1
|
Li H, Shan C, Zhu Y, Yao X, Lin L, Zhang X, Qian Y, Wang Y, Xu J, Zhang Y, Li H, Zhao L, Chen K. Helminth-induced immune modulation in colorectal cancer: exploring therapeutic applications. Front Immunol 2025; 16:1484686. [PMID: 40297577 PMCID: PMC12034720 DOI: 10.3389/fimmu.2025.1484686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer is one of the most lethal tumors, posing a financial and healthcare burden. This study investigates how helminths and pre-existing diseases such as colitis, obesity, diabetes, and gut microbiota issues influence colon cancer development and prognosis. The immune system's protective immunosuppressive response to helminth invasion minimizes inflammation-induced cell damage and DNA mutations, lowering the risk of colorectal cancer precursor lesions. Helminth infection-mediated immunosuppression can hasten colorectal cancer growth and metastasis, which is detrimental to patient outcomes. Some helminth derivatives can activate immune cells to attack cancer cells, making them potentially useful as colorectal cancer vaccines or therapies. This review also covers gene editing approaches. We discovered that using CRISPR/Cas9 to inhibit live helminths modulates miRNA, which limits tumor growth. We propose more multicenter studies into helminth therapy's long-term effects and immune regulation pathways. We hope to treat colorectal cancer patients with helminth therapy and conventional cancer treatments in an integrative setting.
Collapse
Affiliation(s)
- Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Ocean College, Beibu Gulf University, Qinzhou, China
| | - Chaojun Shan
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaodong Yao
- School of Marxism, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijun Lin
- School of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xiaofen Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuncheng Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuqing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jialu Xu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yijie Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hairun Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ling Zhao
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
2
|
Ezediuno LO, Ockiya MA, Awoniyi LO, Sangodare AO, David KB, Robert FO. Developing cancer vaccine with carcinoembryonic antigen and IGF-1R as immunostimulants using immunoinformatics approach. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2025; 21:20-31. [PMID: 40340228 PMCID: PMC12077146 DOI: 10.14216/kjco.24326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 05/10/2025]
Abstract
PURPOSE Colorectal cancer (CRC) remains a significant global health burden, necessitating innovative approaches for prevention and treatment. This study proposes a multiepitope vaccine targeting carcinoembryonic antigen (CEA) and insulin-like growth factor-1 receptor (IGF-1R), two prominent biomarkers associated with CRC progression. METHODS Sequences of CEA and IGF-1R proteins were retrieved from NCBI databank, the sequences were aligned on the MEGA5 tool to identify conserved regions. Immunological and structural predictive analysis which include antigenic potential prediction, cytotoxic T-lymphocytes (CTLs), helper-T lymphocytes (HTLs), B-cell epitopes predictions, and prediction of the vaccine secondary and tertiary structure were performed. The vaccine was evaluated to validate its physiochemical and immunological properties. To determine the binding energy and domain, the tertiary structure of the vaccine was docked to Toll-like receptor 4, and viewed on PyMOL and LigPlot+ tools. RESULTS CEA and IGF-1R were revealed to be highly antigenic, and non-allergens demonstrating the capacity to elicit robust immune responses, which include CTLs, HTLs, and B cells activation. The secondary structure revealed a conformation closely resembling native protein, with alpha helices, beta sheets, and coils, indicative of favorable interactions. Tertiary structure prediction predicted five models, model 0 was selected and validated due its highest confidence, and validation revealed that 87.5% of residues were within favored regions, with a z-score of 4.03. Molecular docking predicted strong binding complex with low binding energy. CONCLUSION Based on our analysis, the proposed multiepitope vaccine holds promise as an effective preventive measure against colorectal cancer development.
Collapse
Affiliation(s)
| | - Michael Asebake Ockiya
- Department of Animal Science, Faculty of Agriculture, Niger Delta University, Amassoma,
Nigeria
| | | | | | - Kehinde Busuyi David
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso,
Nigeria
| | - Faith Owabhel Robert
- Department of Medical Biochemistry, Rivers State University, Port Harcourt,
Nigeria
| |
Collapse
|
3
|
Novakova A, Morris SA, Vaiarelli L, Frank S. Manufacturing and Financial Evaluation of Peptide-Based Neoantigen Cancer Vaccines for Triple-Negative Breast Cancer in the United Kingdom: Opportunities and Challenges. Vaccines (Basel) 2025; 13:144. [PMID: 40006691 PMCID: PMC11860436 DOI: 10.3390/vaccines13020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review evaluates the financial burden of current treatments for triple-negative breast cancer (TNBC) and projects potential financial scenarios to assess the feasibility of introducing a peptide-based neoantigen cancer vaccine (NCV) targeting the disease, using the UK as a healthcare system model. TNBC, the most aggressive breast cancer subtype, is associated with poor prognosis, worsened by the lack of personalised treatment options. Neoantigen cancer vaccine therapies present a personalised alternative with the potential to enhance T-cell responses independently of genetic factors, unlike approved immunotherapies for TNBC. Through a systematic literature review, the underlying science and manufacturing processes of NCVs are explored, the direct medical costs of existing TNBC treatments are enumerated, and two contrasting pricing scenarios for NCV clinical adoption are evaluated. The findings indicate that limited immunogenicity is the main scientific barrier to NCV clinical advancement, alongside production inefficiencies. Financial analysis shows that the UK spends approximately GBP 230 million annually on TNBC treatments, ranging from GBP 2200 to GBP 54,000 per patient. A best-case pricing model involving government-sponsored NCV therapy appears financially viable, while a worst-case, privately funded model exceeds the National Institute for Health and Care Excellence (NICE) cost thresholds. This study concludes that while NCVs show potential clinical benefits for TNBC, uncertainties about their standalone efficacy make their widespread adoption in the UK unlikely without further clinical research.
Collapse
Affiliation(s)
| | | | - Ludovica Vaiarelli
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| |
Collapse
|
4
|
Massariol Pimenta T, Carlos de Souza J, da Silva Martins B, Silva Butzene SM, Simões Padilha JM, Ganho Marçal M, Dos Santos Elias G, Rangel LBA. Emerging strategies to overcome ovarian cancer: advances in immunotherapy. Front Pharmacol 2024; 15:1490896. [PMID: 39564107 PMCID: PMC11573523 DOI: 10.3389/fphar.2024.1490896] [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: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Ovarian cancer is the second most common malignant neoplasm of gynecological origin and the leading cause of death from cancer in the female reproductive system worldwide. This scenario is largely due to late diagnoses, often in advanced stages, and the development of chemoresistance by cancer cells. These challenges highlight the need for alternative treatments, with immunotherapy being a promising option. Cancer immunotherapy involves triggering an anti-tumor immune response and developing immunological memory to eliminate malignant cells, prevent recurrence, and inhibit metastasis. Some ongoing research investigate potentially immunological advancements in the field of cancer vaccines, immune checkpoint blockade, CAR-T cell, and other strategies.
Collapse
Affiliation(s)
- Tatiana Massariol Pimenta
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Josiany Carlos de Souza
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bárbara da Silva Martins
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Solenny Maria Silva Butzene
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - José Matheus Simões Padilha
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Milleny Ganho Marçal
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Guilherme Dos Santos Elias
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Leticia Batista Azevedo Rangel
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
5
|
Cao Q, Fang H, Tian H. mRNA vaccines contribute to innate and adaptive immunity to enhance immune response in vivo. Biomaterials 2024; 310:122628. [PMID: 38820767 DOI: 10.1016/j.biomaterials.2024.122628] [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: 01/06/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Messenger RNA (mRNA) therapeutics have been widely employed as strategies for the treatment and prevention of diseases. Amid the global outbreak of COVID-19, mRNA vaccines have witnessed rapid development. Generally, in the case of mRNA vaccines, the initiation of the innate immune system serves as a prerequisite for triggering subsequent adaptive immune responses. Critical cells, cytokines, and chemokines within the innate immune system play crucial and beneficial roles in coordinating tailored immune reactions towards mRNA vaccines. Furthermore, immunostimulators and delivery systems play a significant role in augmenting the immune potency of mRNA vaccines. In this comprehensive review, we systematically delineate the latest advancements in mRNA vaccine research, present an in-depth exploration of strategies aimed at amplifying the immune effectiveness of mRNA vaccines, and offer some perspectives and recommendations regarding the future advancements in mRNA vaccine development.
Collapse
Affiliation(s)
- Qiannan Cao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China.
| |
Collapse
|
6
|
Moon DO. Advancing Cancer Therapy: The Role of KIF20A as a Target for Inhibitor Development and Immunotherapy. Cancers (Basel) 2024; 16:2958. [PMID: 39272816 PMCID: PMC11393963 DOI: 10.3390/cancers16172958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
The analysis begins with a detailed examination of the gene expression and protein structure of KIF20A, highlighting its interaction with critical cellular components that influence key processes such as Golgi membrane transport and mitotic spindle assembly. The primary focus is on the development of specific KIF20A inhibitors, detailing their roles and the challenges encountered in enhancing their efficacy, such as achieving specificity, overcoming tumor resistance, and optimizing delivery systems. Additionally, it delves into the prognostic value of KIF20A across multiple cancer types, emphasizing its role as a novel tumor-associated antigen, which lays the groundwork for the development of targeted peptide vaccines. The therapeutic efficacy of these vaccines as demonstrated in recent clinical trials is discussed. Future directions are proposed, including the integration of precision medicine strategies to personalize treatments and the use of combination therapies to improve outcomes. By concentrating on the significant potential of KIF20A as both a direct target for inhibitors and an antigen in cancer vaccines, this review sets a foundation for future research aimed at harnessing KIF20A for effective cancer treatment.
