401
|
Lei Y, Hamada Y, Li J, Cong L, Wang N, Li Y, Zheng W, Jiang X. Targeted tumor delivery and controlled release of neuronal drugs with ferritin nanoparticles to regulate pancreatic cancer progression. J Control Release 2016; 232:131-42. [PMID: 27046157 DOI: 10.1016/j.jconrel.2016.03.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/17/2016] [Accepted: 03/14/2016] [Indexed: 01/06/2023]
Abstract
Pancreatic cancer is a lethal malignancy whose progression is highly dependent on the nervous microenvironment. This study develops neural drug-loaded ferritin nanoparticles (Ft NPs) to regulate the nervous microenvironment, in order to control the pancreatic cancer progression. The drug-loaded Ft NPs can target pancreatic tumors via passive targeting of EPR effects of tumors and active targeting via transferrin receptor 1 (TfR1) binding on cancer cells, with a triggered drug release in acidic tumor environment. Two drugs, one activates neural activity (carbachol), the other impairs neural activity (atropine), are encapsulated into the Ft NPs to form two kinds of nano drugs, Nano-Cab NPs and Nano-Ato NPs, respectively. The activation of the nervous microenvironment by Nano-Cab NPs significantly promotes the pancreatic tumor progression, whereas the blockage of neural niche by Nano-Ato NPs remarkably impairs the neurogenesis in tumors and the progression of pancreatic cancer. The Ft-based nanoparticles thus comprise an effective and safe route of delivery of neural drugs for novel anti-cancer therapy.
Collapse
Affiliation(s)
- Yifeng Lei
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China
| | - Yoh Hamada
- Department of Nano-Medical Science, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Jun Li
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China
| | - Liman Cong
- Department of Nano-Medical Science, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Nuoxin Wang
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China
| | - Ying Li
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China
| | - Wenfu Zheng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China.
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China.
| |
Collapse
|
402
|
Sonali, Singh RP, Singh N, Sharma G, Vijayakumar MR, Koch B, Singh S, Singh U, Dash D, Pandey BL, Muthu MS. Transferrin liposomes of docetaxel for brain-targeted cancer applications: formulation and brain theranostics. Drug Deliv 2016; 23:1261-71. [PMID: 26961144 DOI: 10.3109/10717544.2016.1162878] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diagnosis and therapy of brain cancer was often limited due to low permeability of delivery materials across the blood-brain barrier (BBB) and their poor penetration into the brain tissue. This study explored the possibility of utilizing theranostic d-alpha-tocopheryl polyethylene glycol 1000 succinate mono-ester (TPGS) liposomes as nanocarriers for minimally invasive brain-targeted imaging and therapy (brain theranostics). The aim of this work was to formulate transferrin conjugated TPGS coated theranostic liposomes, which contain both docetaxel and quantum dots (QDs) for imaging and therapy of brain cancer. The theranostic liposomes with and without transferrin decoration were prepared and characterized for their particle size, polydispersity, morphology, drug encapsulation efficiency, in-vitro release study and brain theranostics. The particle sizes of the non-targeted and targeted theranostic liposomes were found below 200 nm. Nearly, 71% of drug encapsulation efficiency was achieved with liposomes. The drug release from transferrin conjugated theranostic liposomes was sustained for more than 72 h with 70% of drug release. The in-vivo results indicated that transferrin receptor-targeted theranostic liposomes could be a promising carrier for brain theranostics due to nano-sized delivery and its permeability which provided an improved and prolonged brain targeting of docetaxel and QDs in comparison to the non-targeted preparations.
Collapse
Affiliation(s)
- Sonali
- a Department of Pharmacology
| | | | - Nitesh Singh
- b Department of Biochemistry , Institute of Medical Sciences, Banaras Hindu University , Varanasi, Uttar Pradesh , India
| | - Gunjan Sharma
- c Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh , India
| | - Mahalingam R Vijayakumar
- d Department of Pharmaceutics , Indian Institute of Technology, Banaras Hindu University , Varanasi, Uttar Pradesh , India , and
| | - Biplob Koch
- c Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh , India
| | - Sanjay Singh
- d Department of Pharmaceutics , Indian Institute of Technology, Banaras Hindu University , Varanasi, Uttar Pradesh , India , and
| | - Usha Singh
- e Department of Pathology , Institute of Medical Sciences, Banaras Hindu University , Varanasi, Uttar Pradesh , India
| | - Debabrata Dash
- b Department of Biochemistry , Institute of Medical Sciences, Banaras Hindu University , Varanasi, Uttar Pradesh , India
| | | | - Madaswamy S Muthu
- a Department of Pharmacology .,d Department of Pharmaceutics , Indian Institute of Technology, Banaras Hindu University , Varanasi, Uttar Pradesh , India , and
| |
Collapse
|
403
|
Friberg S, Nyström AM. NANOMEDICINE: will it offer possibilities to overcome multiple drug resistance in cancer? J Nanobiotechnology 2016; 14:17. [PMID: 26955956 PMCID: PMC4784447 DOI: 10.1186/s12951-016-0172-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
This review is written with the purpose to review the current nanomedicine literature and provide an outlook on the developments in utilizing nanoscale drug constructs in treatment of solid cancers as well as in the potential treatment of multi-drug resistant cancers. No specific design principles for this review have been utilized apart from our active choice to avoid results only based on in vitro studies. Few drugs based on nanotechnology have progressed to clinical trials, since most are based only on in vitro experiments which do not give the necessary data for the research to progress towards pre-clinical studies. The area of nanomedicine has indeed spark much attention and holds promise for improved future therapeutics in the treatment of solid cancers. However, despite much investment few targeted therapeutics have successfully progressed to early clinical trials, indicating yet again that the human body is complicated and that much more understanding of the fundamentals of receptor interactions, physics of nanomedical constructs and their circulation in the body is indeed needed. We believe that nanomedical therapeutics can allow for more efficient treatments of resistant cancers, and may well be a cornerstone for RNA based therapeutics in the future given their general need for shielding from the harsh environment in the blood stream.
Collapse
Affiliation(s)
- Sten Friberg
- Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Retzius väg 8, 171 77, Stockholm, Sweden.
| | - Andreas M Nyström
- Institute of Environmental Medicine, Karolinska Institutet, Nobels väg 13, 171 77, Stockholm, Sweden.
| |
Collapse
|
404
|
Wang X, Sun Q, Shen S, Xu Y, Huang L. Nanotrastuzumab in combination with radioimmunotherapy: Can it be a viable treatment option for patients with HER2-positive breast cancer with brain metastasis? Med Hypotheses 2016; 88:79-81. [DOI: 10.1016/j.mehy.2015.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
|
405
|
Masood F. Polymeric nanoparticles for targeted drug delivery system for cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 60:569-578. [DOI: 10.1016/j.msec.2015.11.067] [Citation(s) in RCA: 307] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 10/30/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022]
|
406
|
Jeong SM, Hwang S, Seong RH. Transferrin receptor regulates pancreatic cancer growth by modulating mitochondrial respiration and ROS generation. Biochem Biophys Res Commun 2016; 471:373-9. [PMID: 26869514 DOI: 10.1016/j.bbrc.2016.02.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/06/2016] [Indexed: 12/13/2022]
Abstract
The transferrin receptor (TfR1) is upregulated in malignant cells and its expression is associated with cancer progression. Because of its pre-eminent role in cell proliferation, TfR1 has been an important target for the development of cancer therapy. Although TfR1 is highly expressed in pancreatic cancers, what it carries out in these refractory cancers remains poorly understood. Here we report that TfR1 supports mitochondrial respiration and ROS production in human pancreatic ductal adenocarcinoma (PDAC) cells, which is required for their tumorigenic growth. Elevated TfR1 expression in PDAC cells contributes to oxidative phosphorylation, which allows for the generation of ROS. Importantly, mitochondrial-derived ROS are essential for PDAC growth. However, exogenous iron supplement cannot rescue the defects caused by TfR1 knockdown. Moreover, we found that TfR1 expression determines PDAC cells sensitivity to oxidative stress. Together, our findings reveal that TfR1 can contribute to the mitochondrial respiration and ROS production, which have essential roles in growth and survival of pancreatic cancer.
Collapse
Affiliation(s)
- Seung Min Jeong
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea; Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.
