1
|
Maradani BS, Parameswaran S, Subramanian K. Development of DNA aptamers targeting B7H3 by hybrid-SELEX: an alternative to antibodies for immuno-assays. Sci Rep 2024; 14:13552. [PMID: 38866941 PMCID: PMC11169341 DOI: 10.1038/s41598-024-64559-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Antibodies have been extensively used in numerous applications within proteomics-based technologies, requiring high sensitivity, specificity, a broad dynamic range for detection, and precise, reproducible quantification. Seeking alternatives to antibodies due to several inherent limitations of antibodies is an area of active research of tremendous importance. Recently, aptamers have been receiving increasing attention, because they not only have all of the advantages of antibodies, but also have unique advantages, such as thermal stability, low cost, and unlimited applications. Aptamers are gaining importance in immunological studies and can potentially replace antibodies in immunoassays. B7H3, an immunoregulatory protein belonging to the B7 family, is an attractive and promising target due to its overexpression in several tumor tissues while exhibiting limited expression in normal tissues. This study employed hybrid-SELEX with next-generation sequencing to select ssDNA aptamers specifically binding to the B7H3 protein. These aptamers demonstrated versatility across various assays, including flow cytometry, dot-blot, and immunohistochemistry. Effective performance in sandwich dot-blot assays and western blot analysis suggests their potential for diagnostic applications and demonstrates their adaptability and cost-effectiveness in diverse protein detection techniques.
Collapse
Affiliation(s)
- Bhavani Shankar Maradani
- L&T Ocular Pathology Department, Vision Research Foundation, Sankara Nethralaya, No. 41, College Road, Nungambakkam, Chennai, Tamil Nadu, 600006, India
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Sowmya Parameswaran
- Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Chennai, India
| | - Krishnakumar Subramanian
- L&T Ocular Pathology Department, Vision Research Foundation, Sankara Nethralaya, No. 41, College Road, Nungambakkam, Chennai, Tamil Nadu, 600006, India.
| |
Collapse
|
2
|
Li Y, Liu W, Xu H, Zhou Y, Xie W, Guo Y, Liao Z, Jiang X, Liu J, Ren C. Aptamers combined with immune checkpoints for cancer detection and targeted therapy: A review. Int J Biol Macromol 2024; 262:130032. [PMID: 38342267 DOI: 10.1016/j.ijbiomac.2024.130032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
In recent years, remarkable strides have been made in the field of immunotherapy, which has emerged as a standard treatment for many cancers. As a kind of immunotherapy drug, monoclonal antibodies employed in immune checkpoint therapy have proven beneficial for patients with diverse cancer types. However, owing to the extensive heterogeneity of clinical responses and the complexity and variability of the immune system and tumor microenvironment (TME), accurately predicting its efficacy remains a challenge. Recent advances in aptamers provide a promising approach for monitoring alterations within the immune system and TME, thereby facilitating targeted immunotherapy, particularly focused on immune checkpoint blockade, with enhanced antitumor efficiency. Aptamers have been widely used in tumor cell detection, biosensors, drug discovery, and biomarker screening due to their high specificity and high affinity with their targets. This review aims to comprehensively examine the research status and progress of aptamers in cancer diagnosis and immunotherapy, with a specific emphasis on those related to immune checkpoints. Additionally, we will discuss the future research directions and potential therapeutic targets for aptamer-based immune checkpoint therapy, aiming to provide a theoretical basis for targeting immunotherapy molecules and blocking tumor immune escape.
Collapse
Affiliation(s)
- Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Hongjuan Xu
- NHC Key Laboratory of Biological Nanotechnology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Zhou
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Youwei Guo
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical care medicine, Hainan Hospital of Chinese PLA General Hosptial; project supported by Hainan Province Clinical Medical Center, China.
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
3
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
4
|
Iman M, Moosavian SA, Zamani P, Jaafari MR. Preparation of AS1411 aptamer-modified PEGylated liposomal doxorubicin and evaluation of its anti-cancer effects in vitro and in vivo. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
5
|
Moudgil A, Salve R, Gajbhiye V, Chaudhari BP. Challenges and emerging strategies for next generation liposomal based drug delivery: An account of the breast cancer conundrum. Chem Phys Lipids 2023; 250:105258. [PMID: 36375540 DOI: 10.1016/j.chemphyslip.2022.105258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
The global cancer burden is witnessing an upsurge with breast cancer surpassing other cancers worldwide. Furthermore, an escalation in the breast cancer caseload is also expected in the coming years. The conventional therapeutic regimens practiced routinely are associated with many drawbacks to which nanotechnological interventions offer a great advantage. But how eminent could liposomes and their advantages be in superseding these existing therapeutic modalities? A solution is reflected in this review that draws attention to a decade-long journey embarked upon by researchers in this wake. This text is a comprehensive discussion of liposomes, the front runners of the drug delivery systems, and their active and passive targeting approaches for breast cancer management. Active targeting has been studied over the decade by many receptors overexpressed on the breast cancer cells and passive targeting with many drug combinations. The results converge on the fact that the actively targeted formulations exhibit a superior efficacy over their non-targeted counterparts and the all liposomal formulations are efficacious over the free drugs. This undoubtedly underlines the dominion of liposomal formulations over conventional chemotherapy. These investigations have led to the development of different liposomal formulations with active and passive targeting capacities that could be explored in depth. Acknowledging and getting a deeper insight into the liposomal evolution through time also unveiled many imperfections and unchartered territories that can be explored to deliver dexterous liposomal formulations against breast cancer and more in the clinical trial pipeline.
Collapse
Affiliation(s)
- Aliesha Moudgil
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Rajesh Salve
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Bhushan P Chaudhari
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India.
| |
Collapse
|
6
|
Development and classification of RNA aptamers for therapeutic purposes: an updated review with emphasis on cancer. Mol Cell Biochem 2022; 478:1573-1598. [DOI: 10.1007/s11010-022-04614-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/10/2022] [Indexed: 11/26/2022]
|
7
|
Milošević N, Rütter M, David A. Endothelial Cell Adhesion Molecules- (un)Attainable Targets for Nanomedicines. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:846065. [PMID: 35463298 PMCID: PMC9021548 DOI: 10.3389/fmedt.2022.846065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/15/2022] [Indexed: 01/21/2023] Open
Abstract
Endothelial cell adhesion molecules have long been proposed as promising targets in many pathologies. Despite promising preclinical data, several efforts to develop small molecule inhibitors or monoclonal antibodies (mAbs) against cell adhesion molecules (CAMs) ended in clinical-stage failure. In parallel, many well-validated approaches for targeting CAMs with nanomedicine (NM) were reported over the years. A wide range of potential applications has been demonstrated in various preclinical studies, from drug delivery to the tumor vasculature, imaging of the inflamed endothelium, or blocking immune cells infiltration. However, no NM drug candidate emerged further into clinical development. In this review, we will summarize the most advanced examples of CAM-targeted NMs and juxtapose them with known traditional drugs against CAMs, in an attempt to identify important translational hurdles. Most importantly, we will summarize the proposed strategies to enhance endothelial CAM targeting by NMs, in an attempt to offer a catalog of tools for further development.
Collapse
|
8
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
9
|
Yan J, Gao T, Lu Z, Yin J, Zhang Y, Pei R. Aptamer-Targeted Photodynamic Platforms for Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:27749-27773. [PMID: 34110790 DOI: 10.1021/acsami.1c06818] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Achieving controlled and accurate delivery of photosensitizers (PSs) into tumor sites is a major challenge in conventional photodynamic therapy (PDT). Aptamer is a short oligonucleotide sequence (DNA or RNA) with a folded three-dimensional structure, which can selectively bind to specific small molecules, proteins, or the whole cells. Aptamers could act as ligands and be modified onto PSs or nanocarriers, enabling specific recognition and binding to tumor cells or their membrane proteins. The resultant aptamer-modified PSs or PSs-containing nanocarriers generate amounts of reactive oxygen species with light irradiation and obtain superior photodynamic therapeutic efficiency in tumors. Herein, we overview the recent progress in the designs and applications of aptamer-targeted photodynamic platforms for tumor therapy. First, we focus on the progress on the rational selection of aptamers and summarize the applications of aptamers which have been applied for targeted tumor diagnosis and therapy. Then, aptamer-targeted photodynamic therapies including various aptamer-PSs, aptamer-nanocarriers containing PSs, and aptamer-nano-photosensitizers are highlighted. The aptamer-targeted synergistically therapeutic platforms including PDT, photothermal therapy, and chemotherapy, as well as the imaging-guided theranostics, are also discussed. Finally, we offer an insight into the development trends and future perspectives of aptamer-targeted photodynamic platforms for tumor therapy.
Collapse
Affiliation(s)
- Jincong Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Tian Gao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Zhongzhong Lu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| |
Collapse
|
10
|
Abbasi H, Rahbar N, Kouchak M, Khalil Dezfuli P, Handali S. Functionalized liposomes as drug nanocarriers for active targeted cancer therapy: a systematic review. J Liposome Res 2021; 32:195-210. [PMID: 33729077 DOI: 10.1080/08982104.2021.1903035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a broad term used to describe a group of diseases that have more than 270 types. Today, due to the suffering of patients from the side effects of existing methods in the treatment of cancer such as chemotherapy and radiotherapy, the employment of targeted methods in the treatment of this disease has been received much consideration. In recent years, nanoparticles have revolutionized in the treatment of many diseases such as cancer. Among these nanoparticles, liposomes are more considerable. Active targeted liposomes show an important role in the selective action of the drug on cancer cells. Until now, a variety of anti-cancer agents have been reported for targeted delivery to cancer cells using liposomes. The results of in vitro and studies in vivo have been shown that selective action of the targeted liposomes is increased with reduced side effects and toxicity compared with free drugs or non-targeted liposomes. This systematic review expresses the reports of this type of drug delivery system. Search terms were searched through several online databases including PubMed, Scopus, and Science Direct from 1990 to 2019 and the quality evaluation was performed. Out of 11,676 published articles, 196 articles met the inclusion criteria. The current report reviews developments in the liposomes targeted with aptamer, transferrin, folate, and monoclonal antibodies.
