1
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
2
|
Yi J, Liu L, Gao W, Zeng J, Chen Y, Pang E, Lan M, Yu C. Advances and perspectives in phototherapy-based combination therapy for cancer treatment. J Mater Chem B 2024; 12:6285-6304. [PMID: 38895829 DOI: 10.1039/d4tb00483c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Phototherapy, including photothermal therapy (PTT) and photodynamic therapy (PDT), has the advantages of spatiotemporal selectivity, non-invasiveness, and negligible drug resistance. Phototherapy has been approved for treating superficial epidermal tumors. However, its therapeutic efficacy is limited by the hypoxic tumor microenvironment and the highly expressed heat shock protein. Moreover, poor tissue penetration and focused irradiation laser region in phototherapy make treating deep tissues and metastatic tumors challenging. Combination therapy strategies, which integrate the advantages of each treatment and overcome their disadvantages, can significantly improve the therapeutic efficacy. Recently, many combination therapy strategies have been reported. Our study summarizes the strategies used for combining phototherapy with other cancer treatments such as chemotherapy, immunotherapy, sonodynamic therapy, gas therapy, starvation therapy, and chemodynamic therapy. Some research cases were selected to analyze the combination therapy effect, delivery platform feature, and synergetic anticancer mechanisms. Moreover, additional research cases are summarized in the tables. This review provides strong evidence that phototherapy-based combination strategies can enhance the anticancer effect compared with phototherapy alone. Additionally, the challenges and future perspectives associated with these combinational therapies are discussed.
Collapse
Affiliation(s)
- Jianing Yi
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China.
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Luyao Liu
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China.
| | - Wenjie Gao
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Jie Zeng
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China.
| | - Yongzhi Chen
- Department of Hepatobiliary surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225000, China
| | - E Pang
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China.
| | - Minhuan Lan
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China.
| | - Chunzhao Yu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
3
|
Feng Y, Hu X, Zhang Y, Wang Y. The Role of Microglia in Brain Metastases: Mechanisms and Strategies. Aging Dis 2024; 15:169-185. [PMID: 37307835 PMCID: PMC10796095 DOI: 10.14336/ad.2023.0514] [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: 12/29/2022] [Accepted: 05/14/2023] [Indexed: 06/14/2023] Open
Abstract
Brain metastases and related complications are one of the major fatal factors in cancer. Patients with breast cancer, lung cancer, and melanoma are at a high risk of developing brain metastases. However, the mechanisms underlying the brain metastatic cascade remain poorly understood. Microglia, one of the major resident macrophages in the brain parenchyma, are involved in multiple processes associated with brain metastasis, including inflammation, angiogenesis, and immune modulation. They also closely interact with metastatic cancer cells, astrocytes, and other immune cells. Current therapeutic approaches against metastatic brain cancers, including small-molecule drugs, antibody-coupled drugs (ADCs), and immune-checkpoint inhibitors (ICIs), have compromised efficacy owing to the impermeability of the blood-brain barrier (BBB) and complex brain microenvironment. Targeting microglia is one of the strategies for treating metastatic brain cancer. In this review, we summarize the multifaceted roles of microglia in brain metastases and highlight them as potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Ying Feng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
4
|
Elsori D, Rashid G, Khan NA, Sachdeva P, Jindal R, Kayenat F, Sachdeva B, Kamal MA, Babker AM, Fahmy SA. Nanotube breakthroughs: unveiling the potential of carbon nanotubes as a dual therapeutic arsenal for Alzheimer's disease and brain tumors. Front Oncol 2023; 13:1265347. [PMID: 37799472 PMCID: PMC10548133 DOI: 10.3389/fonc.2023.1265347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/23/2023] [Indexed: 10/07/2023] Open
Abstract
Alzheimer's disease (AD) and brain tumors are debilitating neurological conditions that pose significant challenges in current medical practices. Existing treatment options for AD primarily focus on symptom management, and brain tumors often require aggressive therapeutic approaches. Novel disease-modifying strategies and therapeutic agents are urgently needed to address the underlying causes of AD pathogenesis and improve brain tumor management. In recent years, nanoparticles (NPs) have shown promise as valuable tools in diagnosing and managing various brain disorders, including AD. Among these, carbon nanotubes (CNTs) have garnered attention for their unique properties and biomedical potential. Their ability to cross the blood-brain barrier (BBB) with ease opens up new possibilities for targeted drug delivery and neuroprotection. This literature review aims to explore the versatile nature of CNTs, which can be functionalized with various biomolecules or substances due to their sp2 hybridization. This adaptability enables them to specifically target cells and deliver medications under specific environmental conditions. Moreover, CNTs possess an exceptional capacity to penetrate cell membranes, making them valuable tools in the treatment of AD and brain tumors. By delving into the role of CNTs in biomedicine, this review sheds light on their potential in managing AD, offering a glimpse of hope for effective disease-modifying options. Understanding the mechanisms of CNTs' action and their capabilities in targeting and delivering medication to affected cells will pave the way for innovative therapeutic strategies that can improve the lives of those afflicted with these devastating neurological conditions. The exploration of CNTs as a dual therapeutic arsenal for both brain tumors and Alzheimer's disease holds great promise and may usher in a new era of effective treatment strategies for these challenging conditions.
Collapse
Affiliation(s)
- Deena Elsori
- Faculty of Resillience, Deans Office Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Gowhar Rashid
- Amity Medical School, Amity University Gurgaon, Haryana, India
| | - Nihad Ashraf Khan
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Punya Sachdeva
- Department of Neuropyschology and Neurosciences, Amity University, Noida, UP, India
| | - Riya Jindal
- Department of Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Falak Kayenat
- Department of Biotechnology, Jamia Hamdard University, New Delhi, India
| | - Bhuvi Sachdeva
- Department of Physics and Astrophysics, Bhagini Nivedita College, University of Delhi, New Delhi, India
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Asaad Ma Babker
- Department of Medical Laboratory Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| |
Collapse
|
5
|
Zheng J, Jiang J, Pu Y, Xu T, Sun J, Zhang Q, He L, Liang X. Tumor-associated macrophages in nanomaterial-based anti-tumor therapy: as target spots or delivery platforms. Front Bioeng Biotechnol 2023; 11:1248421. [PMID: 37654704 PMCID: PMC10466823 DOI: 10.3389/fbioe.2023.1248421] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Targeting tumor-associated macrophages (TAMs) has emerged as a promising approach in cancer therapy. This article provides a comprehensive review of recent advancements in the field of nanomedicines targeting TAMs. According to the crucial role of TAMs in tumor progression, strategies to inhibit macrophage recruitment, suppress TAM survival, and transform TAM phenotypes are discussed as potential therapeutic avenues. To enhance the targeting capacity of nanomedicines, various approaches such as the use of ligands, immunoglobulins, and short peptides are explored. The utilization of live programmed macrophages, macrophage cell membrane-coated nanoparticles and macrophage-derived extracellular vesicles as drug delivery platforms is also highlighted, offering improved biocompatibility and prolonged circulation time. However, challenges remain in achieving precise targeting and controlled drug release. The heterogeneity of TAMs and the variability of surface markers pose hurdles in achieving specific recognition. Furthermore, the safety and clinical applicability of these nanomedicines requires further investigation. In conclusion, nanomedicines targeting TAMs hold great promise in cancer therapy, offering enhanced specificity and reduced side effects. Addressing the existing limitations and expanding our understanding of TAM biology will pave the way for the successful translation of these nano-therapies into clinical practice.