Collapse
Affiliation(s)
- Dong Oh Moon
- Department of Biology Education, Daegu University, 201, Daegudae-ro, Gyeongsan-si 38453, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
7
|
Rashid MM, Selvarajoo K. Advancing drug-response prediction using multi-modal and -omics machine learning integration (MOMLIN): a case study on breast cancer clinical data. Brief Bioinform 2024; 25:bbae300. [PMID: 38904542 PMCID: PMC11190965 DOI: 10.1093/bib/bbae300] [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: 03/25/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
The inherent heterogeneity of cancer contributes to highly variable responses to any anticancer treatments. This underscores the need to first identify precise biomarkers through complex multi-omics datasets that are now available. Although much research has focused on this aspect, identifying biomarkers associated with distinct drug responders still remains a major challenge. Here, we develop MOMLIN, a multi-modal and -omics machine learning integration framework, to enhance drug-response prediction. MOMLIN jointly utilizes sparse correlation algorithms and class-specific feature selection algorithms, which identifies multi-modal and -omics-associated interpretable components. MOMLIN was applied to 147 patients' breast cancer datasets (clinical, mutation, gene expression, tumor microenvironment cells and molecular pathways) to analyze drug-response class predictions for non-responders and variable responders. Notably, MOMLIN achieves an average AUC of 0.989, which is at least 10% greater when compared with current state-of-the-art (data integration analysis for biomarker discovery using latent components, multi-omics factor analysis, sparse canonical correlation analysis). Moreover, MOMLIN not only detects known individual biomarkers such as genes at mutation/expression level, most importantly, it correlates multi-modal and -omics network biomarkers for each response class. For example, an interaction between ER-negative-HMCN1-COL5A1 mutations-FBXO2-CSF3R expression-CD8 emerge as a multimodal biomarker for responders, potentially affecting antimicrobial peptides and FLT3 signaling pathways. In contrast, for resistance cases, a distinct combination of lymph node-TP53 mutation-PON3-ENSG00000261116 lncRNA expression-HLA-E-T-cell exclusions emerged as multimodal biomarkers, possibly impacting neurotransmitter release cycle pathway. MOMLIN, therefore, is expected advance precision medicine, such as to detect context-specific multi-omics network biomarkers and better predict drug-response classifications.
Collapse
Affiliation(s)
- Md Mamunur Rashid
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
| | - Kumar Selvarajoo
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
- Synthetic Biology Translational Research Program, Yong Loo Lin School of Medicine, NUS, Singapore 117456, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| |
Collapse
|
8
|
Parhiz H, Atochina-Vasserman EN, Weissman D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 2024; 403:1192-1204. [PMID: 38461842 DOI: 10.1016/s0140-6736(23)02444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024]
Abstract
Recent advances in mRNA technology and its delivery have enabled mRNA-based therapeutics to enter a new era in medicine. The rapid, potent, and transient nature of mRNA-encoded proteins, without the need to enter the nucleus or the risk of genomic integration, makes them desirable tools for treatment of a range of diseases, from infectious diseases to cancer and monogenic disorders. The rapid pace and ease of mass-scale manufacturability of mRNA-based therapeutics supported the global response to the COVID-19 pandemic. Nonetheless, challenges remain with regards to mRNA stability, duration of expression, delivery efficiency, and targetability, to broaden the applicability of mRNA therapeutics beyond COVID-19 vaccines. By learning from the rapidly expanding preclinical and clinical studies, we can optimise the mRNA platform to meet the clinical needs of each disease. Here, we will summarise the recent advances in mRNA technology; its use in vaccines, immunotherapeutics, protein replacement therapy, and genomic editing; and its delivery to desired specific cell types and organs for development of a new generation of targeted mRNA-based therapeutics.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Liu T, Yu S, Hu T, Ji W, Cheng X, Lv L, Shi Z. Comprehensive analyses of genome-wide methylation and RNA epigenetics identify prognostic biomarkers, regulating the tumor immune microenvironment in lung adenocarcinoma. Pathol Res Pract 2023; 248:154621. [PMID: 37336075 DOI: 10.1016/j.prp.2023.154621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
The aim of our study was to identify a signature of immune-regulated molecules and reveal its prognostic role in lung adenocarcinoma (LUAD). We downloaded RNA-Sequencing data and DNA methylation data from the Gene Expression Omnibus (GEO) database. GEO2R was used to analyze differentially expressed mRNAs (DEmRNAs). we used "factoextra" R package to do the principal component analysis (PCA) of DEmRNAs. "Limma" R package was used to identify DEmRNAs, differentially expressed miRNAs (DEmiRNAs), differentially expressed lncRNAs (DElncRNAs) from The Cancer Genome Atlas (TCGA) database. Three R packages "org.Hs.eg.db", "clusterProfiler", "ggplot2″ were used to show enrichment results. Considering about methylation and mutation data, TEK and SOX17 mediated cancer signaling pathways. Through tumor-immune system interactions database (TISIDB) and Tumor Immune Estimation Resource (TIMER), higher methylated and lower expressed TEK may act as a prognostic marker, regulating the tumor immunity in LUAD. Through four databases (MEXPRESS, DNMIVD, MethSurv, Firehose), we further verified the methylation (P = 2.33e-23) and mutation about TEK. A signature of immune-associated TEK to predict survival of LUAD patients was validated. Prognostic, methylation, immune microenvironment analysis showed new light on potential novel therapeutic targets in LUAD.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shuo Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.; Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi 710000, China
| | - Tinghua Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wen Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xue Cheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lin Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhihong Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China..
| |
Collapse
|
10
|
Hari SK, Gauba A, Shrivastava N, Tripathi RM, Jain SK, Pandey AK. Polymeric micelles and cancer therapy: an ingenious multimodal tumor-targeted drug delivery system. Drug Deliv Transl Res 2023; 13:135-163. [PMID: 35727533 DOI: 10.1007/s13346-022-01197-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
Since the beginning of pharmaceutical research, drug delivery methods have been an integral part of it. Polymeric micelles (PMs) have emerged as multifunctional nanoparticles in the current technological era of nanocarriers, and they have shown promise in a range of scientific fields. They can alter the release profile of integrated pharmacological substances and concentrate them in the target zone due to their improved permeability and retention, making them more suitable for poorly soluble medicines. With their ability to deliver poorly soluble chemotherapeutic drugs, PMs have garnered considerable interest in cancer. As a result of their remarkable biocompatibility, improved permeability, and minimal toxicity to healthy cells, while also their capacity to solubilize a wide range of drugs in their micellar core, PMs are expected to be a successful treatment option for cancer therapy in the future. Their nano-size enables them to accumulate in the tumor microenvironment (TME) via the enhanced permeability and retention (EPR) effect. In this review, our major aim is to focus primarily on the stellar applications of PMs in the field of cancer therapeutics along with its mechanism of action and its latest advancements in drug and gene delivery (DNA/siRNA) for cancer, using various therapeutic strategies such as crossing blood-brain barrier, gene therapy, photothermal therapy (PTT), and immunotherapy. Furthermore, PMs can be employed as "smart drug carriers," allowing them to target specific cancer sites using a variety of stimuli (endogenous and exogenous), which improve the specificity and efficacy of micelle-based targeted drug delivery. All the many types of stimulants, as well as how the complex of PM and various anticancer drugs react to it, and their pharmacodynamics are also reviewed here. In conclusion, commercializing engineered micelle nanoparticles (MNPs) for application in therapy and imaging can be considered as a potential approach to improve the therapeutic index of anticancer drugs. Furthermore, PM has stimulated intense interest in research and clinical practice, and in light of this, we have also highlighted a few PMs that have previously been approved for therapeutic use, while the majority are still being studied in clinical trials for various cancer therapies.
Collapse
Affiliation(s)
- Sharath Kumar Hari
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ankita Gauba
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Neeraj Shrivastava
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ravi Mani Tripathi
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India.
| | - Sudhir Kumar Jain
- School of Studies in Microbiology, Vikram University, Ujjain, Madhya Pradesh, 456010, India
| | - Akhilesh Kumar Pandey
- Department of Biological Sciences, Rani Durgavati University, Jabalpur, M.P, 482001, India.,Vikram University, Ujjain, Madhya Pradesh, 456010, India
| |
Collapse
|
11
|
Ladak RJ, He AJ, Huang YH, Ding Y. The Current Landscape of mRNA Vaccines Against Viruses and Cancer-A Mini Review. Front Immunol 2022; 13:885371. [PMID: 35603213 PMCID: PMC9120423 DOI: 10.3389/fimmu.2022.885371] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/14/2022] [Indexed: 12/11/2022] Open
Abstract
Both infectious viral diseases and cancer have historically been some of the most common causes of death worldwide. The COVID-19 pandemic is a decidedly relevant example of the former. Despite progress having been made over past decades, new and improved techniques are still needed to address the limitations faced by current treatment standards, with mRNA-based therapy emerging as a promising solution. Highly flexible, scalable and cost-effective, mRNA therapy is proving to be a compelling vaccine platform against viruses. Likewise, mRNA vaccines show similar promise against cancer as a platform capable of encoding multiple antigens for a diverse array of cancers, including those that are patient specific as a novel form of personalized medicine. In this review, the molecular mechanisms, biotechnological aspects, and clinical developments of mRNA vaccines against viral infections and cancer are discussed to provide an informative update on the current state of mRNA therapy research.