Collapse
Affiliation(s)
- Hanieh Abbasi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parna Khalil Dezfuli
- School of Pharmacy Library, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Hao T, Fu Y, Yang Y, Yang S, Liu J, Tang J, Ridwan KA, Teng Y, Liu Z, Li J, Guo N, Yu P. Tumor vasculature-targeting PEGylated peptide-drug conjugate prodrug nanoparticles improve chemotherapy and prevent tumor metastasis. Eur J Med Chem 2021; 219:113430. [PMID: 33865152 DOI: 10.1016/j.ejmech.2021.113430] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/15/2021] [Accepted: 03/28/2021] [Indexed: 11/27/2022]
Abstract
Metastasis is the main cause of death in cancer patients; therefore, new strategies or technologies that can inhibit the growth of primary tumors and their metastatic spread are extremely valuable. In this study, we selected an E-selectin-binding peptide as a targeting ligand and an inhibitor of metastasis, and conjugated this peptide with SN38 and PEG to produce an amphiphilic PEGylated peptide-drug conjugate (PDC). Novel self-assembled nanoparticles were then formed by the amphiphilic conjugate. The particles were actively targeted to the tumor vasculature by the peptide and passively to the tumor site by the enhanced permeability and retention (EPR) effect. As a nano-prodrug, this multifunctional conjugate (PEG-Pep-SN38) could reduce tumor growth, with an effect similar to that of irinotecan. Moreover, it could prolong the survival of mice bearing primary HCT116 tumors, which was not observed for its parent drug, SN38, nor the clinical prodrug of SN38 (irinotecan). Furthermore, this PDC prodrug prevented B16-F10 colonization in the lungs of mice. This study describes a new tumor vasculature-targeting PDC nano-prodrug with convenient preparation and high potential for cancer therapy, with the potential to be applied to other chemotherapeutic drugs.
Collapse
Affiliation(s)
- Tiantian Hao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Ying Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Yao Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Shuyan Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Jian Liu
- Tianjin Kingyork Group CO., LTD, No.221 Huanghai Road, TEDA, Tianjin, 300457, PR China
| | - Jingjing Tang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Kadir Ahmad Ridwan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China
| | - Jiuyuan Li
- Asymchem Life Science(Tianjin) Co., Ltd, PR China
| | - Na Guo
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, PR China.
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology Tianjin University of Science and Technology, No.29 of 13 th Avenue, TEDA, Tianjin, 300457, PR China.
| |
Collapse
|
12
|
Li Y, Cong H, Wang S, Yu B, Shen Y. Liposomes modified with bio-substances for cancer treatment. Biomater Sci 2020; 8:6442-6468. [DOI: 10.1039/d0bm01531h] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In recent years, liposomes have been used in the field of biomedicine and have achieved many significant results.
Collapse
Affiliation(s)
- Yanan Li
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Song Wang
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Bing Yu
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| |
Collapse
|
13
|
Nsairat H, Mahmoud IS, Odeh F, Abuarqoub D, Al-Azzawi H, Zaza R, Qadri MI, Ismail S, Al Bawab A, Awidi A, Alshaer W. Grafting of anti-nucleolin aptamer into preformed and remotely loaded liposomes through aptamer-cholesterol post-insertion. RSC Adv 2020; 10:36219-36229. [PMID: 35517091 PMCID: PMC9056972 DOI: 10.1039/d0ra07325c] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
A new combination strategy of an active loading and active targeting approach was applied in this work. The liposomes actively loaded with Curcumin (CRM) (LipCRM) were decorated with cholesterol tagged-anti-nucleolin AS1411 aptamer (NCL) via a new post-insertion approach, utilizing the cholesterol as a wedge to incorporate aptamer into the surface of the liposome bilayer. A successful NCL post-insertion was verified by agarose gel electrophoresis and dynamic light scattering (DLS). The cellular uptake of AptNCL-Lip was investigated using flow cytometry and Confocal Laser Scanning Microscopy (CLSM) on two different human breast cancer cell lines (MCF-7 and MDA-MB-231). The uptake and cytotoxicity of loaded CRM were investigated using flow cytometry and MTT assay. Our results showed successful post insertion of NCL aptamer to the surface of Lip. Also, higher cellular uptake was noted for AptNCL-Alexa-LipRhod compared to blank LipRhod in both cell lines. Moreover, CLSM showed prominent endocytosis and uptake of AptNCL-Alexa–LipRhod into the cytoplasm of breast cancer cells. Furthermore, the results showed a significant increase in the uptake and cytotoxicity of AptNCL-LipCRM compared to LipCRM in both cell lines. Overall, our results demonstrate a successful post-insertion of cholesterol-tagged aptamer into liposomes and the possible combination between active loading and active targeting. A new combination strategy of an active loading and active targeting approach was applied in this work.![]()
Collapse
|
14
|
Tran BH, Yu Y, Chang L, Tan B, Jia W, Xiong Y, Dai T, Zhong R, Zhang W, Le VM, Rose P, Wang Z, Mao Y, Zhu YZ. A Novel Liposomal S-Propargyl-Cysteine: A Sustained Release of Hydrogen Sulfide Reducing Myocardial Fibrosis via TGF-β1/Smad Pathway. Int J Nanomedicine 2019; 14:10061-10077. [PMID: 31920303 PMCID: PMC6935304 DOI: 10.2147/ijn.s216667] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/14/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose S-propargyl-cysteine (SPRC; alternatively known as ZYZ-802) is a novel modulator of endogenous tissue H2S concentrations with known cardioprotective and anti-inflammatory effects. However, its rapid metabolism and excretion have limited its clinical application. To overcome these issues, we have developed some novel liposomal carriers to deliver ZYZ-802 to cells and tissues and have characterized their physicochemical, morphological and pharmacological properties. Methods Two liposomal formulations of ZYZ-802 were prepared by thin-layer hydration and the morphological characteristics of each liposome system were assessed using a laser particle size analyzer and transmission electron microscopy. The entrapment efficiency and ZYZ-802 release profiles were determined following ultrafiltration centrifugation, dialysis tube and HPLC measurements. LC-MS/MS was used to evaluate the pharmacokinetic parameters and tissue distribution profiles of each formulation via the measurements of plasma and tissues ZYZ-802 and H2S concentrations. Using an in vivo model of heart failure (HF), the cardio-protective effects of liposomal carrier were determined by echocardiography, histopathology, Western blot and the assessment of antioxidant and myocardial fibrosis markers. Results Both liposomal formulations improved ZYZ-802 pharmacokinetics and optimized H2S concentrations in plasma and tissues. Liposomal ZYZ-802 showed enhanced cardioprotective effects in vivo. Importantly, liposomal ZYZ-802 could inhibit myocardial fibrosis via the inhibition of the TGF-β1/Smad signaling pathway. Conclusion The liposomal formulations of ZYZ-802 have enhanced pharmacokinetic and pharmacological properties in vivo. This work is the first report to describe the development of liposomal formulations to improve the sustained release of H2S within tissues.
Collapse
Affiliation(s)
- Ba Hieu Tran
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,School of Pharmacy, Macau University of Science and Technology, Taipa, Macau.,Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Ying Yu
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,Department of Cardiology, Xinhua Hospital, Shanghai, People's Republic of China
| | - Lingling Chang
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Bo Tan
- Department of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Wanwan Jia
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Ying Xiong
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Tao Dai
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Rui Zhong
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Weiping Zhang
- Department of Hematology, Institute of Hematology of PLA, Changhai Hospital, Shanghai, People's Republic of China
| | - Van Minh Le
- NTT Institute of Hi-Technology (NIH), Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Peter Rose
- School of Biosciences, University of Nottingham, Loughborough, LE12 5RD, UK
| | - Zhijun Wang
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,School of Pharmacy, Macau University of Science and Technology, Taipa, Macau
| | - Yicheng Mao
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yi Zhun Zhu
- School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,School of Pharmacy, Macau University of Science and Technology, Taipa, Macau
| |
Collapse
|
15
|
Odeh F, Nsairat H, Alshaer W, Ismail MA, Esawi E, Qaqish B, Bawab AA, Ismail SI. Aptamers Chemistry: Chemical Modifications and Conjugation Strategies. Molecules 2019; 25:E3. [PMID: 31861277 PMCID: PMC6982925 DOI: 10.3390/molecules25010003] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/14/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Soon after they were first described in 1990, aptamers were largely recognized as a new class of biological ligands that can rival antibodies in various analytical, diagnostic, and therapeutic applications. Aptamers are short single-stranded RNA or DNA oligonucleotides capable of folding into complex 3D structures, enabling them to bind to a large variety of targets ranging from small ions to an entire organism. Their high binding specificity and affinity make them comparable to antibodies, but they are superior regarding a longer shelf life, simple production and chemical modification, in addition to low toxicity and immunogenicity. In the past three decades, aptamers have been used in a plethora of therapeutics and drug delivery systems that involve innovative delivery mechanisms and carrying various types of drug cargos. However, the successful translation of aptamer research from bench to bedside has been challenged by several limitations that slow down the realization of promising aptamer applications as therapeutics at the clinical level. The main limitations include the susceptibility to degradation by nucleases, fast renal clearance, low thermal stability, and the limited functional group diversity. The solution to overcome such limitations lies in the chemistry of aptamers. The current review will focus on the recent arts of aptamer chemistry that have been evolved to refine the pharmacological properties of aptamers. Moreover, this review will analyze the advantages and disadvantages of such chemical modifications and how they impact the pharmacological properties of aptamers. Finally, this review will summarize the conjugation strategies of aptamers to nanocarriers for developing targeted drug delivery systems.