| | - Sunsook Hwang
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea
| | - Rho Hyun Seong
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea
| |
Collapse
|
407
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
408
|
Zhao H, Wang S, Nguyen SN, Elci SG, Kaltashov IA. Evaluation of Nonferrous Metals as Potential In Vivo Tracers of Transferrin-Based Therapeutics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:211-9. [PMID: 26392277 PMCID: PMC4724545 DOI: 10.1007/s13361-015-1267-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 05/10/2023]
Abstract
Transferrin (Tf) is a promising candidate for targeted drug delivery. While development of such products is impossible without the ability to monitor biodistribution of Tf-drug conjugates in tissues and reliable measurements of their levels in blood and other biological fluids, the presence of very abundant endogenous Tf presents a significant impediment to such efforts. Several noncognate metals have been evaluated in this work as possible tracers of exogenous transferrin in complex biological matrices using inductively coupled plasma mass spectrometry (ICP MS) as a detection tool. Placing Ni(II) on a His-tag of recombinant Tf resulted in formation of a marginally stable protein-metal complex, which readily transfers the metal to ubiquitous physiological scavengers, such as serum albumin. An alternative strategy targeted iron-binding pockets of Tf, where cognate Fe(III) was replaced by metal ions known to bind this protein. Both Ga(III) and In(III) were evaluated, with the latter being vastly superior as a tracer (stronger binding to Tf unaffected by the presence of metal scavengers and the retained ability to associate with Tf receptor). Spiking serum with indium-loaded Tf followed by ICP MS detection demonstrated that protein quantities as low as 0.04 nM can be readily detected in animal blood. Combining laser ablation with ICP MS detection allows distribution of exogenous Tf to be mapped within animal tissue cross-sections with spatial resolution exceeding 100 μm. The method can be readily extended to a range of other therapeutics where metalloproteins are used as either carriers or payloads. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Hanwei Zhao
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Shunhai Wang
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Son N Nguyen
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - S Gokhan Elci
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Igor A Kaltashov
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
409
|
Tang HC, Chang PC, Chen YC. Iron depletion strategy for targeted cancer therapy: utilizing the dual roles of neutrophil gelatinase-associated lipocalin protein. J Mol Model 2016; 22:32. [PMID: 26757915 DOI: 10.1007/s00894-015-2897-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/21/2015] [Indexed: 11/24/2022]
Abstract
Decreasing iron uptake and increasing iron efflux may result in cell death by oxidative inactivation of vital enzymes. Applying the dual function of neutrophil gelatinase-associated lipocalin (NGAL) could achieve the goal of iron depletion in the cancer cells. Tyr106, Lys125 or Lys134 was the key binding site for NGAL protein to sequester iron-chelating siderophores. In this study, we employed all bioactive peptides in peptide databank to dock with the siderophore-binding sites of NGAL protein by virtual screening. In addition, we performed molecular dynamics (MD) simulation to observe the molecular character and structural variation of ligand-protein interaction. Glu-Glu-Lys-Glu (EEKE), Glu-Glu-Asp-Cys-Lys (EEDCK), and Gly-Glu-Glu-Cys-Asp (GEECD) were selected preliminarily by rigorous scoring functions for further investigation. GEECD was excluded due to higher binding total energy than the others. Moreover, we also excluded EEKE due to larger influence to the stability of binding residues by the information of root mean square fluctuation (RMSF) and principal component analysis (PCA). Thus, we suggested that EEDCK was the potential bioactive peptide which had been proved to inhibit malignant cells for targeted cancer therapy. Graphical Abstract Perspective drug design of occupying the siderophore-binding sites of NGAL outside the cell temporarily by a potential short peptide until NGAL enters into the cell, and releasing the siderophore-binding sites inside the cell.
Collapse
Affiliation(s)
- Hsin-Chieh Tang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Yu-Chian Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan. .,Human Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, 40402, Taiwan. .,Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
410
|
van der Meel R, Vehmeijer LJC, Kok RJ, Storm G, van Gaal EVB. Ligand-targeted Particulate Nanomedicines Undergoing Clinical Evaluation: Current Status. INTRACELLULAR DELIVERY III 2016. [DOI: 10.1007/978-3-319-43525-1_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
411
|
Gu B, Cai J, Zhang J, Xu X, Luo J, Zhou X, Zheng Y, Zhang Y. 99mTc-labeled and gadolinium-chelated transferrin enhances the sensitivity and specificity of dual-modality SPECT/MR imaging of breast cancer. RSC Adv 2016. [DOI: 10.1039/c5ra22934k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A dual-modal probe 99mTc–Tf–DTPA–Gd could provide high spatial resolution and high sensitivity images of breast tumor.
Collapse
Affiliation(s)
- Bingxin Gu
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| | - Jiali Cai
- Changzheng Hospital
- Secondary Military Medical University
- Shanghai
- China
| | - Jianping Zhang
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| | - Xiaoping Xu
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| | - Jianming Luo
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| | - Xiaobao Zhou
- The Key Laboratory of Resource Chemistry of Ministry of Education
- College of Life and Environmental Science
- Shanghai Normal University
- Shanghai
- China
| | - Yingying Zheng
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| | - Yingjian Zhang
- Department of Nuclear Medicine
- Fudan University Shanghai Cancer Center
- Shanghai
- China
- Center for Biomedical Imaging
| |
Collapse
|
412
|
Dumoga S, Dey N, Kaur A, Singh S, Mishra AK, Kakkar D. Novel biotin-functionalized lipidic nanocarriers for encapsulating BpT and Bp4eT iron chelators: evaluation of potential anti-tumour efficacy by in vitro, in vivo and pharmacokinetic studies in A549 mice models. RSC Adv 2016. [DOI: 10.1039/c6ra03079c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This work proposes a novel strategy for delivery of iron chelators to the tumour cells which is exemplified in A549 mice models by using lipidic nanocarriers and introducing biotin based targeting.
Collapse
Affiliation(s)
- Shweta Dumoga
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
- Department of Chemistry
- University of Delhi
| | - Namit Dey
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | - Anivind Kaur
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | | | - Anil K. Mishra
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| | - Dipti Kakkar
- Institute of Nuclear Medicine and Allied Sciences
- Timarpur
- Delhi-110054
| |
Collapse
|
413
|
Rother M, Nussbaumer MG, Renggli K, Bruns N. Protein cages and synthetic polymers: a fruitful symbiosis for drug delivery applications, bionanotechnology and materials science. Chem Soc Rev 2016; 45:6213-6249. [DOI: 10.1039/c6cs00177g] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protein cages have become essential tools in bionanotechnology due to their well-defined, monodisperse, capsule-like structure. Combining them with synthetic polymers greatly expands their application, giving rise to novel nanomaterials fore.g.drug-delivery, sensing, electronic devices and for uses as nanoreactors.
Collapse
Affiliation(s)
- Martin Rother
- Department of Chemistry
- University of Basel
- CH-4056 Basel
- Switzerland
| | - Martin G. Nussbaumer
- Wyss Institute for Biologically Inspired Engineering
- Harvard University
- Cambridge
- USA
| | - Kasper Renggli
- Department of Biosystems Science and Engineering
- ETH Zürich
- 4058 Basel
- Switzerland
| | - Nico Bruns
- Adolphe Merkle Institute
- University of Fribourg
- CH-1700 Fribourg
- Switzerland
| |
Collapse
|
414
|
Sobot D, Mura S, Couvreur P. How can nanomedicines overcome cellular-based anticancer drug resistance? J Mater Chem B 2016; 4:5078-5100. [DOI: 10.1039/c6tb00900j] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review discusses the mechanisms of anticancer drug resistance according to its cellular level of action and outlines the nanomedicine-based strategies adopted to overcome it.
Collapse
Affiliation(s)
- Dunja Sobot
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| | - Simona Mura
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| | - Patrick Couvreur
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| |
Collapse
|
415
|
CdTe quantum dots as fluorescent probes to study transferrin receptors in glioblastoma cells. Biochim Biophys Acta Gen Subj 2016; 1860:28-35. [DOI: 10.1016/j.bbagen.2015.09.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/18/2015] [Accepted: 09/30/2015] [Indexed: 12/17/2022]
|
416
|
Kuruppu AI, Zhang L, Collins H, Turyanska L, Thomas NR, Bradshaw TD. An Apoferritin-based Drug Delivery System for the Tyrosine Kinase Inhibitor Gefitinib. Adv Healthc Mater 2015; 4:2816-21. [PMID: 26592186 DOI: 10.1002/adhm.201500389] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/21/2015] [Indexed: 12/24/2022]
Abstract
Anticancer drug Gefitinib encapsulated within human heavy chain apoferritin by diffusion allows pH-controlled sustained release of cargo. The combination of increased cellular uptake, and potent and enhanced antitumor activity against the HER2 overexpressing SKBR3 cell line compared to Gefitinib alone, makes it a promising carrier for delivery of drugs to tumor sites.
Collapse
Affiliation(s)
- Anchala I. Kuruppu
- Centre for Biomolecular Sciences; School of Pharmacy; The University of Nottingham; Nottingham NG7 2RD UK
| | - Lei Zhang
- Centre for Biomolecular Sciences; School of Chemistry; The University of Nottingham; Nottingham NG7 2RD UK
| | - Hilary Collins
- Centre for Biomolecular Sciences; School of Pharmacy; The University of Nottingham; Nottingham NG7 2RD UK
| | - Lyudmila Turyanska
- School of Physics and Astronomy; The University of Nottingham; Nottingham NG7 2RD UK
| | - Neil R. Thomas
- Centre for Biomolecular Sciences; School of Chemistry; The University of Nottingham; Nottingham NG7 2RD UK
| | - Tracey D. Bradshaw
- Centre for Biomolecular Sciences; School of Pharmacy; The University of Nottingham; Nottingham NG7 2RD UK
| |
Collapse
|
417
|
Wang S, Meng Y, Li C, Qian M, Huang R. Receptor-Mediated Drug Delivery Systems Targeting to Glioma. NANOMATERIALS 2015; 6:nano6010003. [PMID: 28344260 PMCID: PMC5302535 DOI: 10.3390/nano6010003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/08/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022]
Abstract
Glioma has been considered to be the most frequent primary tumor within the central nervous system (CNS). The complexity of glioma, especially the existence of the blood-brain barrier (BBB), makes the survival and prognosis of glioma remain poor even after a standard treatment based on surgery, radiotherapy, and chemotherapy. This provides a rationale for the development of some novel therapeutic strategies. Among them, receptor-mediated drug delivery is a specific pattern taking advantage of differential expression of receptors between tumors and normal tissues. The strategy can actively transport drugs, such as small molecular drugs, gene medicines, and therapeutic proteins to glioma while minimizing adverse reactions. This review will summarize recent progress on receptor-mediated drug delivery systems targeting to glioma, and conclude the challenges and prospects of receptor-mediated glioma-targeted therapy for future applications.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China.
| | - Ying Meng
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China.
| | - Chengyi Li
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China.
| | - Min Qian
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China.
| | - Rongqin Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China.
| |
Collapse
|
418
|
Bogdan AR, Miyazawa M, Hashimoto K, Tsuji Y. Regulators of Iron Homeostasis: New Players in Metabolism, Cell Death, and Disease. Trends Biochem Sci 2015; 41:274-286. [PMID: 26725301 DOI: 10.1016/j.tibs.2015.11.012] [Citation(s) in RCA: 645] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/16/2015] [Accepted: 11/24/2015] [Indexed: 02/07/2023]
Abstract
Iron is necessary for life, but can also cause cell death. Accordingly, cells evolved a robust, tightly regulated suite of genes for maintaining iron homeostasis. Previous mechanistic studies on iron homeostasis have granted insight into the role of iron in human health and disease. We highlight new regulators of iron metabolism, including iron-trafficking proteins [solute carrier family 39, SLC39, also known as ZRT/IRT-like protein, ZIP; and poly-(rC)-binding protein, PCBP] and a cargo receptor (NCOA4) that is crucial for release of ferritin-bound iron. We also discuss emerging roles of iron in apoptosis and a novel iron-dependent cell death pathway termed 'ferroptosis', the dysregulation of iron metabolism in human pathologies, and the use of iron chelators in cancer therapy.