Collapse
Affiliation(s)
- Reese Jalal Ladak
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Alexander J. He
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, United Kingdom
| | - Yu-Hsun Huang
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Yu Ding
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
12
|
Discovery of Novel 3,4-Dihydro-2(1H)-Quinolinone Sulfonamide Derivatives as New Tubulin Polymerization Inhibitors with Anti-Cancer Activity. Molecules 2022; 27:molecules27051537. [PMID: 35268645 PMCID: PMC8911884 DOI: 10.3390/molecules27051537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 12/10/2022] Open
Abstract
In this paper, a small series of novel quinoline sulfonamide derivatives was synthesized, and their structure of the target compounds were confirmed by 1H NMR and MS. The screening of the news target compounds’ in vitro cytotoxic activities against tumor cell lines by the MTT method was performed. Among them, compound D13 (N-(4-methoxybenzyl)-2-oxo-N-(3,4,5-trimethoxyphenyl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide exhibited the strongest inhibitory effect on the proliferation of HeLa (IC50: 1.34 μM), and this value correlated well with the inhibitory activities of the compound against tubulin polymerization (IC50: 6.74 μM). In summary, a new type of quinoline-sulfonamide derivative with tubulin polymerization inhibitory activity was discovered, and it can be used as a lead compound for further modification.
Collapse
|
13
|
Yu A, Dai X, Wang Z, Chen H, Guo B, Huang L. Recent Advances of Mesoporous Silica as a Platform for Cancer Immunotherapy. BIOSENSORS 2022; 12:109. [PMID: 35200369 PMCID: PMC8869707 DOI: 10.3390/bios12020109] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 05/06/2023]
Abstract
Immunotherapy is a promising modality of treatment for cancer. Immunotherapy is comprised of systemic and local treatments that induce an immune response, allowing the body to fight back against cancer. Systemic treatments such as cancer vaccines harness antigen presenting cells (APCs) to activate T cells with tumor-associated antigens. Small molecule inhibitors can be employed to inhibit immune checkpoints, disrupting tumor immunosuppression and immune evasion. Despite the current efficacy of immunotherapy, improvements to delivery can be made. Nanomaterials such as mesoporous silica can facilitate the advancement of immunotherapy. Mesoporous silica has high porosity, decent biocompatibility, and simple surface functionalization. Mesoporous silica can be utilized as a versatile carrier of various immunotherapeutic agents. This review gives an introduction on mesoporous silica as a nanomaterial, briefly covering synthesis and biocompatibility, and then an overview of the recent progress made in the application of mesoporous silica to cancer immunotherapy.
Collapse
Affiliation(s)
- Albert Yu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Xiaoyong Dai
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Zixian Wang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Huaqing Chen
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China;
| | - Laiqiang Huang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
14
|
Essa N, O'Connell F, Prina-Mello A, O'Sullivan J, Marcone S. Gold nanoparticles and obese adipose tissue microenvironment in cancer treatment. Cancer Lett 2022; 525:1-8. [PMID: 34662546 DOI: 10.1016/j.canlet.2021.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/30/2021] [Accepted: 10/13/2021] [Indexed: 02/07/2023]
Abstract
The epidemiological correlation between obesity and cancer is well characterized, but the biological mechanisms which regulate tumor development and response to therapy in obese cancer patients remain unclear. The tumor microenvironment plays an important role in protecting cancer cells by altering the delivery of anticancer therapy to the tumor tissue, reducing the efficacy of treatment. Obese tumor microenvironment provides additional benefits to the survival of tumor cells against anticancer therapies by altering the extracellular matrix composition, angiogenesis processes and the immune cells profile. Nanotechnology, and in particular gold nanoparticles, are being researched as a theranostic strategy for cancer treatment due to their ability to sensitize cancer cells to radiation and photodynamic therapy, enhance delivery of drugs to tumor cells, and in diagnostic applications. Adipose tissue and the obese tumor microenvironment may alter the activity of nanotherapeutics. In this article, we reviewed the current state of our understanding about the mechanisms by which the obese tumor microenvironment may alter the delivery and efficacy of anti-cancer treatments, and why the use of gold nanoparticles may represent an interesting strategy for cancer treatment in the obesity setting.
Collapse
Affiliation(s)
- Noor Essa
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland; Master in Science Degree in Translational Oncology, Trinity College Dublin, Dublin, Ireland
| | - Fiona O'Connell
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Adriele Prina-Mello
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM) and Nanomedicine Group, Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
15
|
Ren X, Wang Y, Jia L, Guo X, He X, Zhao Z, Gao D, Yang Z. Intelligent Nanomedicine Approaches Using Medical Gas-Mediated Multi-Therapeutic Modalities Against Cancer. J Biomed Nanotechnol 2022; 18:24-49. [PMID: 35180898 DOI: 10.1166/jbn.2022.3224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The emerging area of gas-mediated cancer treatment has received widespread attention in the medical community. Featuring unique physical, chemical, and biological properties, nanomaterials can facilitate the delivery and controllable release of medicinal gases at tumor sites, and also serve as ideal platforms for the integration of other therapeutic modalities with gas therapy to augment cancer therapeutic efficacy. This review presents an overview of anti-cancer mechanisms of several therapeutic gases: nitric oxide (NO), hydrogen sulfide (H₂S), carbon monoxide (CO), oxygen (O₂), and hydrogen (H₂). Controlled release behaviors of gases under different endogenous and exogenous stimuli are also briefly discussed, followed by their synergistic effects with different therapeutic modes. Moreover, the potential challenges and future prospects regarding gas therapy based on nanomaterials are also described, aiming to facilitate the advancement of gas therapeutic nanomedicine in new frontiers for highly efficient cancer treatment.
Collapse
Affiliation(s)
- Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoqing Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhipeng Zhao
- School of Physical Education, Xizang Minzu University, Xianyang, 712000, Shaanxi, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
16
|
Ahmad S, Idris RAM, Wan Hanaffi WN, Perumal K, Boer JC, Plebanski M, Jaafar J, Lim JK, Mohamud R. Cancer Nanomedicine and Immune System—Interactions and Challenges. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.681305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nanoparticles have tremendous therapeutic potential in the treatment of cancer as they increase drug delivery, attenuate drug toxicity, and protect drugs from rapid clearance. Since Doxil®, the first FDA-approved nanomedicine, several other cancer nanomedicines have been approved and have successfully increased the efficacy over their free drug counterparts. Although their mechanisms of action are well established, their effects towards our immune system, particularly in the tumor microenvironment (TME), still warrant further investigation. Herein, we review the interactions between an approved cancer nanomedicine with TME immunology. We also discuss the challenges that need to be addressed for the full clinical potential of ongoing cancer nanomedicines despite the encouraging preclinical data.
Collapse
|
17
|
He JS, Liu SJ, Zhang YR, Chu XD, Lin ZB, Zhao Z, Qiu SH, Guo YG, Ding H, Pan YL, Pan JH. The Application of and Strategy for Gold Nanoparticles in Cancer Immunotherapy. Front Pharmacol 2021; 12:687399. [PMID: 34163367 PMCID: PMC8215714 DOI: 10.3389/fphar.2021.687399] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy of malignant tumor is a verified and crucial anti-tumor strategy to help patients with cancer for prolonging prognostic survival. It is a novel anticancer tactics that activates the immune system to discern and damage cancer cells, thereby prevent them from proliferating. However, immunotherapy still faces many challenges in view of clinical efficacy and safety issues. Various nanomaterials, especially gold nanoparticles (AuNPs), have been developed not only for anticancer treatment but also for delivering antitumor drugs or combining other treatment strategies. Recently, some studies have focused on AuNPs for enhancing cancer immunotherapy. In this review, we summarized how AuNPs applicated as immune agents, drug carriers or combinations with other immunotherapies for anticancer treatment. AuNPs can not only act as immune regulators but also deliver immune drugs for cancer. Therefore, AuNPs are candidates for enhancing the efficiency and safety of cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yun-long Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing-hua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
18
|
Li Y, Yang J, Sun X. Reactive Oxygen Species-Based Nanomaterials for Cancer Therapy. Front Chem 2021; 9:650587. [PMID: 33968899 PMCID: PMC8100441 DOI: 10.3389/fchem.2021.650587] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology advances in cancer therapy applications have led to the development of nanomaterials that generate cytotoxic reactive oxygen species (ROS) specifically in tumor cells. ROS act as a double-edged sword, as they can promote tumorigenesis and proliferation but also trigger cell death by enhancing intracellular oxidative stress. Various nanomaterials function by increasing ROS production in tumor cells and thereby disturbing their redox balance, leading to lipid peroxidation, and oxidative damage of DNA and proteins. In this review, we outline these mechanisms, summarize recent progress in ROS-based nanomaterials, including metal-based nanoparticles, organic nanomaterials, and chemotherapy drug-loaded nanoplatforms, and highlight their biomedical applications in cancer therapy as drug delivery systems (DDSs) or in combination with chemodynamic therapy (CDT), photodynamic therapy (PDT), or sonodynamic therapy (SDT). Finally, we discuss the advantages and limitations of current ROS-mediated nanomaterials used in cancer therapy and speculate on the future progress of this nanotechnology for oncological applications.
Collapse
Affiliation(s)
- Yingbo Li
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jie Yang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xilin Sun
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Chan-Zapata I, Segura-Campos MR. Honey and its protein components: Effects in the cancer immunology. J Food Biochem 2021; 45:e13613. [PMID: 33768550 DOI: 10.1111/jfbc.13613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/23/2022]
Abstract
The immune system plays an important role in cancer development, but some tumor cells can evade or inhibit the processes of innate and adaptive immunity. This review made a description of honey and its proteins effect on diverse mediators from the immune system. Scientific evidence reported that many types of honey (jungle, manuka, pasture, and others) and some isolated proteins enhanced the release of reactive oxygen species (O2 - and H2 O2 ) and cytokines (mostly IL-1β, IL-6, and TNF-α) by innate immune system cells. Furthermore, honey elicited proliferation and functions of T lymphocytes, cells related to specific adaptive immune responses. These studies have established a precedent over the honey and its properties on the immune system, demonstrating that it can promote the innate and adaptive immunity. PRACTICAL APPLICATIONS: Cancer is a genetic illness that represents a world health problem. Recognizing the potential of diet therapy in the prevention and treatment of chronic diseases, the present work summarizes the effects of honey on the immune system and mediators involved in cancer elimination processes, establishing the importance of this natural product as a future anticancer agent.