Collapse
Affiliation(s)
- Fadwa Odeh
- Faculty of Science, The University of Jordan, Amman 11942, Jordan; (F.O.); (H.N.); (A.A.B.)
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| | - Hamdi Nsairat
- Faculty of Science, The University of Jordan, Amman 11942, Jordan; (F.O.); (H.N.); (A.A.B.)
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Mohammad A. Ismail
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (M.A.I.); (E.E.); (B.Q.); (S.I.I.)
| | - Ezaldeen Esawi
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (M.A.I.); (E.E.); (B.Q.); (S.I.I.)
| | - Baraa Qaqish
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (M.A.I.); (E.E.); (B.Q.); (S.I.I.)
| | - Abeer Al Bawab
- Faculty of Science, The University of Jordan, Amman 11942, Jordan; (F.O.); (H.N.); (A.A.B.)
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| | - Said I. Ismail
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (M.A.I.); (E.E.); (B.Q.); (S.I.I.)
- Qatar Genome Project, Qatar Foundation, Doha 5825, Qatar
| |
Collapse
|
16
|
Aptamer-functionalized liposomes for targeted cancer therapy. Cancer Lett 2019; 448:144-154. [PMID: 30763718 DOI: 10.1016/j.canlet.2019.01.045] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Accumulation of chemotherapeutic agents in the tumor tissue while reducing adverse effects and drug resistance are among the major goals in cancer therapy. Among nanocarriers, liposomes have been found to be more effective in the passive targeting of cancer cells. A promising recent development in targeted drug delivery is the use of aptamer-functionalized liposomes for cancer therapy. Aptamer-targeted liposomes have enhanced uptake in tumor cells as shown in vitro and in vivo. Here, we discuss the aptamer-functionalized liposome platforms and review functionalization approaches as well as the factors affecting antitumor efficiency of aptamer-targeted liposomal systems. Finally, we provide a comprehensive overview of aptamer-targeted liposomes based on the molecular targets on the surface of cancer cells.
Collapse
|
17
|
Antipova OM, Zavyalova EG, Golovin AV, Pavlova GV, Kopylov AM, Reshetnikov RV. Advances in the Application of Modified Nucleotides in SELEX Technology. BIOCHEMISTRY (MOSCOW) 2018; 83:1161-1172. [PMID: 30472954 DOI: 10.1134/s0006297918100024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aptamers are widely used as molecular recognition elements for detecting and blocking functional biological molecules. Since the common "alphabet" of DNA and RNA consists of only four letters, the chemical diversity of aptamers is less than the diversity of protein recognition elements built of 20 amino acids. Chemical modification of nucleotides enlarges the potential of DNA/RNA aptamers. This review describes the latest achievements in a variety of approaches to aptamers selection with an extended genetic alphabet.
Collapse
Affiliation(s)
- O M Antipova
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia. .,Apto-Pharm Ltd., Moscow, 115564, Russia
| | - E G Zavyalova
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia.,Apto-Pharm Ltd., Moscow, 115564, Russia
| | - A V Golovin
- Apto-Pharm Ltd., Moscow, 115564, Russia.,Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119234, Russia.,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia
| | - G V Pavlova
- Apto-Pharm Ltd., Moscow, 115564, Russia.,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.,Burdenko National Scientific and Practical Center for Neurosurgery, Ministry of Healthcare of the Russian Federation, Moscow, 125047, Russia
| | - A M Kopylov
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia.,Apto-Pharm Ltd., Moscow, 115564, Russia
| | - R V Reshetnikov
- Apto-Pharm Ltd., Moscow, 115564, Russia.,Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119234, Russia.,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
18
|
Camorani S, Fedele M, Zannetti A, Cerchia L. TNBC Challenge: Oligonucleotide Aptamers for New Imaging and Therapy Modalities. Pharmaceuticals (Basel) 2018; 11:ph11040123. [PMID: 30428522 PMCID: PMC6316260 DOI: 10.3390/ph11040123] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/11/2022] Open
Abstract
Compared to other breast cancers, triple-negative breast cancer (TNBC) usually affects younger patients, is larger in size, of higher grade and is biologically more aggressive. To date, conventional cytotoxic chemotherapy remains the only available treatment for TNBC because it lacks expression of the estrogen receptor (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2), and no alternative targetable molecules have been identified so far. The high biological and clinical heterogeneity adds a further challenge to TNBC management and requires the identification of new biomarkers to improve detection by imaging, thus allowing the specific treatment of each individual TNBC subtype. The Systematic Evolution of Ligands by EXponential enrichment (SELEX) technique holds great promise to the search for novel targetable biomarkers, and aptamer-based molecular approaches have the potential to overcome obstacles of current imaging and therapy modalities. In this review, we highlight recent advances in oligonucleotide aptamers used as imaging and/or therapeutic agents in TNBC, discussing the potential options to discover, image and hit new actionable targets in TNBC.
Collapse
Affiliation(s)
- Simona Camorani
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale G. Salvatore (IEOS), CNR, 80145 Naples, Italy.
| | - Monica Fedele
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale G. Salvatore (IEOS), CNR, 80145 Naples, Italy.
| | | | - Laura Cerchia
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale G. Salvatore (IEOS), CNR, 80145 Naples, Italy.
| |
Collapse
|
19
|
Walss-Bass C, Lokesh GLR, Dyukova E, Gorenstein DG, Roberts DL, Velligan D, Volk DE. X-Aptamer Technology Identifies C4A and ApoB in Blood as Potential Markers for Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 5:52-59. [PMID: 31019918 DOI: 10.1159/000492331] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
The field of proteomics is rapidly gaining territory as a promising alternative to genomic approaches in the efforts to unravel the complex molecular mechanisms underlying schizophrenia and other psychiatric disorders. X-aptamer tech-nology has emerged as a novel proteomic approach for high-sensitivity analyses, and we hypothesized that this technology would identify unique molecular signatures in plasma samples from schizophrenia patients (n = 60) compared to controls (n = 20). Using a combinatorial library of X-aptamer beads, we developed a two-color flow cytometer-based approach to identify specific X-aptamers that bound with high specificity to each target group. Based on this, we synthesized two unique X-aptamer sequences, and specific proteins pulled down from the patient and control groups by these X-aptamers were identified by mass spectrometry. We identified two protein biomarkers, complement component C4A and ApoB, upregulated in plasma samples from schizophrenia patients. ELISA validation suggested that the observed differences in C4 levels in patients are likely due to the presence of the illness itself, while ApoB may be a marker of antipsychotic-induced alterations. These studies highlight the utility of the X-aptamer technology in the identification of biomarkers for schizophrenia that will advance our understanding of the pathophysiological mechanisms of this disorder.
Collapse
Affiliation(s)
- Consuelo Walss-Bass
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ganesh L R Lokesh
- Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Elena Dyukova
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - David G Gorenstein
- Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - David L Roberts
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Dawn Velligan
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David E Volk
- Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
20
|
Kovacevic KD, Gilbert JC, Jilma B. Pharmacokinetics, pharmacodynamics and safety of aptamers. Adv Drug Deliv Rev 2018; 134:36-50. [PMID: 30321620 DOI: 10.1016/j.addr.2018.10.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022]
Abstract
Aptamers are synthetic molecules structured as single-stranded DNA or RNA oligonucleotides that can be designed to mimic the functional properties of monoclonal antibodies. They bind to the target molecules (typically soluble or cell-bound proteins) with high affinity (with picomolar to low nanomolar range) and specificity, and therefore can be an alternative to therapeutic antibodies or peptide ligands. This paper reviews published data regarding pharmacokinetics, pharmacodynamics and safety of aptamers from preclinical and clinical studies. Aptamers have been developed for the treatment of a variety of diseases, including cancer, macular degeneration,g cardiovascular disease, diabetes and anaemia of chronic diseases. There are several preclinical studies with unmodified aptamers, but the vast majority of aptamer trials in humans have been conducted with modified aptamers, because unmodified aptamers demonstrate metabolic instability, as well as rapid renal filtration and elimination. Various strategies have been developed to improve the pharmacokinetic profile of aptamers. Aside from chemical modification of nucleotides in order to stabilize them against nuclease degradation, the main modification to extend the half-life is pegylation. Therefore, the process of pegylation as well as its benefits and possible shortcomings will briefly be discussed.
Collapse
|
21
|
Alshaer W, Hillaireau H, Fattal E. Aptamer-guided nanomedicines for anticancer drug delivery. Adv Drug Deliv Rev 2018; 134:122-137. [PMID: 30267743 DOI: 10.1016/j.addr.2018.09.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/08/2023]
Abstract
Aptamers are versatile nucleic acid-based macromolecules characterized by their high affinity and specificity to a specific target. Taking advantage of such binding properties, several aptamers have been selected to bind tumor biomarkers and have been used as targeting ligands for the functionalization of nanomedicines. Different functionalization methods have been used to link aptamers to the surface drug nanocarriers. The pre-clinical data of such nanomedicines overall show an enhanced and selective delivery of therapeutic payloads to cancer cells, thereby accelerating steps towards more effective therapeutic systems. This review describes the current advances in the use of aptamers as targeting moieties for the delivery of therapeutic and imaging agents to tumors by conjugation to organic and inorganic nanocarriers.