Collapse
Affiliation(s)
- Jixuan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jinting Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yicheng Pu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Tingrui Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiantong Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qiang Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Liang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Liu D, Dai X, Ye L, Wang H, Qian H, Cheng H, Wang X. Nanotechnology meets glioblastoma multiforme: Emerging therapeutic strategies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1838. [PMID: 35959642 DOI: 10.1002/wnan.1838] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/24/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Glioblastoma multiforme (GBM) represents the most common and fatal form of primary invasive brain tumors as it affects a great number of patients each year and has a median overall survival of approximately 14.6 months after diagnosis. Despite intensive treatment, almost all patients with GBM experience recurrence, and their 5-year survival rate is approximately 5%. At present, the main clinical treatment strategy includes surgical resection, radiotherapy, and chemotherapy. However, tumor heterogeneity, blood-brain barrier, glioma stem cells, and DNA damage repair mechanisms hinder efficient GBM treatment. The emergence of nanometer-scale diagnostic and therapeutic approaches in cancer medicine due to the establishment of nanotechnology provides novel and promising tools that will allow us to overcome these difficulties. This review summarizes the application and recent progress in nanotechnology-based monotherapies (e.g., chemotherapy) and combination cancer treatment strategies (chemotherapy-based combined cancer therapy) for GBM and describes the synergistic enhancement between these combination therapies as well as the current standard therapy for brain cancer and its deficiencies. These combination therapies that can reduce individual drug-related toxicities and significantly enhance therapeutic efficiency have recently undergone rapid development. The mechanisms underlying these different nanotechnology-based therapies as well as the application of nanotechnology in GBM (e.g., in photodynamic therapy and chemodynamic therapy) have been systematically summarized here in an attempt to review recent developments and to identify promising directions for future research. This review provides novel and clinically significant insights and directions for the treatment of GBM, which is of great clinical importance. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Dongdong Liu
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China.,Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xingliang Dai
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Koyande NP, Srivastava R, Padmakumar A, Rengan AK. Advances in Nanotechnology for Cancer Immunoprevention and Immunotherapy: A Review. Vaccines (Basel) 2022; 10:1727. [PMID: 36298592 PMCID: PMC9610880 DOI: 10.3390/vaccines10101727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 01/24/2023] Open
Abstract
One of the most effective cancer therapies, cancer immunotherapy has produced outstanding outcomes in the field of cancer treatment. However, the cost is excessive, which limits its applicability. A smart way to address this issue would be to apply the knowledge gained through immunotherapy to develop strategies for the immunoprevention of cancer. The use of cancer vaccines is one of the most popular methods of immunoprevention. This paper reviews the technologies and processes that support the advantages of cancer immunoprevention over traditional cancer immunotherapies. Nanoparticle drug delivery systems and nanoparticle-based nano-vaccines have been employed in the past for cancer immunotherapy. This paper outlines numerous immunoprevention strategies and how nanotechnology can be applied in immunoprevention. To comprehend the non-clinical and clinical evaluation of these cancer vaccines through clinical studies is essential for acceptance of the vaccines.
Collapse
Affiliation(s)
| | | | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, India
| |
Collapse
|
8
|
Dynamics of electric field-controlled methotrexate delivery through membrane nanochannels. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.118525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
9
|
Volovat SR, Ursulescu CL, Moisii LG, Volovat C, Boboc D, Scripcariu D, Amurariti F, Stefanescu C, Stolniceanu CR, Agop M, Lungulescu C, Volovat CC. The Landscape of Nanovectors for Modulation in Cancer Immunotherapy. Pharmaceutics 2022; 14:397. [PMID: 35214129 PMCID: PMC8875018 DOI: 10.3390/pharmaceutics14020397] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy represents a promising strategy for the treatment of cancer, which functions via the reprogramming and activation of antitumor immunity. However, adverse events resulting from immunotherapy that are related to the low specificity of tumor cell-targeting represent a limitation of immunotherapy's efficacy. The potential of nanotechnologies is represented by the possibilities of immunotherapeutical agents being carried by nanoparticles with various material types, shapes, sizes, coated ligands, associated loading methods, hydrophilicities, elasticities, and biocompatibilities. In this review, the principal types of nanovectors (nanopharmaceutics and bioinspired nanoparticles) are summarized along with the shortcomings in nanoparticle delivery and the main factors that modulate efficacy (the EPR effect, protein coronas, and microbiota). The mechanisms by which nanovectors can target cancer cells, the tumor immune microenvironment (TIME), and the peripheral immune system are also presented. A possible mathematical model for the cellular communication mechanisms related to exosomes as nanocarriers is proposed.
Collapse
Affiliation(s)
- Simona-Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Corina Lupascu Ursulescu
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| | - Liliana Gheorghe Moisii
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
- Department of Medical Oncology, “Euroclinic” Center of Oncology, 2 Vasile Conta Str., 700106 Iaşi, Romania
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Dragos Scripcariu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania;
| | - Florin Amurariti
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (S.-R.V.); (D.B.); (F.A.)
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.S.); (C.R.S.)
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.S.); (C.R.S.)
| | - Maricel Agop
- Physics Department, “Gheorghe Asachi” Technical University, Prof. Dr. Docent Dimitrie Mangeron Rd., No. 59A, 700050 Iaşi, Romania;
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Cristian Constantin Volovat
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iaşi, Romania; (C.L.U.); (L.G.M.); (C.C.V.)
| |
Collapse
|
10
|
|
11
|
Ye T, Li F, Ma G, Wei W. Enhancing therapeutic performance of personalized cancer vaccine via delivery vectors. Adv Drug Deliv Rev 2021; 177:113927. [PMID: 34403752 DOI: 10.1016/j.addr.2021.113927] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/29/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022]
Abstract
In recent years, personalized cancer vaccines have gained increasing attention as emerging immunotherapies with the capability to overcome interindividual differences and show great benefits for individual patients in the clinic due to the highly tailored vaccine formulations. A large number of materials have been studied as delivery vectors to enhance the therapeutic performance of personalized cancer vaccines, including artificial materials, engineered microorganisms, cells and cell derivatives. These delivery vectors with distinct features are employed to change antigen biodistributions and to facilitate antigen uptake, processing and presentation, improving the strength, velocity, and duration of the immune response when delivered by different strategies. Here, we provide an overview of personalized cancer vaccine delivery vectors, describing their materials, physicochemical properties, delivery strategies and challenges for clinical transformation.