Collapse
Affiliation(s)
- Ivan Chan-Zapata
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Mérida, México
| | | |
Collapse
|
20
|
Sun H, Wang X, Zhai S. The Rational Design and Biological Mechanisms of Nanoradiosensitizers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E504. [PMID: 32168899 PMCID: PMC7153263 DOI: 10.3390/nano10030504] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy (RT) has been widely used for cancer treatment. However, the intrinsic drawbacks of RT, such as radiotoxicity in normal tissues and tumor radioresistance, promoted the development of radiosensitizers. To date, various kinds of nanoparticles have been found to act as radiosensitizers in cancer radiotherapy. This review focuses on the current state of nanoradiosensitizers, especially the related biological mechanisms, and the key design strategies for generating nanoradiosensitizers. The regulation of oxidative stress, DNA damage, the cell cycle, autophagy and apoptosis by nanoradiosensitizers in vitro and in vivo is highlighted, which may guide the rational design of therapeutics for tumor radiosensitization.
Collapse
Affiliation(s)
- Hainan Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
- Shandong Vocational College of Light Industry, Zibo 255300, Shandong, China
| | - Xiaoling Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| |
Collapse
|
21
|
Liu Q, Zhou Y, Li M, Zhao L, Ren J, Li D, Tan Z, Wang K, Li H, Hussain M, Zhang L, Shen G, Zhu J, Tao J. Polyethylenimine Hybrid Thin-Shell Hollow Mesoporous Silica Nanoparticles as Vaccine Self-Adjuvants for Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:47798-47809. [PMID: 31773941 DOI: 10.1021/acsami.9b19446] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Conventional adjuvants (e.g., aluminum) are insufficient to trigger cell-mediated immunity, which plays a crucial role in triggering specific immunity against cancer. Therefore, developing appropriate adjuvants for cancer vaccines is a central way to stimulate the antitumor immune response. Hollow mesoporous silica nanoparticles (HMSNs) have been proven to stimulate Th1 antitumor immunity in vivo and promote immunological memory in the formulation of novel cancer vaccines. Yet, immune response rates of existing HMSNs for anticancer immunity still remain low. Here, we demonstrate the generation of polyethylenimine (PEI)-incorporated thin-shell HMSNs (THMSNs) through a facile PEI etching strategy for cancer immunotherapy. Interestingly, incorporation of PEI and thin-shell hollow structures of THMSNs not only improved the antigen-loading efficacy and sustained drug release profiles but also enhanced the phagocytosis efficiency by dendritic cells (DCs), enabled DC maturation and Th1 immunity, and sustained immunological memory, resulting in the enhancement of the adjuvant effect of THMSNs. Moreover, THMSNs vaccines without significant side effects can significantly reduce the potentiality of tumor growth and metastasis in tumor challenge and rechallenge models, respectively. THMSNs are considered to be promising vehicles and excellent adjuvants for the formulation of cancer vaccines for immunotherapy.
Collapse
Affiliation(s)
- Qianqian Liu
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Yajie Zhou
- Department of Dermatology, Union Hospital, Tongji Medical College , HUST , Wuhan 430022 , China
| | - Mo Li
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Liang Zhao
- Department of Dermatology, Union Hospital, Tongji Medical College , HUST , Wuhan 430022 , China
| | - Jingli Ren
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Danqi Li
- Department of Dermatology, Union Hospital, Tongji Medical College , HUST , Wuhan 430022 , China
| | - Zhengping Tan
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Ke Wang
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Heli Li
- Department of Immunology, Tongji Medical College , HUST , Wuhan 430030 , China
| | - Mubashir Hussain
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Lianbin Zhang
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College , HUST , Wuhan 430030 , China
| | - Jintao Zhu
- Key Lab of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology (HUST) , Wuhan 430074 , China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College , HUST , Wuhan 430022 , China
| |
Collapse
|
22
|
Xu Y, Liang P, Rashid HU, Wu L, Xie P, Wang H, Zhang S, Wang L, Jiang J. Design, synthesis, and biological evaluation of matrine derivatives possessing piperazine moiety as antitumor agents. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02398-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Rezaian M, Maleki R, Dahri Dahroud M, Alamdari A, Alimohammadi M. pH-Sensitive Co-Adsorption/Release of Doxorubicin and Paclitaxel by Carbon Nanotube, Fullerene, and Graphene Oxide in Combination with N-isopropylacrylamide: A Molecular Dynamics Study. Biomolecules 2018; 8:E127. [PMID: 30380660 PMCID: PMC6316683 DOI: 10.3390/biom8040127] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
Nanotechnology based drug delivery systems for cancer therapy have been the topic of interest for many researchers and scientists. In this research, we have studied the pH sensitive co-adsorption and release of doxorubicin (DOX) and paclitaxel (PAX) by carbon nanotube (CNT), fullerene, and graphene oxide (GO) in combination with N-isopropylacrylamide (PIN). This simulation study has been performed by use of molecular dynamics. Interaction energies, hydrogen bond, and gyration radius were investigated. Results reveal that, compared with fullerene and GO, CNT is a better carrier for the co-adsorption and co-release of DOX and PAX. It can adsorb the drugs in plasma pH and release it in vicinity of cancerous tissues which have acidic pH. Investigating the number of hydrogen bonds revealed that PIN created many hydrogen bonds with water resulting in high hydrophilicity of PIN, hence making it more stable in the bloodstream while preventing from its accumulation. It is also concluded from this study that CNT and PIN would make a suitable combination for the delivery of DOX and PAX, because PIN makes abundant hydrogen bonds and CNT makes stable interactions with these drugs.
Collapse
Affiliation(s)
- Milad Rezaian
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, 19839-63113 Tehran, Iran.
| | - Reza Maleki
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz 71345, Iran.
| | - Mohammad Dahri Dahroud
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Abdolmohammad Alamdari
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz 71345, Iran.
| | | |
Collapse
|
24
|
Zhou Q, Zhang L, Yang T, Wu H. Stimuli-responsive polymeric micelles for drug delivery and cancer therapy. Int J Nanomedicine 2018; 13:2921-2942. [PMID: 29849457 PMCID: PMC5965378 DOI: 10.2147/ijn.s158696] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Polymeric micelles (PMs) have been widely investigated as nanocarriers for drug delivery and cancer treatments due to their excellent physicochemical properties, drug loading and release capacities, facile preparation methods, biocompatibility, and tumor targetability. They can be easily engineered with various functional moieties to further improve their performance in terms of bioavailability, circulation time, tumor specificity, and anticancer activity. The stimuli-sensitive PMs capable of responding to various extra- and intracellular biological stimuli (eg, acidic pH, altered redox potential, and upregulated enzyme), as well as external artificial stimuli (eg, magnetic field, light, temperature, and ultrasound), are considered as “smart” nanocarriers for delivery of anticancer drugs and/or imaging agents for various therapeutic and diagnostic applications. In this article, the recent advances in the development of stimuli-responsive PMs for drug delivery, imaging, and cancer therapy are reviewed. The article covers the generalities of stimuli-responsive PMs with a focus on their major delivery strategies and newly emerging technologies/nanomaterials, discusses their drawbacks and limitations, and provides their future perspectives.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| | - Li Zhang
- State Key Laboratory of Military Stomatology, Air Force Military Medical University, Xi'an, People's Republic of China
| | - TieHong Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| | - Hong Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
25
|
Guo Z, Zhu S, Yong Y, Zhang X, Dong X, Du J, Xie J, Wang Q, Gu Z, Zhao Y. Synthesis of BSA-Coated BiOI@Bi 2 S 3 Semiconductor Heterojunction Nanoparticles and Their Applications for Radio/Photodynamic/Photothermal Synergistic Therapy of Tumor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1704136. [PMID: 29035426 DOI: 10.1002/adma.201704136] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/01/2017] [Indexed: 06/07/2023]
Abstract
Developing an effective theranostic nanoplatform remains a great challenge for cancer diagnosis and treatment. Here, BiOI@Bi2 S3 @BSA (bovine serum albumin) semiconductor heterojunction nanoparticles (SHNPs) for triple-combination radio/photodynamic/photothermal cancer therapy and multimodal computed tomography/photoacoustic (CT/PA) bioimaging are reported. On the one hand, SHNPs possess strong X-ray attenuation capability since they contain high-Z elements, and thus they are anticipated to be a very competent candidate as radio-sensitizing materials for radiotherapy enhancement. On the other hand, as a semiconductor, the as-prepared SHNPs offer an extra approach for reactive oxygen species generation based on electron-hole pair under the irradiation of X-ray through the photodynamic therapy process. This X-ray excited photodynamic therapy obviously has better penetration depth in bio-tissue. What's more, the SHNPs also possess well photothermal conversion efficiency for photothermal therapy, because Bi2 S3 is a thin band semiconductor with strong near-infrared absorption that can cause local overheat. In vivo tumor ablation studies show that synergistic radio/photodynamic/photothermal therapy achieves more significant therapeutic effect than any single treatment. In addition, with the strong X-ray attenuation and high near-infrared absorption, the as-obtained SHNPs can also be applied as a multimodal contrast agent in CT/PA imaging.
