlin R, Poynton F, Prina-Mello A, Donnelly SC. Nanotechnology based therapeutics for lung disease. Thorax 2019; 74:965-976. [PMID: 31285360 DOI: 10.1136/thoraxjnl-2019-213037] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 11/03/2022]
Abstract
Nanomedicine is a multidisciplinary research field with an integration of traditional sciences such as chemistry, physics, biology and materials science. The application of nanomedicine for lung diseases as a relatively new area of interdisciplinary science has grown rapidly over the last 10 years. Promising research outcomes suggest that nanomedicine will revolutionise the practice of medicine, through the development of new approaches in therapeutic agent delivery, vaccine development and nanotechnology-based medical detections. Nano-based approaches in the diagnosis and treatment of lung diseases will, in the not too distant future, change the way we practise medicine. This review will focus on the current trends and developments in the clinical translation of nanomedicine for lung diseases, such as in the areas of lung cancer, cystic fibrosis, asthma, bacterial infections and COPD.
Collapse
Affiliation(s)
- Mohammad Doroudian
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Galway, Ireland.,School of Pharmacy, Royal College of Surgeons, Dublin, Ireland.,School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Fergus Poynton
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- CRANN Institute and AMBER Centre, University of Dublin Trinity College, Dublin, Ireland.,Department of Medicine, Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity College Dublin, Dublin, Ireland.,Nanomedicine Group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
252
|
Larcher LM, Wang T, Veedu RN. Development of Novel antimiRzymes for Targeted Inhibition of miR-21 Expression in Solid Cancer Cells. Molecules 2019; 24:molecules24132489. [PMID: 31284665 PMCID: PMC6651226 DOI: 10.3390/molecules24132489] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that are involved in the regulation of gene expression. Previous reports showed an over-expression of miRNA-21 (miR-21) in various cancer cells, and its up-regulation is closely related to cancer initiation, proliferation and metastasis. In this work, we envisioned the development of novel antimiRzymes (anti-miRNA-DNAzyme) that are capable of selectively targeting and cleaving miR-21 and inhibit its expression in cancer cells using the DNAzyme technique. For this purpose, we have designed different antimiRzyme candidates by systematically targeting different regions of miR-21. Our results demonstrated that RNV541, a potential arm-loop-arm type antimiRzyme, was very efficient (90%) to suppress miR-21 expression in U87MG malignant glioblastoma cell line at 200 nM concentration. In addition, RNV541 also inhibited miR-21 expression (50%) in MDA-MB-231 breast cancer cell line. For targeted delivery, we conjugated RNV541 with a transferrin receptor (TfR) targeting aptamer for TfR-mediated cancer cell delivery. As expected, the developed chimeric structure efficiently delivered the antimiRzyme RNV541 into TfR positive glioblastoma cells. TfR aptamer-RNV541 chimeric construct showed 52% inhibition of miR-21 expression in U87MG glioblastoma cells at 2000 nM concentration, without using any transfection reagents, making it a highly desirable strategy to tackle miR-21 over-expressed malignant cancers. Although these are in vitro based observations, based on our results, we firmly believe that our findings could be beneficial towards the development of targeted cancer therapeutics where conventional therapies face several challenges.
Collapse
Affiliation(s)
- Leon M Larcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| | - Tao Wang
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
- Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia
| | - Rakesh N Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia.
- Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia.
| |
Collapse
|
253
|
Adachi M, Kai K, Yamaji K, Ide T, Noshiro H, Kawaguchi A, Aishima S. Transferrin receptor 1 overexpression is associated with tumour de-differentiation and acts as a potential prognostic indicator of hepatocellular carcinoma. Histopathology 2019; 75:63-73. [PMID: 30811632 DOI: 10.1111/his.13847] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/24/2019] [Indexed: 02/06/2023]
Abstract
AIM Hepatocellular carcinoma (HCC) is the second leading cause of cancer mortality worldwide. An excess of iron in liver tissue causes oxidative stress, leading to hepatocellular carcinogenesis. Iron metabolism, which is regulated by a complex mechanism, is important for cancer cell survival. The aim of this study is to clarify the role of iron regulatory protein in the progression of HCC and in patient outcome. METHODS AND RESULTS We first investigated the mRNA level of iron metabolism-related genes, including hepcidin, ferroportin 1 (FPN-1) and transferrin receptor (TFR)-1/2. TFR-1/2 protein expression was then evaluated in surgical specimens from 210 cases using immunohistochemistry, and we compared clinicopathological factors with TFR-1/2 expression. The mRNA expression levels of TFR-1 were significantly increased in HCC tissues compared with adjacent non-cancerous tissues (P = 0.0013), but there were no differences in other genes. High expression of TFR-1 in HCC was associated with the absence of alcohol abuse (P = 0.0467), liver cirrhosis (P < 0.0001), higher alpha-fetoprotein (AFP; P < 0.0001), smaller tumour size (P = 0.0022), poor histological differentiation (P < 0.0001) and morphological features (P < 0.0001). In contrast, high expression of TFR-2 in HCC was associated with lower AFP (P < 0.0001), well-differentiated histological grade (P < 0.0001) and morphological features (P = 0.0010). Multivariate analysis for both overall survival and recurrence-free survival indicated that high TFR-1 expression was a significant prognostic factor for poor outcome. CONCLUSIONS We found an inverse correlation of TFR-1 and TFR-2 expression in AFP and tumour differentiation. TFR-1 overexpression suggests a higher risk of recurrence and death in HCC patients following liver resection.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Dedifferentiation/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Iron/metabolism
- Kaplan-Meier Estimate
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Middle Aged
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Transferrin/genetics
- Receptors, Transferrin/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Makiko Adachi
- Departments of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
- Department of Pathology, Saga University Hospital, Saga, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, Saga, Japan
| | - Koutaro Yamaji
- Departments of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
- Department of Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takao Ide
- Department of Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirokazu Noshiro
- Department of Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Atsushi Kawaguchi
- Center of Comprehensive Community Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinichi Aishima
- Departments of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
- Department of Pathology, Saga University Hospital, Saga, Japan
| |
Collapse
|
254
|
Zhao Y, Peng J, Yang J, Zhang E, Huang L, Yang H, Kakadiaris E, Li J, Yan B, Shang Z, Jiang N, Zhang X, Han G, Niu Y. Enhancing Prostate-Cancer-Specific MRI by Genetic Amplified Nanoparticle Tumor Homing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900928. [PMID: 31183895 DOI: 10.1002/adma.201900928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/22/2019] [Indexed: 06/09/2023]
Abstract
Precise localization and visualization of early-stage prostate cancer (PCa) is critical to improve the success of focal ablation and reduce cancer mortality. However, it remains challenging under the current imaging techniques due to the heterogeneous nature of PCa and the suboptimal sensitivity of the techniques themselves. Herein, a novel genetic amplified nanoparticle tumor-homing strategy to enhance the MRI accuracy of ultrasmall PCa lesions is reported. This strategy could specifically drive TfR expressions in PCa under PCa-specific DD3 promoter, and thus remarkably increase Tf-USPIONs concentrations in a highly accurate manner while minimizing their non-specific off-target effects on normal tissues. Consequently, this strategy can pinpoint an ultrasmall PCa lesion, which is otherwise blurred in the current MRI, and thereby addresses the unmet key need in MRI imaging for focal therapy. With this proof-of-concept experiment, the synergistic gene-nano strategy holds great promise to boost the MRI effects of a wide variety of commonly used nanoscale and molecular probes that are otherwise limited. In addition, such a strategy may also be translated and applied to MR-specific imaging of other types of cancers by using their respective tumor-specific promoters.
Collapse
Affiliation(s)
- Yang Zhao
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin, 300211, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jing Peng
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jinyi Yang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Enlong Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Ling Huang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Hong Yang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Eugenia Kakadiaris
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jingjin Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Bin Yan
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Zhiqun Shang
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Ning Jiang
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Xuening Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yuanjie Niu
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin, 300211, China
| |
Collapse
|
255
|
Teles CM, Lammoglia LC, Juliano MA, Ruiz ALTG, Candido TZ, de Carvalho JE, Lima CSP, Abbehausen C. Novel anticancer Pd II complexes: The effect of the conjugation of transferrin binding peptide and the nature of halogen coordinated on antitumor activity. J Inorg Biochem 2019; 199:110754. [PMID: 31401348 DOI: 10.1016/j.jinorgbio.2019.110754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/14/2019] [Accepted: 06/23/2019] [Indexed: 01/06/2023]
Abstract
A series of PdII complexes with bis-(2-pyridylmethyl)glycine as a ligand of formula [PdX(bis-(2-pyridylmethyl)glycine)] where X = Cl, Br, I were prepared and the effect of the halogen nature in the antitumor activity of eight tumorigenic and one non-tumorigenic cell line was evaluated. The chloride derivative was further functionalized with a transferrin receptor binding peptide, generating the first PdII based metallopeptide. Its antitumor activity was also evaluated. However, among all the complexes, the chloride and iodine parent compounds showed the lowest GI50 values in the panel evaluated, and lowest GI50 than cisplatin in several cell lines. In contrast, the bromine derivative showed higher values of GI50 than chloride and iodine (around 30 - 50 μM). The same trend was observed for the bovine serum albumin binding constant with higher values for iodine, chlorine, and bromine in this order. In aqueous solution, the chloride is exchanged by water while the bromine and iodine are not. DNA was evaluated as a target and showed no significative interaction for all the compounds. The results suggest sulfur-rich proteins and not DNA as a target. This report represents the first PdII metallopeptide reported, its evaluation in solution and antitumor activity. This work opens the possibilities for further functionalization of PdII complexes and the importance of the halogen coordination in the design of novel metallodrugs.