Collapse
Affiliation(s)
- Alexander R Bogdan
- Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC 27695-7633, USA
| | - Masaki Miyazawa
- Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC 27695-7633, USA
| | - Kazunori Hashimoto
- Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC 27695-7633, USA
| | - Yoshiaki Tsuji
- Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC 27695-7633, USA.
| |
Collapse
|
419
|
Wang CE, Stayton PS, Pun SH, Convertine AJ. Polymer nanostructures synthesized by controlled living polymerization for tumor-targeted drug delivery. J Control Release 2015; 219:345-354. [PMID: 26342661 PMCID: PMC4656053 DOI: 10.1016/j.jconrel.2015.08.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/27/2015] [Accepted: 08/27/2015] [Indexed: 12/21/2022]
Abstract
The development of drug delivery systems based on well-defined polymer nanostructures could lead to significant improvements in the treatment of cancer. The design of these therapeutic nanosystems must account for numerous systemic and circulation obstacles as well as the specific pathophysiology of the tumor. Nanoparticle size and surface charge must also be carefully selected in order to maintain long circulation times, allow tumor penetration, and avoid clearance by the reticuloendothelial system (RES). Targeting ligands such as vitamins, peptides, and antibodies can improve the accumulation of nanoparticle-based therapies in tumor tissue but must be optimized to allow for intratumoral penetration. In this review, we will highlight factors influencing the design of nanoparticle therapies as well as the development of modern controlled "living" polymerization techniques (e.g. ATRP, RAFT, ROMP) that are leading to the creation of sophisticated new polymer architectures with discrete spatially-defined functional modules. These innovative materials (e.g. star polymers, polymer brushes, macrocyclic polymers, and hyperbranched polymers) combine many of the desirable properties of traditional nanoparticle therapies while substantially reducing or eliminating the need for complex formulations.
Collapse
Affiliation(s)
- Christine E Wang
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Patrick S Stayton
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA.
| | - Anthony J Convertine
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
420
|
Szwed M, Wrona D, Kania KD, Koceva-Chyla A, Marczak A. Doxorubicin-transferrin conjugate triggers pro-oxidative disorders in solid tumor cells. Toxicol In Vitro 2015; 31:60-71. [PMID: 26607004 DOI: 10.1016/j.tiv.2015.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/03/2015] [Accepted: 11/16/2015] [Indexed: 12/26/2022]
Abstract
The formation of reactive oxygen species (ROS) is a widely accepted mechanism of doxorubicin (DOX) toxicity toward cancer cells. However, little is known about the potential of new systems, designed for more efficient and targeted doxorubicin delivery (i.e. protein conjugates, polymeric micelles, liposomes, monoclonal antibodies), to induce oxidative stress (OS) in tumors and hematological malignancies. Therefore, the objective of our study was to determine the relation between the toxicity of doxorubicin-transferring (DOX-TRF) conjugate and its capability to generate oxidative/nitrosative stress in solid tumor cells. Our research proves that DOX-TRF conjugate displays higher cytotoxicity towards lung adenocarcinoma epithelial (A549) and hepatocellular carcinoma (HepG2) cell lines than the reference free drug (DOX) and induces more extensive OS, characterized by a significant decrease in the total cellular antioxidant capacity, glutathione level and amount of -SH groups and an increase in hydroperoxide content. The intracellular redox imbalance was accompanied by changes in the transcription of genes encoding key antioxidant enzymes engaged in the sustaining of cellular redox homeostasis: superoxide dismutase (SOD), catalase (CAT), glutathione transferase (GST) and glutathione peroxidase (GP).
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Dominika Wrona
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Katarzyna D Kania
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, PAS, Lodowa St. 106, 93-232 Lodz, Poland.
| | - Aneta Koceva-Chyla
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Agnieszka Marczak
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
421
|
Babu KR, Muckenthaler MU. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J Mol Med (Berl) 2015; 94:347-59. [PMID: 26560875 PMCID: PMC4803811 DOI: 10.1007/s00109-015-1362-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 12/25/2022]
Abstract
Abstract Ferroportin (FPN) exports iron from duodenal enterocytes, macrophages, and hepatocytes to maintain systemic iron homeostasis. In addition, FPN is expressed in various cancer cells. Here, we show that in lung cancer, FPN expression is regulated by miR-20a. Within the FPN-3′-untranslated region (3′UTR), we identify and experimentally validate three evolutionarily conserved target sites for the microRNA (miRNA) members of the miR-17 seed family, including miR-20a. Our analysis of RNA sequencing data from patients with lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) revealed that FPN messenger RNA (mRNA) levels are significantly decreased in tumor compared to matched healthy tissue, while miR-20a levels are increased. A significant negative correlation of miR-20a and FPN expression was observed. Functional studies further demonstrate that FPN is post-transcriptionally regulated by miR-20a in non-small cell lung cancer (NSCLC) cells and that overexpression or knockdown of miR-20a or FPN affects NSCLC proliferation and colony formation. Taken together, our data suggest that increased expression of miR-20 in lung cancer may decrease iron export, leading to intracellular iron retention, which, in turn, favors cell proliferation. Key messages miR-20a controls expression of the iron exporter ferroportin (FPN) by binding to highly conserved target sites in its 3′UTR. Expression of miR-20a is inversely correlated to FPN in lung cancer. Low FPN expression stimulates proliferation and colony formation of non-small cell lung cancer (NSCLC) cells, possibly by increasing iron availability for cancer cell proliferation.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-015-1362-3) contains supplementary material, which is available to authorized users.
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
|
422
|
Shao M, Peng ZX, Shi CY, Tang R, Manzo LM, Liu Y. A convenient method for hTfR1 inclusion body purification. Prep Biochem Biotechnol 2015; 45:743-53. [PMID: 25207990 DOI: 10.1080/10826068.2014.952383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Human transferrin receptor, referred as hTfR1, is ubiquitously expressed at low levels in most normal human tissues; however, the expression level of hTfR1 at the blood-brain barrier (BBB) and in tumor tissues is relatively higher. hTfR1 is a type II homodimeric transmembrane protein. The extracellular domain of hTfR1 consists of three domains: helical domain, apical, and protease-like domain. In order to prepare hTfR1 antibody, which can be utilized to deliver drugs across BBB through receptor-mediated endocytosis, we began to express the nonligand binding domain of hTfR1 in Escherichia coli BL21 Transetta (DE3). The TfR1 gene was first obtained from HepG2 cells by reverse-transcription polymerase chain reaction (RT-PCR) and then inserted into pET 32a(c+) vector. The protein was expressed in the form of inclusion body with extremely high purity by the E. coli BL21 Transetta (DE3), and the purity was further improved by size-exclusion chromatography. The Western blot test indicated that the recombinant protein was TfR1 as expected. Above all, this report provided a convenient protocol that could be fulfilled in order to prepare hTfR1 inclusion body, which failed to be purified by an Ni(2+) affinity column.
Collapse
Affiliation(s)
- Ming Shao
- a State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University , Nanjing , China
| | | | | | | | | | | |
Collapse
|
423
|
Sriraman SK, Geraldo V, Luther E, Degterev A, Torchilin V. Cytotoxicity of PEGylated liposomes co-loaded with novel pro-apoptotic drug NCL-240 and the MEK inhibitor cobimetinib against colon carcinoma in vitro. J Control Release 2015; 220:160-168. [PMID: 26497930 DOI: 10.1016/j.jconrel.2015.10.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/08/2015] [Accepted: 10/19/2015] [Indexed: 01/21/2023]
Abstract
The overactivation of signaling pathways, such as the PI3K and MAPK, which are crucial to cell growth and survival, is a common feature in many cancer types. Though a number of advances have been made in the development of molecular agents targeting these pathways, their application as monotherapies has not significantly improved clinical outcome. A novel liposomal preparation was developed, co-loaded with NCL-240, a small-molecule inhibitor of the PI3K/mTOR pathway, along with cobimetinib, a MEK/ERK pathway inhibitor. This combination drug-loaded nanocarrier, (N+C)-LP, was able to significantly enhance the cytotoxicity of these drugs against colon carcinoma cells in vitro demonstrating a clear synergistic effect (combination index of 0.79). The (N+C)-LP was also able to induce cell cycle arrest of the cells, specifically in the G1 phase thereby preventing their progression to the S-phase, typical of the action of MEK inhibitors. Analyzing the apoptotic events, it was found that this effect on cell cycle regulation is followed by the induction of apoptosis. The quantified distribution of apoptotic events showed that the (N+C)-LP induced apoptosis significantly by over 3-4 fold (P<0.001) compared to other treatment groups. The co-loaded liposomal preparation was also targeted to the transferrin receptor of cancer cells by modifying the surface of the liposome with transferrin. FACS analysis showed that transferrin-mediated targeting enhanced the association of liposomes to HCT 116 cells by almost 5-fold. This could potentially allow for cancer cell-specific effects in vivo thereby minimizing any non-specific interactions of the liposomes with non-cancerous cells. Taken together, this study clearly shows that the combined inhibition of the PI3K and MEK pathways correlates with a significant anti-proliferative effect, due to cell-cycle regulation leading to the induction of apoptosis.