Collapse
|
12
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
13
|
Jin H, Gao S, Song D, Liu Y, Chen X. Intratumorally CpG immunotherapy with carbon nanotubes inhibits local tumor growth and liver metastasis by suppressing the epithelial-mesenchymal transition of colon cancer cells. Anticancer Drugs 2021; 32:278-285. [PMID: 32976213 PMCID: PMC7861496 DOI: 10.1097/cad.0000000000001000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/26/2020] [Indexed: 02/07/2023]
Abstract
Colon cancer liver metastasis accounts for the major cause of death of colon cancer patients. Previous study reported a carbon nanotubes (CNT)-conjugated CpG complex (CNT-CpG), which displayed a significant antitumor effect in gliomas. However, whether CNT-CpG could limit colon tumor growth and suppress the colon cancer liver metastasis has not been evaluated. In this study, we report CNT enhances CpG uptake in mouse colon cancer cells. Results demonstrated only CpG with CNT conjugation showed significant activation of NF-κB signal. Moreover, intratumorally delivery of CNT-CpG successfully suppressed local xenograft tumor growth and liver metastasis. CNT-CpG treatments cured 75% of mice and inhibited local tumor growth, significantly prolonged survival outcomes and limited liver metastatic tumor nodules from colon cancer cells. Using human colon cancer cell line, HCT116, we observed significantly inhibitory effects of CNT-CpG on cell growth, invasion and migration. Importantly, CNT-CpG treatment blocked the epithelial to mesenchymal transition (EMT). We compared the mRNA levels of EMT markers of colon cancer cells without or with CNT-CpG treatment from in-vitro and in-vivo models. Consistent results demonstrated expression of epithelial marker, E-cadherin was upregulated by CNT-CpG. In contrast, three mesenchymal markers, snail, fibronectin and vimentin were significantly suppressed by CNT-CpG treatment compared with control or free CpG. In summary, our data suggest CNT-CpG is an effective therapeutic approach against local colon tumor and their liver metastasis. This study presents the CNT-CpG complex as a promising therapeutic target for developing novel therapies against both local colon tumors and liver metastatic tumors.
Collapse
Affiliation(s)
| | - Sujie Gao
- Department of Anesthesia, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province
| | | | - Yiting Liu
- Department of Radiology, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, People’s Republic of China
| | | |
Collapse
|
14
|
Nonsurgical treatment of skin cancer with local delivery of bioadhesive nanoparticles. Proc Natl Acad Sci U S A 2021; 118:2020575118. [PMID: 33526595 DOI: 10.1073/pnas.2020575118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Keratinocyte-derived carcinomas, including squamous cell carcinoma (SCC), comprise the most common malignancies. Surgical excision is the therapeutic standard but is not always clinically feasible, and currently available alternatives are limited to superficial tumors. To address the need for a nonsurgical treatment for nodular skin cancers like SCC, we developed a bioadhesive nanoparticle (BNP) drug delivery system composed of biodegradable polymer, poly(lactic acid)-hyperbranched polyglycerol (PLA-HPG), encapsulating camptothecin (CPT). Nanoparticles (NPs) of PLA-HPG are nonadhesive NPs (NNPs), which are stealthy in their native state, but we have previously shown that conversion of the vicinal diols of HPG to aldehydes conferred NPs the ability to form strong covalent bonds with amine-rich surfaces. Herein, we show that these BNPs have significantly enhanced binding to SCC tumor cell surfaces and matrix proteins, thereby significantly enhancing the therapeutic efficacy of intratumoral drug delivery. Tumor injection of BNP-CPT resulted in tumor retention of CPT at ∼50% at 10 d postinjection, while CPT was undetectable in NNP-CPT or free (intralipid) CPT-injected tumors at that time. BNP-CPT also significantly reduced tumor burden, with a portion (∼20%) of BNP-CPT-treated established tumors showing histologic cure. Larger, more fully established PDV SCC tumors treated with a combination of BNP-CPT and immunostimulating CpG oligodeoxynucleotides exhibited enhanced survival relative to controls, revealing the potential for BNP delivery to be used along with local tumor immunotherapy. Taken together, these results indicate that percutaneous delivery of a chemotherapeutic agent via BNPs, with or without adjuvant immunostimulation, represents a viable, nonsurgical alternative for treating cutaneous malignancy.
Collapse
|
15
|
Burn OK, Prasit KK, Hermans IF. Modulating the Tumour Microenvironment by Intratumoural Injection of Pattern Recognition Receptor Agonists. Cancers (Basel) 2020; 12:E3824. [PMID: 33352882 PMCID: PMC7765936 DOI: 10.3390/cancers12123824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Signalling through pattern recognition receptors (PRRs) leads to strong proinflammatory responses, enhancing the activity of antigen presenting cells and shaping adaptive immune responses against tumour associated antigens. Unfortunately, toxicities associated with systemic administration of these agonists have limited their clinical use to date. Direct injection of PRR agonists into the tumour can enhance immune responses by directly modulating the cells present in the tumour microenvironment. This can improve local antitumour activity, but importantly, also facilitates systemic responses that limit tumour growth at distant sites. As such, this form of therapy could be used clinically where metastatic tumour lesions are accessible, or as neoadjuvant therapy. In this review, we summarise current preclinical data on intratumoural administration of PRR agonists, including new strategies to optimise delivery and impact, and combination studies with current and promising new cancer therapies.
Collapse
Affiliation(s)
- Olivia K. Burn
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| | - Kef K. Prasit
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| |
Collapse
|
16
|
|
17
|
Far-reaching advances in the role of carbon nanotubes in cancer therapy. Life Sci 2020; 257:118059. [PMID: 32659368 DOI: 10.1016/j.lfs.2020.118059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022]
Abstract
Cancer includes a group of diseases involving unregulated cell growth with the potential to invade or expand to other parts of the body, resulting in an estimate of 9.6 million deaths worldwide in 2018. Manifold studies have been conducted to design more efficacious techniques for cancer therapy due to the inadequacy of conventional treatments including chemotherapy, surgery, and radiation therapy. With the advances in the biomedical applications of nanotechnology-based systems, nanomaterials have gained increasing attention as promising vehicles for targeted cancer therapy and optimizing treatment outcomes. Owing to their outstanding thermal, electrical, optical and chemical properties, carbon nanotubes (CNTs) have been profoundly studied to explore the various perspectives of their application in cancer treatment. The current study aims to review the role of CNTs whether as a carrier or mediator in cancer treatment for enhancing the efficacy as well as the specificity of therapy and reducing adverse side effects. This comprehensive review indicates that CNTs have the capability to be the next generation nanomaterials to actualize noninvasive targeted eradication of tumors. However, further studies are needed to evaluate the consequences of their biomedical application before the transition into clinical trials, since possible adverse effects of CNTs on biological systems have not been clearly understood.
Collapse
|
18
|
Wang X, Li B, Jing H, Dong X, Leng X. MWCNT-mediated combinatorial photothermal ablation and chemo-immunotherapy strategy for the treatment of melanoma. J Mater Chem B 2020; 8:4245-4258. [DOI: 10.1039/c9tb02238d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
DOX and CpG loaded MWCNT with NIR irradiation could destroy tumor cells by photothermal and chemotherapy and release tumor-associated antigens, thus generating melanoma specific immune response to achieve synergistic therapeutic effect.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Tianjin Key Laboratory of Biomedical Materials
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Tianjin 300192
- P. R. China
| | - Binhan Li
- Tianjin Key Laboratory of Biomedical Materials
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Tianjin 300192
- P. R. China
| | - Huimin Jing
- Tianjin Key Laboratory of Biomedical Materials
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Tianjin 300192
- P. R. China
| | - Xia Dong
- Tianjin Key Laboratory of Biomedical Materials
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Tianjin 300192
- P. R. China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomedical Materials
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Tianjin 300192
- P. R. China
| |
Collapse
|
19
|
Ovais M, Guo M, Chen C. Tailoring Nanomaterials for Targeting Tumor-Associated Macrophages. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1808303. [PMID: 30883982 DOI: 10.1002/adma.201808303] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/07/2019] [Indexed: 05/17/2023]
Abstract
Advances in the field of nanotechnology together with an increase understanding of tumor immunology have paved the way for the development of more personalized cancer immuno-nanomedicines. Nanovehicles, due to their specific physicochemical properties, are emerging as key translational moieties in tackling tumor-promoting, M2-like tumor-associated macrophages (TAMs). Cancer immuno-nanomedicines target TAMs primarily by blocking M2-like TAM survival or affecting their signaling cascades, restricting macrophage recruitment to tumors and re-educating tumor-promoting M2-like TAMs to the tumoricidal, M1-like phenotype. Here, the TAM effector mechanisms and strategies for targeting TAMs are summarized, followed by a focus on the mechanistic considerations in the development of novel immuno-nanomedicines. Furthermore, imaging TAMs with nanoparticles so as to forecast a patient's clinical outcome, describing treatment options, and observing therapy responses is also discussed. At present, strategies that target TAMs are being investigated not only at the basic research level but also in early clinical trials. The significance of TAM-targeting biomaterials is highlighted, with the goal of facilitating future clinical translation.