Collapse
Affiliation(s)
- C M Teles
- Institute of Chemistry, University of Campinas - UNICAMP, PO Box 6154, 13083-970 Campinas, SP, Brazil
| | - L C Lammoglia
- Faculty of Pharmaceutical Sciences, University of Campinas, UNICAMP, 13083-871 Campinas, SP, Brazil
| | - M A Juliano
- Universidade Federal de São Paulo, Escola Paulista de Medicina, UNIFESP, 04063-062 São Paulo, SP, Brazil
| | - A L T G Ruiz
- Faculty of Pharmaceutical Sciences, University of Campinas, UNICAMP, 13083-871 Campinas, SP, Brazil
| | - T Z Candido
- Faculty of Medical Sciences, University of Campinas, UNICAMP, 13083-970 Campinas, SP, Brazil
| | - J E de Carvalho
- Faculty of Pharmaceutical Sciences, University of Campinas, UNICAMP, 13083-871 Campinas, SP, Brazil
| | - C S P Lima
- Faculty of Medical Sciences, University of Campinas, UNICAMP, 13083-970 Campinas, SP, Brazil
| | - C Abbehausen
- Institute of Chemistry, University of Campinas - UNICAMP, PO Box 6154, 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
256
|
Lu X, Liu J, Wu X, Ding B. Multifunctional DNA Origami Nanoplatforms for Drug Delivery. Chem Asian J 2019; 14:2193-2202. [PMID: 31125182 DOI: 10.1002/asia.201900574] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Indexed: 12/11/2022]
Abstract
DNA nanotechnology has been employed in the construction of self-assembled nano-biomaterials with uniform size and shape for various biological applications, such as bioimaging, diagnosis, or therapeutics. Herein, recent successful efforts to utilize multifunctional DNA origami nanoplatforms as drug-delivery vehicles are reviewed. Diagnostic and therapeutic strategies based on gold nanorods, chemotherapeutic drugs, cytosine-phosphate-guanine, functional proteins, gene drugs, and their combinations for optoacoustic imaging, photothermal therapy, chemotherapy, immunological therapy, gene therapy, and coagulation-based therapy are summarized. The challenges and opportunities for DNA-based nanocarriers for biological applications are also discussed.
Collapse
Affiliation(s)
- Xuehe Lu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, P.R. China.,CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing, 100190, P.R. China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing, 100190, P.R. China
| | - Xiaohui Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Baoquan Ding
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, P.R. China.,CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| |
Collapse
|
257
|
Křivohlavá R, Neuhӧferová E, Jakobsen KQ, Benson V. Knockdown of microRNA-135b in Mammary Carcinoma by Targeted Nanodiamonds: Potentials and Pitfalls of In Vivo Applications. NANOMATERIALS 2019; 9:nano9060866. [PMID: 31181619 PMCID: PMC6632128 DOI: 10.3390/nano9060866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/26/2022]
Abstract
Nanodiamonds (ND) serve as RNA carriers with potential for in vivo application. ND coatings and their administration strategy significantly change their fate, toxicity, and effectivity within a multicellular system. Our goal was to develop multiple ND coating for effective RNA delivery in vivo. Our final complex (NDA135b) consisted of ND, polymer, antisense RNA, and transferrin. We aimed (i) to assess if a tumor-specific coating promotes NDA135b tumor accumulation and effective inhibition of oncogenic microRNA-135b and (ii) to outline off-targets and immune cell interactions. First, we tested NDA135b toxicity and effectivity in tumorospheres co-cultured with immune cells ex vivo. We found NDA135b to target tumor cells, but it binds also to granulocytes. Then, we followed with NDA135b intravenous and intratumoral applications in tumor-bearing animals in vivo. Application of NDA135b in vivo led to the effective knockdown of microRNA-135b in tumor tissue regardless administration. Only intravenous application resulted in NDA135b circulation in peripheral blood and urine and the decreased granularity of splenocytes. Our data show that localized intratumoral application of NDA135b represents a suitable and safe approach for in vivo application of nanodiamond-based constructs. Systemic intravenous application led to an interaction of NDA135b with bio-interface, and needs further examination regarding its safety.
Collapse
Affiliation(s)
- Romana Křivohlavá
- Institute of Microbiology of the CAS, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Eva Neuhӧferová
- Institute of Microbiology of the CAS, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Katrine Q Jakobsen
- Institute of Microbiology of the CAS, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Veronika Benson
- Institute of Microbiology of the CAS, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
258
|
Abstract
An FDA-approved iron oxide nanoparticle used for the treatment of anaemia can be repurposed for leukaemia therapy.
Collapse
Affiliation(s)
- Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
259
|
Sakpakdeejaroen I, Somani S, Laskar P, Mullin M, Dufès C. Transferrin-bearing liposomes entrapping plumbagin for targeted cancer therapy. JOURNAL OF INTERDISCIPLINARY NANOMEDICINE 2019; 4:54-71. [PMID: 31341642 PMCID: PMC6619241 DOI: 10.1002/jin2.56] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/08/2019] [Indexed: 12/30/2022]
Abstract
The therapeutic potential of plumbagin, a naphthoquinone extracted from the officinal leadwort with anticancer properties, is hampered by its failure to specifically reach tumours at a therapeutic concentration after intravenous administration, without secondary effects on normal tissues. Its use in clinic is further limited by its poor aqueous solubility, its spontaneous sublimation, and its rapid elimination in vivo. We hypothesize that the entrapment of plumbagin within liposomes grafted with transferrin, whose receptors are overexpressed on many cancer cells, could result in a selective delivery to tumours after intravenous administration. The objectives of this study were therefore to prepare and characterize transferrin-targeted liposomes entrapping plumbagin and to evaluate their therapeutic efficacy in vitro and in vivo. The entrapment of plumbagin in transferrin-bearing liposomes led to an increase in plumbagin uptake by cancer cells and improved antiproliferative efficacy and apoptosis activity in B16-F10, A431, and T98G cell lines compared with that observed with the drug solution. In vivo, the intravenous injection of transferrin-bearing liposomes entrapping plumbagin led to tumour suppression for 10% of B16-F10 tumours and tumour regression for a further 10% of the tumours. By contrast, all the tumours treated with plumbagin solution or left untreated were progressive. The animals did not show any signs of toxicity. Transferrin-bearing liposomes entrapping plumbagin are therefore highly promising therapeutic systems that should be further optimized as a therapeutic tool for cancer treatment.
Collapse
Affiliation(s)
- Intouch Sakpakdeejaroen
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of Strathclyde161 Cathedral StreetGlasgowG4 0REUK
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of Strathclyde161 Cathedral StreetGlasgowG4 0REUK
| | - Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of Strathclyde161 Cathedral StreetGlasgowG4 0REUK
| | - Margaret Mullin
- College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowG12 8QQUK
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of Strathclyde161 Cathedral StreetGlasgowG4 0REUK
| |
Collapse
|
260
|
NAMI-A and KP1019/1339, Two Iconic Ruthenium Anticancer Drug Candidates Face-to-Face: A Case Story in Medicinal Inorganic Chemistry. Molecules 2019; 24:molecules24101995. [PMID: 31137659 PMCID: PMC6571951 DOI: 10.3390/molecules24101995] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 01/23/2023] Open
Abstract
NAMI-A ((ImH)[trans-RuCl4(dmso-S)(Im)], Im = imidazole) and KP1019/1339 (KP1019 = (IndH)[trans-RuCl4(Ind)2], Ind = indazole; KP1339 = Na[trans-RuCl4(Ind)2]) are two structurally related ruthenium(III) coordination compounds that have attracted a lot of attention in the medicinal inorganic chemistry scientific community as promising anticancer drug candidates. This has led to a considerable amount of studies on their respective chemico-biological features and to the eventual admission of both to clinical trials. The encouraging pharmacological performances qualified KP1019 mainly as a cytotoxic agent for the treatment of platinum-resistant colorectal cancers, whereas the non-cytotoxic NAMI-A has gained the reputation of being a very effective antimetastatic drug. A critical and strictly comparative analysis of the studies conducted so far on NAMI-A and KP1019 allows us to define the state of the art of these experimental ruthenium drugs in terms of the respective pharmacological profiles and potential clinical applications, and to gain some insight into the inherent molecular mechanisms. Despite their evident structural relatedness, deeply distinct biological and pharmacological profiles do emerge. Overall, these two iconic ruthenium complexes form an exemplary and unique case in the field of medicinal inorganic chemistry.
Collapse
|
261
|
Jafari B, Pourseif MM, Barar J, Rafi MA, Omidi Y. Peptide-mediated drug delivery across the blood-brain barrier for targeting brain tumors. Expert Opin Drug Deliv 2019; 16:583-605. [DOI: 10.1080/17425247.2019.1614911] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Behzad Jafari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia,
Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Mohammad A. Rafi
- Department of Neurology, College of Medicine, Thomas Jefferson University, Philadelphia,
PA, USA
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| |
Collapse
|
262
|
Roberti A, Valdes AF, Torrecillas R, Fraga MF, Fernandez AF. Epigenetics in cancer therapy and nanomedicine. Clin Epigenetics 2019; 11:81. [PMID: 31097014 PMCID: PMC6524244 DOI: 10.1186/s13148-019-0675-4] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022] Open
Abstract
The emergence of nanotechnology applied to medicine has revolutionized the treatment of human cancer. As in the case of classic drugs for the treatment of cancer, epigenetic drugs have evolved in terms of their specificity and efficiency, especially because of the possibility of using more effective transport and delivery systems. The use of nanoparticles (NPs) in oncology management offers promising advantages in terms of the efficacy of cancer treatments, but it is still unclear how these NPs may be affecting the epigenome such that safe routine use is ensured. In this work, we summarize the importance of the epigenetic alterations identified in human cancer, which have led to the appearance of biomarkers or epigenetic drugs in precision medicine, and we describe the transport and release systems of the epigenetic drugs that have been developed to date.