Collapse
Affiliation(s)
- Shravan Kumar Sriraman
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Vananelia Geraldo
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil
| | - Ed Luther
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Alexei Degterev
- Department of Biochemistry, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Vladimir Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
424
|
Preclinical evaluation of ⁸⁹Zr-labeled human antitransferrin receptor monoclonal antibody as a PET probe using a pancreatic cancer mouse model. Nucl Med Commun 2015; 36:286-94. [PMID: 25460304 DOI: 10.1097/mnm.0000000000000245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Pancreatic cancer is aggressive and its prognosis remains poor; thus, effective therapy is urgently needed. Transferrin receptor (TfR) is highly expressed in pancreatic cancer and is considered to be a good candidate for molecular-targeted therapy. We radiolabeled and evaluated fully human anti-TfR monoclonal antibodies as a new PET probe for evaluating the biodistribution of the anti-TfR antibody in pancreatic cancer. MATERIALS AND METHODS TfR expression was evaluated in four human pancreatic cancer (MIAPaCa-2, PANC-1, BxPC-3, and AsPC-1) and murine A4 cell lines. The binding of 125I-labeled anti-TfR antibodies (TSP-A01, TSP-A02, TSP-A03, and TSP-A04) to MIAPaCa-2 cells was compared. 125I-labeled, 67Ga-labeled, and 89Zr-labeled TSP-A01 were evaluated by cell binding, competitive inhibition, and internalization assays. Biodistribution studies of 125I-labeled and 89Zr-labeled TSP-A01 were conducted in mice bearing MIAPaCa-2 and A4 tumors. PET imaging with [89Zr]TSP-A01 was carried out. RESULTS MIAPaCa-2 cells showed the highest TfR expression in vitro and in vivo, whereas A4 cells showed no expression. Of the four antibodies, [125I]TSP-A01 showed the highest binding to MIAPaCa-2 cells, but not to A4 cells. The dissociation constant of TSP-A01 was 0.29 nmol/l. Uptake of radiolabeled TSP-A01, especially [89Zr]TSP-A01, was significantly higher in MIAPaCa-2 tumors than in A4 tumors. PET with [89Zr]TSP-A01 clearly visualized MIAPaCa-2 xenografts but not A4 xenografts. CONCLUSION [89Zr]TSP-A01 is a promising PET probe for evaluating the accumulation of anti-TfR antibody in pancreatic cancer and has the potential to facilitate the selection of appropriate patients who would benefit from anti-TfR antibody therapy.
Collapse
|
425
|
Szwed M, Kania KD, Jozwiak Z. Toxicity of doxorubicin-transferrin conjugate is connected to the modulation of Wnt/β-catenin pathway in human leukemia cells. Leuk Res 2015; 39:1096-102. [DOI: 10.1016/j.leukres.2015.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/26/2015] [Accepted: 07/09/2015] [Indexed: 01/17/2023]
|
426
|
Shen S, Xia JX, Wang J. Nanomedicine-mediated cancer stem cell therapy. Biomaterials 2015; 74:1-18. [PMID: 26433488 DOI: 10.1016/j.biomaterials.2015.09.037] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/19/2022]
Abstract
Circumstantial evidence suggests that most tumours are heterogeneous and contain a small population of cancer stem cells (CSCs) that exhibit distinctive self-renewal, proliferation and differentiation capabilities, which are believed to play a crucial role in tumour progression, drug resistance, recurrence and metastasis in multiple malignancies. Given that the existence of CSCs is a primary obstacle to cancer therapy, a tremendous amount of effort has been put into the development of anti-CSC strategies, and several potential approaches to kill therapeutically-resistant CSCs have been explored, including inhibiting ATP-binding cassette transporters, blocking essential signalling pathways involved in self-renewal and survival of CSCs, targeting CSCs surface markers and destroying the tumour microenvironment. Meanwhile, an increasing number of therapeutic agents (e.g. small molecule drugs, nucleic acids and antibodies) to selectively target CSCs have been screened or proposed in recent years. Drug delivery technology-based approaches hold great potential for tackling the limitations impeding clinical applications of CSC-specific agents, such as poor water solubility, short circulation time and inconsistent stability. Properly designed nanocarrier-based therapeutic agents (or nanomedicines) offer new possibilities of penetrating CSC niches and significantly increasing therapeutic drug accumulation in CSCs, which are difficult for free drug counterparts. In addition, intelligent nanomedicine holds great promise to overcome pump-mediated multidrug resistance which is driven by ATP and to decrease detrimental effects on normal somatic stem cells. In this review, we summarise the distinctive biological processes related to CSCs to highlight strategies against inherently drug-resistant CSCs. We then focus on some representative examples that give a glimpse into state-of-the-art nanomedicine approaches developed for CSCs elimination. A perspective on innovative therapeutic strategies and the potential direction of nanomedicine-based CSC therapy in the near future is also presented.
Collapse
Affiliation(s)
- Song Shen
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui 230027, PR China
| | - Jin-Xing Xia
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui 230027, PR China.
| | - Jun Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui 230027, PR China; Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui 230027, PR China; High Magnetic Field Laboratory of CAS, University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| |
Collapse
|
427
|
Moody PR, Sayers EJ, Magnusson JP, Alexander C, Borri P, Watson P, Jones AT. Receptor Crosslinking: A General Method to Trigger Internalization and Lysosomal Targeting of Therapeutic Receptor:Ligand Complexes. Mol Ther 2015; 23:1888-98. [PMID: 26412588 PMCID: PMC4700114 DOI: 10.1038/mt.2015.178] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023] Open
Abstract
A major unmet clinical need is a universal method for subcellular targeting of bioactive molecules to lysosomes. Delivery to this organelle enables either degradation of oncogenic receptors that are overexpressed in cancers, or release of prodrugs from antibody–drug conjugates. Here, we describe a general method that uses receptor crosslinking to trigger endocytosis and subsequently redirect trafficking of receptor:cargo complexes from their expected route, to lysosomes. By incubation of plasma membrane receptors with biotinylated cargo and subsequent addition of streptavidin to crosslink receptor:cargo–biotin complexes, we achieved rapid and selective lysosomal targeting of transferrin, an anti-MHC class I antibody, and the clinically approved anti-Her2 antibody trastuzumab. These three protein ligands each target a receptor with a distinct cellular function and intracellular trafficking profile. Importantly, we confirmed that crosslinking of trastuzumab increased lysosomal degradation of its cognate oncogenic receptor Her2 in breast cancer cell lines SKBR3 and BT474. These data suggest that crosslinking could be exploited for a wide range of target receptors, for navigating therapeutics through the endolysosomal pathway, for significant therapeutic benefit.
Collapse
Affiliation(s)
- Paul R Moody
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales
| | - Edward J Sayers
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales
| | | | | | - Paola Borri
- School of Biosciences, Cardiff University, Cardiff, Wales
| | - Peter Watson
- School of Biosciences, Cardiff University, Cardiff, Wales
| | - Arwyn T Jones
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales
| |
Collapse
|
428
|
Nogueira E, Gomes AC, Preto A, Cavaco-Paulo A. Design of liposomal formulations for cell targeting. Colloids Surf B Biointerfaces 2015; 136:514-26. [PMID: 26454541 DOI: 10.1016/j.colsurfb.2015.09.034] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/16/2015] [Accepted: 09/18/2015] [Indexed: 01/04/2023]
Abstract
Liposomes have gained extensive attention as carriers for a wide range of drugs due to being both nontoxic and biodegradable as they are composed of substances naturally occurring in biological membranes. Active targeting for cells has explored specific modification of the liposome surface by functionalizing it with specific targeting ligands in order to increase accumulation and intracellular uptake into target cells. None of the Food and Drug Administration-licensed liposomes or lipid nanoparticles are coated with ligands or target moieties to delivery for homing drugs to target tissues, cells or subcellular organelles. Targeted therapies (with or without controlled drug release) are an emerging and relevant research area. Despite of the numerous liposomes reviews published in the last decades, this area is in constant development. Updates urgently needed to integrate new advances in targeted liposomes research. This review highlights the evolution of liposomes from passive to active targeting and challenges in the development of targeted liposomes for specific therapies.
Collapse
Affiliation(s)
- Eugénia Nogueira
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; CEB-Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Andreia C Gomes
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Ana Preto
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Artur Cavaco-Paulo
- CEB-Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
429
|
Zhang C, Qu X, Li J, Hong H, Li J, Ren J, Payne GF, Liu C. Biofabricated nanoparticle coating for liver-cell targeting. Adv Healthc Mater 2015; 4:1972-81. [PMID: 26138108 DOI: 10.1002/adhm.201500202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/04/2015] [Indexed: 11/08/2022]
Abstract
Biology routinely uses noncovalent interactions to perform complex functions that range from the molecular recognition of ligand-receptor binding to the reversible self-assembly/disassembly of hierarchical nanostructures (e.g., virus particles). Potentially, biological materials that offer such recognition and reversible self-assembly functionality can be applied to nanomedicine. Here, polysaccharides with the multifunctional polysaccharide-binding protein Concanavalin A (Con A) are coupled to create a functional nanoparticle coating. This coating is self-assembled in a layer-by-layer format by sequentially contacting a nanoparticle with Con A and the polysaccharide glycogen. In the final assembly step, a galactomannan targeting ligand is self-assembled into the coating. Evidence indicates that the mannose residues of the galactomannan backbone are responsible for assembly into the coating by Con A binding, while the galactose side chain residues are responsible for targeting to the liver-specific asialoglycoprotein receptor (ASGP-R). Binding to ASGP-R induces endocytic uptake, while the low endosomal pH triggers disassembly of the coating and release of the nanoparticle-entrapped drug. In vitro cell studies indicate that the coating confers liver-cell-specific function for both nanoparticle uptake and drug delivery. These studies extend the use of Con A to sugar-mediated and organ-specific targeting, and further illustrate the potential of biologically based fabrication for generating functional materials.