Collapse
Affiliation(s)
- Muhammad Ovais
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
20
|
Benbenishty A, Gadrich M, Cottarelli A, Lubart A, Kain D, Amer M, Shaashua L, Glasner A, Erez N, Agalliu D, Mayo L, Ben-Eliyahu S, Blinder P. Prophylactic TLR9 stimulation reduces brain metastasis through microglia activation. PLoS Biol 2019; 17:e2006859. [PMID: 30921319 PMCID: PMC6469801 DOI: 10.1371/journal.pbio.2006859] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 04/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.
Collapse
Affiliation(s)
- Amit Benbenishty
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Meital Gadrich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Azzurra Cottarelli
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Alisa Lubart
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - David Kain
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
| | - Malak Amer
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lee Shaashua
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ariella Glasner
- The Lautenberg Centre for General and Tumor Immunology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Lior Mayo
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Pablo Blinder
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
21
|
Kadiyala P, Li D, Nuñez FM, Altshuler D, Doherty R, Kuai R, Yu M, Kamran N, Edwards M, Moon JJ, Lowenstein PR, Castro MG, Schwendeman A. High-Density Lipoprotein-Mimicking Nanodiscs for Chemo-immunotherapy against Glioblastoma Multiforme. ACS NANO 2019; 13:1365-1384. [PMID: 30721028 PMCID: PMC6484828 DOI: 10.1021/acsnano.8b06842] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive primary brain tumor, for which there is no cure. Treatment effectiveness for GBM has been limited due to tumor heterogeneity, an immunosuppressive tumor microenvironment (TME), and the presence of the blood-brain barrier, which hampers the transport of chemotherapeutic compounds to the central nervous system (CNS). High-density lipoprotein (HDL)-mimicking nanodiscs hold considerable promise to achieve delivery of bioactive compounds into tumors. Herein, we tested the ability of synthetic HDL nanodiscs to deliver chemotherapeutic agents to the GBM microenvironment and elicit tumor regression. To this end, we developed chemo-immunotherapy delivery vehicles based on sHDL nanodiscs loaded with CpG, a Toll-like receptor 9 (TLR9) agonist, together with docetaxel (DTX), a chemotherapeutic agent, for targeting GBM. Our data show that delivery of DTX-sHDL-CpG nanodiscs into the tumor mass elicited tumor regression and antitumor CD8+ T cell responses in the brain TME. We did not observe any overt off-target side effects. Furthermore, the combination of DTX-sHDL-CpG treatment with radiation (IR), which is the standard of care for GBM, resulted in tumor regression and long-term survival in 80% of GBM-bearing animals. Mice remained tumor-free upon tumor cell rechallenge in the contralateral hemisphere, indicating the development of anti-GBM immunological memory. Collectively, these data indicate that sHDL nanodiscs constitute an effective drug delivery platform for the treatment of GBM, resulting in tumor regression, long-term survival, and immunological memory when used in combination with IR. The proposed delivery platform has significant potential for clinical translation.
Collapse
Affiliation(s)
- Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fernando M. Nuñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David Altshuler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Robert Doherty
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Neha Kamran
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Lead Contacts
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Lead Contacts
| |
Collapse
|
22
|
Zhang R, Billingsley MM, Mitchell MJ. Biomaterials for vaccine-based cancer immunotherapy. J Control Release 2018; 292:256-276. [PMID: 30312721 PMCID: PMC6355332 DOI: 10.1016/j.jconrel.2018.10.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 12/28/2022]
Abstract
The development of therapeutic cancer vaccines as a means to generate immune reactivity against tumors has been explored since the early discovery of tumor-specific antigens by Georg Klein in the 1960s. However, challenges including weak immunogenicity, systemic toxicity, and off-target effects of cancer vaccines remain as barriers to their broad clinical translation. Advances in the design and implementation of biomaterials are now enabling enhanced efficacy and reduced toxicity of cancer vaccines by controlling the presentation and release of vaccine components to immune cells and their microenvironment. Here, we discuss the rational design and clinical status of several classes of cancer vaccines (including DNA, mRNA, peptide/protein, and cell-based vaccines) along with novel biomaterial-based delivery technologies that improve their safety and efficacy. Further, strategies for designing new platforms for personalized cancer vaccines are also considered.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
23
|
Saleem J, Wang L, Chen C. Carbon-Based Nanomaterials for Cancer Therapy via Targeting Tumor Microenvironment. Adv Healthc Mater 2018; 7:e1800525. [PMID: 30073803 DOI: 10.1002/adhm.201800525] [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: 05/14/2018] [Revised: 07/04/2018] [Indexed: 12/12/2022]
Abstract
Cancer remains one of the major health problems all over the world and conventional therapeutic approaches have failed to attain an effective cure. Tumor microenvironments (TME) present a unique challenge in tumor therapy due to their complex structures and multiple components, which also serve as the soil for tumor growth, development, invasion, and migration. The complex TME includes immune cells, fibrous collagen structures, and tortuous blood vessels, in which conventional therapeutic approaches are rendered useless. State-of-the-art nanotechnologies have potential to cope with the threats of malignant tumors. With unique physiochemical properties, carbon nanomaterials (CNMs), including graphene, fullerenes, carbon nanotubes, and carbon quantum dots, offer opportunities to resolve the hurdles, by targeting not only cancer cells but also the TME. This review summarizes the progress about CNM-based cancer therapy strategies, which mainly focuses on both the treatment for cancer cells and TME-targeted modulation. In the last, the challenges for TME-based therapy via CNMs are discussed, which will be important in guiding current basic research to clinical translation in the future.
Collapse
Affiliation(s)
- Jabran Saleem
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology of China; Beijing 100190 P. R. China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Chunying Chen
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology of China; Beijing 100190 P. R. China
| |
Collapse
|
24
|
Gao H, Zhang IY, Zhang L, Song Y, Liu S, Ren H, Liu H, Zhou H, Su Y, Yang Y, Badie B. S100B suppression alters polarization of infiltrating myeloid-derived cells in gliomas and inhibits tumor growth. Cancer Lett 2018; 439:91-100. [PMID: 30076898 PMCID: PMC7048242 DOI: 10.1016/j.canlet.2018.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 01/03/2023]
Abstract
S100B, a member of the multigene family of Ca2+-binding proteins, is overexpressed by most malignant gliomas but its biological role in gliomagenesis is unclear. Recently, we demonstrated that low concentrations of S100B attenuated microglia activation through the induction of STAT3. Furthermore, S100B downregulation in a murine glioma model inhibited macrophage trafficking and tumor growth. Based on these observations, we hypothesized that S100B inhibitors may have antiglioma properties through modulation of tumor microenvironment. To discover novel S100B inhibitors, we developed a high-throughput screening cell-based S100B promoter-driven luciferase reporter assay. Initial screening of 768 compounds in the NIH library identified 36 hits with >85% S100B inhibitory activity. Duloxetine (Dul, an SNRI) was selected for the initial proof-of-concept studies. At low concentrations (1–5 μM) Dul inhibited S100B and CCL2 production in mouse GL261 glioma cells, but had minimal cytotoxic activity in vitro. In vivo, however, Dul (30 mg/kg/14 days) inhibited S100B production, altered the polarization and trafficking of tumor-associated myeloid-derived cells, and inhibited the growth of intracranial GL261 gliomas. Dul therapeutic efficacy, however, was not observed in the K-Luc glioma model that expresses low levels of S100B. These findings affirm the role of S100B in gliomagenesis and justify the development of more potent S100B inhibitors for glioma therapy.