Collapse
|
22
|
Avitabile E, Bedognetti D, Ciofani G, Bianco A, Delogu LG. How can nanotechnology help the fight against breast cancer? NANOSCALE 2018; 10:11719-11731. [PMID: 29917035 DOI: 10.1039/c8nr02796j] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this review we provide a broad overview on the use of nanotechnology for the fight against breast cancer (BC). Nowadays, detection, diagnosis, treatment, and prevention may be possible thanks to the application of nanotechnology to clinical practice. Taking into consideration the different forms of BC and the disease status, nanomaterials can be designed to meet the most forefront objectives of modern therapy and diagnosis. We have analyzed in detail three main groups of nanomaterial applications for BC treatment and diagnosis. We have identified several types of drugs successfully conjugated with nanomaterials. We have analyzed the main important imaging techniques and all nanomaterials used to help the non-invasive, early detection of the lesions. Moreover, we have examined theranostic nanomaterials as unique tools, combining imaging, detection, and therapy for BC. This state of the art review provides a useful guide depicting how nanotechnology can be used to overcome the current barriers in BC clinical practice, and how it will shape the future scenario of treatments, prevention, and diagnosis, revolutionizing the current approaches, e.g., reducing the suffering related to chemotherapy.
Collapse
Affiliation(s)
- Elisabetta Avitabile
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy.
| | | | | | | | | |
Collapse
|
23
|
Abstract
PURPOSE Cancer remains a significant cause of morbidity and mortality across the globe. A recent report suggests around 14.1 million new cases and 8.2 million cancer-related deaths, which are expected to reach 21.7 million and 13 million by 2030 worldwide, respectively. MATERIALS AND METHODS Because of highly complex mechanisms of cancer progression, it is important to explore and develop new innovative technologies which are more efficient compared with presently available treatment options. RESULTS Currently, chemotherapy, radiation and surgery are the most commonly used cancer treatment methods. In the last decade, nanomedicine emerged as an alternative treatment option that uses specific drug-delivery systems, improves efficacy of drugs and reduces detrimental side effects to normal tissues. CONCLUSION In this review, we have summarized cancer nanomedicines (active and passive drug delivery) available in the market. We have also discussed other nanomedicines that are at different stages of clinical trials.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- a King Fahd Medical Research Center , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Khalid Anwar
- b School of Life Sciences , Jawaharlal Nehru University , New Delhi , India
| | - Chelapram K Firoz
- a King Fahd Medical Research Center , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Mohammad Oves
- c Center of Excellence in Environmental Studies , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Mohammad Amjad Kamal
- a King Fahd Medical Research Center , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Shams Tabrez
- a King Fahd Medical Research Center , King Abdulaziz University , Jeddah , Saudi Arabia
| |
Collapse
|
24
|
Abstract
Aptamers and second generation analogs, such as X-Aptamers (XAs), SOMAmers, locked nucleic acids (LNAs), and others are increasingly being used for molecular pathway targeting, biomarker discovery, or disease diagnosis by interacting with protein targets on the surface of cells or in solution. Such targeting is being used for imaging, diagnostic evaluation, interference of protein function, or delivery of therapeutic agents. Selection of aptamers using the original SELEX method is cumbersome and time-consuming, often requiring 10-15 rounds of selection, and provides aptamers with a limited number of functional groups, namely four bases of DNA or RNA, although newer SELEX methods have increased this diversity. In contrast, X-Aptamers provide an unlimited number of functional groups and thus are superior targeting agents. Here, we discuss the X-Aptamer selection process.
Collapse
|
25
|
Morita Y, Leslie M, Kameyama H, Volk DE, Tanaka T. Aptamer Therapeutics in Cancer: Current and Future. Cancers (Basel) 2018; 10:cancers10030080. [PMID: 29562664 PMCID: PMC5876655 DOI: 10.3390/cancers10030080] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Aptamer-related technologies represent a revolutionary advancement in the capacity to rapidly develop new classes of targeting ligands. Structurally distinct RNA and DNA oligonucleotides, aptamers mimic small, protein-binding molecules and exhibit high binding affinity and selectivity. Although their molecular weight is relatively small—approximately one-tenth that of monoclonal antibodies—their complex tertiary folded structures create sufficient recognition surface area for tight interaction with target molecules. Additionally, unlike antibodies, aptamers can be readily chemically synthesized and modified. In addition, aptamers’ long storage period and low immunogenicity are favorable properties for clinical utility. Due to their flexibility of chemical modification, aptamers are conjugated to other chemical entities including chemotherapeutic agents, siRNA, nanoparticles, and solid phase surfaces for therapeutic and diagnostic applications. However, as relatively small sized oligonucleotides, aptamers present several challenges for successful clinical translation. Their short plasma half-lives due to nuclease degradation and rapid renal excretion necessitate further structural modification of aptamers for clinical application. Since the US Food and Drug Administration (FDA) approval of the first aptamer drug, Macugen® (pegaptanib), which treats wet-age-related macular degeneration, several aptamer therapeutics for oncology have followed and shown promise in pre-clinical models as well as clinical trials. This review discusses the advantages and challenges of aptamers and introduces therapeutic aptamers under investigation and in clinical trials for cancer treatments.
Collapse
Affiliation(s)
- Yoshihiro Morita
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - Macall Leslie
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - Hiroyasu Kameyama
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - David E Volk
- McGovern Medical School, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston, TX 77030, USA.
| | - Takemi Tanaka
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, 940 SL Young Blvd, Oklahoma City, OK 73104, USA.
| |
Collapse
|
26
|
Woodside DG, Tanifum EA, Ghaghada KB, Biediger RJ, Caivano AR, Starosolski ZA, Khounlo S, Bhayana S, Abbasi S, Craft JW, Maxwell DS, Patel C, Stupin IV, Bakthavatsalam D, Market RV, Willerson JT, Dixon RAF, Vanderslice P, Annapragada AV. Magnetic Resonance Imaging of Atherosclerotic Plaque at Clinically Relevant Field Strengths (1T) by Targeting the Integrin α4β1. Sci Rep 2018; 8:3733. [PMID: 29487319 PMCID: PMC5829217 DOI: 10.1038/s41598-018-21893-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Inflammation drives the degradation of atherosclerotic plaque, yet there are no non-invasive techniques available for imaging overall inflammation in atherosclerotic plaques, especially in the coronary arteries. To address this, we have developed a clinically relevant system to image overall inflammatory cell burden in plaque. Here, we describe a targeted contrast agent (THI0567-targeted liposomal-Gd) that is suitable for magnetic resonance (MR) imaging and binds with high affinity and selectivity to the integrin α4β1(very late antigen-4, VLA-4), a key integrin involved in recruiting inflammatory cells to atherosclerotic plaques. This liposomal contrast agent has a high T1 relaxivity (~2 × 105 mM-1s-1 on a particle basis) resulting in the ability to image liposomes at a clinically relevant MR field strength. We were able to visualize atherosclerotic plaques in various regions of the aorta in atherosclerosis-prone ApoE-/- mice on a 1 Tesla small animal MRI scanner. These enhanced signals corresponded to the accumulation of monocyte/macrophages in the subendothelial layer of atherosclerotic plaques in vivo, whereas non-targeted liposomal nanoparticles did not demonstrate comparable signal enhancement. An inflammatory cell-targeted method that has the specificity and sensitivity to measure the inflammatory burden of a plaque could be used to noninvasively identify patients at risk of an acute ischemic event.
Collapse
Affiliation(s)
- Darren G Woodside
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.
| | - Eric A Tanifum
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ronald J Biediger
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Amy R Caivano
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Zbigniew A Starosolski
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Sayadeth Khounlo
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Saakshi Bhayana
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Shahrzad Abbasi
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - John W Craft
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.,Department of Biology and Chemistry, University of Houston, 4800 Calhoun Road, Houston, Texas, 77004, USA
| | - David S Maxwell
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA.,Department of Institutional Analytics and Informatics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chandreshkumar Patel
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Igor V Stupin
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | | | - Robert V Market
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - James T Willerson
- Division of Cardiology Research, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Richard A F Dixon
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Peter Vanderslice
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Ananth V Annapragada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA.
| |
Collapse
|
27
|
Prusty DK, Adam V, Zadegan RM, Irsen S, Famulok M. Supramolecular aptamer nano-constructs for receptor-mediated targeting and light-triggered release of chemotherapeutics into cancer cells. Nat Commun 2018; 9:535. [PMID: 29416033 PMCID: PMC5803212 DOI: 10.1038/s41467-018-02929-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/09/2018] [Indexed: 12/01/2022] Open
Abstract
Platforms for targeted drug-delivery must simultaneously exhibit serum stability, efficient directed cell internalization, and triggered drug release. Here, using lipid-mediated self-assembly of aptamers, we combine multiple structural motifs into a single nanoconstruct that targets hepatocyte growth factor receptor (cMet). The nanocarrier consists of lipidated versions of a cMet-binding aptamer and a separate lipidated GC-rich DNA hairpin motif loaded with intercalated doxorubicin. Multiple 2',6'-dimethylazobenzene moieties are incorporated into the doxorubicin-binding motif to trigger the release of the chemotherapeutics by photoisomerization. The lipidated DNA scaffolds self-assemble into spherical hybrid-nanoconstructs that specifically bind cMet. The combined features of the nanocarriers increase serum nuclease resistance, favor their import into cells presumably mediated by endocytosis, and allow selective photo-release of the chemotherapeutic into the targeted cells. cMet-expressing H1838 tumor cells specifically internalize drug-loaded nanoconstructs, and subsequent UV exposure enhances cell mortality. This modular approach thus paves the way for novel classes of powerful aptamer-based therapeutics.