Collapse
Affiliation(s)
- Annalisa Roberti
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-FINBA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Avenida de Roma, 33011, Oviedo, Asturias, Spain
| | - Adolfo F Valdes
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC)-Universidad de Oviedo-Principado de Asturias, Avenida de Roma, 33011, Oviedo, Asturias, Spain
| | - Ramón Torrecillas
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC)-Universidad de Oviedo-Principado de Asturias, Avenida de Roma, 33011, Oviedo, Asturias, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC)-Universidad de Oviedo-Principado de Asturias, Avenida de Roma, 33011, Oviedo, Asturias, Spain.
| | - Agustin F Fernandez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-FINBA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Avenida de Roma, 33011, Oviedo, Asturias, Spain.
| |
Collapse
|
263
|
Biffi S, Voltan R, Bortot B, Zauli G, Secchiero P. Actively targeted nanocarriers for drug delivery to cancer cells. Expert Opin Drug Deliv 2019; 16:481-496. [DOI: 10.1080/17425247.2019.1604679] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stefania Biffi
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Rebecca Voltan
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Barbara Bortot
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery, Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
264
|
Moosavian SA, Sahebkar A. Aptamer-functionalized liposomes for targeted cancer therapy. Cancer Lett 2019; 448:144-154. [PMID: 30763718 DOI: 10.1016/j.canlet.2019.01.045] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Accumulation of chemotherapeutic agents in the tumor tissue while reducing adverse effects and drug resistance are among the major goals in cancer therapy. Among nanocarriers, liposomes have been found to be more effective in the passive targeting of cancer cells. A promising recent development in targeted drug delivery is the use of aptamer-functionalized liposomes for cancer therapy. Aptamer-targeted liposomes have enhanced uptake in tumor cells as shown in vitro and in vivo. Here, we discuss the aptamer-functionalized liposome platforms and review functionalization approaches as well as the factors affecting antitumor efficiency of aptamer-targeted liposomal systems. Finally, we provide a comprehensive overview of aptamer-targeted liposomes based on the molecular targets on the surface of cancer cells.
Collapse
Affiliation(s)
- Seyedeh Alia Moosavian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
265
|
Riaz MK, Zhang X, Wong KH, Chen H, Liu Q, Chen X, Zhang G, Lu A, Yang Z. Pulmonary delivery of transferrin receptors targeting peptide surface-functionalized liposomes augments the chemotherapeutic effect of quercetin in lung cancer therapy. Int J Nanomedicine 2019; 14:2879-2902. [PMID: 31118613 PMCID: PMC6503309 DOI: 10.2147/ijn.s192219] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/15/2019] [Indexed: 01/03/2023] Open
Abstract
Purpose: Lung cancer has a high incidence rate worldwide with a 5-year survival rate of 18%, and is the leading cause of cancer-related deaths. The aim of this study is to augment therapeutic efficacy of quercetin (QR) for lung cancer therapy by targeting transferrin receptors, which are overexpressed and confined to tumor cells. Methods: In this study, T7 surface-functionalized liposomes loaded with QR (T7-QR-lip) having different T7 peptide densities (0.5%, 1% and 2%) were prepared by the film hydration method. T7 surface-functionalized liposomes were characterized and evaluated in terms of in vitro cytotoxicity and cellular uptake, 3D tumor spheroid penetration and inhibition capabilities, in vivo biodistribution and therapeutic efficacy in mice with orthotopic lung-tumor implantation by fluorescent and bioluminescent imaging via pulmonary administration. Results: In vitro, 2% T7-QR-lip exhibited significantly augmented cytotoxicity (~3-fold), higher apoptosis induction and S-phase cell-cycle arrest. A prominent peak right-shift and enhanced mean fluorescence intensity was observed in A549 cells treated with T7 Coumarin-6 liposomes (T7-Cou6-lip), confirming the target specificity of T7 targeted liposomes; while, after treatment with T7-QR-lip and non-targeted QR-lip, no significant difference was observed in cellular uptake and in vitro cytotoxicity studies in MRC-5 (normal lung fibroblast) cells. T7-Cou6-lip showed higher fluorescence intensity in A549 cells and a significantly deeper penetration depth of 120 µm in the core of the tumor spheroids and T7-QR-lip produced significantly higher tumor-spheroid growth inhibition. The in vivo biodistribution study via pulmonary delivery of T7 1,1'-dioctadecyltetramethyl-indotricarbocyanine iodide liposomes demonstrated liposome accumulation in the lungs and sustained-release behavior up to 96 h. Further, T7-QR-lip significantly enhanced the anticancer activity of QR and lifespan of mice (p<0.01, compared with saline) in orthotopic lung tumor-bearing mice via pulmonary administration. Conclusion: T7 surface-functionalized liposomes provide a potential drug delivery system for a range of anticancer drugs to enhance their therapeutic efficacy by localized (pulmonary) administration and targeted delivery.
Collapse
Affiliation(s)
- Muhammad Kashif Riaz
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Xue Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Huoji Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Ge Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu, Jiangsu Province, People’s Republic of China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu, Jiangsu Province, People’s Republic of China
| |
Collapse
|
266
|
Kim JS, Sirois AR, Cegla AJV, Jumai’an E, Murata N, Buck ME, Moore SJ. Protein-Polymer Conjugates Synthesized Using Water-Soluble Azlactone-Functionalized Polymers Enable Receptor-Specific Cellular Uptake toward Targeted Drug Delivery. Bioconjug Chem 2019; 30:1220-1231. [PMID: 30920802 PMCID: PMC6608588 DOI: 10.1021/acs.bioconjchem.9b00155] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conjugation of proteins to drug-loaded polymeric structures is an attractive strategy for facilitating target-specific drug delivery for a variety of clinical needs. Polymers currently available for conjugation to proteins generally have limited chemical versatility for subsequent drug loading. Many polymers that do have chemical functionality useful for drug loading are often insoluble in water, making it difficult to synthesize functional protein-polymer conjugates for targeted drug delivery. In this work, we demonstrate that reactive, azlactone-functionalized polymers can be grafted to proteins, conjugated to a small-molecule fluorophore, and subsequently internalized into cells in a receptor-specific manner. Poly(2-vinyl-4,4-dimethylazlactone), synthesized using reversible addition-fragmentation chain transfer polymerization, was modified post-polymerization with substoichiometric equivalents of triethylene glycol monomethyl ether to yield reactive water-soluble, azlactone-functionalized copolymers. These reactive polymers were then conjugated to proteins holo-transferrin and ovotransferrin. Protein gel analysis verified successful conjugation of proteins to polymer, and protein-polymer conjugates were subsequently purified from unreacted proteins and polymers using size exclusion chromatography. Internalization experiments using a breast cancer cell line that overexpresses the transferrin receptor on its surface showed that the holo-transferrin-polymer conjugate was successfully internalized by cells in a mechanism consistent with receptor-mediated endocytosis. Internalization of protein-polymer conjugate demonstrated that the protein ligand maintained its overall structure and function following conjugation to polymer. Our approach to protein-polymer conjugate synthesis offers a simple, tailorable strategy for preparing bioconjugates of interest for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Julia S. Kim
- Biochemistry Program, Smith College, Northampton, Massachusetts 01063, United States
| | - Allison R. Sirois
- Picker Engineering Program, Smith College, Northampton, Massachusetts 01063, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | | | - Eugenie Jumai’an
- Picker Engineering Program, Smith College, Northampton, Massachusetts 01063, United States
| | - Naomi Murata
- Neuroscience Program, Smith College, Northampton, Massachusetts 01063, United States
| | - Maren E. Buck
- Department of Chemistry, Smith College, Northampton, Massachusetts 01063, United States
| | - Sarah J. Moore
- Picker Engineering Program, Smith College, Northampton, Massachusetts 01063, United States
- Department of Biological Sciences, Smith College, Northampton, Massachusetts 01063, United States
| |
Collapse
|
267
|
Wang X, Yang J, Guo G, Feng R, Chen K, Liao Y, Zhang L, Sun L, Huang S, Chen JL. Novel lncRNA-IUR suppresses Bcr-Abl-induced tumorigenesis through regulation of STAT5-CD71 pathway. Mol Cancer 2019; 18:84. [PMID: 30961617 PMCID: PMC6454664 DOI: 10.1186/s12943-019-1013-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs), defined as the transcripts longer than 200 nt without protein-coding capacity, have been found to be aberrantly expressed in diverse human diseases including cancer. A reciprocal translocation between chromosome 9 and 22 generates the chimeric Bcr-Abl oncogene, which is associated with several hematological malignancies. However, the functional relevance between aberrantly expressed lncRNAs and Bcr-Abl-mediated leukemia remains obscure. Methods LncRNA cDNA microarray was used to identify novel lncRNAs involved in Bcr-Abl-mediated cellular transformation. To study the functional relevance of novel imatinib-upregulated lncRNA (IUR) family in Abl-induced tumorigenesis, Abl-transformed cell survival and xenografted tumor growth in mice was evaluated. Primary bone marrow transformation and in vivo leukemia transplant using lncRNA-IUR knockdown (KD) transgenic mice were further conducted to corroborate the role of lncRNA-IUR in Abl-induced tumorigenesis. Transcriptome RNA-seq, Western blot, RNA pull down and RNA Immunoprecipitation (RIP) were employed to determine the mechanisms by which lncRNA-IUR-5 regulates Bcr-Abl-mediated tumorigenesis. Results We identified a conserved lncRNA-IUR family as a key negative regulator of Bcr-Abl-induced tumorigenesis. Increased expression of lncRNA-IUR was detected in both human and mouse Abl-transformed cells upon imatinib treatment. In contrast, reduced expression of lncRNA-IUR was observed in the peripheral blood lymphocytes derived from Bcr-Abl-positive acute lymphoblastic leukemia (ALL) patients compared to normal subjects. Knockdown of lncRNA-IUR remarkably promoted Abl-transformed leukemic cell survival and xenografted tumor growth in mice, whereas overexpression of lncRNA-IUR had opposite effects. Also, silencing murine lncRNA-IUR promoted Bcr-Abl-mediated primary bone marrow transformation and Abl-transformed leukemia cell survival in vivo. Besides, knockdown of murine lncRNA-IUR in transgenic mice provided a favorable microenvironment for development of Abl-mediated leukemia. Finally, we demonstrated that lncRNA-IUR-5 suppressed Bcr-Abl-mediated tumorigenesis by negatively regulating STAT5-mediated expression of CD71. Conclusions The results suggest that lncRNA-IUR may act as a critical tumor suppressor in Bcr-Abl-mediated tumorigenesis by suppressing the STAT5-CD71 pathway. This study provides new insights into functional involvement of lncRNAs in leukemogenesis. Electronic supplementary material The online version of this article (10.1186/s12943-019-1013-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuefei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jianling Yang