Collapse
Affiliation(s)
- Cheng Zhang
- The State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai 200237 China
- Key Laboratory for Ultrafine Materials of Ministry of Education; East China University of Science and Technology; Shanghai 200237 China
| | - Xue Qu
- The State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai 200237 China
- Key Laboratory for Ultrafine Materials of Ministry of Education; East China University of Science and Technology; Shanghai 200237 China
| | - Jinyang Li
- The State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai 200237 China
- Key Laboratory for Ultrafine Materials of Ministry of Education; East China University of Science and Technology; Shanghai 200237 China
| | - Hua Hong
- The State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai 200237 China
- Key Laboratory for Ultrafine Materials of Ministry of Education; East China University of Science and Technology; Shanghai 200237 China
| | - Jianbo Li
- Institute of Nano and Bio-polymeric Materials; School of Materials Science and Engineering; Tongji University; 4800 Caoan Road Shanghai 201804 China
| | - Jie Ren
- Institute of Nano and Bio-polymeric Materials; School of Materials Science and Engineering; Tongji University; 4800 Caoan Road Shanghai 201804 China
| | - Gregory F. Payne
- Institute for Biosystems and Biotechnology Research and Fischell Department of Engineering; 5115 Plant Sciences Building College Park MD 20742 USA
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai 200237 China
- Key Laboratory for Ultrafine Materials of Ministry of Education; East China University of Science and Technology; Shanghai 200237 China
| |
Collapse
|
430
|
Patil R, Koronyo Y, Ljubimov AV, Salumbides B, Mamelak A, Gangalum PR, Ding H, Portilla-Arias J, Holler E, Butte P, Koronyo-Hamaoui M, Ljubimova JY, Black KL. Advances in Imaging: Brain Tumors to Alzheimer's Disease. THE BANGKOK MEDICAL JOURNAL 2015; 10:83-97. [PMID: 29142857 PMCID: PMC5683733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Professor Black and colleagues have been working to improve the quality and sensitivity of imaging in the early detection of conditions from brain tumors to Alzheimer's disease to enhance treatment protocols and patient management. Professor Black et al introduced nanoparticles to improve MRI imaging. These nanoparticles consist of poly (b-L- malic acid (PMLA)) conjugates with monoclonal antibodies ((mAbs)) and Gd-DOTA. These are known as MRI nano-imaging agents (NIA). Most importantly, they can penetrate the endothelial blood-brain barrier (BBB) to reach brain tumors (primary or metastasis). This is effective in cases of brain tumors or breast cancer or other cancers such as lung cancer and gastric cancer having HER2 and/or EGFR positive crossing BBB. By the covalent conjugation of MR contrast (NIA), the MRI virtual biopsy can differentiate brain tumors from infections or other brain pathological conditions. The brain's intrinsic natural fluorescence such as NADH, FAD, lipopigments and porphyrin in the brain tissue can be identified by using time resolved fluorescence spectroscopy (TRFS) which is operated through the use of ultra-short laser. TRFS produces various color bands to differentiate the tumor from normal brain tissue in real time and registers the data on a 3D map. This is significant, as this will provide a greatly improved assessment methodology of tissue type. Consequently, this will potentially result in shorter operation times as well as more satisfactory tumor removal. In the detection of Alzheimer disease, amyloid plaque is deposited in retina tissue (including the RGC, RNFL and inner plexiform layer) which can produce a fluorescence effect by using curcumin as a contrast. This is then shown by human retina amyloid imaging device. Immunotherapies with glatiramer acetate (GA) have been shown to reduce amyloid deposits in brain and retinal AB deposits in mice. The study of advanced imaging technology and techniques including NIA, TRFS and the detection of amyloid plaque in Alzheimer disease are very important approaches to create a new era for diagnostic and therapeutic management of brain tumors and other cancers (HER2 and/or EGFR positive). This pioneering work by Professor Black, and colleagues, gives rise to a new hope for cancer patients for targeted therapy and for immunotherapies in Alzheimer's disease.
Collapse
Affiliation(s)
- Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alexander V. Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Brenda Salumbides
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Adam Mamelak
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Pallavi R. Gangalum
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hui Ding
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jose Portilla-Arias
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Eggehard Holler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Pramod Butte
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Julia Y. Ljubimova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
431
|
Levina A, McLeod AI, Gasparini SJ, Nguyen A, De Silva WGM, Aitken JB, Harris HH, Glover C, Johannessen B, Lay PA. Reactivity and Speciation of Anti-Diabetic Vanadium Complexes in Whole Blood and Its Components: The Important Role of Red Blood Cells. Inorg Chem 2015; 54:7753-66. [PMID: 26230577 DOI: 10.1021/acs.inorgchem.5b00665] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Reactions with blood components are crucial for controlling the antidiabetic, anticancer, and other biological activities of V(V) and V(IV) complexes. Despite extensive studies of V(V) and V(IV) reactions with the major blood proteins (albumin and transferrin), reactions with whole blood and red blood cells (RBC) have been studied rarely. A detailed speciation study of Na3[V(V)O4] (A), K4[V(IV)2O2(citr)2]·6H2O (B; citr = citrato(4-)); [V(IV)O(ma)2] (C; ma = maltolato(-)), and (NH4)[V(V)(O)2(dipic)] (D; dipic = pyridine-2,6-dicarboxylato(2-)) in whole rat blood, freshly isolated rat plasma, and commercial bovine serum using X-ray absorption near-edge structure (XANES) spectroscopy is reported. The latter two compounds are potential oral antidiabetic drugs, and the former two are likely to represent their typical decomposition products in gastrointestinal media. XANES spectral speciation was performed by principal component analysis and multiple linear regression techniques, and the distribution of V between RBC and plasma fractions was measured by electrothermal atomic absorption spectroscopy. Reactions of A, C, or D with whole blood (1.0 mM V, 1-6 h at 310 K) led to accumulation of ∼50% of total V in the RBC fraction (∼10% in the case of B), which indicated that RBC act as V carriers to peripheral organs. The spectra of V products in RBC were independent of the initial V complex, and were best fitted by a combination of V(IV)-carbohydrate (2-hydroxyacid moieties) and/or citrate (65-85%) and V(V)-protein (15-35%) models. The presence of RBC created a more reducing environment in the plasma fraction of whole blood compared with those in isolated plasma or serum, as shown by the differences in distribution of V(IV) and V(V) species in the reaction products of A-D in these media. At physiologically relevant V concentrations (<50 μM), this role of RBC may promote the formation of V(III)-transferrin as a major V carrier in the blood plasma. The results reported herein have broad implications for the roles of RBC in the transport and speciation of metal pro-drugs that have broad applications across medicine.
Collapse
Affiliation(s)
- Aviva Levina
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Andrew I McLeod
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Sylvia J Gasparini
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Annie Nguyen
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | | | - Jade B Aitken
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia.,‡Australian Synchrotron, 800 Blackburn Rd., Clayton VIC 3168, Australia
| | - Hugh H Harris
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Chris Glover
- ‡Australian Synchrotron, 800 Blackburn Rd., Clayton VIC 3168, Australia
| | - Bernt Johannessen
- ‡Australian Synchrotron, 800 Blackburn Rd., Clayton VIC 3168, Australia
| | - Peter A Lay
- †School of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| |
Collapse
|
432
|
Leoh LS, Daniels-Wells TR, Martínez-Maza O, Penichet ML. Insights into the effector functions of human IgG3 in the context of an antibody targeting transferrin receptor 1. Mol Immunol 2015; 67:407-15. [PMID: 26232328 DOI: 10.1016/j.molimm.2015.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 06/19/2015] [Accepted: 07/01/2015] [Indexed: 12/29/2022]
Abstract
The transferrin receptor 1 (TfR1) is involved in cellular iron uptake and regulation of cell proliferation. The increased expression of TfR1 observed in malignant cells, compared to normal cells, together with its extracellular accessibility, make this receptor an attractive target for antibody-mediated cancer therapy. We have developed a mouse/human chimeric IgG3 specific for human TfR1 (ch128.1), which shows anti-tumor activity against certain malignant B cells in vitro through TfR1 degradation and iron deprivation, and in vivo through a mechanism yet to be defined. To further explore potential mechanisms of action of ch128.1, we examined its ability to induce antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-mediated cytotoxicity (CDC). We now report that ch128.1 is capable of mediating ADCC and CDC against malignant B cells, which is consistent with its ability to bind FcγRI, FcγRIIIa, and the complement component C1q. To delineate the residues involved in these effector functions, we developed a panel of three constructs with mutations in the lower hinge region and CH2 domain: 1) L234A/L235A, 2) P331S, and 3) L234A/L235A/P331S. The triple mutant consistently displayed a significant reduction in ADCC, while the L234A/L235A mutant exhibited less reduction in ADCC, and the P331S mutant did not show reduced ADCC. However, all three mutants exhibited impaired binding to FcγRI and FcγRIIIa. These results suggest that all three residues contribute to ADCC, although to different degrees. The P331S mutant showed drastically decreased C1q binding and abolished CDC, confirming the critical role of this residue in complement activation, while the other residues play a less important role in CDC. Our study provides insights into the effector functions of human IgG3 in the context of an antibody targeting TfR1.