Collapse
Affiliation(s)
- Deepak K Prusty
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany
- Stiftung Caesar, Max-Planck-Fellowship Group Chemical Biology, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Volker Adam
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany
| | - Reza M Zadegan
- Nanoscale Materials & Device Group, Micron School of Materials Science and Engineering, Boise State University, Boise, USA
| | - Stephan Irsen
- Stiftung Caesar, Elektronenmikroskopie und Analytik, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Michael Famulok
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany.
- Stiftung Caesar, Max-Planck-Fellowship Group Chemical Biology, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
| |
Collapse
|
28
|
Leonard F, Ha NP, Sule P, Alexander JF, Volk DE, Lokesh GLR, Liu X, Cirillo JD, Gorenstein DG, Yuan J, Chatterjee S, Graviss EA, Godin B. Thioaptamer targeted discoidal microparticles increase self immunity and reduce Mycobacterium tuberculosis burden in mice. J Control Release 2017; 266:238-247. [PMID: 28987879 DOI: 10.1016/j.jconrel.2017.09.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/29/2017] [Indexed: 12/25/2022]
Abstract
Worldwide, tuberculosis (TB) remains one of the most prevalent infectious diseases causing morbidity and death in >1.5 million patients annually. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, usually resides in the alveolar macrophages. Current tuberculosis treatment methods require more than six months, and low compliance often leads to therapeutic failure and multidrug resistant strain development. Critical to improving TB-therapy is shortening treatment duration and increasing therapeutic efficacy. In this study, we sought to determine if lung hemodynamics and pathological changes in Mtb infected cells can be used for the selective targeting of microparticles to infected tissue(s). Thioaptamers (TA) with CD44 (CD44TA) targeting moiety were conjugated to discoidal silicon mesoporous microparticles (SMP) to enhance accumulation of these agents/carriers in the infected macrophages in the lungs. In vitro, CD44TA-SMP accumulated in macrophages infected with mycobacteria efficiently killing the infected cells and decreasing survival of mycobacteria. In vivo, increased accumulations of CD44TA-SMP were recorded in the lung of M. tuberculosis infected mice as compared to controls. TA-targeted carriers significantly diminished bacterial load in the lungs and caused recruitment of T lymphocytes. Proposed mechanism of action of the designed vector accounts for a combination of increased uptake of particles that leads to infected macrophage death, as well as, activation of cellular immunity by the TA, causing increased T-cell accumulation in the treated lungs. Based on our data with CD44TA-SMP, we anticipate that this drug carrier can open new avenues in TB management.
Collapse
Affiliation(s)
- Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Ngan P Ha
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Preeti Sule
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - Jenolyn F Alexander
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - David E Volk
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Ganesh L R Lokesh
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Jeffrey D Cirillo
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - David G Gorenstein
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Jinyun Yuan
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Soumya Chatterjee
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States.
| |
Collapse
|
29
|
Tang X, Loc WS, Dong C, Matters GL, Butler PJ, Kester M, Meyers C, Jiang Y, Adair JH. The use of nanoparticulates to treat breast cancer. Nanomedicine (Lond) 2017; 12:2367-2388. [PMID: 28868970 DOI: 10.2217/nnm-2017-0202] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is a major ongoing public health issue among women in both developing and developed countries. Significant progress has been made to improve the breast cancer treatment in the past decades. However, the current clinical approaches are invasive, of low specificity and can generate severe side effects. As a rapidly developing field, nanotechnology brings promising opportunities to human cancer diagnosis and treatment. The use of nanoparticulate-based platforms overcomes biological barriers and allows prolonged blood circulation time, simultaneous tumor targeting and enhanced accumulation of drugs in tumors. Currently available and clinically applicable innovative nanoparticulate-based systems for breast cancer nanotherapies are discussed in this review.
Collapse
Affiliation(s)
- Xiaomeng Tang
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA.,Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Welley S Loc
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA.,Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Cheng Dong
- Department of Bioengineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Gail L Matters
- Department of Biochemistry & Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Peter J Butler
- Department of Bioengineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Craig Meyers
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Yixing Jiang
- Marlene & Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James H Adair
- Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
30
|
Volk DE, Lokesh GLR. Development of Phosphorothioate DNA and DNA Thioaptamers. Biomedicines 2017; 5:E41. [PMID: 28703779 PMCID: PMC5618299 DOI: 10.3390/biomedicines5030041] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/03/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid aptamers are short RNA- or DNA-based affinity reagents typically selected from combinatorial libraries to bind to a specific target such as a protein, a small molecule, whole cells or even animals. Aptamers have utility in the development of diagnostic, imaging and therapeutic applications due to their size, physico-chemical nature and ease of synthesis and modification to suit the application. A variety of oligonucleotide modifications have been used to enhance the stability of aptamers from nuclease degradation in vivo. The non-bridging oxygen atoms of the phosphodiester backbones of RNA and DNA aptamers can be substituted with one or two sulfur atoms, resulting in thioaptamers with phosphorothioate or phosphorodithioate linkages, respectively. Such thioaptamers are known to have increased binding affinity towards their target, as well as enhanced resistance to nuclease degradation. In this review, we discuss the development of phosphorothioate chemistry and thioaptamers, with a brief review of selection methods.
Collapse
Affiliation(s)
- David E Volk
- McGovern Medical School, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Ganesh L R Lokesh
- McGovern Medical School, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
31
|
Morph-X-Select: Morphology-based tissue aptamer selection for ovarian cancer biomarker discovery. Biotechniques 2016; 61:249-259. [PMID: 27839510 DOI: 10.2144/000114473] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/09/2016] [Indexed: 12/28/2022] Open
Abstract
High affinity aptamer-based biomarker discovery has the advantage of simultaneously discovering an aptamer affinity reagent and its target biomarker protein. Here, we demonstrate a morphology-based tissue aptamer selection method that enables us to use tissue sections from individual patients and identify high-affinity aptamers and their associated target proteins in a systematic and accurate way. We created a combinatorial DNA aptamer library that has been modified with thiophosphate substitutions of the phosphate ester backbone at selected 5´dA positions for enhanced nuclease resistance and targeting. Based on morphological assessment, we used image-directed laser microdissection (LMD) to dissect regions of interest bound with the thioaptamer (TA) library and further identified target proteins for the selected TAs. We have successfully identified and characterized the lead candidate TA, V5, as a vimentin-specific sequence that has shown specific binding to tumor vasculature of human ovarian tissue and human microvascular endothelial cells. This new Morph-X-Select method allows us to select high-affinity aptamers and their associated target proteins in a specific and accurate way, and could be used for personalized biomarker discovery to improve medical decision-making and to facilitate the development of targeted therapies to achieve more favorable outcomes.
Collapse
|
32
|
Whiting N, Hu J, Zacharias NM, Lokesh GLR, Volk DE, Menter DG, Rupaimoole R, Previs R, Sood AK, Bhattacharya P. Developing hyperpolarized silicon particles for in vivo MRI targeting of ovarian cancer. J Med Imaging (Bellingham) 2016; 3:036001. [PMID: 27547777 DOI: 10.1117/1.jmi.3.3.036001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/18/2016] [Indexed: 11/14/2022] Open
Abstract
Silicon-based nanoparticles are ideally suited for use as biomedical imaging agents due to their biocompatibility, biodegradability, and simple surface chemistry that facilitates drug loading and targeting. A method of hyperpolarizing silicon particles using dynamic nuclear polarization, which increases magnetic resonance imaging signals by several orders-of-magnitude through enhanced nuclear spin alignment, has recently been developed to allow silicon particles to function as contrast agents for in vivo magnetic resonance imaging. The enhanced spin polarization of silicon lasts significantly longer than other hyperpolarized agents (tens of minutes, whereas [Formula: see text] for other species at room temperature), allowing a wide range of potential applications. We report our recent characterizations of hyperpolarized silicon particles, with the ultimate goal of targeted, noninvasive, and nonradioactive molecular imaging of various cancer systems. A variety of particle sizes (20 nm to [Formula: see text]) were found to have hyperpolarized relaxation times ranging from [Formula: see text] to 50 min. The addition of various functional groups to the particle surface had no effect on the hyperpolarization buildup or decay rates and allowed in vivo imaging over long time scales. Additional in vivo studies examined a variety of particle administration routes in mice, including intraperitoneal injection, rectal enema, and oral gavage.