Collapse
Affiliation(s)
- Zhao Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Yong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Chemistry and Environment Protection Engineering, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Xiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xinghua Dong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiangfeng Du
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jiani Xie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Wang
- School of Material Science and Engineering, Institute of Nano Engineering, Shandong University of Science and Technology, Qingdao, Shandong, 266590, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
26
|
Song G, Cheng L, Chao Y, Yang K, Liu Z. Emerging Nanotechnology and Advanced Materials for Cancer Radiation Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700996. [PMID: 28643452 DOI: 10.1002/adma.201700996] [Citation(s) in RCA: 493] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/11/2017] [Indexed: 05/22/2023]
Abstract
Radiation therapy (RT) including external beam radiotherapy (EBRT) and internal radioisotope therapy (RIT) has been widely used for clinical cancer treatment. However, owing to the low radiation absorption of tumors, high doses of ionizing radiations are often needed during RT, leading to severe damages to normal tissues adjacent to tumors. Meanwhile, the RT efficacies are limited by different mechanisms, among which the tumor hypoxia-associated radiation resistance is a well-known one, as there exists hypoxia inside most solid tumors while oxygen is essential to enhance radiation-induced DNA damages. With the development in nanotechnology, there have been great interests in using nanomedicine strategies to enhance radiation responses of tumors. Nanomaterials containing high-Z elements to absorb radiation rays (e.g. X-ray) can act as radio-sensitizers to deposit radiation energy within tumors and promote treatment efficacy. Nanoscale carriers are able to deliver therapeutic radioisotopes into tumors for internal RIT, or chemotherapeutic drugs for synergistically combined chemo-radiotherapy. As uncovered in recent studies, the tumor microenvironment could be modulated by various nanomedicine approaches to overcome hypoxia-associated radiation resistance. Herein, the authors will summarize the applications of nanomedicine for RT cancer treatment, and pay particular attention to the latest development of 'advanced materials' for enhanced cancer RT.
Collapse
Affiliation(s)
- Guosheng Song
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Liang Cheng
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
27
|
Carabineiro SAC. Applications of Gold Nanoparticles in Nanomedicine: Recent Advances in Vaccines. Molecules 2017; 22:E857. [PMID: 28531163 PMCID: PMC6154615 DOI: 10.3390/molecules22050857] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 11/20/2022] Open
Abstract
Nowadays, gold is used in (nano-)medicine, usually in the form of nanoparticles, due to the solid proofs given of its therapeutic effects on several diseases. Gold also plays an important role in the vaccine field as an adjuvant and a carrier, reducing toxicity, enhancing immunogenic activity, and providing stability in storage. An even brighter golden future is expected for gold applications in this area.
Collapse
Affiliation(s)
- Sónia Alexandra Correia Carabineiro
- Laboratório de Catálise e Materiais (LCM), Laboratório Associado LSRE-LCM, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal.
| |
Collapse
|
28
|
Molino NM, Neek M, Tucker JA, Nelson EL, Wang SW. Display of DNA on Nanoparticles for Targeting Antigen Presenting Cells. ACS Biomater Sci Eng 2017; 3:496-501. [PMID: 28989957 DOI: 10.1021/acsbiomaterials.7b00148] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Efficient delivery of antigens is of paramount concern in immunotherapies. We aimed to target antigen presenting cells (APCs) by conjugating CpG oligonucleotides to an E2 protein nanoparticle surface (CpG-PEG-E2). Compared to E2 alone, we observed ~4-fold increase of in vitro APC uptake of both CpG-PEG-E2 and E2 conjugated to non-CpG DNA. Furthermore, compared to E2-alone or E2 functionalized solely with polyethylene glycol (PEG), the CpG-PEG-E2 showed enhanced lymph node retention up to at least 48 hr and 2-fold increase in APC uptake in vivo, parameters which are advantageous for vaccine success. This suggests that enhanced APC uptake of nanoparticles mediated by oligonucleotide display may help overcome delivery barriers in vaccine development.
Collapse
Affiliation(s)
- Nicholas M Molino
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697 USA
| | - Medea Neek
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697 USA
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697 USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697 USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697 USA.,Institute for Immunology, University of California, Irvine, CA 92697 USA
| | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697 USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697 USA.,Department of Biomedical Engineering, University of California, Irvine, CA 92697 USA
| |
Collapse
|
29
|
Mannucci S, Calderan L, Quaranta P, Antonini S, Mosca F, Longoni B, Marzola P, Boschi F. Quantum dots labelling allows detection of the homing of mesenchymal stem cells administered as immunomodulatory therapy in an experimental model of pancreatic islets transplantation. J Anat 2016; 230:381-388. [PMID: 27861845 DOI: 10.1111/joa.12563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2016] [Indexed: 12/28/2022] Open
Abstract
Cell transplantation is considered a promising therapeutic approach in several pathologies but still needs innovative and non-invasive imaging technologies to be validated. The use of mesenchymal stem cells (MSCs) attracts major interest in clinical transplantation thanks to their regenerative properties, low immunogenicity and ability to regulate immune responses. In several animal models, MSCs are used in co-transplantation with pancreatic islets (PIs) for the treatment of type I diabetes, supporting graft survival and prolonging normal glycaemia levels. In this study we investigated the homing of systemically administered MSCs in a rat model of pancreatic portal vein transplantation. MSCs labelled with quantum dots (Qdots) were systemically injected by tail vein and monitored by optical fluorescence imaging. The fluorescence signal of the liver in animals co-transplanted with MSCs and PIs was significantly higher than in control animals in which MSCs alone were transplanted. By using magnetic labelling of PIs, the homing of PIs into liver was independently confirmed. These results demonstrate that MSCs injected in peripheral blood vessels preferentially accumulate into liver when PIs are transplanted in the same organ. Moreover, we prove that bimodal MRI-fluorescence imaging allows specific monitoring of the fate of two types of cells.
Collapse
Affiliation(s)
- Silvia Mannucci
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Laura Calderan
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Paola Quaranta
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Sara Antonini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Franco Mosca
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Biancamaria Longoni
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Pasquina Marzola
- Department of Computer Science, University of Verona, Verona, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Verona, Italy
| |
Collapse
|
30
|
Lybaert L, Vanparijs N, Fierens K, Schuijs M, Nuhn L, Lambrecht BN, De Geest BG. A Generic Polymer-Protein Ligation Strategy for Vaccine Delivery. Biomacromolecules 2016; 17:874-81. [PMID: 26812240 DOI: 10.1021/acs.biomac.5b01571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although the field of cancer immunotherapy is intensively investigated, there is still a need for generic strategies that allow easy, mild and efficient formulation of vaccine antigens. Here we report on a generic polymer-protein ligation strategy to formulate protein antigens into reversible polymeric conjugates for enhanced uptake by dendritic cells and presentation to CD8 T-cells. A N-hydroxypropylmethacrylamide (HPMA)-based copolymer was synthesized via RAFT polymerization followed by introduction of pyridyldisulfide moieties. To enhance ligation efficiency to ovalbumin, which is used as a model protein antigen, protected thiols were introduced onto lysine residues and deprotected in situ in the presence of the polymer. The ligation efficiency was compared for both the thiol-modified versus unmodified ovalbumin, and the reversibility was confirmed. Furthermore, the obtained nanoconjugates were tested in vitro for their interaction and association with dendritic cells, showing enhanced cellular uptake and antigen cross-presentation to CD8 T-cells.
Collapse
Affiliation(s)
- Lien Lybaert
- Department of Pharmaceutics, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Nane Vanparijs
- Department of Pharmaceutics, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kaat Fierens
- VIB Inflammation Research Center, Ghent University , Technologiepark 927, 9052 Ghent Belgium
| | - Martijn Schuijs
- VIB Inflammation Research Center, Ghent University , Technologiepark 927, 9052 Ghent Belgium
| | - Lutz Nuhn
- Department of Pharmaceutics, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center, Ghent University , Technologiepark 927, 9052 Ghent Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
31
|
Patel JM, Vartabedian VF, Bozeman EN, Caoyonan BE, Srivatsan S, Pack CD, Dey P, D'Souza MJ, Yang L, Selvaraj P. Plasma membrane vesicles decorated with glycolipid-anchored antigens and adjuvants via protein transfer as an antigen delivery platform for inhibition of tumor growth. Biomaterials 2015; 74:231-44. [PMID: 26461116 DOI: 10.1016/j.biomaterials.2015.09.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 09/19/2015] [Accepted: 09/23/2015] [Indexed: 02/05/2023]
Abstract
Antigen delivered within particulate materials leads to enhanced antigen-specific immunity compared to soluble administration of antigen. However, current delivery approaches for antigen encapsulated in synthetic particulate materials are limited by the complexity of particle production that affects stability and immunogenicity of the antigen. Herein, we describe a protein delivery system that utilizes plasma membrane vesicles (PMVs) derived from biological materials such as cultured cells or isolated tissues and a simple protein transfer technology. We show that these particulate PMVs can be easily modified within 4 h by a protein transfer process to stably incorporate a glycosylphosphatidylinositol (GPI)-anchored form of the breast cancer antigen HER-2 onto the PMV surface. Immunization of mice with GPI-HER-2-modified-PMVs induced strong HER-2-specific antibody responses and protection from tumor challenge in two different breast cancer models. Further incorporation of the immunostimulatory molecules IL-12 and B7-1 onto the PMVs by protein transfer enhanced tumor protection and induced beneficial Th1 and Th2-type HER-2-specific immune responses. Since protein antigens can be easily converted to GPI-anchored forms, these results demonstrate that isolated plasma membrane vesicles can be modified with desired antigens along with immunostimulatory molecules by protein transfer and used as a vaccine delivery vehicle to elicit potent antigen-specific immunity.