Collapse
Affiliation(s)
- Hang Gao
- Department of Bone and Joint Surgery, No.1 Hospital of Jilin University, Changchun, Jilin Province, PR China.
| | - Ian Y Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, USA.
| | - Leying Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, USA.
| | - Yanyan Song
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin Province, PR China.
| | - Shunan Liu
- Department of Pharmacology, The Pharmacy School of Jilin University, Changchun, Jilin Province, PR China.
| | - Hui Ren
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, PR China.
| | - Huili Liu
- Division of Neurosurgery, City of Hope Beckman Research Institute, USA.
| | - Hui Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang Province, PR China.
| | - Yanping Su
- College of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, PR China.
| | - Yihang Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, PR China.
| | - Behnam Badie
- Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA.
| |
Collapse
|
25
|
Alizadeh D, White EE, Sanchez TC, Liu S, Zhang L, Badie B, Berlin JM. Immunostimulatory CpG on Carbon Nanotubes Selectively Inhibits Migration of Brain Tumor Cells. Bioconjug Chem 2018. [PMID: 29526082 DOI: 10.1021/acs.bioconjchem.8b00146] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Even when treated with aggressive current therapies, patients with glioblastoma usually survive less than two years and exhibit a high rate of recurrence. CpG is an oligonucleotide that activates the innate immune system via Toll-like receptor 9 (TLR9) activation. Injection of CpG into glioblastoma tumors showed promise as an immunotherapy in mouse models but proved disappointing in human trials. One aspect of glioma that is not addressed by CpG therapy alone is the highly invasive nature of glioma cells, which is associated with resistance to radiation and chemotherapy. Here, we demonstrate that single-walled carbon nanotubes noncovalently functionalized with CpG (SWNT/CpG), which retain the immunostimulatory property of the CpG, selectively inhibit the migration of glioma cells and not macrophages without affecting cell viability or proliferation. SWNT/CpG also selectively decreased NF-κB activation in glioma cells, while activating macrophages by induction of the TLR9/NF-κB pathway, as we have previously reported. The migration inhibition of glioma cells was correlated with selective reduction of intracellular levels of reactive oxygen species (ROS), suggesting that an antioxidant-based mechanism mediates the observed effects. To the best of our knowledge, SWNT/CpG is the first nanomaterial that inhibits the migration of cancer cells while stimulating the immune system.
Collapse
|
26
|
Synthetic Poly(L-Glutamic Acid)-conjugated CpG Exhibits Antitumor Efficacy With Increased Retention in Tumor and Draining Lymph Nodes After Intratumoral Injection in a Mouse Model of Melanoma. J Immunother 2018; 40:11-20. [PMID: 27681378 DOI: 10.1097/cji.0000000000000145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is an urgent need for new clinically applicable drug-delivery methods to enhance accumulation of immune-activating drugs in tumors. We synthesized a poly(L-glutamic acid)-CpG ODN2216 conjugate (PG-CpG) and injected it intratumorally into C57BL/6 mice bearing subcutaneous B16-ovalbumin melanoma. PG-CpG elicited the same potent antitumoral activity as CpG with respect to reducing tumor growth and triggering antigen-specific CD8 T-cell responses in this well-established solid tumor model. Moreover, PG-CpG was retained significantly longer in both tumor and draining lymph nodes than was free CpG after intratumoral injection. Specifically, 48 hours after injection, 26.5%±16.9% of the injected PG-CpG dose versus 4.72%±2.61% of free CpG remained at the tumor, and 1.53%±1.22% of the injected PG-CpG versus 0.37%±0.33% of free CpG was retained in the draining inguinal lymph nodes. These findings indicate that PG is an effective synthetic polymeric carrier for delivery of immunostimulatory agents to tumors and lymph nodes.
Collapse
|
27
|
Binnemars-Postma K, Storm G, Prakash J. Nanomedicine Strategies to Target Tumor-Associated Macrophages. Int J Mol Sci 2017; 18:E979. [PMID: 28471401 PMCID: PMC5454892 DOI: 10.3390/ijms18050979] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
In recent years, the influence of the tumor microenvironment (TME) on cancer progression has been better understood. Macrophages, one of the most important cell types in the TME, exist in different subtypes, each of which has a different function. While classically activated M1 macrophages are involved in inflammatory and malignant processes, activated M2 macrophages are more involved in the wound-healing processes occurring in tumors. Tumor-associated macrophages (TAM) display M2 macrophage characteristics and support tumor growth and metastasis by matrix remodeling, neo-angiogenesis, and suppressing local immunity. Due to their detrimental role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer may prove to be beneficial in the treatment of cancer. Due to the plastic nature of macrophages, their activities may be altered to inhibit tumor growth. In this review, we will discuss the therapeutic options for the modulation and targeting of TAM. Different therapeutic strategies to deplete, inhibit recruitment of, or re-educate TAM will be discussed. Current strategies for the targeting of TAM using nanomedicine are reviewed. Passive targeting using different nanoparticle systems is described. Since TAM display a number of upregulated surface proteins compared to non-TAM, specific targeting using targeting ligands coupled to nanoparticles is discussed in detail.
Collapse
Affiliation(s)
- Karin Binnemars-Postma
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| | - Gert Storm
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
- Department of Pharmaceutics, Utrecht University, 3584CS Utrecht, The Netherlands.