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Guijie Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Riyue Feng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Ke Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Yuan Liao
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lianfeng Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, China
| | - Liping Sun
- Department of Blood Transfusion, Chinese PLA General Hospital, Beijing, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
268
|
Abstract
Nanotechnology offers new solutions for the development of cancer therapeutics that display improved efficacy and safety. Although several nanotherapeutics have received clinical approval, the most promising nanotechnology applications for patients still lie ahead. Nanoparticles display unique transport, biological, optical, magnetic, electronic, and thermal properties that are not apparent on the molecular or macroscale, and can be utilized for therapeutic purposes. These characteristics arise because nanoparticles are in the same size range as the wavelength of light and display large surface area to volume ratios. The large size of nanoparticles compared to conventional chemotherapeutic agents or biological macromolecule drugs also enables incorporation of several supportive components in addition to active pharmaceutical ingredients. These components can facilitate solubilization, protection from degradation, sustained release, immunoevasion, tissue penetration, imaging, targeting, and triggered activation. Nanoparticles are also processed differently in the body compared to conventional drugs. Specifically, nanoparticles display unique hemodynamic properties and biodistribution profiles. Notably, the interactions that occur at the bio-nano interface can be exploited for improved drug delivery. This review discusses successful clinically approved cancer nanodrugs as well as promising candidates in the pipeline. These nanotherapeutics are categorized according to whether they predominantly exploit multifunctionality, unique electromagnetic properties, or distinct transport characteristics in the body. Moreover, future directions in nanomedicine such as companion diagnostics, strategies for modifying the microenvironment, spatiotemporal nanoparticle transitions, and the use of extracellular vesicles for drug delivery are also explored.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida 32224, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
269
|
He L, Liu J, Li S, Feng X, Wang C, Zhuang X, Ding J, Chen X. Polymer Nanoplatforms at Work in Prostate Cancer Therapy. ADVANCED THERAPEUTICS 2019; 2. [DOI: 10.1002/adtp.201800122] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 12/13/2022]
Abstract
AbstractProstate cancer (PCa) is the most common male urogenital malignancy worldwide. Surgery, endocrine therapy, radiotherapy, and chemotherapy are the main clinical management options for PCa. However, these three therapies each have limitations. For example, surgery is not suitable for the advanced PCa patients with extensive metastases, and radiotherapy causes serious side effects. Primary endocrine therapy promotes the progression of hormone‐sensitive PCa into the castration‐resistant prostate cancer. Therefore, considering these drawbacks, chemotherapy has become an effective and extensive treatment for PCa. Among the modern therapeutic strategies against advanced PCa, polymer‐nanocarrier‐incorporated formulations have gradually emerged due to their well‐controlled release profiles and improved tumor targeting abilities. The drug delivery systems based on polymer nanoplatforms passively target tumors via the enhanced permeability and retention effect. Simultaneously, stimuli‐responsive polymer nanoplatforms unload cargoes in response to certain stimuli in the tumor area. Furthermore, the active targeting ligand‐conjugated polymer nanoformulations against PCa‐specific markers have also achieved great success in PCa therapies. Herein, the advanced polymer nanoplatforms for PCa therapy are reviewed, while the future development of polymer nanoplatforms for PCa therapy is also predicted.
Collapse
Affiliation(s)
- Liang He
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Jianhua Liu
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Shengxian Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Chunxi Wang
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
270
|
Boondireke S, Léonard M, Durand A, Thanomsub Wongsatayanon B. Encapsulation of monomyristin into polymeric nanoparticles improved its in vitro antiproliferative activity against cervical cancer cells. Colloids Surf B Biointerfaces 2019; 176:9-17. [DOI: 10.1016/j.colsurfb.2018.12.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/09/2018] [Accepted: 12/20/2018] [Indexed: 01/20/2023]
|
271
|
Kerkhofs M, Bultynck G, Vervliet T, Monaco G. Therapeutic implications of novel peptides targeting ER-mitochondria Ca 2+-flux systems. Drug Discov Today 2019; 24:1092-1103. [PMID: 30910738 DOI: 10.1016/j.drudis.2019.03.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/16/2019] [Accepted: 03/18/2019] [Indexed: 01/03/2023]
Abstract
Intracellular Ca2+-flux systems located at the ER-mitochondrial axis govern mitochondrial Ca2+ balance and cell fate. Multiple yet incurable pathologies are characterized by insufficient or excessive Ca2+ fluxes toward the mitochondria, in turn leading to aberrant cell life or death dynamics. The discovery and ongoing molecular characterization of the main interorganellar Ca2+ gateways have resulted in a novel class of peptide tools able to regulate relevant protein-protein interactions (PPIs) underlying this signaling scenario. Here, we review peptides, molecularly derived from Ca2+-flux systems or their accessory proteins. We discuss how they alter Ca2+-signaling protein complexes and modulate cell survival in light of their forthcoming therapeutic applications.
Collapse
Affiliation(s)
- Martijn Kerkhofs
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium.
| | - Tim Vervliet
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Giovanni Monaco
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
272
|
Montalvo-Quiros S, Aragoneses-Cazorla G, Garcia-Alcalde L, Vallet-Regí M, González B, Luque-Garcia JL. Cancer cell targeting and therapeutic delivery of silver nanoparticles by mesoporous silica nanocarriers: insights into the action mechanisms using quantitative proteomics. NANOSCALE 2019; 11:4531-4545. [PMID: 30806414 PMCID: PMC6667342 DOI: 10.1039/c8nr07667g] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
An approach for safely delivering AgNPs to cancer cells and the evaluation of the affected cellular mechanism are presented. The use of mesoporous silica nanoparticles (MSNs) as nanovehicles decorated with transferrin (Tf, targeting agent) provides a nanoplatform for the nucleation and immobilization of AgNPs (MSNs-Tf-AgNPs). We performed the physico-chemical characterization of the nanosystems and evaluated their therapeutic potential using bioanalytical strategies to estimate the efficiency of the targeting, the degree of cellular internalization in two cell lines with different TfR expression, and the cytotoxic effects of the delivered AgNPs. In addition, cellular localization of the nanosystems in cells has been evaluated by a transmission electron microscopy analysis of ultrathin sections of human hepatocarcinoma (HepG2) cells exposed to MSNs-Tf-AgNPs. The in vitro assays demonstrate that only the nanosystem functionalized with Tf is able to transport the AgNPs inside the cells which overexpress transferrin receptors. Therefore, this novel nanosystem is able to deliver AgNPs specifically to cancer cells overexpressing Tf receptors and offers the possibility of a targeted therapy using reduced doses of silver nanoparticles as cytotoxic agents. Then, a quantitative proteomic experiment validated through the analysis of gene expression has been performed to identify the molecular mechanisms of action associated with the chemotherapeutic potential of the MSNs-Tf-AgNP nanocarriers.
Collapse
Affiliation(s)
- Sandra Montalvo-Quiros
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
273
|
Martinelli C, Pucci C, Ciofani G. Nanostructured carriers as innovative tools for cancer diagnosis and therapy. APL Bioeng 2019; 3:011502. [PMID: 31069332 PMCID: PMC6481740 DOI: 10.1063/1.5079943] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer accounts for millions of deaths every year and, due to the increase and aging of the world population, the number of new diagnosed cases is continuously rising. Although many progresses in early diagnosis and innovative therapeutic protocols have been already set in clinical practice, still a lot of critical aspects need to be addressed in order to efficiently treat cancer and to reduce several drawbacks caused by conventional therapies. Nanomedicine has emerged as a very promising approach to support both early diagnosis and effective therapy of tumors, and a plethora of different inorganic and organic multifunctional nanomaterials have been ad hoc designed to meet the constant demand for new solutions in cancer treatment. Given their unique features and extreme versatility, nanocarriers represent an innovative and easily adaptable tool both for imaging and targeted therapy purposes, in order to improve the specific delivery of drugs administered to cancer patients. The current review reports an in-depth analysis of the most recent research studies aiming at developing both inorganic and organic materials for nanomedical applications in cancer diagnosis and therapy. A detailed overview of different approaches currently undergoing clinical trials or already approved in clinical practice is provided.
Collapse
Affiliation(s)
- Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Pontedera (Pisa) 56025, Italy
| | - Carlotta Pucci
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Pontedera (Pisa) 56025, Italy
| | - Gianni Ciofani
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
274
|
Zhong L, Xu L, Liu Y, Li Q, Zhao D, Li Z, Zhang H, Zhang H, Kan Q, Wang Y, Sun J, He Z. Transformative hyaluronic acid-based active targeting supramolecular nanoplatform improves long circulation and enhances cellular uptake in cancer therapy. Acta Pharm Sin B 2019; 9:397-409. [PMID: 30972285 PMCID: PMC6437598 DOI: 10.1016/j.apsb.2018.11.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/02/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic acid (HA) is a natural ligand of tumor-targeted drug delivery systems (DDS) due to the relevant CD44 receptor overexpressed on tumor cell membranes. However, other HA receptors (HARE and LYVE-1) are also overexpressing in the reticuloendothelial system (RES). Therefore, polyethylene glycol (PEG) modification of HA-based DDS is necessary to reduce RES capture. Unfortunately, pegylation remarkably inhibits tumor cellular uptake and endosomal escapement, significantly compromising the in vivo antitumor efficacy. Herein, we developed a Dox-loaded HA-based transformable supramolecular nanoplatform (Dox/HCVBP) to overcome this dilemma. Dox/HCVBP contains a tumor extracellular acidity-sensitive detachable PEG shell achieved by a benzoic imine linkage. The in vitro and in vivo investigations further demonstrated that Dox/HCVBP could be in a "stealth" state at blood stream for a long circulation time due to the buried HA ligands and the minimized nonspecific interaction by PEG shell. However, it could transform into a "recognition" state under the tumor acidic microenvironment for efficient tumor cellular uptake due to the direct exposure of active targeting ligand HA following PEG shell detachment. Such a transformative concept provides a promising strategy to resolve the dilemma of natural ligand-based DDS with conflicting two processes of tumor cellular uptake and in vivo nonspecific biodistribution.