Collapse
Affiliation(s)
- Nicholas Whiting
- University of Texas MD Anderson Cancer Center , Department of Cancer Systems Imaging, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Jingzhe Hu
- University of Texas MD Anderson Cancer Center, Department of Cancer Systems Imaging, 1515 Holcombe Boulevard, Houston, Texas 77030, United States; Rice University, Department of Bioengineering, 6100 Main Street, Houston, Texas 770005-1892, United States
| | - Niki M Zacharias
- University of Texas MD Anderson Cancer Center , Department of Cancer Systems Imaging, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Ganesh L R Lokesh
- University of Texas Health Science Center at Houston , Department of NanoMedicine and Biomedical Engineering and the Institute of Molecular Medicine, 7000 Fannin, Houston, Texas 77030, United States
| | - David E Volk
- University of Texas Health Science Center at Houston , Department of NanoMedicine and Biomedical Engineering and the Institute of Molecular Medicine, 7000 Fannin, Houston, Texas 77030, United States
| | - David G Menter
- University of Texas MD Anderson Cancer Center , Department of Gastrointestinal Medical Oncology, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Rajesha Rupaimoole
- University of Texas MD Anderson Cancer Center , Department of Gynecologic Oncology and Reproductive Medicine, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Rebecca Previs
- University of Texas MD Anderson Cancer Center , Department of Gynecologic Oncology and Reproductive Medicine, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Anil K Sood
- University of Texas MD Anderson Cancer Center, Department of Gynecologic Oncology and Reproductive Medicine, 1515 Holcombe Boulevard, Houston, Texas 77030, United States; University of Texas MD Anderson Cancer Center, Center for RNA Interference and Non-Coding RNA, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Pratip Bhattacharya
- University of Texas MD Anderson Cancer Center , Department of Cancer Systems Imaging, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
33
|
Li X, Zhu X, Qiu L. Constructing aptamer anchored nanovesicles for enhanced tumor penetration and cellular uptake of water soluble chemotherapeutics. Acta Biomater 2016; 35:269-79. [PMID: 26873366 DOI: 10.1016/j.actbio.2016.02.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 02/01/2016] [Accepted: 02/08/2016] [Indexed: 12/20/2022]
Abstract
Polymersomes represent a promising pharmaceutical vehicle for the delivery of hydrophilic therapeutic agents. However, modification of polymersomes with molecules that confer targeting functions remains challenging because of the strict requirements regarding the weight fractions of the hydrophilic and hydrophobic block polymers. In this study, based on the compatibility between cholesterol and polymeric carriers, polymersomes self-assembled by amphiphilic graft polyphosphazenes were endowed with a targeting function by incorporating the cholesterol-linked aptamer through a simple dialysis method. The aqueous interior of the polymersomes was employed to encapsulate water-soluble doxorubicin hydrochloride. In vivo experiments in tumor-bearing mice showed that the aptamer-anchored vesicle targeted accumulation at the tumor site, favorable penetration through tumor tissue, and incremental endocytosis into tumor cells. Correspondingly, the aptamer-anchored vesicle decreased systemic toxicity and effectively suppressed the growth of subcutaneous MCF-7 xenografts. These findings suggested that vesicles modified with targeted groups via hydrophobic supermolecular interactions could provide a platform for selective delivery of hydrophilic drug. STATEMENT OF SIGNIFICANCE Polymersomes have represented a promising type of pharmaceutical vehicles due to their predominant physical properties. However, it is still a challenge to endow polymersomes with active target function because of strict requirements of the weight fractions of hydrophilic polymer block to hydrophobic one. In this research, by taking advantage of the supermolecular interactions between amphiphilic graft polyphosphazene and cholesterol which was linked to aptamer AS1411, we prepared a targeted functional polymersome (PEP-DOX·HCl-Ap) through a simple method with high loading of water soluble anti-cancer drug doxorubicin hydrochloride. The in vivo experiments in MCF-7 tumor-bearing mice demonstrated several advantages of PEP-DOX·HCl-Ap vesicle such as prolonged circulation time in blood, targeted accumulation at tumor site, permeation through the tumor tissue and incremental endocytosis by tumor cells, which consequently resulted in the significantly improved anti-cancer efficacy. Moreover, this novel polymersome designed in this study has built a research platform to achieve targeted delivery of hydrophilic chemotherapeutics for cancer therapy.
Collapse
Affiliation(s)
- Xin Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiumei Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 38 Zheda Road, Hangzhou 310027, China; Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
34
|
Riese SB, Buscher K, Enders S, Kuehne C, Tauber R, Dernedde J. Structural requirements of mono- and multivalent L-selectin blocking aptamers for enhanced receptor inhibition in vitro and in vivo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:901-908. [PMID: 26772426 DOI: 10.1016/j.nano.2015.12.379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/28/2015] [Accepted: 12/24/2015] [Indexed: 01/08/2023]
Abstract
UNLABELLED L-selectin mediates extravasation of leukocytes from blood into the surrounding tissue during inflammation and is therefore a therapeutical target in certain overwhelming immune reactions. In this study, we characterized an L-selectin specific blocking DNA aptamer with respect to nucleotide composition and target binding. Introduction of deletions and nucleotide exchanges resulted in an optimized DNA sequence but preservation of the IC50 in the low nanomolar range. The inhibitory potential was significantly increased when the aptamer was displayed as a di- and trimer connected via appropriate linker length. Similar to monoclonal antibodies, trimer yielded picomolar IC50 values in a competitive binding assay. In comparison to the monovalent aptamer, the trivalent assembly reduced PBMC interactions to L-selectin ligands 90-fold under shear and exerted superior inhibition of PBMC rolling in vivo. In conclusion, our work demonstrates the feasibility of optimizing aptamer sequences and shows that multivalent ligand presentation enables superior adhesion receptor targeting. FROM THE CLINICAL EDITOR During inflammation, leukocytes extravasate from blood vessels under chemotaxic signals. The presence of L-selectin on endothelium acts as a mediator for the extravasation process. In this study, the authors investigated an L-selectin specific blocking DNA aptamer in various forms, as inhibitors to leukocyte binding and extravasation. This new approach confirmed the potential use of aptamers in clinical setting.
Collapse
Affiliation(s)
- Sebastian B Riese
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Konrad Buscher
- Institute for Physiological Chemistry Pathobiochemistry, University of Muenster, Muenster, Germany; Department of Nephrology and Rheumatology, University Hospital Muenster, Muenster, Germany.
| | - Sven Enders
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Christian Kuehne
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Rudolf Tauber
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Jens Dernedde
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
35
|
Conjugate-SELEX: A High-throughput Screening of Thioaptamer-liposomal Nanoparticle Conjugates for Targeted Intracellular Delivery of Anticancer Drugs. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e382. [DOI: 10.1038/mtna.2016.81] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/17/2016] [Indexed: 01/03/2023]
|
36
|
Moosavian SA, Abnous K, Badiee A, Jaafari MR. Improvement in the drug delivery and anti-tumor efficacy of PEGylated liposomal doxorubicin by targeting RNA aptamers in mice bearing breast tumor model. Colloids Surf B Biointerfaces 2015; 139:228-36. [PMID: 26722819 DOI: 10.1016/j.colsurfb.2015.12.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/22/2015] [Accepted: 12/03/2015] [Indexed: 12/12/2022]
Abstract
Targeted delivery by ligands such as aptamers, is a promising method to increase the efficiency of PEGylated-liposomal doxorubicin (PL-Dox). In this study, we have successfully conjugated our recently developed anti-breast cancer RNA aptamer (TSA14) to the surface of PL-Dox and characterized for their size, zeta potential, Dox percent encapsulation and release properties in the presence of fetal bovine serum. In vitro experiments showed that aptamer could improve cellular uptake and cytotoxicity of PL-Dox in TUBO breast cell line. In mice bearing TUBO breast tumor, although, the doxorubicin plasma level of liposomal doxorubicin did not significantly change after modification of nanoparticles with aptamer, however, much higher tumor accumulation of Dox as compared with non-targeted liposomes proved the tumor-targeting capability of aptamers. In the same way, aptamer-PL-Dox improved anti-tumor efficiency of liposomes in TUBO breast tumor in mice compared to non-targeted liposomes. Overall, the results showed that aptamer decoration of PL-Dox could significantly improve selectivity and the therapeutic efficacy of liposomal DOX and merits further investigation.
Collapse
Affiliation(s)
- Seyedeh Alia Moosavian
- Biotechnology Research Center, Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Badiee
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Ma H, Liu J, Ali MM, Mahmood MAI, Labanieh L, Lu M, Iqbal SM, Zhang Q, Zhao W, Wan Y. Nucleic acid aptamers in cancer research, diagnosis and therapy. Chem Soc Rev 2015; 44:1240-56. [PMID: 25561050 DOI: 10.1039/c4cs00357h] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aptamers are single-stranded DNA or RNA oligomers, identified from a random sequence pool, with the ability to form unique and versatile tertiary structures that bind to cognate molecules with superior specificity. Their small size, excellent chemical stability and low immunogenicity enable them to rival antibodies in cancer imaging and therapy applications. Their facile chemical synthesis, versatility in structural design and engineering, and the ability for site-specific modifications with functional moieties make aptamers excellent recognition motifs for cancer biomarker discovery and detection. Moreover, aptamers can be selected or engineered to regulate cancer protein functions, as well as to guide anti-cancer drug design or screening. This review summarizes their applications in cancer, including cancer biomarker discovery and detection, cancer imaging, cancer therapy, and anti-cancer drug discovery. Although relevant applications are relatively new, the significant progress achieved has demonstrated that aptamers can be promising players in cancer research.
Collapse
Affiliation(s)
- Haitao Ma
- The Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Nanoparticle imaging agents for vascular pathologies are in development, and some agents are already in clinical trials. Untargeted agents, with long circulation, are excellent blood-pool agents, but molecularly targeted agents have significant advantages due to the signal enhancement possible with nanoparticle presentation of the contrast agent molecules. Molecular targets that are accessible directly from the vasculature are optimal for such agents. Targets that are removed from the vasculature, such as those on tumor cell surfaces, have limited accessibility owing to the enhanced permeation and retention effect. Yet, efforts at molecular targeting have tested small molecules, peptides, antibodies, and most recently aptamers as possible targeting ligands. The future is bright for nanoparticle-based imaging of vascular pathologies.