| | - Jai Prakash
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| |
Collapse
|
28
|
Kumar S, Rani R, Dilbaghi N, Tankeshwar K, Kim KH. Carbon nanotubes: a novel material for multifaceted applications in human healthcare. Chem Soc Rev 2017; 46:158-196. [DOI: 10.1039/c6cs00517a] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Remarkable advances achieved in modern material technology, especially in device fabrication, have facilitated diverse materials to expand the list of their application fields.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Bio and Nano Technology
- Guru Jambheshwar University of Science and Technology
- Hisar
- India
| | - Ruma Rani
- Department of Bio and Nano Technology
- Guru Jambheshwar University of Science and Technology
- Hisar
- India
| | - Neeraj Dilbaghi
- Department of Bio and Nano Technology
- Guru Jambheshwar University of Science and Technology
- Hisar
- India
| | - K. Tankeshwar
- Department of Bio and Nano Technology
- Guru Jambheshwar University of Science and Technology
- Hisar
- India
- Department of Physics
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering
- Hanyang University
- Seoul 04763
- Republic of Korea
| |
Collapse
|
29
|
Abstract
Carbon nanotubes (CNTs) have received increasing attention in biomedical fields because of their unique structures and properties, including high aspect ratios, large surface areas, rich surface chemical functionalities, and size stability on the nanoscale. Particularly, they are attractive as carriers and mediators for cancer therapy. Through appropriate functionalization, CNTs have been used as nanocarriers for anticancer drugs including doxorubicin, camptothecin, carboplatin, cisplatin, paclitaxel, Pt(II), and Pt(IV), and genes including plasmid DNA, small-interfering RNA, oligonucleotides, and RNA/DNA aptamers. CNTs can also deliver proteins and immunotherapy components. Using combinations of light energy, they have also been applied as mediators for photothermal therapy and photodynamic therapy to directly destroy cancer cells without severely damaging normal tissue. If limitations such as a long-term cytotoxicity in the body, lack of size uniformity during the synthetic process, loading deviations for drug–CNT complexes, and release controllability at the target point are overcome, CNTs will become one of the strongest tools that are available for various other biomedical fields as well as for cancer therapy.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center
| | | | - Jin Woo Lee
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
30
|
Hassan HAFM, Smyth L, Wang JTW, Costa PM, Ratnasothy K, Diebold SS, Lombardi G, Al-Jamal KT. Dual stimulation of antigen presenting cells using carbon nanotube-based vaccine delivery system for cancer immunotherapy. Biomaterials 2016; 104:310-22. [PMID: 27475727 PMCID: PMC4993816 DOI: 10.1016/j.biomaterials.2016.07.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 06/23/2016] [Accepted: 07/05/2016] [Indexed: 12/14/2022]
Abstract
Although anti-cancer immuno-based combinatorial therapeutic approaches have shown promising results, efficient tumour eradication demands further intensification of anti-tumour immune response. With the emerging field of nanovaccinology, multi-walled carbon nanotubes (MWNTs) have manifested prominent potentials as tumour antigen nanocarriers. Nevertheless, the utilization of MWNTs in co-delivering antigen along with different types of immunoadjuvants to antigen presenting cells (APCs) has not been investigated yet. We hypothesized that harnessing MWNT for concurrent delivery of cytosine-phosphate-guanine oligodeoxynucleotide (CpG) and anti-CD40 Ig (αCD40), as immunoadjuvants, along with the model antigen ovalbumin (OVA) could potentiate immune response induced against OVA-expressing tumour cells. We initially investigated the effective method to co-deliver OVA and CpG using MWNT to the APC. Covalent conjugation of OVA and CpG prior to loading onto MWNTs markedly augmented the CpG-mediated adjuvanticity, as demonstrated by the significantly increased OVA-specific T cell responses in vitro and in C57BL/6 mice. αCD40 was then included as a second immunoadjuvant to further intensify the immune response. Immune response elicited in vitro and in vivo by OVA, CpG and αCD40 was significantly potentiated by their co-incorporation onto the MWNTs. Furthermore, MWNT remarkably improved the ability of co-loaded OVA, CpG and αCD40 in inhibiting the growth of OVA-expressing B16F10 melanoma cells in subcutaneous or lung pseudo-metastatic tumour models. Therefore, this study suggests that the utilization of MWNTs for the co-delivery of tumour-derived antigen, CpG and αCD40 could be a competent approach for efficient tumours eradication.
Collapse
Affiliation(s)
- Hatem A F M Hassan
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Lesley Smyth
- Immunoregulation Laboratory, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Julie T-W Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Pedro M Costa
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Kulachelvy Ratnasothy
- Immunoregulation Laboratory, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sandra S Diebold
- Division of Immunology, Infection, and Inflammatory Diseases, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom.
| |
Collapse
|
31
|
Zhang H, Gao XD. Nanodelivery systems for enhancing the immunostimulatory effect of CpG oligodeoxynucleotides. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 70:935-946. [PMID: 27772724 DOI: 10.1016/j.msec.2016.03.045] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/29/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022]
Abstract
Synthetic oligodeoxynucleotides containing immunostimulatory CpG motif mimic bacterial DNA and are potent activator of innate and adaptive immune responses. Therefore, CpG ODNs have significant potentials as immunotherapeutic agent for treatment of infectious diseases, allergy and cancer. Many clinical trials involving CpG ODNs either used alone or as adjuvant have been initiated. However, delivery of CpG ODNs to target sites still remains a great challenge due to their extreme susceptibility to nuclease degradation in serum and poor cellular uptake. Chemical modification of CpG ODNs backbone can protect them against degradation by nucleases, but have raised concern regarding several severe side effects. Development of efficient CpG ODNs delivery systems to address these issues and enhance their immunostimulatory effect are highly desirable. In recent years, the emergence of nanotechnology has provided unprecedented opportunities to encapsulate CpG ODN into various nanocarriers or synthesize CpG ODNs nanostructures. This review provides an overview of the delivery systems based on nanomaterials and nanostructures newly developed for enhancing the immunostimulatory effect of CpG ODNs, together with a brief discussion on perspectives for future studies in this field.
Collapse
Affiliation(s)
- Huijie Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
32
|
Toll-like receptor-5 agonist, entolimod, suppresses metastasis and induces immunity by stimulating an NK-dendritic-CD8+ T-cell axis. Proc Natl Acad Sci U S A 2016; 113:E874-83. [PMID: 26831100 DOI: 10.1073/pnas.1521359113] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Activation of an anticancer innate immune response is highly desirable because of its inherent ability to generate an adaptive antitumor T-cell response. However, insufficient safety of innate immune modulators limits clinical use to topical applications. Toll-like receptor 5 (TLR5) agonists are favorably positioned as potential systemic immunotherapeutic agents because of unusual tissue specificity of expression, uniquely safe profile of induced cytokines, and antitumor efficacy demonstrated in a number of animal models. Here, we decipher the molecular and cellular events underlying the metastasis suppressive activity of entolimod, a clinical stage TLR5 agonist that activates NF-κB-, AP-1-, and STAT3-driven immunomodulatory signaling pathways specifically within the liver. Used as a single agent in murine colon and mammary metastatic cancer models, entolimod rapidly induces CXCL9 and -10 that support homing of blood-borne CXCR3-expressing NK cells to the liver predominantly through an IFN-γ signaling independent mechanism. NK cell-dependent activation of dendritic cells is followed by stimulation of a CD8(+) T-cell response, which exert both antimetastatic effect of entolimod and establishment of tumor-specific and durable immune memory. These results define systemically administered TLR5 agonists as organ-specific immunoadjuvants, enabling efficient antitumor vaccination that does not depend on identification of tumor-specific antigens.
Collapse
|
33
|
Ouyang M, White EE, Ren H, Guo Q, Zhang I, Gao H, Yanyan S, Chen X, Weng Y, Da Fonseca A, Shah S, Manuel ER, Zhang L, Vonderfecht SL, Alizadeh D, Berlin JM, Badie B. Metronomic Doses of Temozolomide Enhance the Efficacy of Carbon Nanotube CpG Immunotherapy in an Invasive Glioma Model. PLoS One 2016; 11:e0148139. [PMID: 26829221 PMCID: PMC4734656 DOI: 10.1371/journal.pone.0148139] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/13/2016] [Indexed: 12/25/2022] Open
Abstract
Even when treated with aggressive current therapies, most patients with glioblastoma survive less than two years. Rapid tumor growth, an invasive nature, and the blood-brain barrier, which limits the penetration of large molecules into the brain, all contribute to the poor tumor response associated with conventional therapies. Immunotherapy has emerged as a therapeutic approach that may overcome these challenges. We recently reported that single-walled carbon nanotubes (SWCNTs) can be used to dramatically increase the immunotherapeutic efficacy of CpG oligonucleotides in a mouse model of glioma. Following implantation in the mouse brain, the tumor cell line used in these previous studies (GL261) tends to form a spherical tumor with limited invasion into healthy brain. In order to evaluate SWCNT/CpG therapy under more clinically-relevant conditions, here we report the treatment of a more invasive mouse glioma model (K-Luc) that better recapitulates human disease. In addition, a CpG sequence previously tested in humans was used to formulate the SWCNT/CpG which was combined with temozolomide, the standard of care chemotherapy for glioblastoma patients. We found that, following two intracranial administrations, SWCNT/CpG is well-tolerated and improves the survival of mice bearing invasive gliomas. Interestingly, the efficacy of SWCNT/CpG was enhanced when combined with temozolomide. This enhanced anti-tumor efficacy was correlated to an increase of tumor-specific cytotoxic activity in splenocytes. These results reinforce the emerging understanding that immunotherapy can be enhanced by combining it with chemotherapy and support the continued development of SWCNT/CpG.