Collapse
Key Words
- AD-B-PEG, the pH-responsive adamantane-PEG conjugate
- AD-O-PEG, the non-pH sensitive adamantane-PEG conjugate
- ADA, 1-adamantane carboxylic acid
- AUC, area under the plasma concentration—time curve
- Active-targeting
- Benzoic imine linkage
- CLSM, confocal laser scanning microscope
- Cancer therapy
- DAPI, 2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride
- DCC, N,N′-dicyclohexylcarbodiimide
- DCM, dichloromethane
- DDS, drug delivery systems
- DL, drug-loading content
- DLS, dynamic light scattering
- DMAP, 4-dimethylaminopyrideine
- DMEM, Dulbecco׳s modified Eagle׳s medium
- DiR, 1,1′-dioctadecyltetramethyl indotricarbocyanine iodide
- Dox/HCVBP, Dox-loaded hyaluronic acid-based transformable supramolecular nanoplatform
- Dox/HCVOP, Dox-loaded hyaluronic acid-based untransformable supramolecular nanoplatform
- Dox·HCl, doxorubicin hydrochloride
- EDC, 1-ethyl-3-(3-dimethyalminopropl) carbodiimide
- EE, encapsulation efficiency
- FBS, fetal bovine serum
- H&E, hematoxylin and eosin
- HA, hyaluronic acid
- HA-CD, hydroxypropyl-β-cyclodextrin grafted hyaluronic acid polymer
- HCBP, hydroxypropyl-β-cyclodextrin grafted hyaluronic acid polymer and pH-responsive adamantane-PEG conjugate inclusion complex
- HCPs, hydroxypropyl-β-cyclodextrin grafted hyaluronic acid polymer and adamantane-PEG conjugate inclusion complexes
- HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesul-fonic acid
- HOBT, 1-hydroxybenzotriazole
- HPCD, hydroxypropyl-β-cyclodextrin
- Hyaluronic acid
- MW, molecular weight
- NPs, nanoparticles
- Natural ligand
- PCC, Pearson׳s correlation coefficient
- PDI, polydispersity index
- PEG dilemma
- RES, reticuloendothelial system
- RPMI-1640, Roswell Park Memorial Institute-1640
- Supramolecular nanoplat-form
- THF, tetrahydrofuran
- TUNEL, terminal deoxynucleotidyl transferased dUTP nick end labeling
- Transformative nanoparti-cles
- VES, vitamin E succinate
- pHe, the extracellular pH
Collapse
Affiliation(s)
- Lu Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qingsong Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongyang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhenbao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huicong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haotian Zhang
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiming Kan
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongjun Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
275
|
Alonso-García FJ, Blanco-González E, Montes-Bayón M. An inductively coupled plasma-mass spectrometry (ICP-MS) linked immunoassay by means of iodinated antibodies for transferrin quantitative analysis in breast cancer cell lines. Talanta 2019; 194:336-342. [DOI: 10.1016/j.talanta.2018.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022]
|
276
|
Chen SJ, Sinsuebphon N, Rudkouskaya A, Barroso M, Intes X, Michalet X. In vitro and in vivo phasor analysis of stoichiometry and pharmacokinetics using short-lifetime near-infrared dyes and time-gated imaging. JOURNAL OF BIOPHOTONICS 2019; 12:e201800185. [PMID: 30421551 PMCID: PMC6559731 DOI: 10.1002/jbio.201800185] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/08/2018] [Accepted: 11/11/2018] [Indexed: 05/22/2023]
Abstract
We introduce a simple new approach for time-resolved multiplexed analysis of complex systems using near-infrared (NIR) dyes, applicable to in vitro and in vivo studies. We show that fast and precise in vitro quantification of NIR fluorophores' short (subnanosecond) lifetime and stoichiometry can be done using phasor analysis, a computationally efficient and user-friendly representation of complex fluorescence intensity decays obtained with pulsed laser excitation and time-gated camera imaging. We apply this approach to the study of binding equilibria by Förster resonant energy transfer using two different model systems: primary/secondary antibody binding in vitro and ligand/receptor binding in cell cultures. We then extend it to dynamic imaging of the pharmacokinetics of transferrin engagement with the transferrin receptor in live mice, elucidating the kinetics of differential transferrin accumulation in specific organs, straightforwardly differentiating specific from nonspecific binding. Our method, implemented in a freely-available software, has the advantage of time-resolved NIR imaging, including better tissue penetration and background-free imaging, but simplifies and considerably speeds up data processing and interpretation, while remaining quantitative. These advances make this method attractive and of broad applicability for in vitro and in vivo molecular imaging and could be extended to applications as diverse as image-guided surgery or optical tomography.
Collapse
Affiliation(s)
- Sez-Jade Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Nattawut Sinsuebphon
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Xavier Michalet
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
277
|
Gasparetto M, Pei S, Minhajuddin M, Stevens B, Smith CA, Seligman P. Low ferroportin expression in AML is correlated with good risk cytogenetics, improved outcomes and increased sensitivity to chemotherapy. Leuk Res 2019; 80:1-10. [PMID: 30852438 DOI: 10.1016/j.leukres.2019.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/31/2022]
Abstract
Iron metabolism is altered in a variety of cancers; however, little is known about the role of iron metabolism in the biology and response to therapy of acute myeloid leukemia (AML). Here we show that SLC40A1, the gene encoding the iron exporter ferroportin (FPN), is variably expressed among primary AMLs and that low levels are associated with good prognosis and improved outcomes. In particular, core binding factor (CBF) AMLs, which are associated with good outcomes with chemotherapy, consistently have low level of SLC40A1 expression. AML cell lines that expressed relatively low levels of FPN endogenously, or were engineered via gene knockdown, had an increased sensitivity to chemotherapy relative to controls expressing high levels of FPN. Primary FPNlow AML bulk cells also had increased sensitivity to Ara-C treatment, iron treatment and the combination of Ara-C and iron relative to FPNhigh cells. FPNlow leukemic stem cells (LSCs) had decreased viability following addition of iron alone and in combination with Ara-C treatment relative to FPNhigh LSCs. Together these observations suggest a model where FPN mediated iron metabolism may play a role in chemosensitivity and outcome to therapy in AML.
Collapse
Affiliation(s)
- Maura Gasparetto
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA.
| | - Shanshan Pei
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | | | - Brett Stevens
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | - Clayton A Smith
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| | - Paul Seligman
- Division of Hematology, University of Colorado Medical Center, Aurora, CO, USA
| |
Collapse
|
278
|
Babu KR, Muckenthaler MU. miR-148a regulates expression of the transferrin receptor 1 in hepatocellular carcinoma. Sci Rep 2019; 9:1518. [PMID: 30728365 PMCID: PMC6365501 DOI: 10.1038/s41598-018-35947-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/10/2018] [Indexed: 02/07/2023] Open
Abstract
Transferrin receptor 1 (TFR1) is a transmembrane glycoprotein that allows for transferrin-bound iron uptake in mammalian cells. It is overexpressed in various cancers to satisfy the high iron demand of fast proliferating cells. Here we show that in hepatocellular carcinoma (HCC) TFR1 expression is regulated by miR-148a. Within the TFR1 3′UTR we identified and experimentally validated two evolutionarily conserved miRNA response elements (MREs) for miR-148/152 family members, including miR-148a. Interestingly, analyses of RNA sequencing data from patients with liver hepatocellular carcinoma (LIHC) revealed a significant inverse correlation of TFR1 mRNA levels and miR-148a. In addition, TFR1 mRNA levels were significantly increased in the tumor compared to matched normal healthy tissue, while miR-148a levels are decreased. Functional analysis demonstrated post-transcriptional regulation of TFR1 by miR-148a in HCC cells as well as decreased HCC cell proliferation upon either miR-148a overexpression or TFR1 knockdown. We hypothesize that decreased expression of miR-148a in HCC may elevate transferrin-bound iron uptake, increasing cellular iron levels and cell proliferation.
Collapse
Affiliation(s)
- Kamesh R Babu
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany. .,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
279
|
Zhang Z, Yang J, Min Q, Ling C, Maiti D, Xu J, Qin L, Yang K. Holo-Lactoferrin Modified Liposome for Relieving Tumor Hypoxia and Enhancing Radiochemotherapy of Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1803703. [PMID: 30645056 DOI: 10.1002/smll.201803703] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/05/2018] [Indexed: 05/23/2023]
Abstract
Hypoxic microenvironments in the solid tumor play a negative role in radiotherapy. Holo-lactoferrin (holo-Lf) is a natural protein, which acts as a potential ligand of transferrin receptor (TfR). In this work, an anticancer drug, doxorubicin (Dox)-loaded liposome-holo-Lf nanocomposites, is developed for tumor targeting and imaging guided combined radiochemotherapy. Dox-loaded liposome-holo-Lf (Lf-Liposome-Dox) nanocomposites exhibit significant cellular uptake likely owing to the TfR receptor-mediated targeting accumulation of Lf-Liposome-Dox nanocomposites. Additionally, the nanocomposites exhibit high accumulation in the tumor site after intravenous injection as evidenced from in vivo fluorescence imaging. More importantly, it is found that the holo-Lf has the ability to catalyze the conversion of hydrogen peroxide (H2 O2 ) to oxygen for relieving the tumor hypoxic microenvironment. Photoacoustic imaging further confirms the abundant generation of oxygen in the presence of Lf-Liposome-Dox nanocomposites. Based on these findings, in vivo combined radiochemotherapy is performed using Lf-Liposome-Dox as therapeutic agent, achieving excellent cancer treatment effect. The study further promotes the potential biomedical application of holo-Lf in cancer treatment.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jingrong Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qingqing Min
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chenjie Ling
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Debabrata Maiti
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jiaying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Liqiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
280
|
Breen AF, Wells G, Turyanska L, Bradshaw TD. Development of novel apoferritin formulations for antitumour benzothiazoles. Cancer Rep (Hoboken) 2019; 2:e1155. [PMID: 32721126 PMCID: PMC7941424 DOI: 10.1002/cnr2.1155] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The benzothiazole structure is important in medicinal chemistry, and 5-fluoro-2-(3,4-dimethoxyphenyl) benzothiazole (GW 610) is of particular interest as it shows outstanding anticancer activity in sensitive breast and colorectal carcinoma cell lines via generation of lethal DNA adducts in sensitive cancer cells. Despite promising activity, poor water solubility limits its applications. The apoferritin (AFt) protein cage has been proposed as a robust and biocompatible drug delivery vehicle. AIMS Here, we aim to enhance solubility of GW 610 by developing amino acid prodrug conjugates and utilizing the AFt capsule as drug delivery vessel. METHODS AND RESULTS The potent experimental antitumour agent, GW 610, has been successfully encapsulated within AFt with more than 190 molecules per AFt cage. The AFt-GW 610 complex exhibits dose-dependent growth inhibition and is more potent than GW 610 alone in 5/7 cancer cell lines. To enhance both aqueous solubility and encapsulation efficiency, a series of amino acid esters of GW 608 prodrug were synthesized via N,N'-dicyclohexylcarbodiimide ester coupling to produce molecules with different polarity. A dramatic increase in encapsulation efficiency was achieved, with more than 380 molecules of GW 608-Lys molecules per AFt cage. Release studies show sustained release of the cargo over 12 hours at physiologically relevant pH. The AFt-encapsulated amino acid modified GW 608 complexes are sequestered more rapidly and exhibit more potent anticancer activity than unencapsulated agent. CONCLUSION These results indicate that AFt-encapsulation of GW 610 prodrug provides a biocompatible delivery option for this potent, selective experimental antitumour agent and for amino acid-modified GW 608. Of particular interest is the encapsulation efficiency and in vitro antitumour activity of AFt-GW 608-Lys, which warrants further preclinical evaluation.