Collapse
Affiliation(s)
- Ananth Annapragada
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas 77030;
| |
Collapse
|
39
|
Shamay Y, Raviv L, Golan M, Voronov E, Apte RN, David A. Inhibition of primary and metastatic tumors in mice by E-selectin-targeted polymer-drug conjugates. J Control Release 2015; 217:102-12. [PMID: 26297207 DOI: 10.1016/j.jconrel.2015.08.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 12/20/2022]
Abstract
There is currently no effective means to prevent or control metastatic dissemination of cancer cells. E-selectin, an adhesion molecule expressed exclusively on inflamed and angiogenic blood vessels, plays an important role in several rate-limiting steps of cancer metastasis. In this study, we assessed the in vivo antitumor efficacy of N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers conjugated to an E-selectin binding peptide (Esbp, DITWDQLWDLMK) and equipped with the chemotherapeutic drug doxorubicin (P-(Esbp)-DOX) or with the proapoptotic peptide D(KLAKLAK)2 (P-(Esbp)-KLAK). Following a single intravenous injection, P-(Esbp)-DOX reduced tumor growth rate and prolonged the survival of mice bearing primary Lewis lung carcinoma (3LL) tumors significantly more than treatment with a non-targeted copolymer (P-DOX) or with free DOX. In an experimental B16-F10 lung metastasis model, a single intravenous dose of P-(Esbp)-DOX or P-(Esbp)-KLAK prolonged mice survival time significantly more than the non-targeted copolymers or the free drugs, and the percentage of complete tumor regression increased with increasing doses and with dosing frequency. In addition, mice pretreated with an E-selectin-targeted "drug-free" copolymer (P-(Esbp)-FITC) exhibited significantly fewer B16-F10 tumor foci in the lungs as compared with non-treated mice, demonstrating the anti-metastatic properties of the copolymer and its ability to control cancer spread through E-selectin-mediated interactions. Biodistribution analysis further confirmed the preferential accumulation of the E-selectin-targeted near-infrared fluorescently-labeled copolymer P-(Esbp)-IR783 in B16-F10 lung metastases. Taken together, this study demonstrates, for the first time, that the E-selectin targeted copolymer-drug conjugates can inhibit primary tumor growth and prevent metastases in vivo.
Collapse
Affiliation(s)
- Yosi Shamay
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lior Raviv
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moran Golan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, The Cancer Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, The Cancer Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
40
|
Varner CT, Rosen T, Martin JT, Kane RS. Recent advances in engineering polyvalent biological interactions. Biomacromolecules 2015; 16:43-55. [PMID: 25426695 PMCID: PMC4294584 DOI: 10.1021/bm5014469] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/11/2014] [Indexed: 12/21/2022]
Abstract
Polyvalent interactions, where multiple ligands and receptors interact simultaneously, are ubiquitous in nature. Synthetic polyvalent molecules, therefore, have the ability to affect biological processes ranging from protein-ligand binding to cellular signaling. In this review, we discuss recent advances in polyvalent scaffold design and applications. First, we will describe recent developments in the engineering of polyvalent scaffolds based on biomolecules and novel materials. Then, we will illustrate how polyvalent molecules are finding applications as toxin and pathogen inhibitors, targeting molecules, immune response modulators, and cellular effectors.
Collapse
Affiliation(s)
- Chad T. Varner
- The Howard P. Isermann Department
of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Tania Rosen
- The Howard P. Isermann Department
of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Jacob T. Martin
- The Howard P. Isermann Department
of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Ravi S. Kane
- The Howard P. Isermann Department
of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| |
Collapse
|
41
|
Sakurai Y, Kajimoto K, Hatakeyama H, Harashima H. Advances in an active and passive targeting to tumor and adipose tissues. Expert Opin Drug Deliv 2014; 12:41-52. [PMID: 25376864 DOI: 10.1517/17425247.2015.955847] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Data reported during the last decade of the twentieth century indicate that passive targeting is an efficient strategy for delivering nanocarrier systems to tumor tissues. The focus of this review is on active targeting as a next-generation strategy for extending the capacity of a drug delivery system (DDS). AREAS COVERED Tumor vasculature targeting was achieved using arginine- glycine-aspartic acid, asparagine-glycine-arginine and other peptides, which are well-known peptides, as ligand against tumor vasculature. An efficient system for delivering small interfering RNA to the tumor vasculature involved the use of a multifunctional envelope-type nanodevice based on a pH-modified cationic lipid and targeting ligands. The active-targeting system was extended from tumor delivery to adipose tissue delivery, where endothelial cells are tightly linked and are impermeable to nanocarriers. In mice, prohibitin-targeted nanoparticles can be used to successfully deliver macromolecules to induce anti-obese effects. Finally, the successful delivery of nanocarriers to adipose tissue in obese mice via the enhanced permeability and retention-effect is reported, which can be achieved in tumor tissue. EXPERT OPINION Unlike tumor tissues, only a few reports have appeared on how liposomal carriers accumulate in adipose tissues after systemic injection. This finding, as well as active targeting to the adipose vasculature, promises to extend the capacity of DDS to adipose tissue. Since the site of action of nucleic acids is the cytosol, the intracellular trafficking of carriers and their cargoes as well as cellular uptake must be taken into consideration.
Collapse
Affiliation(s)
- Yu Sakurai
- Hokkaido University, Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences , Kita 12, Nishi 6, Kita-ku, Sapporo, Hokkaido 060-0812 , Japan
| | | | | | | |
Collapse
|
42
|
Sun H, Zhu X, Lu PY, Rosato RR, Tan W, Zu Y. Oligonucleotide aptamers: new tools for targeted cancer therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e182. [PMID: 25093706 PMCID: PMC4221593 DOI: 10.1038/mtna.2014.32] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Aptamers are a class of small nucleic acid ligands that are composed of RNA or single-stranded DNA oligonucleotides and have high specificity and affinity for their targets. Similar to antibodies, aptamers interact with their targets by recognizing a specific three-dimensional structure and are thus termed “chemical antibodies.” In contrast to protein antibodies, aptamers offer unique chemical and biological characteristics based on their oligonucleotide properties. Hence, they are more suitable for the development of novel clinical applications. Aptamer technology has been widely investigated in various biomedical fields for biomarker discovery, in vitro diagnosis, in vivo imaging, and targeted therapy. This review will discuss the potential applications of aptamer technology as a new tool for targeted cancer therapy with emphasis on the development of aptamers that are able to specifically target cell surface biomarkers. Additionally, we will describe several approaches for the use of aptamers in targeted therapeutics, including aptamer-drug conjugation, aptamer-nanoparticle conjugation, aptamer-mediated targeted gene therapy, aptamer-mediated immunotherapy, and aptamer-mediated biotherapy.
Collapse
Affiliation(s)
- Hongguang Sun
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Xun Zhu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Jilin, China
| | | | - Roberto R Rosato
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Wen Tan
- School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
43
|
Babu A, Templeton AK, Munshi A, Ramesh R. Nanodrug delivery systems: a promising technology for detection, diagnosis, and treatment of cancer. AAPS PharmSciTech 2014; 15:709-21. [PMID: 24550101 DOI: 10.1208/s12249-014-0089-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/17/2014] [Indexed: 01/15/2023] Open
Abstract
Nanotechnology has enabled the development of novel therapeutic and diagnostic strategies, such as advances in targeted drug delivery systems, versatile molecular imaging modalities, stimulus responsive components for fabrication, and potential theranostic agents in cancer therapy. Nanoparticle modifications such as conjugation with polyethylene glycol have been used to increase the duration of nanoparticles in blood circulation and reduce renal clearance rates. Such modifications to nanoparticle fabrication are the initial steps toward clinical translation of nanoparticles. Additionally, the development of targeted drug delivery systems has substantially contributed to the therapeutic efficacy of anti-cancer drugs and cancer gene therapies compared with nontargeted conventional delivery systems. Although multifunctional nanoparticles offer numerous advantages, their complex nature imparts challenges in reproducibility and concerns of toxicity. A thorough understanding of the biological behavior of nanoparticle systems is strongly warranted prior to testing such systems in a clinical setting. Translation of novel nanodrug delivery systems from the bench to the bedside will require a collective approach. The present review focuses on recent research efforts citing relevant examples of advanced nanodrug delivery and imaging systems developed for cancer therapy. Additionally, this review highlights the newest technologies such as microfluidics and biomimetics that can aid in the development and speedy translation of nanodrug delivery systems to the clinic.
Collapse
|
44
|
Liu Q, Jin C, Wang Y, Fang X, Zhang X, Chen Z, Tan W. Aptamer-conjugated nanomaterials for specific cancer cell recognition and targeted cancer therapy. NPG ASIA MATERIALS 2014; 6:e95. [PMID: 29619132 PMCID: PMC5880215 DOI: 10.1038/am.2014.12] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Based on their unique advantages, increasing interest has been shown in the use of aptamers as target ligands for specific cancer cell recognition and targeted cancer therapy. Recently, the development of aptamer-conjugated nanomaterials has offered new therapeutic opportunities for cancer treatment with better efficacy and lower toxicity. We highlight some of the promising classes of aptamer-conjugated nanomaterials for the specific recognition of cancer cells and targeted cancer therapy. Recent developments in the use of novel strategies that enable sensitive and selective cancer cell recognition are introduced. In addition to targeted drug delivery for chemotherapy, we also review how aptamer-conjugated nanomaterials are being incorporated into emerging technologies with significant improvement in efficiency and selectivity in cancer treatment.