Collapse
Affiliation(s)
- Mao Ouyang
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha Hunan, P.R. China
| | - Ethan E. White
- Irell & Manella Graduate School of Biological Sciences at City of Hope, Duarte, California, 91010, United States of America
- Department of Molecular Medicine, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Hui Ren
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, P.R. China
| | - Qin Guo
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha Hunan, P.R. China
| | - Ian Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Hang Gao
- Department of Bone and Joint Surgery, No.1 Hospital of Jilin University, Changchun, Jilin Province, P.R. China
| | - Song Yanyan
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin Province, P.R. China
| | - Xuebo Chen
- Department of General Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin Province, P.R. China
| | - Yiming Weng
- Department of Molecular Medicine, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Anna Da Fonseca
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sunny Shah
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Edwin R. Manuel
- Division of Translational Vaccine Research, Department of Virology, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Leying Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Steven L. Vonderfecht
- Division of Comparative Medicine, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Darya Alizadeh
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
| | - Jacob M. Berlin
- Irell & Manella Graduate School of Biological Sciences at City of Hope, Duarte, California, 91010, United States of America
- Department of Molecular Medicine, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
- * E-mail: (BB); (JB)
| | - Behnam Badie
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
- Department of Cancer Immunotherapeutics & Tumor Immunology City of Hope Beckman Research Institute, Duarte, California, 91010, United States of America
- * E-mail: (BB); (JB)
| |
Collapse
|
34
|
Current Perspective of Carbon Nanotubes Application in Neurology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 130:229-63. [DOI: 10.1016/bs.irn.2016.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Zhang F, Lin YA, Kannan S, Kannan RM. Targeting specific cells in the brain with nanomedicines for CNS therapies. J Control Release 2015; 240:212-226. [PMID: 26686078 DOI: 10.1016/j.jconrel.2015.12.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Treatment of Central Nervous System (CNS) disorders still remains a major clinical challenge. The Blood-Brain Barrier (BBB), known as the major hindrance, greatly limits therapeutics penetration into the brain. Moreover, even though some therapeutics can cross BBB based on their intrinsic properties or via the use of proper nanoscale delivery vehicles, their therapeutic efficacy is still often limited without the specific uptake of drugs by the cancer or disease-associated cells. As more studies have started to elucidate the pathological roles of major cells in the CNS (for example, microglia, neurons, and astrocytes) for different disorders, nanomedicines that can enable targeting of specific cells in these diseases may provide great potential to boost efficacy. In this review, we aim to briefly cover the pathological roles of endothelial cells, microglia, tumor-associated microglia/macrophage, neurons, astrocytes, and glioma in CNS disorders and to highlight the recent advances in nanomedicines that can target specific disease-associated cells. Furthermore, we summarized some strategies employed in nanomedicine to achieve specific cell targeting or to enhance the drug neuroprotective effects in the CNS. The specific targeting at the cellular level by nanotherapy can be a more precise and effective means not only to enhance the drug availability but also to reduce side effects.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.,Department of Material Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yi-An Lin
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, MD, 21287 USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
36
|
White EE, Pai A, Weng Y, Suresh AK, Van Haute D, Pailevanian T, Alizadeh D, Hajimiri A, Badie B, Berlin JM. Functionalized iron oxide nanoparticles for controlling the movement of immune cells. NANOSCALE 2015; 7:7780-9. [PMID: 25848983 PMCID: PMC4409571 DOI: 10.1039/c3nr04421a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Immunotherapy is currently being investigated for the treatment of many diseases, including cancer. The ability to control the location of immune cells during or following activation would represent a powerful new technique for this field. Targeted magnetic delivery is emerging as a technique for controlling cell movement and localization. Here we show that this technique can be extended to microglia, the primary phagocytic immune cells in the central nervous system. The magnetized microglia were generated by loading the cells with iron oxide nanoparticles functionalized with CpG oligonucleotides, serving as a proof of principle that nanoparticles can be used to both deliver an immunostimulatory cargo to cells and to control the movement of the cells. The nanoparticle-oligonucleotide conjugates are efficiently internalized, non-toxic, and immunostimulatory. We demonstrate that the in vitro migration of the adherent, loaded microglia can be controlled by an external magnetic field and that magnetically-induced migration is non-cytotoxic. In order to capture video of this magnetically-induced migration of loaded cells, a novel 3D-printed "cell box" was designed to facilitate our imaging application. Analysis of cell movement velocities clearly demonstrate increased cell velocities toward the magnet. These studies represent the initial step towards our final goal of using nanoparticles to both activate immune cells and to control their trafficking within the diseased brain.
Collapse
Affiliation(s)
- Ethan E White
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Irell & Manella Graduate School of Biological Sciences at City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Alex Pai
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
| | - Yiming Weng
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Anil K. Suresh
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Desiree Van Haute
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Irell & Manella Graduate School of Biological Sciences at City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Torkom Pailevanian
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
| | - Darya Alizadeh
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Ali Hajimiri
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| | - Behnam Badie
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| | - Jacob M. Berlin
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| |
Collapse
|
37
|
Carvalho da Fonseca AC, Wang H, Fan H, Chen X, Zhang I, Zhang L, Lima FRS, Badie B. Increased expression of stress inducible protein 1 in glioma-associated microglia/macrophages. J Neuroimmunol 2014; 274:71-7. [PMID: 25042352 DOI: 10.1016/j.jneuroim.2014.06.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/25/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
Factors released by glioma-associated microglia/macrophages (GAMs) play an important role in the growth and infiltration of tumors. We have previously demonstrated that the co-chaperone stress-inducible protein 1 (STI1) secreted by microglia promotes proliferation and migration of human glioblastoma (GBM) cell lines in vitro. In the present study, in order to investigate the role of STI1 in a physiological context, we used a glioma model to evaluate STI1 expression in vivo. Here, we demonstrate that STI1 expression in both the tumor and in the infiltrating GAMs and lymphocytes significantly increased with tumor progression. Interestingly, high expression of STI1 was observed in macrophages and lymphocytes that infiltrated brain tumors, whereas STI1 expression in the circulating blood monocytes and lymphocytes remained unchanged. Our results correlate, for the first time, the expression of STI1 and glioma progression, and suggest that STI1 expression in GAMs and infiltrating lymphocytes is modulated by the brain tumor microenvironment.