Collapse
Affiliation(s)
- Alastair F Breen
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Geoffrey Wells
- School of Pharmacy, University College London, London, UK
| | - Lyudmila Turyanska
- School of Physics and Astronomy, University of Nottingham, Nottingham, UK.,School of Chemistry, University of Lincoln, Lincoln, UK
| | - Tracey D Bradshaw
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
281
|
Jung M, Mertens C, Tomat E, Brüne B. Iron as a Central Player and Promising Target in Cancer Progression. Int J Mol Sci 2019; 20:ijms20020273. [PMID: 30641920 PMCID: PMC6359419 DOI: 10.3390/ijms20020273] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 02/07/2023] Open
Abstract
Iron is an essential element for virtually all organisms. On the one hand, it facilitates cell proliferation and growth. On the other hand, iron may be detrimental due to its redox abilities, thereby contributing to free radical formation, which in turn may provoke oxidative stress and DNA damage. Iron also plays a crucial role in tumor progression and metastasis due to its major function in tumor cell survival and reprogramming of the tumor microenvironment. Therefore, pathways of iron acquisition, export, and storage are often perturbed in cancers, suggesting that targeting iron metabolic pathways might represent opportunities towards innovative approaches in cancer treatment. Recent evidence points to a crucial role of tumor-associated macrophages (TAMs) as a source of iron within the tumor microenvironment, implying that specifically targeting the TAM iron pool might add to the efficacy of tumor therapy. Here, we provide a brief summary of tumor cell iron metabolism and updated molecular mechanisms that regulate cellular and systemic iron homeostasis with regard to the development of cancer. Since iron adds to shaping major hallmarks of cancer, we emphasize innovative therapeutic strategies to address the iron pool of tumor cells or cells of the tumor microenvironment for the treatment of cancer.
Collapse
Affiliation(s)
- Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| |
Collapse
|
282
|
Naves MA, Graminha AE, Vegas LC, Luna-Dulcey L, Honorato J, Menezes ACS, Batista AA, Cominetti MR. Transport of the Ruthenium Complex [Ru(GA)(dppe)2]PF6 into Triple-Negative Breast Cancer Cells Is Facilitated by Transferrin Receptors. Mol Pharm 2019; 16:1167-1183. [DOI: 10.1021/acs.molpharmaceut.8b01154] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | - Antônio C. S. Menezes
- Campus of Exact Sciences and Technology (CCET), State University of Goiás, CEP 75132-903 Anápolis, GO, Brazil
| | | | | |
Collapse
|
283
|
Nguyen HT, Soe ZC, Yang KY, Phung CD, Nguyen LTT, Jeong JH, Jin SG, Choi HG, Ku SK, Yong CS, Kim JO. Transferrin-conjugated pH-sensitive platform for effective delivery of porous palladium nanoparticles and paclitaxel in cancer treatment. Colloids Surf B Biointerfaces 2019; 176:265-275. [PMID: 30623814 DOI: 10.1016/j.colsurfb.2019.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/26/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Porous palladium (Pd) nanoparticles have garnered great research attention due to their potential anticancer activity and photothermal effect. In this study, a transferrin-conjugated pH-sensitive platform (Tf-PPP), comprising porous Pd nanoparticles (PdNPs) and paclitaxel (PTX), was successfully developed for combined chemo-phototherapy. Tf-PPPs have a small size of 164.6 ± 8.7 nm, PDI of 0.278 ± 0.029, and negative charge (-13.2 ± 1.8 mV). Poly(acrylic acid)-poly(ethylene oxide) (PAA-PEO), a pH sensitive polymer, was used to achieve pH-dependent drug release from nanoparticles. Transferrin (Tf) conjugated on the surface of nanoplatforms could enhance the cellular uptake and prolong nanoparticle accumulation in the tumor site. The combination of phototherapy induced by PdNPs and chemotherapeutic agent (PTX) could exhibit synergistic anticancer activities. Consistent findings were observed in both in vitro experiments including cytotoxicity, live/dead assay, and assessment of apoptotic protein levels, and in vivo antitumor study in MCF-7 tumor-bearing mice, with results decreasing in the following order: Tf-PPPs + NIR > Tf-PPPs > PPPs + NIR > PPPs > PTX > PdNPs. These findings suggest that the administration of Tf-PPPs, followed by NIR irradiation could be a promising strategy in the treatment of cancer.
Collapse
Affiliation(s)
- Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea; National Institute of Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Zar Chi Soe
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Kwan Yeol Yang
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Cao Dai Phung
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Lan Thi-Trinh Nguyen
- Department of Pharmaceutical Industry, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan, 426-791, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 712-715, Republic of Korea.
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea.
| |
Collapse
|
284
|
Quilles Junior JC, Carlos FDRR, Montanari A, Leitão A, Mignone VW, Arruda MA, Turyanska L, Bradshaw TD. Apoferritin encapsulation of cysteine protease inhibitors for cathepsin L inhibition in cancer cells. RSC Adv 2019; 9:36699-36706. [PMID: 35539052 PMCID: PMC9075514 DOI: 10.1039/c9ra07161j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/04/2019] [Indexed: 11/21/2022] Open
Abstract
Cysteine proteases play a key role in tumorigenesis causing protein degradation and promoting invasive tumour growth. Cathepsin L is overexpressed in cancer cells and could provide a specific target for delivery of anticancer agents. We encapsulated novel dipeptidyl nitrile based cysteine protease inhibitors (Neq0551, Neq0554 and Neq0568) into biocompatible apoferritin (AFt) protein nanocages to achieve specific delivery to tumours and pH-induced drug release. AFt-encapsulated Neq0554 demonstrated ∼3-fold enhanced in vitro activity (GI50 = 79 μM) compared to naked agent against MiaPaCa-2 pancreatic carcinoma cells. Selectivity for cancer cells was confirmed by comparing their activity to non-tumourigenic human fibroblasts (GI50 > 200 μM). Transferrin receptor (TfR-1) expression, detected only in lysates prepared from carcinoma cells, may contribute to the cancer-selectivity. The G1 cell cycle arrest caused by AFt-Neq0554 resulting in cytostasis was corroborated by clonogenic assays. Superior and more persistent inhibition of cathepsin L up to 80% was achieved with AFt-encapsulated agent in HCT-116 cells following 6 h exposure to 50 μM agent. The selective anticancer activity of AFt-encapsulated cysteine protease inhibitor Neq0554 reported here warrants further preclinical in vivo evaluation. Novel apoferritin encapsulated cysteine protease inhibitors are developed with enhanced and selective uptake by cancer cells, and sustained pH-induced release of the agent. The persistent inhibition of cathepsin L is demonstrated in vitro.![]()
Collapse
Affiliation(s)
- José C. Quilles Junior
- Centre for Biomolecular Sciences
- School of Pharmacy
- University of Nottingham
- UK
- Medicinal Chemistry Group (NEQUIMED)
| | | | - A. Montanari
- Medicinal Chemistry Group (NEQUIMED)
- São Carlos Institute of Chemistry (IQSC)
- University of São Paulo
- Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group (NEQUIMED)
- São Carlos Institute of Chemistry (IQSC)
- University of São Paulo
- Brazil
| | | | | | | | - Tracey D. Bradshaw
- Centre for Biomolecular Sciences
- School of Pharmacy
- University of Nottingham
- UK
| |
Collapse
|
285
|
Das M, Musetti S, Huang L. RNA Interference-Based Cancer Drugs: The Roadblocks, and the "Delivery" of the Promise. Nucleic Acid Ther 2018; 29:61-66. [PMID: 30562145 DOI: 10.1089/nat.2018.0762] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Nucleic acid-based therapeutics like synthetic small interfering RNAs have been exploited to modulate gene function, taking advantage of RNA interference (RNAi), an evolutionally conserved biological process. Recently, the world's first RNAi drug was approved for a rare genetic disorder in the liver. However, there are significant challenges that need to be resolved before RNAi can be translated in other genetic diseases like cancer. Current drug delivery platforms for therapeutic silencing RNAs are tailored to hepatic targets. RNAi therapies for nonhepatic conditions are still at early clinical phases. In this study, we discuss the critical design considerations in anticancer RNAi drug development, insights gained from initial clinical trials, and new strategies that are entering clinical development, shaping the future of RNAi in cancer.