Collapse
Affiliation(s)
- Jaina M Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vincent F Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erica N Bozeman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brianne E Caoyonan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjay Srivatsan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher D Pack
- Metaclipse Therapeutics Corporation, 3175 Presidential Drive, Atlanta, GA 30340, USA
| | - Paulami Dey
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin J D'Souza
- Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
32
|
Markov OV, Mironova NL, Sennikov SV, Vlassov VV, Zenkova MA. Prophylactic Dendritic Cell-Based Vaccines Efficiently Inhibit Metastases in Murine Metastatic Melanoma. PLoS One 2015; 10:e0136911. [PMID: 26325576 PMCID: PMC4556596 DOI: 10.1371/journal.pone.0136911] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022] Open
Abstract
Recent data on the application of dendritic cells (DCs) as anti-tumor vaccines has shown their great potential in therapy and prophylaxis of cancer. Here we report on a comparison of two treatment schemes with DCs that display the models of prophylactic and therapeutic vaccination using three different experimental tumor models: namely, Krebs-2 adenocarcinoma (primary tumor), melanoma (B16, metastatic tumor without a primary node) and Lewis lung carcinoma (LLC, metastatic tumor with a primary node). Dendritic cells generated from bone marrow-derived DC precursors and loaded with lysate of tumor cells or transfected with the complexes of total tumor RNA with cationic liposomes were used for vaccination. Lipofectamine 2000 and liposomes consisting of helper lipid DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) and cationic lipid 2D3 (1,26-Bis(1,2-de-O-tetradecyl-rac-glycerol)-7,11,16,20-tetraazahexacosan tetrahydrocloride) were used for RNA transfection. It was shown that DCs loaded with tumor lysate were ineffective in contrast to tumor-derived RNA. Therapeutic vaccination with DCs loaded by lipoplexes RNA/Lipofectamine 2000 was the most efficient for treatment of non-metastatic Krebs-2, where a 1.9-fold tumor growth retardation was observed. Single prophylactic vaccination with DCs loaded by lipoplexes RNA/2D3 was the most efficient to treat highly aggressive metastatic tumors LLC and B16, where 4.7- and 10-fold suppression of the number of lung metastases was observed, respectively. Antimetastatic effect of single prophylactic DC vaccination in metastatic melanoma model was accompanied by the reductions in the levels of Th2-specific cytokines however the change of the levels of Th1/Th2/Th17 master regulators was not found. Failure of double prophylactic vaccination is explained by Th17-response polarization associated with autoimmune and pro-inflammatory reactions. In the case of therapeutic DC vaccine the polarization of Th1-response was found nevertheless the antimetastatic effect was less effective in comparison with prophylactic DC vaccine.
Collapse
Affiliation(s)
- Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Nadezhda L. Mironova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey V. Sennikov
- Federal State Budgetary Institution "Research Institute of Clinical Immunology", Siberian Branch of Russian Academy of Medical Sciences, Novosibirsk, Russia
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- * E-mail:
| |
Collapse
|
33
|
Allison RR. Radiobiological modifiers in clinical radiation oncology: current reality and future potential. Future Oncol 2015; 10:2359-79. [PMID: 25525845 DOI: 10.2217/fon.14.174] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy can successfully ablate tumors. However, the same ionization process that destroys a cancer can also permanently damage surrounding organs resulting in unwanted clinical morbidity. Therefore, modern radiation therapy attempts to minimize dose to normal tissue to prevent side effects. Still, as tumors and normal tissues intercalate, the risk of normal tissue injury often may prevent tumoricidal doses of radiation therapy to be delivered. This paper will review current outcomes and limitations of radiobiological modifiers that may selectively enhance the radiosensitivity of tumors as well as parallel techniques that may protect normal tissues from radiation injury. Future endeavors based in part upon newly elucidated genetic pathways will be highlighted.
Collapse
|
34
|
Golovastova MO, Bazhin AV, Philippov PP. Cancer-retina antigens -- a new group of tumor antigens. BIOCHEMISTRY (MOSCOW) 2015; 79:733-9. [PMID: 25365483 DOI: 10.1134/s000629791408001x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Some photoreceptor proteins normally specific for the eye retina are aberrantly expressed in malignant tumors. These proteins include recoverin, visual rhodopsin, transducin, cGMP-phosphodiesterase 6 (PDE 6), cGMP-dependent cationic channels, guanylyl cyclase 1, rhodopsin kinase, and arrestin. By analogy with cancer-testis antigens, these photoreceptor proteins form the group of cancer-retina antigens. It is shown that an aberrant demethylation of the promoter region of recoverin is involved in the aberrant expression of this protein. The cascade Wnt5a → Frizzled-2 → transducin → PDE 6 is shown to function in skin melanoma cells, and this suggests that these cancer-retina antigens can play a functional role. The events accompanying the signal transduction in this cascade, including those involving calcium ions and cGMP-dependent protein kinase (protein kinase G), are discussed.
Collapse
Affiliation(s)
- M O Golovastova
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia
| | | | | |
Collapse
|
35
|
Kwon S, Kim YE, Park JA, Kim DS, Kwon HJ, Lee Y. Therapeutic effect of a TM4SF5-specific peptide vaccine against colon cancer in a mouse model. BMB Rep 2015; 47:215-20. [PMID: 24286311 PMCID: PMC4163885 DOI: 10.5483/bmbrep.2014.47.4.157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 07/24/2013] [Accepted: 07/26/2014] [Indexed: 12/14/2022] Open
Abstract
Molecular-targeted therapy has gained attention because of its high efficacy and weak side effects. Previously, we confirmed that transmembrane 4 superfamily member 5 protein (TM4SF5) can serve as a molecular target to prevent or treat hepatocellular carcinoma (HCC). We recently extended the application of the peptide vaccine, composed of CpG-DNA, liposome complex, and TM4SF5 peptide, to prevent colon cancer in a mouse model. Here, we first implanted mice with mouse colon cancer cells and then checked therapeutic effects of the vaccine against tumor growth. Immunization with the peptide vaccine resulted in robust production of TM4SF5-specific antibodies, alleviated tumor growth, and reduced survival rate of the tumor-bearing mice. We also found that serum levels of VEGF were markedly reduced in the mice immunized with the peptide vaccine. Therefore, we suggest that the TM4SF5-specific peptide vaccine has a therapeutic effect against colon cancer in a mouse model. [BMB Reports 2014; 47(4): 215-220]
Collapse
Affiliation(s)
- Sanghoon Kwon
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Young-Eun Kim
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Jeong-A Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Doo-Sik Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Hyung-Joo Kwon
- Center for Medical Science Research; Department of Microbiology, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 361-763, Korea
| |
Collapse
|
36
|
Brewer KD, Lake K, Pelot N, Stanford MM, DeBay DR, Penwell A, Weir GM, Karkada M, Mansour M, Bowen CV. Clearance of depot vaccine SPIO-labeled antigen and substrate visualized using MRI. Vaccine 2014; 32:6956-6962. [PMID: 25444822 DOI: 10.1016/j.vaccine.2014.10.058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/16/2014] [Accepted: 10/20/2014] [Indexed: 01/26/2023]
Abstract
Immunotherapies, including peptide-based vaccines, are a growing area of cancer research, and understanding their mechanism of action is crucial for their continued development and clinical application. Exploring the biodistribution of vaccine components may be key to understanding this action. This work used magnetic resonance imaging (MRI) to characterize the in vivo biodistribution of the antigen and oil substrate of the vaccine delivery system known as DepoVax(TM). DepoVax uses a novel adjuvanted lipid-in-oil based formulation to solubilise antigens and promote a depot effect. In this study, antigen or oil were tagged with superparamagnetic iron oxide (SPIO), making them visible on MR images. This enables tracking of individual vaccine components to determine changes in biodistribution. Mice were injected with SPIO-labeled antigen or SPIO-labeled oil, and imaged to examine clearance of labeled components from the vaccine site. The SPIO-antigen was steadily cleared, with nearly half cleared within two months post-vaccination. In contrast, the SPIO-oil remained relatively unchanged. The biodistribution of the SPIO-antigen component within the vaccine site was heterogeneous, indicating the presence of active clearance mechanisms, rather than passive diffusion or drainage. Mice injected with SPIO-antigen also showed MRI contrast for several weeks post-vaccination in the draining inguinal lymph node. These results indicate that MRI can visualize the in vivo longitudinal biodistribution of vaccine components. The sustained clearance is consistent with antigen up-take and trafficking by immune cells, leading to accumulation in the draining lymph node, which corresponds to the sustained immune responses and reduced tumor burden observed in vaccinated mice.