Collapse
Affiliation(s)
- Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Otoniel Martínez-Maza
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA; UCLA AIDS Institute, Los Angeles, CA, USA
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA; UCLA AIDS Institute, Los Angeles, CA, USA; The Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
433
|
Abstract
Theranostics is a promising field that combines therapeutics and diagnostics into single multifunctional formulations. This field is driven by advancements in nanoparticle systems capable of providing the necessary functionalities. By utilizing these powerful nanomedicines, the concept of personalized medicine can be realized by tailoring treatment strategies to the individual. This review gives a brief overview of the components of a theranostic system and the challenges that designing truly multifunctional nanoparticles present. Considerations when choosing a class of nanoparticle include the size, shape, charge, and surface chemistry, while classes of nanoparticles discussed are polymers, liposomes, dendrimers, and polymeric micelles. Targeting to disease states can be achieved either through passive or active targeting which uses specific ligands to target receptors that are overexpressed in tumors and common targeting elements are presented. To image the interactions with disease states, contrast agents are included in the nanoparticle formulation. Imaging options include optical imaging techniques, computed tomography, nuclear based, and magnetic resonance imaging. The interplay between all of these components needs to be carefully considered when designing a theranostic system.
Collapse
|
434
|
Sun L, Wu Q, Peng F, Liu L, Gong C. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surf B Biointerfaces 2015; 135:56-72. [PMID: 26241917 DOI: 10.1016/j.colsurfb.2015.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
In order to achieve long circulation time and high drug accumulation in the tumor sites via the EPR effects, anticancer drugs have to be protected by non-fouling polymers such as poly(ethylene glycol) (PEG), poly(ethylene oxide) (PEO), dextran, and poly(acrylic acid) (PAA). However, the dense layer of stealth polymer also prohibits efficient uptake of anticancer drugs by target cancer cells. For cancer therapy, it is often more desirable to accomplish rapid cellular uptake after anticancer drugs arriving at the pathological site, which could on one hand maximize the therapeutic efficacy and on the other hand reduce probability of drug resistance in cells. In this review, special attention will be focused on the recent potential strategies that can enable drug-loaded polymeric nanoparticles to rapidly recognize cancer cells, leading to enhanced internalization.
Collapse
Affiliation(s)
- Lu Sun
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qinjie Wu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Feng Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
435
|
Pourcelot E, Lénon M, Mobilia N, Cahn JY, Arnaud J, Fanchon E, Moulis JM, Mossuz P. Iron for proliferation of cell lines and hematopoietic progenitors: Nailing down the intracellular functional iron concentration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1596-605. [DOI: 10.1016/j.bbamcr.2015.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/25/2015] [Accepted: 03/19/2015] [Indexed: 12/29/2022]
|
436
|
Friberg S, Nyström AM. Nanotechnology in the war against cancer: new arms against an old enemy – a clinical view. Future Oncol 2015; 11:1961-75. [DOI: 10.2217/fon.15.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ABSTRACT Clinical oncology is facing a paradigm shift. A new treatment philosophy is emerging and new targets are appearing that require new active agents. The medical use of nanotechnology – nanomedicine – holds several promising possibilities in the war against cancer. Some of these include: new formats for old drugs, that is, increasing efficacy while diminishing side effects; and new administration routes – that is, dermal, oral and pulmonary. In this overview, we describe some nanoparticles and their medical uses as well as highlight advantages of nanoparticles compared with conventional pharmaceuticals. We also point to some of the many technical challenges and potential risks with using nanotechnology for oncological applications.
Collapse
Affiliation(s)
- Sten Friberg
- Swedish Medical Nanoscience Center, Department of Neuroscience, Retzius väg 8, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Andreas M Nyström
- Institute of Environmental Medicine, Nobels väg 13, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
437
|
Saçlıgil D, Şenel S, Yavuz H, Denizli A. Purification of transferrin by magnetic immunoaffinity beads. J Sep Sci 2015; 38:2729-36. [PMID: 25980364 DOI: 10.1002/jssc.201500216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 12/26/2022]
Abstract
Immunoaffinity adsorbent for transferrin (Tf) purification was prepared by immobilizing anti-transferrin (Anti-Tf) antibody on magnetic monosizepoly(glycidyl methacrylate) beads, which were synthesized by dispersion polymerization technique in the presence of Fe3 O4 nanopowder and obtained with an average size of 2.0 μm. The magnetic poly(glycidyl methacrylate) (mPGMA) beads were characterized by Fourier transform infrared spectroscopy, swelling tests, scanning electron microscopy, electron spin resonance spectroscopy, thermogravimetric analysis and zeta sizing analysis. The density and swelling ratio of the beads were 1.08 g/cm(3) and 52%, respectively. Anti-Tf molecules were covalently coupled through epoxy groups of mPGMA. Optimum binding of anti-Tf was 2.0 mg/g. Optimum Tf binding from aqueous Tf solutions was determined as 1.65 mg/g at pH 6.0 and initial Tf concentration of 1.0 mg/mL. There was no remarkable loss in the Tf adsorption capacity of immunoaffinity beads after five adsorption-desorption cycles. Tf adsorption from artificial plasma was also investigated and the purity of the Tf molecules was shown with gel electrophoresis studies.
Collapse
Affiliation(s)
- Dilara Saçlıgil
- Hacettepe University, Department of Chemistry, Beytepe, Ankara, Turkey
| | - Serap Şenel
- Hacettepe University, Department of Chemistry, Beytepe, Ankara, Turkey
| | - Handan Yavuz
- Hacettepe University, Department of Chemistry, Beytepe, Ankara, Turkey
| | - Adil Denizli
- Hacettepe University, Department of Chemistry, Beytepe, Ankara, Turkey
| |
Collapse
|
438
|
Legendre C, Garcion E. Iron metabolism: a double-edged sword in the resistance of glioblastoma to therapies. Trends Endocrinol Metab 2015; 26:322-31. [PMID: 25936466 DOI: 10.1016/j.tem.2015.03.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM), the deadliest primary tumor of the central nervous system (CNS), is a clear illustration of the resistance of cancer cells to conventional therapies. Application of combinatorial strategies able to overcome pivotal factors of GBM resistance, particularly within the resection margins, represents an essential issue. This review focuses on the role of iron metabolism in GBM progression and resistance to therapy, and the impact of its pharmaceutical modulation on the disease. Iron, through its involvement in many biological processes, is a key factor in the control of cell behavior and cancer biology. Therefore, targeting cellular iron signaling or taking advantage of its dysregulation in cancer cells may lead to new opportunities for improving treatments and drug delivery in GBM.
Collapse
Affiliation(s)
- Claire Legendre
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1066, Bio-Inspired Micro and Nanomedicines (MINT), Angers, France; L'Université Nantes Angers Le Mans (LUNAM), Université d'Angers, Angers, France
| | - Emmanuel Garcion
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1066, Bio-Inspired Micro and Nanomedicines (MINT), Angers, France; L'Université Nantes Angers Le Mans (LUNAM), Université d'Angers, Angers, France.
| |
Collapse
|
439
|
Schneider LS, von Schwarzenberg K, Lehr T, Ulrich M, Kubisch-Dohmen R, Liebl J, Trauner D, Menche D, Vollmar AM. Vacuolar-ATPase Inhibition Blocks Iron Metabolism to Mediate Therapeutic Effects in Breast Cancer. Cancer Res 2015; 75:2863-74. [DOI: 10.1158/0008-5472.can-14-2097] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
|
440
|
Patil R, Ljubimov AV, Gangalum PR, Ding H, Portilla-Arias J, Wagner S, Inoue S, Konda B, Rekechenetskiy A, Chesnokova A, Markman JL, Ljubimov VA, Li D, Prasad RS, Black KL, Holler E, Ljubimova JY. MRI virtual biopsy and treatment of brain metastatic tumors with targeted nanobioconjugates: nanoclinic in the brain. ACS NANO 2015; 9:5594-608. [PMID: 25906400 PMCID: PMC4768903 DOI: 10.1021/acsnano.5b01872] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Differential diagnosis of brain magnetic resonance imaging (MRI) enhancement(s) remains a significant problem, which may be difficult to resolve without biopsy, which can be often dangerous or even impossible. Such MRI enhancement(s) can result from metastasis of primary tumors such as lung or breast, radiation necrosis, infections, or a new primary brain tumor (glioma, meningioma). Neurological symptoms are often the same on initial presentation. To develop a more precise noninvasive MRI diagnostic method, we have engineered a new class of poly(β-l-malic acid) polymeric nanoimaging agents (NIAs). The NIAs carrying attached MRI tracer are able to pass through the blood-brain barrier (BBB) and specifically target cancer cells for efficient imaging. A qualitative/quantitative "MRI virtual biopsy" method is based on a nanoconjugate carrying MRI contrast agent gadolinium-DOTA and antibodies recognizing tumor-specific markers and extravasating through the BBB. In newly developed double tumor xenogeneic mouse models of brain metastasis this noninvasive method allowed differential diagnosis of HER2- and EGFR-expressing brain tumors. After MRI diagnosis, breast and lung cancer brain metastases were successfully treated with similar tumor-targeted nanoconjugates carrying molecular inhibitors of EGFR or HER2 instead of imaging contrast agent. The treatment resulted in a significant increase in animal survival and markedly reduced immunostaining for several cancer stem cell markers. Novel NIAs could be useful for brain diagnostic MRI in the clinic without currently performed brain biopsies. This technology shows promise for differential MRI diagnosis and treatment of brain metastases and other pathologies when biopsies are difficult to perform.