Collapse
Affiliation(s)
- Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sara Musetti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
286
|
Choudhury H, Pandey M, Chin PX, Phang YL, Cheah JY, Ooi SC, Mak KK, Pichika MR, Kesharwani P, Hussain Z, Gorain B. Transferrin receptors-targeting nanocarriers for efficient targeted delivery and transcytosis of drugs into the brain tumors: a review of recent advancements and emerging trends. Drug Deliv Transl Res 2018; 8:1545-1563. [PMID: 29916012 DOI: 10.1007/s13346-018-0552-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Treatment of glioblastoma multiforme (GBM) is a predominant challenge in chemotherapy due to the existence of blood-brain barrier (BBB) which restricts delivery of chemotherapeutic agents to the brain together with the problem of drug penetration through hard parenchyma of the GBM. With the structural and mechanistic elucidation of the BBB under both physiological and pathological conditions, it is now viable to target central nervous system (CNS) disorders utilizing the presence of transferrin (Tf) receptors (TfRs). However, overexpression of these TfRs on the GBM cell surface can also help to avoid restrictions of GBM cells to deliver chemotherapeutic agents within the tumor. Therefore, targeting of TfR-mediated delivery could counteract drug delivery issues in GBM and create a delivery system that could cross the BBB effectively to utilize ligand-conjugated drug complexes through receptor-mediated transcytosis. Hence, approach towards successful delivery of antitumor agents to the gliomas has been making possible through targeting these overexpressed TfRs within the CNS and glioma cells. This review article presents a thorough analysis of current understanding on Tf-conjugated nanocarriers as efficient drug delivery system.
Collapse
Affiliation(s)
- Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia.
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Pei Xin Chin
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Yee Lin Phang
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Jeng Yuen Cheah
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Shu Chien Ooi
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Kit-Kay Mak
- School of Postgraduate Studies and Research, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Prashant Kesharwani
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300, Puncak Alam, Malaysia
| | - Bapi Gorain
- Faculty of Pharmacy, Lincoln University College, Petalling Jaya, 47301, Kuala Lumpur, Selangor, Malaysia
| |
Collapse
|
287
|
Multimodal highly fluorescent-magnetic nanoplatform to target transferrin receptors in cancer cells. Biochim Biophys Acta Gen Subj 2018; 1862:2788-2796. [DOI: 10.1016/j.bbagen.2018.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/31/2018] [Accepted: 08/17/2018] [Indexed: 01/16/2023]
|
288
|
Perring J, Crawshay-Williams F, Huang C, Townley HE. Bio-inspired melanin nanoparticles induce cancer cell death by iron adsorption. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:181. [PMID: 30506101 PMCID: PMC6267116 DOI: 10.1007/s10856-018-6190-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/09/2018] [Indexed: 05/04/2023]
Abstract
Dysregulation of iron metabolism is a common characteristic of cancer cells. The rapid proliferation of the tumour cells means that there is an increased dependence upon iron compared to healthy cells. Chelation of iron can be undertaken with a number of different compounds, however, simply lowering systemic iron levels to control tumour growth is not possible since iron is essential for cellular metabolism in the rest of the body. Nanoparticulate iron chelators could overcome this difficulty by targeting to the tumour either by the passive enhanced permeation and retention effect, or by targeting ligands on the surface. Nanoparticles were prepared from melanin, which is a naturally occurring pigment that is widely distributed within the body, but that can chelate iron. The prepared nanoparticles were shown to be ~220 nm, and could adsorb 16.45 mmoles iron/g melanin. The nanoparticles showed no affect on control fibroblast cells at a concentration of 200 μM, whereas the immortalised cancer cell lines showed at least 56% reduction in cell growth. At a concentration of 1 mM melanin nanoparticles the cell growth could be reduced by 99% compared to the control. The nanoparticles also show no significant haemotoxicity, even at concentration of 500 μM. Melanin nanoparticles are therefore a viable prospect for destroying cancer cells via iron starvation.
Collapse
Affiliation(s)
- James Perring
- Department of Medical Sciences, Oxford University, Oxford, Oxfordshire, UK
| | | | - Cindy Huang
- Department of Women's and Reproductive Health, Oxford University, Oxford, Oxfordshire, UK
| | - Helen E Townley
- Department of Women's and Reproductive Health, Oxford University, Oxford, Oxfordshire, UK.
- Department of Engineering Science, Oxford University, Oxford, Oxfordshire, UK.
| |
Collapse
|
289
|
Alhajj N, Chee CF, Wong TW, Rahman NA, Abu Kasim NH, Colombo P. Lung cancer: active therapeutic targeting and inhalational nanoproduct design. Expert Opin Drug Deliv 2018; 15:1223-1247. [DOI: 10.1080/17425247.2018.1547280] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Nasser Alhajj
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Chin Fei Chee
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Noorsaadah Abd Rahman
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Wellness Research Cluster, Institute of Research Management & Monitoring, University of Malaya, Kuala Lumpur, Malaysia
| | - Paolo Colombo
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| |
Collapse
|
290
|
Sherman HG, Jovanovic C, Stolnik S, Baronian K, Downard AJ, Rawson FJ. New Perspectives on Iron Uptake in Eukaryotes. Front Mol Biosci 2018; 5:97. [PMID: 30510932 PMCID: PMC6254016 DOI: 10.3389/fmolb.2018.00097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
All eukaryotic organisms require iron to function. Malfunctions within iron homeostasis have a range of physiological consequences, and can lead to the development of pathological conditions that can result in an excess of non-transferrin bound iron (NTBI). Despite extensive understanding of iron homeostasis, the links between the “macroscopic” transport of iron across biological barriers (cellular membranes) and the chemistry of redox changes that drive these processes still needs elucidating. This review draws conclusions from the current literature, and describes some of the underlying biophysical and biochemical processes that occur in iron homeostasis. By first taking a broad view of iron uptake within the gut and subsequent delivery to tissues, in addition to describing the transferrin and non-transferrin mediated components of these processes, we provide a base of knowledge from which we further explore NTBI uptake. We provide concise up-to-date information of the transplasma electron transport systems (tPMETSs) involved within NTBI uptake, and highlight how these systems are not only involved within NTBI uptake for detoxification but also may play a role within the reduction of metabolic stress through regeneration of intracellular NAD(P)H/NAD(P)+ levels. Furthermore, we illuminate the thermodynamics that governs iron transport, namely the redox potential cascade and electrochemical behavior of key components of the electron transport systems that facilitate the movement of electrons across the plasma membrane to the extracellular compartment. We also take account of kinetic changes that occur to transport iron into the cell, namely membrane dipole change and their consequent effects within membrane structure that act to facilitate transport of ions.
Collapse
Affiliation(s)
- Harry G Sherman
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | | | - Snow Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Kim Baronian
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Alison J Downard
- MacDiarmid Institute for Advanced Materials and Nanotechnology, School of Physical and Chemical Sciences, University of Canterbury, Christchurch, New Zealand
| | - Frankie J Rawson
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
291
|
Sutar YB, Telvekar VN. Chitosan based copolymer-drug conjugate and its protein targeted polyelectrolyte complex nanoparticles to enhance the efficiency and specificity of low potency anticancer agent. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:393-406. [DOI: 10.1016/j.msec.2018.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 06/05/2018] [Accepted: 07/01/2018] [Indexed: 11/25/2022]
|
292
|
Gonda A, Kabagwira J, Senthil GN, Ferguson Bennit HR, Neidigh JW, Khan S, Wall NR. Exosomal survivin facilitates vesicle internalization. Oncotarget 2018; 9:34919-34934. [PMID: 30405884 PMCID: PMC6201849 DOI: 10.18632/oncotarget.26182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 09/15/2018] [Indexed: 12/11/2022] Open
Abstract
Survivin, a member of the inhibitor of apoptosis (IAP) protein family plays a significant role in cell fate and function. It is significantly overexpressed in tumor cells and has been identified in most cancer cell types. A novel extracellular population has recently been identified and its function is still unknown. Emerging evidence continues to shed light on the important role the tumor microenvironment (TME) has on tumor survival and progression. This new population of survivin has been seen to enhance the tumor phenotype when internalized by recipient cells. In this paper, we sought to better understand the mechanism by which survivin is taken up by cancer cells and the possible role it plays in this phenomenon. We isolated the exosomal carriers of extracellular survivin and using a lipophilic stain, PKH67, we tracked their uptake with immunofluorescence and flow cytometry. We found that by blocking exosomal survivin, exosome internalization is reduced, signifying a novel function for this protein. We also discovered that the common membrane receptors, transferrin receptor, endothelin B receptor, insulin receptor alpha, and membrane glucocorticoid receptor all facilitate exosomal internalization. This understanding further clarifies the protein-protein interactions in the TME that may influence tumor progression and identifies additional potential chemotherapeutic targets.
Collapse
Affiliation(s)
- Amber Gonda
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
- Department of Basic Sciences, Division of Anatomy, Loma Linda University, Loma Linda, California, 92350, USA
| | - Janviere Kabagwira
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, California, 92350, USA
| | - Girish N. Senthil
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
| | - Heather R. Ferguson Bennit
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, California, 92350, USA
| | - Jonathan W. Neidigh
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, California, 92350, USA
| | - Salma Khan
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, California, 92350, USA
| | - Nathan R. Wall
- Center for Health Disparities Research and Molecular Medicine, Loma Linda University, Loma Linda, California, 92350, USA
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, California, 92350, USA
| |
Collapse
|
293
|
Chen Q, Liu J. Transferrin and folic acid co-modified bufalin-loaded nanoliposomes: preparation, characterization, and application in anticancer activity. Int J Nanomedicine 2018; 13:6009-6018. [PMID: 30323588 PMCID: PMC6179727 DOI: 10.2147/ijn.s176012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim The aim of this study was to prepare transferrin (Tf) and folic acid (FA) co-modified bufalin (BF) liposomes for lung cancer treatment. Method In this study, (FA+Tf) BF-LPs were prepared using the high-pressure homogenization method. Results The EE% and DL% of prepared LPs were 82.3% and 10.7%, respectively, and the mean diameter was 120.4 nm from three batches. In vitro release showed that the release of BF from (FA+Tf) BF-LPs was slow with burst effects at an early stage. In vitro cytotoxicity assay showed that (FA+Tf) BF-LPs had a superior antiproliferative effect on A549 cells. An in vivo imaging study indicated that (FA+Tf) BF-LPs had obvious targeting characteristics on subcutaneous tumor, with the potential to actively deliver drugs to tumor tissues. In terms of the in vivo antitumor activity, (FA+Tf) BF-LPs treated mice showed a significantly suppressed tumor growth and no systemic toxicity in the body. Conclusion Through this study, it was found that the Tf and FA co-modified BF could be a very promising lung target preparation.