Collapse
Affiliation(s)
- Qiaoling Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Chen Jin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Yanyue Wang
- Departments of Chemistry, Physiology and Functional Genomics, Molecular Genetics and Microbiology, and Pathology and Laboratory Medicine, Shands Cancer Center, Center for Research at the Bio/Nano Interface, University of Florida, Gainesville, FL, USA
| | - Xiaohong Fang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- Departments of Chemistry, Physiology and Functional Genomics, Molecular Genetics and Microbiology, and Pathology and Laboratory Medicine, Shands Cancer Center, Center for Research at the Bio/Nano Interface, University of Florida, Gainesville, FL, USA
| |
Collapse
|
45
|
Liu Q, Jin C, Wang Y, Fang X, Zhang X, Chen Z, Tan W. Aptamer-conjugated nanomaterials for specific cancer cell recognition and targeted cancer therapy. NPG ASIA MATERIALS 2014; 6:e95. [PMID: 29619132 DOI: 10.1038/am.2013.75] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Based on their unique advantages, increasing interest has been shown in the use of aptamers as target ligands for specific cancer cell recognition and targeted cancer therapy. Recently, the development of aptamer-conjugated nanomaterials has offered new therapeutic opportunities for cancer treatment with better efficacy and lower toxicity. We highlight some of the promising classes of aptamer-conjugated nanomaterials for the specific recognition of cancer cells and targeted cancer therapy. Recent developments in the use of novel strategies that enable sensitive and selective cancer cell recognition are introduced. In addition to targeted drug delivery for chemotherapy, we also review how aptamer-conjugated nanomaterials are being incorporated into emerging technologies with significant improvement in efficiency and selectivity in cancer treatment.
Collapse
Affiliation(s)
- Qiaoling Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Chen Jin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Yanyue Wang
- Departments of Chemistry, Physiology and Functional Genomics, Molecular Genetics and Microbiology, and Pathology and Laboratory Medicine, Shands Cancer Center, Center for Research at the Bio/Nano Interface, University of Florida, Gainesville, FL, USA
| | - Xiaohong Fang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- Departments of Chemistry, Physiology and Functional Genomics, Molecular Genetics and Microbiology, and Pathology and Laboratory Medicine, Shands Cancer Center, Center for Research at the Bio/Nano Interface, University of Florida, Gainesville, FL, USA
| |
Collapse
|
46
|
Kong HY, Byun J. Nucleic Acid aptamers: new methods for selection, stabilization, and application in biomedical science. Biomol Ther (Seoul) 2014; 21:423-34. [PMID: 24404332 PMCID: PMC3879913 DOI: 10.4062/biomolther.2013.085] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/05/2013] [Accepted: 11/05/2013] [Indexed: 12/19/2022] Open
Abstract
The adoption of oligonucleotide aptamer is well on the rise, serving an ever increasing demand for versatility in biomedical field. Through the SELEX (Systematic Evolution of Ligands by EXponential enrichment), aptamer that can bind to specific target with high affinity and specificity can be obtained. Aptamers are single-stranded nucleic acid molecules that can fold into complex threedimensional structures, forming binding pockets and clefts for the specific recognition and tight binding of any given molecular target. Recently, aptamers have attracted much attention because they not only have all of the advantages of antibodies, but also have unique merits such as thermal stability, ease of synthesis, reversibility, and little immunogenicity. The advent of novel technologies is revolutionizing aptamer applications. Aptamers can be easily modified by various chemical reactions to introduce functional groups and/or nucleotide extensions. They can also be conjugated to therapeutic molecules such as drugs, drug containing carriers, toxins, or photosensitizers. Here, we discuss new SELEX strategies and stabilization methods as well as applications in drug delivery and molecular imaging.
Collapse
Affiliation(s)
- Hoon Young Kong
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Yongin 448-701, Republic of Korea
| | - Jonghoe Byun
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Yongin 448-701, Republic of Korea
| |
Collapse
|
47
|
Ding Y, Li S, Nie G. Nanotechnological strategies for therapeutic targeting of tumor vasculature. Nanomedicine (Lond) 2014; 8:1209-22. [PMID: 23837858 DOI: 10.2217/nnm.13.106] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neovascularization plays fundamental roles in tumor growth and metastasis. Tumor blood vessels are highly accessible and express various angiogenic markers that are either not present or are expressed at low levels in normal vessels, thereby serving as favorable targets for cancer therapy. Cancer nanotechnology, as an integrated platform, offers great opportunities for optimizing drug efficacy and pharmacokinetics while reducing side effects. Nanoparticles with tunable size, shape and surface modification have been exploited to achieve effective tumor vascular targeting. Here, we briefly introduce the signatures of tumor neovascularization and the review investigations on vascular-targeted anti-tumor nanomedicines. We also provide our perspectives on the promising fields of combination therapy and theranostic nanomedicines, as well as the challenges of nanotechnology-based cancer therapy. Furthermore, introducing new functionality would significantly consolidate the current development of nanomaterials based on tumor vasculature targeting.
Collapse
Affiliation(s)
- Yanping Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, Beijing, China
| | | | | |
Collapse
|
48
|
Thiviyanathan V, Gorenstein DG. Aptamers and the next generation of diagnostic reagents. Proteomics Clin Appl 2014; 6:563-73. [PMID: 23090891 DOI: 10.1002/prca.201200042] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/17/2012] [Accepted: 09/20/2012] [Indexed: 01/06/2023]
Abstract
Antibodies have been extensively used as capture and detection reagents in diagnostic applications of proteomics-based technologies. Proteomic assays need high sensitivity and specificity, a wide dynamic range for detection, and accurate, reproducible quantification with small confidence values. However, several inherent limitations of monoclonal antibodies in meeting the emerging challenges of proteomics led to the development of a new class of oligonucleotide-based reagents. Natural and derivatized nucleic acid aptamers are emerging as promising alternatives to monoclonal antibodies. Aptamers can be effectively used to simultaneously detect thousands of proteins in multiplex discovery platforms, where antibodies often fail due to cross-reactivity problems. Through chemical modification, vast range of additional functional groups can be added at any desired position in the oligonucleotide sequence, therefore the best features of small molecule drugs, proteins, and antibodies can be brought together into aptamers, making aptamers the most versatile reagent in proteomics. In this review, we discuss the recent developments in aptamer technology, including new selection methods and the aptamers' application in proteomics.
Collapse
Affiliation(s)
- Varatharasa Thiviyanathan
- Centers for Proteomics & Systems Biology, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, USA
| | | |
Collapse
|
49
|
Kraft JC, Freeling JP, Wang Z, Ho RJY. Emerging research and clinical development trends of liposome and lipid nanoparticle drug delivery systems. J Pharm Sci 2014; 103:29-52. [PMID: 24338748 PMCID: PMC4074410 DOI: 10.1002/jps.23773] [Citation(s) in RCA: 360] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/11/2013] [Accepted: 10/14/2013] [Indexed: 12/13/2022]
Abstract
Liposomes are spherical-enclosed membrane vesicles mainly constructed with lipids. Lipid nanoparticles are loaded with therapeutics and may not contain an enclosed bilayer. The majority of those clinically approved have diameters of 50-300 nm. The growing interest in nanomedicine has fueled lipid-drug and lipid-protein studies, which provide a foundation for developing lipid particles that improve drug potency and reduce off-target effects. Integrating advances in lipid membrane research has enabled therapeutic development. At present, about 600 clinical trials involve lipid particle drug delivery systems. Greater understanding of pharmacokinetics, biodistribution, and disposition of lipid-drug particles facilitated particle surface hydration technology (with polyethylene glycol) to reduce rapid clearance and provide sufficient blood circulation time for drug to reach target tissues and cells. Surface hydration enabled the liposome-encapsulated cancer drug doxorubicin (Doxil) to gain clinical approval in 1995. Fifteen lipidic therapeutics are now clinically approved. Although much research involves attaching lipid particles to ligands selective for occult cells and tissues, preparation procedures are often complex and pose scale-up challenges. With emerging knowledge in drug target and lipid-drug distribution in the body, a systems approach that integrates knowledge to design and scale lipid-drug particles may further advance translation of these systems to improve therapeutic safety and efficacy.
Collapse
Affiliation(s)
- John C Kraft
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | | | | |
Collapse
|
50
|
Falvo E, Tremante E, Fraioli R, Leonetti C, Zamparelli C, Boffi A, Morea V, Ceci P, Giacomini P. Antibody-drug conjugates: targeting melanoma with cisplatin encapsulated in protein-cage nanoparticles based on human ferritin. NANOSCALE 2013; 5:12278-12285. [PMID: 24150593 DOI: 10.1039/c3nr04268e] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
A novel antibody-drug conjugate (ADC) was synthesized incorporating ferritin-based nanoparticles. An average of three molecules of monoclonal antibody (mAb) Ep1 to the human melanoma-specific antigen CSPG4 were conjugated to a single ferritin cage encapsulating about 50 cisplatin molecules (HFt-Pt-Ep1). The HFt-Pt-Ep1 nanoparticle had an estimated molecular size of about 900 kD and 33 nm, and flow cytometry demonstrated specific binding to a CSPG4(+) melanoma cell line, but not to a CSPG4(-) breast carcinoma cell line. As compared to the cisplatin-containing ferritin nanoparticle alone (HFt-Pt), which inhibited thymidine incorporation more efficiently in breast carcinoma than melanoma cells, the mAb-derivatized HFt-Pt-Ep1 nanoparticle had a 25-fold preference for the latter. A similar preference for melanoma was observed upon systemic intravenous administration of HFt-Pt-Ep1 to nude mice xenotransplanted with pre-established, palpable melanoma and breast carcinoma tumors. Thus, we have been able to determine precise combinations and stoichiometric relationships between mAbs and nanoparticle protein cages, whereby the latter lose their tropism for ubiquitously distributed cellular receptors, and acquire instead remarkably lineage-selective binding. HFt-Pt-Ep1 is therefore an interesting model to improve the therapeutic index of antiblastic therapy in a tumor such as melanoma, which at its advanced stages is totally refractory to mono- and combination-chemotherapy.
Collapse
Affiliation(s)
- Elisabetta Falvo
- CNR - National Research Council of Italy, Institute of Molecular Biology and Pathology, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|