Collapse
Affiliation(s)
- Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Alexander V. Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Arrogene Inc., Los Angeles, California, United States
| | - Pallavi R. Gangalum
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Hui Ding
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jose Portilla-Arias
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Shawn Wagner
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Satoshi Inoue
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Bindu Konda
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Arthur Rekechenetskiy
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Alexandra Chesnokova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Janet L. Markman
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Vladimir A. Ljubimov
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Debiao Li
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Ravi S. Prasad
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Arrogene Inc., Los Angeles, California, United States
| | - Eggehard Holler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Arrogene Inc., Los Angeles, California, United States
| | - Julia Y. Ljubimova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Arrogene Inc., Los Angeles, California, United States
- Address correspondence to
| |
Collapse
|
441
|
Transferrin: Endocytosis and Cell Signaling in Parasitic Protozoa. BIOMED RESEARCH INTERNATIONAL 2015; 2015:641392. [PMID: 26090431 PMCID: PMC4450279 DOI: 10.1155/2015/641392] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022]
Abstract
Iron is the fourth most abundant element on Earth and the most abundant metal in the human body. This element is crucial for life because almost all organisms need iron for several biological activities. This is the case with pathogenic organisms, which are at the vanguard in the battle with the human host for iron. The latest regulates Fe concentration through several iron-containing proteins, such as transferrin. The transferrin receptor transports iron to each cell that needs it and maintains it away from pathogens. Parasites have developed several strategies to obtain iron as the expression of specific transferrin receptors localized on plasma membrane, internalized through endocytosis. Signal transduction pathways related to the activation of the receptor have functional importance in proliferation. The study of transferrin receptors and other proteins with action in the signaling networks is important because these proteins could be used as therapeutic targets due to their specificity or to differences with the human counterpart. In this work, we describe proteins that participate in signal transduction processes, especially those that involve transferrin endocytosis, and we compare these processes with those found in T. brucei, T. cruzi, Leishmania spp., and E. histolytica parasites.
Collapse
|
442
|
Fernandes E, Ferreira JA, Andreia P, Luís L, Barroso S, Sarmento B, Santos LL. New trends in guided nanotherapies for digestive cancers: A systematic review. J Control Release 2015; 209:288-307. [PMID: 25957905 DOI: 10.1016/j.jconrel.2015.05.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
Digestive tract tumors are among the most common and deadliest malignancies worldwide, mainly due to late diagnosis and lack of efficient therapeutics. Current treatments essentially rely on surgery associated with (neo)adjuvant chemotherapy agents. Despite an upfront response, conventional drugs often fail to eliminate highly aggressive clones endowed with chemoresistant properties, which are responsible for tumor recurrence and disease dissemination. Synthetic drugs also present severe adverse systemic effects, hampering the administration of biologically effective dosages. Nanoencapsulation of chemotherapeutic agents within biocompatible polymeric or lipid matrices holds great potential to improve the pharmacokinetics and efficacy of conventional chemotherapy while reducing systemic toxicity. Tagging nanoparticle surfaces with specific ligands for cancer cells, namely monoclonal antibodies or antibody fragments, has provided means to target more aggressive clones, further improving the selectivity and efficacy of nanodelivery vehicles. In fact, over the past twenty years, significant research has translated into a wide array of guided nanoparticles, providing the molecular background for a new generation of intelligent and more effective anti-cancer agents. Attempting to bring awareness among the medical community to emerging targeted nanopharmaceuticals and foster advances in the field, we have conducted a systematic review about this matter. Emphasis was set on ongoing preclinical and clinical trials for liver, colorectal, gastric and pancreatic cancers. To the best of our knowledge this is the first systematic and integrated overview on this field. Using a specific query, 433 abstracts were gathered and narrowed to 47 manuscripts when matched against inclusion/exclusion criteria. All studies showed that active targeting improves the effectiveness of the nanodrugs alone, while lowering its side effects. The main focus has been on hepatocarcinomas, mainly by exploring glycans as homing molecules. Other ligands such as peptides/small proteins and antibodies/antibody fragments, with affinity to either tumor vasculature or tumor cells, have also been widely and successfully applied to guide nanodrugs to gastrointestinal carcinomas. Conversely, few solutions have been presented for pancreatic tumors. To this date only three nanocomplexes have progressed beyond pre-clinical stages: i) PK2, a galactosamine-functionalized polymeric-DOX formulation for hepatocarcinomas; ii) MCC-465, an anti-(myosin heavy chain a) immunoliposome for advanced stage metastatic solid tumors; and iii) MBP-426, a transferrin-liposome-oxaliplatin conjugate, also for advanced stage tumors. Still, none has been approved for clinical use. However, based on the high amount of pre-clinical studies showing enthusiastic results, the number of clinical trials is expected to increase in the near future. A more profound understanding about the molecular nature of chemoresistant clones and cancer stem cell biology will also contribute to boost the field of guided nanopharmacology towards more effective solutions.
Collapse
Affiliation(s)
- Elisabete Fernandes
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| | - Peixoto Andreia
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Lima Luís
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Nucleo de Investigação em Farmácia - Centro de Investigação em Saúde e Ambiente (CISA), Health School of the Polytechnic Institute of Porto, Porto, Portugal
| | - Sérgio Barroso
- Serviço de Oncologia, Hospital de Évora, Évora, Portugal
| | - Bruno Sarmento
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Health School of University of Fernando Pessoa, Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| |
Collapse
|
443
|
Bazak R, Houri M, Achy SE, Kamel S, Refaat T. Cancer active targeting by nanoparticles: a comprehensive review of literature. J Cancer Res Clin Oncol 2015; 141:769-84. [PMID: 25005786 PMCID: PMC4710367 DOI: 10.1007/s00432-014-1767-3] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022]
Abstract
PURPOSE Cancer is one of the leading causes of death, and thus, the scientific community has but great efforts to improve cancer management. Among the major challenges in cancer management is development of agents that can be used for early diagnosis and effective therapy. Conventional cancer management frequently lacks accurate tools for detection of early tumors and has an associated risk of serious side effects of chemotherapeutics. The need to optimize therapeutic ratio as the difference with which a treatment affects cancer cells versus healthy tissues lead to idea that it is needful to have a treatment that could act a the "magic bullet"-recognize cancer cells only. Nanoparticle platforms offer a variety of potentially efficient solutions for development of targeted agents that can be exploited for cancer diagnosis and treatment. There are two ways by which targeting of nanoparticles can be achieved, namely passive and active targeting. Passive targeting allows for the efficient localization of nanoparticles within the tumor microenvironment. Active targeting facilitates the active uptake of nanoparticles by the tumor cells themselves. METHODS Relevant English electronic databases and scientifically published original articles and reviews were systematically searched for the purpose of this review. RESULTS In this report, we present a comprehensive review of literatures focusing on the active targeting of nanoparticles to cancer cells, including antibody and antibody fragment-based targeting, antigen-based targeting, aptamer-based targeting, as well as ligand-based targeting. CONCLUSION To date, the optimum targeting strategy has not yet been announced, each has its own advantages and disadvantages even though a number of them have found their way for clinical application. Perhaps, a combination of strategies can be employed to improve the precision of drug delivery, paving the way for a more effective personalized therapy.
Collapse
Affiliation(s)
- Remon Bazak
- Department of Otorhinolaryngology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamad Houri
- Department of Ophthalmology, Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Samar El Achy
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Serag Kamel
- House Officer, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Tamer Refaat
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| |
Collapse
|
444
|
Magro G, Salvatorelli L, Puzzo L, Musumeci G, Bisceglia M, Parenti R. Oncofetal expression of Wilms' tumor 1 (WT1) protein in human fetal, adult and neoplastic skeletal muscle tissues. Acta Histochem 2015; 117:492-504. [PMID: 25800978 DOI: 10.1016/j.acthis.2015.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/10/2015] [Accepted: 02/21/2015] [Indexed: 12/12/2022]
Abstract
There is increasing evidence that WT1 protein expression is found not only at nuclear, but also at cytoplasmic, level in several developing and neoplastic tissues. In order to better understand the possible role of WT1 protein in human skeletal myogenesis and oncogenesis of rhabdomyosarcoma, we assessed immunohistochemically its comparative expression in a large series of human developing, adult and neoplastic skeletal muscle tissues. The present study shows that WT1 protein is developmentally expressed in the cytoplasm of human myoblasts from the 6 weeks of gestational age. This expression was maintained in the myotubes of developing muscles of the trunk, head, neck, and extremities, while it was down-regulated in fetal skeletal fibers from 20 weeks of gestational age as well as in adult normal skeletal muscle. Notably, WT1 immunostaining disappeared from rhabdomyomas, whereas it was strongly and diffusely re-expressed in all cases (27/27) of embryonal and alveolar rhabdomyosarcoma. The comparative evaluation of the immunohistochemical findings revealed that WT1 cytoplasmic expression in rhabdomyosarcoma may represent an ontogenetic reversal, and this nuclear transcription factor can also be considered an oncofetal protein which can be exploitable as an additional, highly sensitive immunomarker, together with desmin, myogenin and MyoD1, of this tumor. Moreover, our observations support the rationale for the use of WT1 protein-based target therapy in high risk rhabdomyosarcomas in children and adolescents.