Collapse
Affiliation(s)
- Qiankun Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Liu
- Department of Anesthesia, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China,
| |
Collapse
|
294
|
Quantum dot-based fluorescent probes for targeted imaging of the EJ human bladder urothelial cancer cell line. Exp Ther Med 2018; 16:4779-4783. [PMID: 30546399 DOI: 10.3892/etm.2018.6805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 03/16/2018] [Indexed: 12/28/2022] Open
Abstract
QDs are a type of inorganic nanoparticle with unique optical properties. As a fluorescent label, QDs are widely used in biomedical fields. In the present study, fluorescent probes of quantum dots (QDs) conjugated with a prostate stem cell antigen (PSCA) monoclonal antibody (QD-PSCA) were prepared to study the targeted imaging of QD-PSCA probes in EJ human bladder urothelial cancer cells and analyze the feasibility of QD-based non-invasive tumor-targeted imaging in vivo. QDs with an emission wavelength of 605 nm (QD605) were conjugated with PSCA to prepare QD605-PSCA fluorescent probes by chemical covalent coupling. The optical properties of the probes coupled and uncoupled with PSCA were measured and assessed using an ultraviolet spectrophotometer and a fluorescence spectrophotometer. Direct immune-fluorescent labeling was utilized to detect and analyze imaging of the probes for EJ cells. The results revealed that QD605-PSCA probes retained the fluorescent properties of QD605 and the immunogenicity of the PSCA protein. The probes were able to specifically recognize the PSCA protein expressed in bladder cancer cells, while fluorescence was stable and had a long duration. The present study suggests that QD-PSCA fluorescent probes may be useful for specific targeted labeling and imaging in bladder urothelial cancer cells. Furthermore, the probes possess good optical stability and may be useful for research into non-invasive targeted imaging, early diagnosis and targeted in vivo tumor therapy.
Collapse
|
295
|
Henry KE, Dacek MM, Dilling TR, Caen JD, Fox IL, Evans MJ, Lewis JS. A PET Imaging Strategy for Interrogating Target Engagement and Oncogene Status in Pancreatic Cancer. Clin Cancer Res 2018; 25:166-176. [PMID: 30228208 DOI: 10.1158/1078-0432.ccr-18-1485] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/17/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers, with a 5-year survival rate of less than 10%. Physicians often rely on biopsy or CT to guide treatment decisions, but these techniques fail to reliably measure the actions of therapeutic agents in PDAC. KRAS mutations are present in >90% of PDAC and are connected to many signaling pathways through its oncogenic cascade, including extracellular regulated kinase (ERK) and MYC. A key downstream event of MYC is transferrin receptor (TfR), which has been identified as a biomarker for cancer therapeutics and imaging. EXPERIMENTAL DESIGN In this study, we aimed to test whether zirconium-89 transferrin ([89Zr]Zr-Tf) could measure changes in MYC depending on KRAS status of PDAC, and assess target engagement of anti-MYC and anti-ERK-targeted therapies. RESULTS Mice bearing iKras*p53* tumors showed significantly higher (P < 0.05) uptake of [89Zr]Zr-Tf in mice withdrawn from inducible oncogenic KRAS. A therapy study with JQ1 showed a statistically significant decrease (P < 0.05) of [89Zr]Zr-Tf uptake in drug versus vehicle-treated mice bearing Capan-2 and Suit-2 xenografts. IHC analysis of resected PDAC tumors reflects the data observed via PET imaging and radiotracer biodistribution. CONCLUSIONS Our study demonstrates that [89Zr]Zr-Tf is a valuable tool to noninvasively assess oncogene status and target engagement of small-molecule inhibitors downstream of oncogenic KRAS, allowing a quantitative assessment of drug delivery.
Collapse
Affiliation(s)
- Kelly E Henry
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Megan M Dacek
- Program of Molecular Pharmacology and Chemistry, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill Cornell Medical College, New York, New York
| | - Thomas R Dilling
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan D Caen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian L Fox
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Evans
- Departments of Radiology and Biomedical Imaging, and Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Program of Molecular Pharmacology and Chemistry, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill Cornell Medical College, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
296
|
Transferrin as a thermosensitizer in radiofrequency hyperthermia for cancer treatment. Sci Rep 2018; 8:13505. [PMID: 30202000 PMCID: PMC6131143 DOI: 10.1038/s41598-018-31232-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/06/2018] [Indexed: 01/05/2023] Open
Abstract
One of the main characteristics of cancer tissues is poor development of neovascularization that results in a limited blood circulation. Because of this phenomenon, it is harder for cancer tissues to diffuse their elevated heat into other parts of the body. The scientific principle of radiofrequency hyperthermia relies on this quality of cancer tissues which with higher temperature becomes more apparent. Despite the obvious necessity to selectively heat the cancer tissue for radiofrequency hyperthermia, a proper thermosensitizer has not been developed until now. Here, we show that transferrin containing ferric ion could be an ideal thermosensitizer for the increased efficiency of radiofrequency hyperthermia. In our result, the ferric ion-enriched cancer tissues dramatically react with 13.56 MHz radiofrequency wave to cause cancer-selective dielectric temperature increment. The overall anticancer efficacy of a 13.56 MHz radiofrequency hyperthermia using transferrin as a thermosensitizer was much higher than the oncotherapeutic efficacy of paclitaxel, successfully eradicating cancer in a tumor-xenografted mouse experiment.
Collapse
|
297
|
Kumari P, Rompicharla SVK, Muddineti OS, Ghosh B, Biswas S. Transferrin-anchored poly(lactide) based micelles to improve anticancer activity of curcumin in hepatic and cervical cancer cell monolayers and 3D spheroids. Int J Biol Macromol 2018; 116:1196-1213. [DOI: 10.1016/j.ijbiomac.2018.05.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/06/2018] [Accepted: 05/08/2018] [Indexed: 12/29/2022]
|
298
|
Deshpande P, Jhaveri A, Pattni B, Biswas S, Torchilin V. Transferrin and octaarginine modified dual-functional liposomes with improved cancer cell targeting and enhanced intracellular delivery for the treatment of ovarian cancer. Drug Deliv 2018; 25:517-532. [PMID: 29433357 PMCID: PMC6058534 DOI: 10.1080/10717544.2018.1435747] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Off-target effects of drugs severely limit cancer therapy. Targeted nanocarriers are promising to enhance the delivery of therapeutics to tumors. Among many approaches for active tumor-targeting, arginine-rich cell penetrating peptides (AR-CPP) and ligands specific to target over-expressed receptors on cancer-cell surfaces, are popular. Earlier, we showed that the attachment of an AR-CPP octaarginine (R8) to the surface of DOXIL® (Doxorubicin encapsulated PEGylated liposomes) improved cytoplasmic and nuclear DOX delivery that enhanced the cytotoxic effect in vitro and improved therapeutic efficacy in vivo. Here, we report on DOX-loaded liposomes, surface-modified with, R8 and transferrin (Tf) (Dual DOX-L), to improve targeting of A2780 ovarian carcinoma cells via the over-expressed transferrin receptors (TfRs) with R8-mediated intracellular DOX delivery. Flow cytometry analysis with fluorescently labeled DualL (without DOX) showed two-fold higher cancer-cell association than other treatments after 4 h treatment. Blocking entry pathways of R8 (macropinocytosis) and Tf (receptor-mediated endocytosis, RME) resulted in a decreased cancer-cell association of DualL. Confocal microscopy confirmed involvement of both entry pathways and cytoplasmic liposome accumulation with nuclear DOX delivery for Dual DOX-L. Dual DOX-L exhibited enhanced cytotoxicity in vitro and was most effective in controlling tumor growth in vivo in an A2780 ovarian xenograft model compared to other treatments. A pilot biodistribution study showed improved DOX accumulation in tumors after Dual DOX-L treatment. All results collectively presented a clear advantage of the R8 and Tf combination to elevate the therapeutic potential of DOX-L by exploiting TfR over-expression imparting specificity followed by endosomal escape and intracellular delivery via R8.
Collapse
Affiliation(s)
- Pranali Deshpande
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Aditi Jhaveri
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Bhushan Pattni
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Swati Biswas
- b Department of Pharmacy , Birla Institute of Technology & Science-Pilani, Hyderabad Campus , Hyderabad , India
| | - Vladimir Torchilin
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| |
Collapse
|
299
|
Understanding Ovarian Cancer: iTRAQ-Based Proteomics for Biomarker Discovery. Int J Mol Sci 2018; 19:ijms19082240. [PMID: 30065196 PMCID: PMC6121953 DOI: 10.3390/ijms19082240] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023] Open
Abstract
Despite many years of studies, ovarian cancer remains one of the top ten cancers worldwide. Its high mortality rate is mainly due to lack of sufficient diagnostic methods. For this reason, our research focused on the identification of blood markers whose appearance would precede the clinical manifestation of the disease. ITRAQ-tagging (isobaric Tags for Relative and Absolute Quantification) coupled with mass spectrometry technology was applied. Three groups of samples derived from patients with: ovarian cancer, benign ovarian tumor, and healthy controls, were examined. Mass spectrometry analysis allowed for highlighting the dysregulation of several proteins associated with ovarian cancer. Further validation of the obtained results indicated that five proteins (Serotransferrin, Amyloid A1, Hemopexin, C-reactive protein, Albumin) were differentially expressed in ovarian cancer group. Interestingly, the addition of Albumin, Serotransferrin, and Amyloid A1 to CA125 (cancer antigen 125) and HE4 (human epididymis protein4) improved the diagnostic performance of the model discriminating between benign and malignant tumors. Identified proteins shed light on the molecular signaling pathways that are associated with ovarian cancer development and should be further investigated in future studies. Our findings indicate five proteins with a strong potential to use in a multimarker test for screening and detection of ovarian cancer.
Collapse
|
300
|
Artemisinins—a Promising New Treatment for Systemic Lupus Erythematosus: a Descriptive Review. Curr Rheumatol Rep 2018; 20:55. [DOI: 10.1007/s11926-018-0764-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|