Collapse
Affiliation(s)
- Kimberly D Brewer
- Immunovaccine Inc., Halifax, NS, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Kerry Lake
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Nicole Pelot
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Marianne M Stanford
- Immunovaccine Inc., Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Drew R DeBay
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | | | - Genevieve M Weir
- Immunovaccine Inc., Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Mohan Karkada
- Immunovaccine Inc., Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Chris V Bowen
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada; Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada; Departments of Radiology and Physics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
37
|
Koido S, Ito M, Sagawa Y, Okamoto M, Hayashi K, Nagasaki E, Kan S, Komita H, Kamata Y, Homma S. Vaccination with vascular progenitor cells derived from induced pluripotent stem cells elicits antitumor immunity targeting vascular and tumor cells. Cancer Immunol Immunother 2014; 63:459-68. [PMID: 24627093 PMCID: PMC11028528 DOI: 10.1007/s00262-014-1531-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/25/2014] [Indexed: 01/12/2023]
Abstract
Vaccination of BALB/c mice with dendritic cells (DCs) loaded with the lysate of induced vascular progenitor (iVP) cells derived from murine-induced pluripotent stem (iPS) cells significantly suppressed the tumor of CMS-4 fibrosarcomas and prolonged the survival of CMS-4-inoculated mice. This prophylactic antitumor activity was more potent than that of immunization with DCs loaded with iPS cells or CMS-4 tumor cells. Tumors developed slowly in mice vaccinated with DCs loaded with iVP cells (DC/iVP) and exhibited a limited vascular bed. Immunohistochemistry and a tomato-lectin perfusion study demonstrated that the tumors that developed in the iVP-immunized mice showed a marked decrease in tumor vasculature. Immunization with DC/iVP induced a potent suppressive effect on vascular-rich CMS-4 tumors, a weaker effect on BNL tumors with moderate vasculature, and nearly no effect on C26 tumors with poor vasculature. Treatment of DC/iVP-immunized mice with a monoclonal antibody against CD4 or CD8, but not anti-asialo GM1, inhibited the antitumor activity. CD8(+) T cells from DC/iVP-vaccinated mice showed significant cytotoxic activity against murine endothelial cells and CMS-4 cells, whereas CD8(+) T cells from DC/iPS-vaccinated mice did not. DNA microarray analysis showed that the products of 29 vasculature-associated genes shared between genes upregulated by differentiation from iPS cells into iVP cells and genes shared by iVP cells and isolated Flk-1(+) vascular cells in CMS-4 tumor tissue might be possible targets in the immune response. These results suggest that iVP cells from iPS cells could be used as a cancer vaccine targeting tumor vascular cells and tumor cells.
Collapse
Affiliation(s)
- Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
- Institute of Clinical Medicine and Research, Jikei University School of Medicine, Chiba, Japan
| | - Masaki Ito
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
| | - Yukiko Sagawa
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
| | - Masato Okamoto
- Division of Cellular Signaling, Institute of Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kazumi Hayashi
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Eijiro Nagasaki
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Shin Kan
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
| | - Hideo Komita
- Shimbashi Medical Checkup Office, Jikei University Hospital, Tokyo, Japan
| | - Yuko Kamata
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
| | - Sadamu Homma
- Department of Oncology, Institute of DNA Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461 Japan
| |
Collapse
|
38
|
Cicchelero L, de Rooster H, Sanders NN. Various ways to improve whole cancer cell vaccines. Expert Rev Vaccines 2014; 13:721-35. [PMID: 24758597 DOI: 10.1586/14760584.2014.911093] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunotherapy based on whole cancer cell vaccines is regarded as a promising avenue for cancer treatment. However, limited efficacy in the first human clinical trials calls for more optimized whole cancer cell vaccines and better patient selection. It is suggested that whole cancer cell vaccines consist preferably of immunogenically killed autologous cancer stem cells associated with dendritic cells. Adjuvants should stimulate both immune effector cells and memory cells, which could be achieved through their correct dosage and timing of administration. There are indications that whole cancer cell vaccination is less effective in patients who are immunocompromised, who have specific genetic defects in their immune or cancer cells, as well as in patients in an advanced cancer stage. However, such patients form the bulk of enrolled patients in clinical trials, prohibiting an objective evaluation of the true potential of whole cancer cell immunotherapy. Each key point will be discussed.
Collapse
Affiliation(s)
- Laetitia Cicchelero
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | | | | |
Collapse
|
39
|
Ye W, Xing Y, Paustian C, van de Ven R, Moudgil T, Hilton TL, Fox BA, Urba WJ, Zhao W, Hu HM. Cross-presentation of viral antigens in dribbles leads to efficient activation of virus-specific human memory T cells. J Transl Med 2014; 12:100. [PMID: 24735498 PMCID: PMC4021424 DOI: 10.1186/1479-5876-12-100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 04/03/2014] [Indexed: 12/11/2022] Open
Abstract
Background Autophagy regulates innate and adaptive immune responses to pathogens and tumors. We have reported that autophagosomes derived from tumor cells after proteasome inhibition, DRibbles (Defective ribosomal products in blebs), were excellent sources of antigens for efficient cross priming of tumor-specific CD8+ T cells, which mediated regression of established tumors in mice. But the activity of DRibbles in human has not been reported. Methods DRibbles or cell lysates derived from HEK293T or UbiLT3 cell lines expressing cytomegalovirus (CMV) pp65 protein or transfected with a plasmid encoding dominant HLA-A2 restricted CMV, Epstein-Barr virus (EBV), and Influenza (Flu) epitopes (CEF) were loaded onto human monocytes or PBMCs and the response of human CMV pp65 or CEF antigen-specific CD4+ and CD8+ memory T cells was detected by intracellular staining. The effect of cytokines (GM-CSF, IL-4, IL-12, TNF-α, IFN-α and IFN-γ) TLR agonists (Lipopolysaccharide, Polyinosinic-polycytidylic acid (poly(I:C), M52-CpG, R848, TLR2 ligand) and CD40 ligand on the cross-presentation of antigens contained in DRibbles or cell lysates was explored. Results In this study we showed that purified monocytes, or human PBMCs, loaded with DRibbles isolated from cells expressing CMV or CEF epitopes, could activate CMV- or CEF-specific memory T cells. DRibbles were significantly more efficient at stimulating CD8+ memory T cells compared to cell lysates expressing the same antigenic epitopes. We optimized the conditions for T-cell activation and IFN-γ production following direct loading of DRibbles onto PBMCs. We found that the addition of Poly(I:C), CD40 ligand, and GM-CSF to the PBMCs together with DRibbles significantly increased the level of CD8+ T cell responses. Conclusions DRibbles containing specific viral antigens are an efficient ex vivo activator of human antigen-specific memory T cells specific for those antigens. This function could be enhanced by combining with Poly(I:C), CD40 ligand, and GM-CSF. This study provides proof-of-concept for applying this strategy to activate memory T cells against other antigens, including tumor-specific T cells ex vivo for immunological monitoring and adoptive immunotherapy, and in vivo as vaccines for patients with cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wei Zhao
- Medical School, Southeast University, 87 Dingjiaqiao Street, 210009 Nanjing, Jiangsu, PR China.
| | | |
Collapse
|
40
|
Long TTA, Kalantarov G, Chudner A, Trakht I. A dual-chamber system for screening cytotoxic effects of hybridoma-produced antibodies. Monoclon Antib Immunodiagn Immunother 2014; 32:246-54. [PMID: 23909417 DOI: 10.1089/mab.2013.0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There are many methods for evaluating the cytotoxic effect of monoclonal antibodies (MAbs) against cancer cells. Most of these methods require either purified MAbs or biological solutions (e.g., cell culture supernatants, ascitic fluids) containing high concentrations of MAbs. This makes the primary screening of antibody-producing hybridomas for specific cytotoxic antibodies a challenging task. Addressing this issue, this work introduces a high throughput screening method, which enables the identification of cytotoxic antibodies using primary hybridoma populations without prior antibody concentration and/or purification. The method is comprised of a dual-chamber system, where antibody-producing hybridomas and target cancer cells are co-cultured but separated by a porous membrane in which the pore size is sufficient for the diffusion of antibody molecules. The MAbs produced in the system continuously diffuse through the membrane between the two chambers and interact with the target cells placed on the other side of a membrane, resulting in death or proliferation arrest of these cells, if MAbs are cytotoxic or cytostatic. The cytotoxic/cytostatic effect can be registered by measuring the viability of target cells. The advantage of this method is that purification or concentration of antibodies secreted by hybridomas is not required. In addition, this method does not require MAb-secreting hybridomas, which are subcloned or have a high level of MAb production. The method may serve as an effective primary high throughput screening for cytotoxic antibodies.
Collapse
Affiliation(s)
- Ton That Ai Long
- Division of Experimental Therapeutics, Department of Medicine, College of Physicians and Surgeons, Columbia University in New York City, New York 10032, USA
| | | | | | | |
Collapse
|
41
|
Abstract
Several reports have described the use of tumor-extracted RNA as source of tumor antigen for the preparation of vaccines based on dendritic cells (DC) and its potential use for antigen-specific or polyvalent tumor vaccination. Upon transfection, RNA is transcribed into proteins that enter the cytoplasmic degradation pathway and can be presented by DC through class I major histocompatibility complex (MHC)-I, thus inducing specific T cell cytotoxic responses. In this chapter, we present a protocol to transfect murine dendritic cells with tumor mRNA by means of electroporation.
Collapse
Affiliation(s)
- Fabian Benencia
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, OH, USA
| |
Collapse
|
42
|
Marconato L, Frayssinet P, Rouquet N, Comazzi S, Leone VF, Laganga P, Rossi F, Vignoli M, Pezzoli L, Aresu L. Randomized, Placebo-Controlled, Double-Blinded Chemoimmunotherapy Clinical Trial in a Pet Dog Model of Diffuse Large B-cell Lymphoma. Clin Cancer Res 2013; 20:668-77. [DOI: 10.1158/1078-0432.ccr-13-2283] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Molino NM, Anderson AKL, Nelson EL, Wang SW. Biomimetic protein nanoparticles facilitate enhanced dendritic cell activation and cross-presentation. ACS NANO 2013; 7:9743-52. [PMID: 24090491 PMCID: PMC3893022 DOI: 10.1021/nn403085w] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Many current cancer vaccine strategies suffer from the inability to mount a CD8 T cell response that is strong enough to overcome the low immunogenicity of tumors. Viruses naturally possess the sizes, geometries, and physical properties for which the immune system has evolved to recognize, and mimicking those properties with nanoparticles can produce robust platforms for vaccine design. Using the nonviral E2 core of pyruvate dehydrogenase, we have engineered a viral-mimicking vaccine platform capable of encapsulating dendritic cell (DC)-activating CpG molecules in an acid-releasable manner and displaying MHC I-restricted SIINFEKL peptide epitopes. Encapsulated CpG activated bone marrow-derived DCs at a 25-fold lower concentration in vitro when delivered with the E2 nanoparticle than with unbound CpG alone. Combining CpG and SIINFEKL within a single multifunctional particle induced ∼3-fold greater SIINFEKL display on MHC I by DCs over unbound peptide. Importantly, combining CpG and SIINFEKL to the E2 nanoparticle for simultaneous temporal and spatial delivery to DCs showed increased and prolonged CD8 T cell activation, relative to free peptide or peptide-bound E2. By codelivering peptide epitopes and CpG activator in a particle of optimal DC-uptake size, we demonstrate the ability of a noninfectious protein nanoparticle to mimic viral properties and facilitate enhanced DC activation and cross-presentation.