Collapse
|
445
|
Evaluation of Efficacy of Radioimmunotherapy with 90Y-Labeled Fully Human Anti-Transferrin Receptor Monoclonal Antibody in Pancreatic Cancer Mouse Models. PLoS One 2015; 10:e0123761. [PMID: 25893775 PMCID: PMC4404254 DOI: 10.1371/journal.pone.0123761] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/21/2015] [Indexed: 11/24/2022] Open
Abstract
Objective Pancreatic cancer is an aggressive tumor and the prognosis remains poor. Therefore, development of more effective therapy is needed. We previously reported that 89Zr-labeled TSP-A01, an antibody against transferrin receptor (TfR), is highly accumulated in a pancreatic cancer xenograft, but not in major normal organs. In the present study, we evaluated the efficacy of radioimmunotherapy (RIT) with 90Y-TSP-A01 in pancreatic cancer mouse models. Methods TfR expression in pancreatic cancer cell lines (AsPC-1, BxPC-3, MIAPaCa-2) was evaluated by immunofluorescence staining. 111In-labeled anti-TfR antibodies (TSP-A01, TSP-A02) were evaluated in vitro by cell binding assay with the three cell lines and by competitive inhibition assay with MIAPaCa-2. In vivo biodistribution was evaluated in mice bearing BxPC-3 and MIAPaCa-2 xenografts. Tumor volumes of BxPC-3 and MIAPaCa-2 were sequentially measured after 90Y-TSP-A01 injection and histological analysis of tumors was conducted. Results MIAPaCa-2 cells showed the highest TfR expression, followed by AsPC-1 and BxPC-3 cells. 111In-TSP-A01 and 111In-TSP-A02 bound specifically to the three cell lines according to TfR expression. The dissociation constants for TSP-A01, DOTA-TSP-A01, TSP-A02, and DOTA-TSP-A02 were 0.22, 0.28, 0.17, and 0.22 nM, respectively. 111In-TSP-A01 was highly accumulated in tumors, especially in MIAPaCa-2, but this was not true of 111In-TSP-A02. The absorbed dose for 90Y-TSP-A01 was estimated to be 8.3 Gy/MBq to BxPC-3 and 12.4 Gy/MBq to MIAPaCa-2. MIAPaCa-2 tumors treated with 3.7 MBq of 90Y-TSP-A01 had almost completely disappeared around 3 weeks after injection and regrowth was not observed. Growth of BxPC-3 tumors was inhibited by 3.7 MBq of 90Y-TSP-A01, but the tumor size was not reduced. Conclusion 90Y-TSP-A01 treatment achieved an almost complete response in MIAPaCa-2 tumors, whereas it merely inhibited the growth of BxPC-3 tumors. 90Y-TSP-A01 is a promising RIT agent for pancreatic cancer, although further investigation is necessary to improve the efficacy for the radioresistant types like BxPC-3.
Collapse
|
446
|
Du W, Fan Y, He B, Zheng N, Yuan L, Dai W, Zhang H, Wang X, Wang J, Zhang X, Zhang Q. Bionano Interactions of MCF-7 Breast Tumor Cells with a Transferrin Receptor Targeted Nanoparticle. Mol Pharm 2015; 12:1467-76. [DOI: 10.1021/mp500796d] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Wenwen Du
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
- Department
of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yuchen Fan
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Bing He
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Nan Zheng
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | | | - Wenbing Dai
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Hua Zhang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Xueqing Wang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Jiancheng Wang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Xuan Zhang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| | - Qiang Zhang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, and ‡Medical and Healthy Analytical Center, Peking University, Beijing 100191, China
| |
Collapse
|
447
|
Wang G, Wang J, Wu W, Tony To SS, Zhao H, Wang J. Advances in lipid-based drug delivery: enhancing efficiency for hydrophobic drugs. Expert Opin Drug Deliv 2015; 12:1475-99. [PMID: 25843160 DOI: 10.1517/17425247.2015.1021681] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Many drug candidates with high therapeutic efficacy have low water solubility, which limits the administration and transport across physiological barriers, for example, the tumor tissue barrier. Therefore, strategies are needed to permeabilize the physiological barriers safely so that hydrophobic drugs may be delivered efficiently. AREAS COVERED This review focuses on prospects for therapeutic application of lipid-based drug delivery carriers that increase hydrophobic drugs to improve their solubility, bioavailability, drug release, targeting and absorption. Moreover, novel techniques to prepare for lipid-based drug delivery to extend pharmaceuticals with poor bioavailability such as surface modifications of lipid-based drug delivery are presented. Industrial developments of several drug candidates employing these strategies are discussed, as well as applications and clinical trials. EXPERT OPINION Overall, hydrophobic drugs can be encapsulated in the lipid-based drug delivery systems, represent a relatively safe and promising strategy to extend drug retention, lengthen the lifetime in the circulation, and allow active targeting to specific tissues and controllable drug release in the desirable sites. However, there are still noticeable gaps that need to be filled before the theoretical advantage of these formulations may truly be realized such as investigation on the use of lipid-based drug delivery for administration routes. This research may provide further interest within the area of lipid-based systems, both in industry and in the clinic.
Collapse
Affiliation(s)
- Gang Wang
- Shanghai Eighth People's Hospital, Department of Pharmaceutics , Shanghai , China
| | | | | | | | | | | |
Collapse
|
448
|
Li Y, Lin R, Wang L, Huang J, Wu H, Cheng G, Zhou Z, MacDonald T, Yang L, Mao H. PEG- b-AGE Polymer Coated Magnetic Nanoparticle Probes with Facile Functionalization and Anti-fouling Properties for Reducing Non-specific Uptake and Improving Biomarker Targeting. J Mater Chem B 2015; 3:3591-3603. [PMID: 26594360 DOI: 10.1039/c4tb01828a] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-specific surface adsorption of bio-macromolecules (e.g. proteins) on nanoparticles, known as biofouling, and the uptake of nanoparticles by the mononuclear phagocyte system (MPS) and reticuloendothelial system (RES) lead to substantial reduction in the efficiency of target-directed imaging and delivery in biomedical applications of engineered nanomaterials in vitro and in vivo. In this work, a novel copolymer consisting of blocks of poly ethylene glycol and allyl glycidyl ether (PEG-b-AGE) was developed for coating magnetic iron oxide nanoparticles (IONPs) to reduce non-specific protein adhesion that leads to formation of "protein corona" and uptake by macrophages. The facile surface functionalization was demonstrated by using targeting ligands of a small peptide of RGD or a whole protein of transferrin (Tf). The PEG-b-AGE coated IONPs exhibited anti-biofouling properties with significantly reduced protein corona formation and non-specific uptake by macrophages before and after the surface functionalization, thus improving targeting of RGD-conjugated PEG-b-AGE coated IONPs to integrins in U87MG glioblastoma and MDA-MB-231 breast cancer cells that overexpress αvβ3 integrins, and Tf-conjugated PEG-b-AGE coated IONPs to transferrin receptor (TfR) in D556 and Daoy medulloblastoma cancer cells with high overexpression of transferrin receptor, compared to respective control cell lines. Magnetic resonance imaging (MRI) of cancer cells treated with targeted IONPs with or without anti-biofouling PEG-b-AGE coating polymers demonstrated the target specific MRI contrast change using anti-biofouling PEG-b-AGE coated IONP with minimal off-targeted background compared to the IONPs without anti-biofouling coating, promising the highly efficient active targeting of nanoparticle imaging probes and drug delivery systems and potential applications of imaging quantification of targeted biomarkers.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Run Lin
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Guojun Cheng
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhengyang Zhou
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Radiology, Nanjing University College of Medicine and Affiliated Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Tobey MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
449
|
Endocytosis of a functionally enhanced GFP-tagged transferrin receptor in CHO cells. PLoS One 2015; 10:e0122452. [PMID: 25803700 PMCID: PMC4372551 DOI: 10.1371/journal.pone.0122452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
The endocytosis of transferrin receptor (TfR) has served as a model to study the receptor-targeted cargo delivery system for cancer therapy for many years. To accurately evaluate and optically measure this TfR targeting delivery in vitro, a CHO cell line with enhanced green fluorescent protein (EGFP)-tagged human TfR was established. A chimera of the hTfR and EGFP was engineered by fusing EGFP to the amino terminus of hTfR. Data were provided to demonstrate that hTfR-EGFP chimera was predominantly localized on the plasma membrane with some intracellular fluorescent structures on CHO cells and the EGFP moiety did not affect the endocytosis property of hTfR. Receptor internalization occurred similarly to that of HepG2 cells expressing wild-type hTfR. The internalization percentage of this chimeric receptor was about 81±3% of wild type. Time-dependent co-localization of hTfR-EGFP and PE-conjugated anti-hTfR mAb in living cells demonstrated the trafficking of mAb-receptor complexes through the endosomes followed by segregation of part of the mAb and receptor at the late stages of endocytosis. The CHO-hTfR cells preferentially took up anti-hTfR mAb conjugated nanoparticles. This CHO-hTfR cell line makes it feasible for accurate evaluation and visualization of intracellular trafficking of therapeutic agents conjugated with transferrin or Abs targeting the hTfRs.
Collapse
|
450
|
Dai Q, Yan Y, Ang CS, Kempe K, Kamphuis MMJ, Dodds SJ, Caruso F. Monoclonal antibody-functionalized multilayered particles: targeting cancer cells in the presence of protein coronas. ACS NANO 2015; 9:2876-85. [PMID: 25712076 DOI: 10.1021/nn506929e] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Engineered particles adsorb biomolecules (e.g., proteins) when introduced in a biological medium to form a layer called a "corona". Coronas, in particular the protein corona, play an important role in determining the surface properties of particles and their targeting abilities. This study examines the influence of protein coronas on the targeting ability of layer-by-layer (LbL)-assembled polymer capsules and core-shell particles functionalized with monoclonal antibodies. Upon exposure of humanized A33 monoclonal antibody (huA33 mAb)-functionalized poly(methacrylic acid) (PMA) capsules or huA33 mAb-PMA particles to human serum, a total of 83 or 65 proteins were identified in the protein coronas, respectively. Human serum of varying concentrations altered the composition of the protein corona. The antibody-driven specific cell membrane binding was qualitatively and quantitatively assessed by flow cytometry and fluorescence microscopy in both the absence and presence of a protein corona. The findings show that although different protein coronas formed in human serum (at different concentrations), the targeting ability of both the huA33 mAb-functionalized PMA capsules and particles toward human colon cancer cells was retained, demonstrating no significant difference compared with capsules and particles in the absence of protein coronas: ∼70% and ∼90% A33-expressing cells were targeted by the huA33 mAb-PMA capsules and particles, respectively, in a mixed cell population. This result demonstrates that the formation of protein coronas did not significantly influence the targeting ability of antibody-functionalized LbL-polymer carriers, indicating that the surface functionality of engineered particles in the presence of protein coronas can be preserved.
Collapse
Affiliation(s)
- Qiong Dai
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yan Yan
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- ‡Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kristian Kempe
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marloes M J Kamphuis
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Sarah J Dodds
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frank Caruso
- †ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|