Collapse
Affiliation(s)
| | - Huaqing Wang
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Haitao Fan
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Xuebo Chen
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, PR China
| | - Ian Zhang
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States
| | - Leying Zhang
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States
| | - Flavia Regina Souza Lima
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Behnam Badie
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States.
| |
Collapse
|
38
|
Guo L, Yan DD, Yang D, Li Y, Wang X, Zalewski O, Yan B, Lu W. Combinatorial photothermal and immuno cancer therapy using chitosan-coated hollow copper sulfide nanoparticles. ACS NANO 2014; 8:5670-81. [PMID: 24801008 PMCID: PMC4072412 DOI: 10.1021/nn5002112] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 05/06/2014] [Indexed: 05/19/2023]
Abstract
Near-infrared light-responsive inorganic nanoparticles have been shown to enhance the efficacy of cancer photothermal ablation therapy. However, current nanoparticle-mediated photothermal ablation is more effective in treating local cancer at the primary site than metastatic cancer. Here, we report the design of a near-infrared light-induced transformative nanoparticle platform that combines photothermal ablation with immunotherapy. The design is based on chitosan-coated hollow CuS nanoparticles that assemble the immunoadjuvants oligodeoxynucleotides containing the cytosine-guanine (CpG) motifs. Interestingly, these structures break down after laser excitation, reassemble, and transform into polymer complexes that improve tumor retention of the immunotherapy. In this "photothermal immunotherapy" approach, photothermal ablation-induced tumor cell death reduces tumor growth and releases tumor antigens into the surrounding milieu, while the immunoadjuvants potentiate host antitumor immunity. Our results indicated that combined photothermal immunotherapy is more effective than either immunotherapy or photothermal therapy alone against primary treated and distant untreated tumors in a mouse breast cancer model. These hollow CuS nanoparticles are biodegradable and can be eliminated from the body after laser excitation.
Collapse
Affiliation(s)
- Liangran Guo
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Daisy D. Yan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Dongfang Yang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Yajuan Li
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Xiaodong Wang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Olivia Zalewski
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Bingfang Yan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Wei Lu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
- School of Pharmacy, Fudan University, Shanghai 201203, China
- Address correspondence to
| |
Collapse
|
39
|
Zhou S, Hashida Y, Kawakami S, Mihara J, Umeyama T, Imahori H, Murakami T, Yamashita F, Hashida M. Preparation of immunostimulatory single-walled carbon nanotube/CpG DNA complexes and evaluation of their potential in cancer immunotherapy. Int J Pharm 2014; 471:214-23. [PMID: 24861942 DOI: 10.1016/j.ijpharm.2014.05.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/30/2014] [Accepted: 05/19/2014] [Indexed: 01/10/2023]
Abstract
Carbon nanotubes (CNTs) have many interesting properties. In particular, their photohyperthermic effect by near-infrared (NIR) irradiation could be used to kill cancer cells, and could thus be applied in photohyperthermic therapy. However, the solubility of CNTs must be improved before they can be used in biological applications. As DNA is reported to disperse the CNTs in aqueous solution with π-π interactions, we hypothesis that immunostimulatory CpG DNA may also disperse the CNTs in aqueous solution. In this study, we used CpG DNA to disperse single-walled CNTs (SWCNTs) in aqueous solution, in order to combine photohyperthermic effect and immunoactivation together to achieve a more effective cancer therapy. As expected, CpG DNA effectively dispersed the SWCNTs in aqueous solution via the formation of SWCNT/CpG DNA complexes. Moreover, the immunoreactivity of the SWCNT/CpG DNA complexes was investigated. The results showed that intratumoral administration of the SWCNT/CpG DNA complexes in mice enhanced the production level of inflammatory cytokines in tumor tissues. Finally, we evaluated the antitumor effects of the SWCNT/CpG DNA complexes in tumor-bearing mice. The result indicated that intratumoral administration of the SWCNT/CpG DNA complexes combined with NIR irradiation was a more effective approach to prevent the proliferation of tumor growth.
Collapse
Affiliation(s)
- Shuwen Zhou
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; The Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Tokyo 102-8471, Japan
| | - Yasuhiko Hashida
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Junya Mihara
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Tomokazu Umeyama
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Hiroshi Imahori
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Tatsuya Murakami
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
40
|
Immunotherapy applications of carbon nanotubes: from design to safe applications. Trends Biotechnol 2014; 32:198-209. [DOI: 10.1016/j.tibtech.2014.02.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 01/20/2014] [Accepted: 02/07/2014] [Indexed: 02/05/2023]
|
41
|
Battigelli A, Ménard-Moyon C, Bianco A. Carbon nanomaterials as new tools for immunotherapeutic applications. J Mater Chem B 2014; 2:6144-6156. [DOI: 10.1039/c4tb00563e] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The possibility to exploit carbon-based nanostructures such as carbon nanotubes and graphene as immunotherapeutic agents has interesting future prospects. In particular, their applications for anticancer treatment, imaging and vaccine development, together with their immunomodulator properties are highlighted.
Collapse
Affiliation(s)
- Alessia Battigelli
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| | - Alberto Bianco
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| |
Collapse
|
42
|
|
43
|
Wang H, Zhang L, Zhang IY, Chen X, Da Fonseca A, Wu S, Ren H, Badie S, Sadeghi S, Ouyang M, Warden CD, Badie B. S100B promotes glioma growth through chemoattraction of myeloid-derived macrophages. Clin Cancer Res 2013; 19:3764-75. [PMID: 23719262 DOI: 10.1158/1078-0432.ccr-12-3725] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE S100B is member of a multigenic family of Ca(2+)-binding proteins, which is overexpressed by gliomas. Recently, we showed that low concentrations of S100B attenuated microglia activation through the induction of Stat3. We hypothesized that overexpression of S100B in gliomas could promote tumor growth by modulating the activity of tumor-associated macrophages (TAM). EXPERIMENTAL DESIGN We stably transfected GL261 glioma cell lines with constructs that overexpressed (S100B(high)) or underexpressed (S100B(low)) S100B and compared their growth characteristics to intracranial wild-type (S100B(wt)) tumors. RESULTS Downregulation of S100B in gliomas had no impact on cell division in vitro but abrogated tumor growth in vivo. Interestingly, compared to S100B(low) tumors, S100B(wt) and S100B(high) intracranial gliomas exhibited higher infiltration of TAMs, stronger inflammatory cytokine expression, and increased vascularity. To identify the potential mechanisms involved, the expression of the S100B receptor, receptor for advanced glycation end products (RAGE), was evaluated in gliomas. Although S100B expression induced RAGE in vivo, RAGE ablation in mice did not significantly inhibit TAM infiltration into gliomas, suggesting that other pathways were involved in this process. To evaluate other mechanisms responsible for TAM chemoattraction, we then examined chemokine pathways and found that C-C motif ligand 2 (CCL2) was upregulated in S100B(high) tumors. Furthermore, analysis of The Cancer Genome Atlas's glioma data bank showed a positive correlation between S100B and CCL2 expression in human proneural and neural glioma subtypes, supporting our finding. CONCLUSIONS These observations suggest that S100B promotes glioma growth by TAM chemoattraction through upregulation of CCL2 and introduces the potential utility of S100B inhibitors for glioma therapy.
Collapse
Affiliation(s)
- Huaqing Wang
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bosi S, Ballerini L, Prato M. Carbon Nanotubes in Tissue Engineering. MAKING AND EXPLOITING FULLERENES, GRAPHENE, AND CARBON NANOTUBES 2013; 348:181-204. [DOI: 10.1007/128_2013_474] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|