Collapse
Affiliation(s)
- Nicholas M. Molino
- Department of Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697-2575
| | | | | | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697-2575
| |
Collapse
|
44
|
Benencia F. RNA vaccines for anti-tumor therapy. World J Exp Med 2013; 3:62-73. [DOI: 10.5493/wjem.v3.i4.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/27/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
The immune system is able to recognize tumor antigens and this has been the basis for the development of cancer immunotherapies. The immune system can be instructed to recognize and attack tumor cells by means of vaccination strategies. One such strategy involves the delivery of tumor antigen as genetic material. Herewith we describe the use of RNA encoding tumor antigens for vaccination purposes in tumor settings. RNA has features that are interesting for vaccination. Upon transfection, the RNA has no possibility of integration into the genome, and the tumor translated proteins enter the intrinsic antigen processing pathway thus enabling presentation by MHC-I molecules. This can specifically activate cytotoxic CD8 T cells that can attack and kill tumor cells. RNA can be delivered as a naked molecule for vaccination purposes or can be used to transfect dendritic cells. The combination of RNA technology with dendritic cell vaccination provides a powerful tool for cancer immunotherapies.
Collapse
|
45
|
Fenoglio D, Traverso P, Parodi A, Kalli F, Zanetti M, Filaci G. Generation of more effective cancer vaccines. Hum Vaccin Immunother 2013; 9:2543-7. [PMID: 23978951 DOI: 10.4161/hv.26147] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cancer vaccines represent a promising therapeutic approach for which prime time is imminent. However, clinical efficacy must be improved in order for cancer vaccines to become a valid alternative or complement to traditional cancer treatments. Considerable efforts have been undertaken so far to better understand the fundamental requirements for clinically-effective cancer vaccines. Recent data emphasize that important requirements, among others, are (1) the use of multi-epitope immunogens, possibly deriving from different tumor antigens; (2) the selection of effective adjuvants; (3) the association of cancer vaccines with agents able to counteract the regulatory milieu present in the tumor microenvironment; and (4) the need to choose the definitive formulation and regimen of a vaccine after accurate preliminary tests comparing different antigen formulations. The first requirement deals with issues related to HLA restriction of tumor antigen presentation, as well as usefulness of tumor antigen spreading and counteraction of immune escape phenomena, linked to tumor antigen down-modulation, for an effective anti-cancer immune response. The second point underscores the necessity of optimal activation of innate immunity to achieve an efficient adaptive anti-cancer immune response. The third point focuses on the importance to inhibit subsets of regulatory cells. The last requirement stresses the concept that the regimen and formulation of the vaccine impacts profoundly on cancer vaccine efficacy. A new generation of cancer vaccines, provided with both immunological and clinical efficacy, will hopefully soon address these requirements.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Surgical Sciences; University of Genoa; Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Maurizio Zanetti
- The Laboratory of Immunology; Department of Medicine and Cancer Center; University of California; San Diego, CA USA
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| |
Collapse
|
46
|
Antitumor activity of human hydatid cyst fluid in a murine model of colon cancer. ScientificWorldJournal 2013; 2013:230176. [PMID: 24023528 PMCID: PMC3759250 DOI: 10.1155/2013/230176] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 07/14/2013] [Indexed: 12/11/2022] Open
Abstract
This study evaluates the antitumor immune response induced by human hydatic cyst fluid (HCF) in an animal model of colon carcinoma. We found that anti-HCF antibodies were able to identify cell surface and intracellular antigens in CT26 colon cancer cells. In prophylactic tumor challenge experiments, HCF vaccination was found to be protective against tumor formation for 40% of the mice (P = 0.01). In the therapeutic setting, HCF vaccination induced tumor regression in 40% of vaccinated mice (P = 0.05). This vaccination generated memory immune responses that protected surviving mice from tumor rechallenge, implicating the development of an adaptive immune response in this process. We performed a proteomic analysis of CT26 antigens recognized by anti-HCF antibodies to analyze the immune cross-reactivity between E. granulosus (HCF) and CT26 colon cancer cells. We identified two proteins: mortalin and creatine kinase M-type. Interestingly, CT26 mortalin displays 60% homology with E. granulosus hsp70. In conclusion, our data demonstrate the capacity of HCF vaccination to induce antitumor immunity which protects from tumor growth in an animal model. This new antitumor strategy could open new horizons in the development of highly immunogenic anticancer vaccines.
Collapse
|
47
|
Arens R, van Hall T, van der Burg SH, Ossendorp F, Melief CJM. Prospects of combinatorial synthetic peptide vaccine-based immunotherapy against cancer. Semin Immunol 2013; 25:182-90. [PMID: 23706598 DOI: 10.1016/j.smim.2013.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/10/2013] [Accepted: 04/19/2013] [Indexed: 01/15/2023]
Abstract
The insight that the immune system is involved in tumor resistance is gaining momentum and this has led to the development of immunotherapeutic strategies aiming at enhancement of immune-mediated tumor destruction. Although some of these strategies have moderate clinical benefit, most stand-alone therapies fail to significantly affect progressive disease and survival or do so only in a minority of patients. Research on the mechanisms underlying the generation of immune responses against tumors and the immune evasion by tumors has emphasized that various mechanisms simultaneously prevent effective immunity against cancer including inefficient presentation of tumor antigens by dendritic cells and induction of negative immune regulation by regulatory T-cells (Tregs) and myeloid derived suppressor cells (MDSCs). Thus the design of therapies that simultaneously improve effective tumor immunity and counteract immune evasion by tumors seems most desirable for clinical efficacy. As it is unlikely that a single immunotherapeutic strategy addresses all necessary requirements, combinatorial strategies that act synergistically need to be developed. Here we discuss the current knowledge and prospects of treatment with synthetic peptide vaccines that stimulate tumor-specific T-cell responses combined with adjuvants, immune modulating antibodies, cytokines and chemotherapy. We conclude that combinatorial approaches have the best potency to accomplish the most significant tumor destruction but further research is required to optimize such approaches.
Collapse
Affiliation(s)
- Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
48
|
Pouyanfard S, Bamdad T, Hashemi H, Bandehpour M, Kazemi B. Induction of protective anti-CTL epitope responses against HER-2-positive breast cancer based on multivalent T7 phage nanoparticles. PLoS One 2012; 7:e49539. [PMID: 23166703 PMCID: PMC3499470 DOI: 10.1371/journal.pone.0049539] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 10/10/2012] [Indexed: 02/05/2023] Open
Abstract
We report here the development of multivalent T7 bacteriophage nanoparticles displaying an immunodominant H-2kd-restricted CTL epitope derived from the rat HER2/neu oncoprotein. The immunotherapeutic potential of the chimeric T7 nanoparticles as anti-cancer vaccine was investigated in BALB/c mice in an implantable breast tumor model. The results showed that T7 phage nanoparticles confer a high immunogenicity to the HER-2-derived minimal CTL epitope, as shown by inducing robust CTL responses. Furthermore, the chimeric nanoparticles protected mice against HER-2-positive tumor challenge in both prophylactic and therapeutic setting. In conclusion, these results suggest that CTL epitope-carrying T7 phage nanoparticles might be a promising approach for development of T cell epitope-based cancer vaccines.
Collapse
Affiliation(s)
- Somayeh Pouyanfard
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taravat Bamdad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- * E-mail: (TB); (BK)
| | - Hamidreza Hashemi
- Department of Medical Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandehpour
- Biotechnology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center (CMRC), School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Biotechnology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center (CMRC), School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- * E-mail: (TB); (BK)
| |
Collapse
|
49
|
Kwon S, Kim D, Park BK, Wu G, Park MC, Ha YW, Kwon HJ, Lee Y. Induction of immunological memory response by vaccination with TM4SF5 epitope-CpG-DNA-liposome complex in a mouse hepatocellular carcinoma model. Oncol Rep 2012; 29:735-40. [PMID: 23138455 DOI: 10.3892/or.2012.2130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/08/2012] [Indexed: 11/06/2022] Open
Abstract
The innovation of a peptide vaccine strategy may contribute to the development of efficacious and convenient cancer vaccines. Recently, we formulated an efficacious peptide vaccine without carriers using the natural phosphodiester bond CpG-DNA and a special liposome complex [Lipoplex(O)]. The peptide vaccine targeting a tumor antigen, transmembrane 4 superfamily member 5 protein (TM4SF5), was confirmed to have preventive and therapeutic effects in a mouse hepatocellular carcinoma (HCC) model. In this study, we demonstrated that the isotype-switched (IgM(-)IgD(-)) B cell population increased after immunization and that the functional memory response persisted for at least 70 days after the final immunization of mice. Delayed implantation of BNL-HCC cells significantly induced the peptide-specific IgG2a production in the immunized mice. Accordingly, tumor growth was inhibited and the survival rate increased. These results suggest that our peptide vaccine induces memory response, which is essential for cancer vaccine application.
Collapse
Affiliation(s)
- Sanghoon Kwon
- Center for Medical Science Research, College of Medicine, Hallym University, Gangwon, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|